1
|
Kim JH, Ha EK, Lee GC, Han B, Shin J, Han MY, Rhie S. Diverse weaning foods and diet patterns at multiple time points during infancy period and their association with neurodevelopmental outcomes in 6-year-old children. Eur J Clin Nutr 2024:10.1038/s41430-024-01528-3. [PMID: 39424987 DOI: 10.1038/s41430-024-01528-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND/OBJECTIVES Understanding the impact of early-life nutritional choices on neurodevelopment in children is a growing area of research. To investigate the association between dietary patterns at multiple timelines and neurodevelopmental outcomes in 6-year-old children. SUBJECTS/METHODS This administrative observational study utilized a merged data from the national health insurance database and the health screening program for children. Information on the diet patterns from infancy to 3 years of age was obtained from parent-administered questionnaires. Dietary pattern clusters of the participants were identified using Polytomous Latent Class Analysis. The outcome was neurodevelopment using the Korean Developmental Screening Test (K-DST) at the age of 6 years. RESULTS The study identified four distinct clusters among with the 133,243 eligible children (49.6% male, birth weight 3.22 kg, head circumference 42.7 cm at 4 months). The control cluster (53.4%) exhibited a diet including breast milk feeding and a variety of dietary patterns at the age of 1 year. In contrast, cluster 1 (36.0%) showed a skewed dietary pattern at the same age. Cluster 2 (6.6%) displayed diverse dietary patterns at one year but primarily consumed formula at four months, while cluster 3 (4.0%) had reduced dietary diversity and formula feeding. Compared with the control cluster, the adjusted odds ratio for unfavorable development was 1.209 (95% CI, 1.156-1.266) in cluster 1, 1.418 (95% CI, 1.312-1.532) in cluster 2, and 1.741 (95% CI, 1.593-1.903) in cluster 3. These findings remained consistent across individual domains of the K-DST. CONCLUSIONS Dietary patterns during infancy and early childhood may be associated with neurodevelopment at the age of 6 years.
Collapse
Affiliation(s)
- Ju Hee Kim
- Department of Pediatrics, College of Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul, South Korea
| | - Eun Kyo Ha
- Department of Pediatrics, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea
| | - Gi Chun Lee
- Department of Computer Science and Engineering, College of Engineering, Konkuk University, Seoul, South Korea
| | - Boeun Han
- Department of Pediatrics, Bundang CHA Medical Center, CHA University School of Medicine, Seongnam, South Korea
| | - Jeewon Shin
- Department of Pediatrics, Ilsan CHA Medical Center, CHA University School of Medicine, Goyang, South Korea
| | - Man Yong Han
- Department of Pediatrics, Bundang CHA Medical Center, CHA University School of Medicine, Seongnam, South Korea.
| | - Seonkyeong Rhie
- Department of Pediatrics, Bundang CHA Medical Center, CHA University School of Medicine, Seongnam, South Korea.
| |
Collapse
|
2
|
Abate F, Adu-Amankwah A, Ae-Ngibise KA, Agbokey F, Agyemang VA, Agyemang CT, Akgun C, Ametepe J, Arichi T, Asante KP, Balaji S, Baljer L, Basser PJ, Beauchemin J, Bennallick C, Berhane Y, Boateng-Mensah Y, Bourke NJ, Bradford L, Bruchhage M, Lorente RC, Cawley P, Cercignani M, D Sa V, Canha AD, Navarro ND, Dean DC, Delarosa J, Donald KA, Dvorak A, Edwards AD, Field D, Frail H, Freeman B, George T, Gholam J, Guerrero-Gonzalez J, Hajnal JV, Haque R, Hollander W, Hoodbhoy Z, Huentelman M, Jafri SK, Jones DK, Joubert F, Karaulanov T, Kasaro MP, Knackstedt S, Kolind S, Koshy B, Kravitz R, Lafayette SL, Lee AC, Lena B, Lepore N, Linguraru M, Ljungberg E, Lockart Z, Loth E, Mannam P, Masemola KM, Moran R, Murphy D, Nakwa FL, Nankabirwa V, Nelson CA, North K, Nyame S, O Halloran R, O'Muircheartaigh J, Oakley BF, Odendaal H, Ongeti CM, Onyango D, Oppong SA, Padormo F, Parvez D, Paus T, Pepper MS, Phiri KS, Poorman M, Ringshaw JE, Rogers J, Rutherford M, Sabir H, Sacolick L, Seal M, Sekoli ML, Shama T, Siddiqui K, Sindano N, Spelke MB, Springer PE, Suleman FE, Sundgren PC, Teixeira R, Terekegn W, Traughber M, Tuuli MG, Rensburg JV, Váša F, Velaphi S, Velasco P, Viljoen IM, Vokhiwa M, Webb A, Weiant C, Wiley N, Wintermark P, Yibetal K, Deoni S, Williams S. UNITY: A low-field magnetic resonance neuroimaging initiative to characterize neurodevelopment in low and middle-income settings. Dev Cogn Neurosci 2024; 69:101397. [PMID: 39029330 PMCID: PMC11315107 DOI: 10.1016/j.dcn.2024.101397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 07/21/2024] Open
Abstract
Measures of physical growth, such as weight and height have long been the predominant outcomes for monitoring child health and evaluating interventional outcomes in public health studies, including those that may impact neurodevelopment. While physical growth generally reflects overall health and nutritional status, it lacks sensitivity and specificity to brain growth and developing cognitive skills and abilities. Psychometric tools, e.g., the Bayley Scales of Infant and Toddler Development, may afford more direct assessment of cognitive development but they require language translation, cultural adaptation, and population norming. Further, they are not always reliable predictors of future outcomes when assessed within the first 12-18 months of a child's life. Neuroimaging may provide more objective, sensitive, and predictive measures of neurodevelopment but tools such as magnetic resonance (MR) imaging are not readily available in many low and middle-income countries (LMICs). MRI systems that operate at lower magnetic fields (< 100mT) may offer increased accessibility, but their use for global health studies remains nascent. The UNITY project is envisaged as a global partnership to advance neuroimaging in global health studies. Here we describe the UNITY project, its goals, methods, operating procedures, and expected outcomes in characterizing neurodevelopment in sub-Saharan Africa and South Asia.
Collapse
Affiliation(s)
- F Abate
- Addis Continental Institute of Public Health, Addis Ababa, Ethiopia; Waisman Research Center, Madison, WI, USA
| | - A Adu-Amankwah
- Korle-Bu Teaching Hospital, Accra, Ghana; Waisman Research Center, Madison, WI, USA
| | - K A Ae-Ngibise
- Kintampo Health Research Centre, Research and Development Division, Ghana Health Service, Kintampo North Municipality, Bono East Region, Ghana; Waisman Research Center, Madison, WI, USA
| | - F Agbokey
- Kintampo Health Research Centre, Research and Development Division, Ghana Health Service, Kintampo North Municipality, Bono East Region, Ghana; Waisman Research Center, Madison, WI, USA
| | - V A Agyemang
- Kintampo Health Research Centre, Research and Development Division, Ghana Health Service, Kintampo North Municipality, Bono East Region, Ghana; Waisman Research Center, Madison, WI, USA
| | - C T Agyemang
- Kintampo Health Research Centre, Research and Development Division, Ghana Health Service, Kintampo North Municipality, Bono East Region, Ghana; Waisman Research Center, Madison, WI, USA
| | - C Akgun
- flywheel.io Minneapolis, MN, USA; Waisman Research Center, Madison, WI, USA
| | - J Ametepe
- Cardiff University Brain Research Imaging Center, Cardiff University, Cardiff, UK; Waisman Research Center, Madison, WI, USA
| | - T Arichi
- Centre for the Developing Brain, Kings College London, London, UK; Waisman Research Center, Madison, WI, USA
| | - K P Asante
- Kintampo Health Research Centre, Research and Development Division, Ghana Health Service, Kintampo North Municipality, Bono East Region, Ghana; Waisman Research Center, Madison, WI, USA
| | - S Balaji
- Dept. of Neurology, University of British Columbia, Vancouver, BC, Canada; Waisman Research Center, Madison, WI, USA
| | - L Baljer
- Centre for Neuroimaging Sciences, King's College London, London, UK; Waisman Research Center, Madison, WI, USA
| | - P J Basser
- National Institutes of Health, Washington, DC, USA; Waisman Research Center, Madison, WI, USA
| | - J Beauchemin
- Advanced Baby Imaging Lab, Providence, RI, USA; Waisman Research Center, Madison, WI, USA
| | - C Bennallick
- Centre for Neuroimaging Sciences, King's College London, London, UK; Waisman Research Center, Madison, WI, USA
| | - Y Berhane
- Addis Continental Institute of Public Health, Addis Ababa, Ethiopia; Waisman Research Center, Madison, WI, USA
| | - Y Boateng-Mensah
- Korle-Bu Teaching Hospital, Accra, Ghana; Waisman Research Center, Madison, WI, USA
| | - N J Bourke
- Centre for Neuroimaging Sciences, King's College London, London, UK; Waisman Research Center, Madison, WI, USA
| | - L Bradford
- Division of Developmental Paediatrics, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital and the Neuroscience Institute, University of Cape Town, Cape Town, South Africa; Waisman Research Center, Madison, WI, USA
| | - Mmk Bruchhage
- Dept. of Psychology, Stavanger University, Norway; Waisman Research Center, Madison, WI, USA
| | - R Cano Lorente
- Advanced Baby Imaging Lab, Providence, RI, USA; Waisman Research Center, Madison, WI, USA
| | - P Cawley
- Centre for the Developing Brain, Kings College London, London, UK; Waisman Research Center, Madison, WI, USA
| | - M Cercignani
- Cardiff University Brain Research Imaging Center, Cardiff University, Cardiff, UK; Waisman Research Center, Madison, WI, USA
| | - V D Sa
- Advanced Baby Imaging Lab, Providence, RI, USA; Waisman Research Center, Madison, WI, USA
| | - A de Canha
- Institute for Cellular and Molecular Medicine, Department of Medical Immunology, University of Pretoria, Pretoria, South Africa; Waisman Research Center, Madison, WI, USA
| | - N de Navarro
- Collective Minds Radiology, Sweden; Waisman Research Center, Madison, WI, USA
| | - D C Dean
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA; Waisman Research Center, Madison, WI, USA
| | - J Delarosa
- PATH, Seattle, WA, USA; Waisman Research Center, Madison, WI, USA
| | - K A Donald
- Division of Developmental Paediatrics, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital and the Neuroscience Institute, University of Cape Town, Cape Town, South Africa; Waisman Research Center, Madison, WI, USA
| | - A Dvorak
- Dept. of Neurology, University of British Columbia, Vancouver, BC, Canada; Waisman Research Center, Madison, WI, USA
| | - A D Edwards
- Centre for the Developing Brain, Kings College London, London, UK; Waisman Research Center, Madison, WI, USA
| | - D Field
- Collective Minds Radiology, Sweden; Waisman Research Center, Madison, WI, USA
| | - H Frail
- Hyperfine.io, Guilford, CT, USA; Waisman Research Center, Madison, WI, USA
| | - B Freeman
- University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, USA; Waisman Research Center, Madison, WI, USA
| | - T George
- Department of Radiology, Faculty of Health Sciences, Chris Hani Baragwanath Academic Hospital, University; Waisman Research Center, Madison, WI, USA
| | - J Gholam
- Cardiff University Brain Research Imaging Center, Cardiff University, Cardiff, UK; Waisman Research Center, Madison, WI, USA
| | - J Guerrero-Gonzalez
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA; Waisman Research Center, Madison, WI, USA
| | - J V Hajnal
- Centre for the Developing Brain, Kings College London, London, UK; Waisman Research Center, Madison, WI, USA
| | - R Haque
- International Centre for Diarrheal Disease Research, Bangladesh (Icddr,b), Dhaka, Bangladesh; Waisman Research Center, Madison, WI, USA
| | - W Hollander
- CaliberMRI, Boulder CO USA; Waisman Research Center, Madison, WI, USA
| | - Z Hoodbhoy
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan; Waisman Research Center, Madison, WI, USA
| | - M Huentelman
- TGen, Phoenix, AZ, USA; Waisman Research Center, Madison, WI, USA
| | - S K Jafri
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan; Waisman Research Center, Madison, WI, USA
| | - D K Jones
- Cardiff University Brain Research Imaging Center, Cardiff University, Cardiff, UK; Waisman Research Center, Madison, WI, USA
| | - F Joubert
- Centre for Bioinformatics and Computational Biology, Department of Biochemistry, Microbiology and Genetics, University of Pretoria, Pretoria, South Africa; Waisman Research Center, Madison, WI, USA
| | - T Karaulanov
- CaliberMRI, Boulder CO USA; Waisman Research Center, Madison, WI, USA
| | - M P Kasaro
- University of North Carolina - Global Projects Zambia, Lusaka, Zambia; Waisman Research Center, Madison, WI, USA
| | - S Knackstedt
- PATH, Seattle, WA, USA; Waisman Research Center, Madison, WI, USA
| | - S Kolind
- Dept. of Neurology, University of British Columbia, Vancouver, BC, Canada; Waisman Research Center, Madison, WI, USA
| | - B Koshy
- Developmental Paediatrics, Christian Medical College, Vellore, India; Waisman Research Center, Madison, WI, USA
| | - R Kravitz
- International Society for Magnetic Resonance in Medicine, San Fransisco, CA, USA; Waisman Research Center, Madison, WI, USA
| | - S Lecurieux Lafayette
- Centre for the Developing Brain, Kings College London, London, UK; Waisman Research Center, Madison, WI, USA
| | - A C Lee
- Brigham and Women's Hospital, Department of Pediatrics; Harvard Medical School; Boston, MA, USA; Waisman Research Center, Madison, WI, USA
| | - B Lena
- Dept. of Radiology, Leiden University, Leiden, the Netherlands; Waisman Research Center, Madison, WI, USA
| | - N Lepore
- Dept. of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Waisman Research Center, Madison, WI, USA
| | - M Linguraru
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC, USA; Waisman Research Center, Madison, WI, USA
| | - E Ljungberg
- Medical Radiation Physics, Lund University, Lund, Sweden; Waisman Research Center, Madison, WI, USA
| | - Z Lockart
- Department of Radiology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa; Waisman Research Center, Madison, WI, USA
| | - E Loth
- Department of Forensic and Neurodevelopemental Science, Institute of Psychatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Waisman Research Center, Madison, WI, USA
| | - P Mannam
- Developmental Paediatrics, Christian Medical College, Vellore, India; Waisman Research Center, Madison, WI, USA
| | - K M Masemola
- Department of Paediatrics and Child Health, Kalafong Hospital and Faculty of Health Sciences, University of Pretoria, South Africa; Waisman Research Center, Madison, WI, USA
| | - R Moran
- Centre for Neuroimaging Sciences, King's College London, London, UK; Waisman Research Center, Madison, WI, USA
| | - D Murphy
- Department of Forensic and Neurodevelopemental Science, Institute of Psychatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Waisman Research Center, Madison, WI, USA
| | - F L Nakwa
- Department of Paediatrics and Child Health, Chris Hani Baragwanath Academic Hospital and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Waisman Research Center, Madison, WI, USA
| | - V Nankabirwa
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University. Kampala, Uganda; Waisman Research Center, Madison, WI, USA
| | - C A Nelson
- Laboratories of Cognitive Neuroscience, Division of Developmental Medicine, Department of Medicine, Boston Children's Hospital, Boston, MA, USA; Waisman Research Center, Madison, WI, USA
| | - K North
- Brigham and Women's Hospital, Department of Pediatrics; Harvard Medical School; Boston, MA, USA; Waisman Research Center, Madison, WI, USA
| | - S Nyame
- Kintampo Health Research Centre, Research and Development Division, Ghana Health Service, Kintampo North Municipality, Bono East Region, Ghana; Waisman Research Center, Madison, WI, USA
| | - R O Halloran
- Hyperfine.io, Guilford, CT, USA; Waisman Research Center, Madison, WI, USA
| | - J O'Muircheartaigh
- Centre for the Developing Brain, Kings College London, London, UK; Waisman Research Center, Madison, WI, USA
| | - B F Oakley
- Department of Forensic and Neurodevelopemental Science, Institute of Psychatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Waisman Research Center, Madison, WI, USA
| | - H Odendaal
- Dept Obstet Gynaecol, Stellenbosch University, South Africa; Waisman Research Center, Madison, WI, USA
| | - C M Ongeti
- Jaramogi Oginga Odinga Teaching and Referral Hospital, Kisumu, Kenya; Waisman Research Center, Madison, WI, USA
| | - D Onyango
- Jaramogi Oginga Odinga Teaching and Referral Hospital, Kisumu, Kenya; Waisman Research Center, Madison, WI, USA
| | - S A Oppong
- Korle-Bu Teaching Hospital, Accra, Ghana; Waisman Research Center, Madison, WI, USA
| | - F Padormo
- Hyperfine.io, Guilford, CT, USA; Waisman Research Center, Madison, WI, USA
| | - D Parvez
- Collective Minds Radiology, Sweden; Waisman Research Center, Madison, WI, USA
| | - T Paus
- Departments of Psychiatry and Neuroscience, Faculty of Medicine and Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, Quebec, Canada; Waisman Research Center, Madison, WI, USA
| | - M S Pepper
- Institute for Cellular and Molecular Medicine, Department of Medical Immunology, University of Pretoria, Pretoria, South Africa; Waisman Research Center, Madison, WI, USA
| | - K S Phiri
- Training and Research Unit of Excellence (TRUE), Zomba Malawi; Waisman Research Center, Madison, WI, USA
| | - M Poorman
- Hyperfine.io, Guilford, CT, USA; Waisman Research Center, Madison, WI, USA
| | - J E Ringshaw
- Division of Developmental Paediatrics, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital and the Neuroscience Institute, University of Cape Town, Cape Town, South Africa; Waisman Research Center, Madison, WI, USA
| | - J Rogers
- Hyperfine.io, Guilford, CT, USA; Waisman Research Center, Madison, WI, USA
| | - M Rutherford
- Centre for the Developing Brain, Kings College London, London, UK; Waisman Research Center, Madison, WI, USA
| | - H Sabir
- Experimental Neonatology, University Hospitals Bonn, Bonn, Germany; Waisman Research Center, Madison, WI, USA
| | - L Sacolick
- Hyperfine.io, Guilford, CT, USA; Waisman Research Center, Madison, WI, USA
| | - M Seal
- Murdoch Children's Research Institute, Melbourne, AUS; Waisman Research Center, Madison, WI, USA
| | - M L Sekoli
- Institute for Cellular and Molecular Medicine, Department of Medical Immunology, University of Pretoria, Pretoria, South Africa; Waisman Research Center, Madison, WI, USA
| | - T Shama
- International Centre for Diarrheal Disease Research, Bangladesh (Icddr,b), Dhaka, Bangladesh; Waisman Research Center, Madison, WI, USA
| | - K Siddiqui
- Hyperfine.io, Guilford, CT, USA; Waisman Research Center, Madison, WI, USA
| | - N Sindano
- University of North Carolina - Global Projects Zambia, Lusaka, Zambia; Waisman Research Center, Madison, WI, USA
| | - M B Spelke
- University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, USA; Waisman Research Center, Madison, WI, USA
| | - P E Springer
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa; Waisman Research Center, Madison, WI, USA
| | - F E Suleman
- Department of Radiology, Faculty of Health Sciences, Kalafong Hospital, University of Pretoria, Pretoria, South Africa; Waisman Research Center, Madison, WI, USA
| | - P C Sundgren
- Section of Diagnostic Radiology,Department of Clinical Sciences Lund, Lund University, Lund, Sweden; Waisman Research Center, Madison, WI, USA
| | - R Teixeira
- Hyperfine.io, Guilford, CT, USA; Waisman Research Center, Madison, WI, USA
| | - W Terekegn
- Addis Continental Institute of Public Health, Addis Ababa, Ethiopia; Waisman Research Center, Madison, WI, USA
| | - M Traughber
- Hyperfine.io, Guilford, CT, USA; Waisman Research Center, Madison, WI, USA
| | - M G Tuuli
- Jaramogi Oginga Odinga Teaching and Referral Hospital, Kisumu, Kenya; Waisman Research Center, Madison, WI, USA
| | - J van Rensburg
- Institute for Cellular and Molecular Medicine, Department of Medical Immunology, University of Pretoria, Pretoria, South Africa; Waisman Research Center, Madison, WI, USA
| | - F Váša
- Centre for Neuroimaging Sciences, King's College London, London, UK; Waisman Research Center, Madison, WI, USA
| | - S Velaphi
- Department of Paediatrics and Child Health, Chris Hani Baragwanath Academic Hospital and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Waisman Research Center, Madison, WI, USA
| | - P Velasco
- flywheel.io Minneapolis, MN, USA; Waisman Research Center, Madison, WI, USA
| | - I M Viljoen
- Department of Radiology, Faculty of Health Sciences, Chris Hani Baragwanath Academic Hospital, University; Waisman Research Center, Madison, WI, USA
| | - M Vokhiwa
- Training and Research Unit of Excellence (TRUE), Zomba Malawi; Waisman Research Center, Madison, WI, USA
| | - A Webb
- Dept. of Radiology, Leiden University, Leiden, the Netherlands; Waisman Research Center, Madison, WI, USA
| | - C Weiant
- CaliberMRI, Boulder CO USA; Waisman Research Center, Madison, WI, USA
| | - N Wiley
- Dept. of Neurology, University of British Columbia, Vancouver, BC, Canada; Waisman Research Center, Madison, WI, USA
| | - P Wintermark
- Division of Newborn Medicine, Department of Pediatrics, Montreal Children's Hospital, McGill University, Montreal, QC, Canada; Waisman Research Center, Madison, WI, USA
| | - K Yibetal
- Addis Continental Institute of Public Health, Addis Ababa, Ethiopia; Waisman Research Center, Madison, WI, USA
| | - Scl Deoni
- Bill & Melinda Gates Foundation, MNCH D&T, Seattle, WA, USA; Waisman Research Center, Madison, WI, USA
| | - Scr Williams
- Centre for Neuroimaging Sciences, King's College London, London, UK; Waisman Research Center, Madison, WI, USA.
| |
Collapse
|
3
|
Normann SS, Beck IH, Nielsen F, Andersen MS, Bilenberg N, Jensen TK, Andersen HR. Prenatal exposure to pyrethroids and chlorpyrifos and IQ in 7-year-old children from the Odense Child Cohort. Neurotoxicol Teratol 2024; 103:107352. [PMID: 38636567 DOI: 10.1016/j.ntt.2024.107352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 03/07/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Organophosphates and pyrethroids are two major groups of insecticides used for crop protection worldwide. They are neurotoxicants and exposure during vulnerable windows of brain development may have long-term impact on human neurodevelopment. Only few longitudinal studies have investigated associations between prenatal exposure to these substances and intelligence quotient (IQ) at school age in populations with low, mainly dietary, exposure. OBJECTIVE To investigate associations between maternal urinary concentrations of insecticide metabolites at gestational week 28 and IQ in offspring at 7-years of age. MATERIALS AND METHODS Data was derived from the Odense Child Cohort (OCC). Metabolites of chlorpyrifos (TCPy) and pyrethroids (3-PBA, cis- and trans-DCCA, 4-F-3PBA, cis-DBCA) were measured in maternal urine collected at gestational week (GW) 28. An abbreviated version of the Danish Wechsler Intelligence Scale for Children fifth edition (WISC-V) consisting of four subtests to estimate full scale IQ (FSIQ) was administered by trained psychologists. Data were analyzed by use of multiple linear regression and adjusted for confounders. RESULTS 812 mother/child-pairs were included. Median concentrations were 0.21 μg/L for 3-PBA, 1.67 μg/L for TCPy and the mean IQ for children were 99.4. Null association between maternal 3-PBA and child IQ at 7 years was seen, but with trends suggesting an inverse association. There was a significant association for maternal TCPy and child IQ at mid-level exposure. Trans-DCCA above the level of detection (LOD) was also associated with slightly lower child IQ, but the association was also not statistically significant. CONCLUSIONS We found no significant associations between maternal 3-PBA metabolites and child IQ at 7 years, but with trends suggesting an inverse association. A non-significant trend between maternal TCPy exposure and child IQ in 7-year-children was seen even in this low exposed population. Given the widespread exposure and increasing use of insecticides, this should be elaborated in future studies.
Collapse
Affiliation(s)
- Stine Søgaard Normann
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark.
| | - Iben Have Beck
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Flemming Nielsen
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | | | - Niels Bilenberg
- Department of Child and Adolescent Psychiatry, Mental Health Services in Region of Southern Denmark, University of Southern Denmark, Odense, Denmark
| | - Tina Kold Jensen
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark; Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark; OPEN Patient data Explorative Network, Odense, Denmark
| | - Helle Raun Andersen
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
4
|
Smolińska K, Szopa A, Sobczyński J, Serefko A, Dobrowolski P. Nutritional Quality Implications: Exploring the Impact of a Fatty Acid-Rich Diet on Central Nervous System Development. Nutrients 2024; 16:1093. [PMID: 38613126 PMCID: PMC11013435 DOI: 10.3390/nu16071093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/05/2024] [Accepted: 04/07/2024] [Indexed: 04/14/2024] Open
Abstract
Given the comprehensive examination of the role of fatty acid-rich diets in central nervous system development in children, this study bridges significant gaps in the understanding of dietary effects on neurodevelopment. It delves into the essential functions of fatty acids in neurodevelopment, including their contributions to neuronal membrane formation, neuroinflammatory modulation, neurogenesis, and synaptic plasticity. Despite the acknowledged importance of these nutrients, this review reveals a lack of comprehensive synthesis in current research, particularly regarding the broader spectrum of fatty acids and their optimal levels throughout childhood. By consolidating the existing knowledge and highlighting critical research gaps, such as the effects of fatty acid metabolism on neurodevelopmental disorders and the need for age-specific dietary guidelines, this study sets a foundation for future studies. This underscores the potential of nutritional strategies to significantly influence neurodevelopmental trajectories, advocating an enriched academic and clinical understanding that can inform dietary recommendations and interventions aimed at optimizing neurological health from infancy.
Collapse
Affiliation(s)
- Katarzyna Smolińska
- Chronic Wounds Laboratory, Medical University of Lublin, Chodźki St. 7, 20-093 Lublin, Poland;
| | - Aleksandra Szopa
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Lublin, Chodźki St. 1, 20-093 Lublin, Poland; (A.S.); (J.S.); (A.S.)
| | - Jan Sobczyński
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Lublin, Chodźki St. 1, 20-093 Lublin, Poland; (A.S.); (J.S.); (A.S.)
| | - Anna Serefko
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Lublin, Chodźki St. 1, 20-093 Lublin, Poland; (A.S.); (J.S.); (A.S.)
| | - Piotr Dobrowolski
- Department of Functional Anatomy and Cytobiology, Maria Curie Sklodowska University, Akademicka St. 19, 20-033 Lublin, Poland
| |
Collapse
|
5
|
Petrican R, Fornito A, Boyland E. Lifestyle Factors Counteract the Neurodevelopmental Impact of Genetic Risk for Accelerated Brain Aging in Adolescence. Biol Psychiatry 2024; 95:453-464. [PMID: 37393046 DOI: 10.1016/j.biopsych.2023.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/30/2023] [Accepted: 06/19/2023] [Indexed: 07/03/2023]
Abstract
BACKGROUND The transition from childhood to adolescence is characterized by enhanced neural plasticity and a consequent susceptibility to both beneficial and adverse aspects of one's milieu. METHODS To understand the implications of the interplay between protective and risk-enhancing factors, we analyzed longitudinal data from the Adolescent Brain Cognitive Development (ABCD) Study (n = 834; 394 female). We probed the maturational correlates of positive lifestyle variables (friendships, parental warmth, school engagement, physical exercise, healthy nutrition) and genetic vulnerability to neuropsychiatric disorders (major depressive disorder, Alzheimer's disease, anxiety disorders, bipolar disorder, schizophrenia) and sought to further elucidate their implications for psychological well-being. RESULTS Genetic risk factors and lifestyle buffers showed divergent relationships with later attentional and interpersonal problems. These effects were mediated by distinguishable functional neurodevelopmental deviations spanning the limbic, default mode, visual, and control systems. More specifically, greater genetic vulnerability was associated with alterations in the normative maturation of areas rich in dopamine (D2), glutamate, and serotonin receptors and of areas with stronger expression of astrocytic and microglial genes, a molecular signature implicated in the brain disorders discussed here. Greater availability of lifestyle buffers predicted deviations in the normative functional development of higher density GABAergic (gamma-aminobutyric acidergic) receptor regions. The two profiles of neurodevelopmental alterations showed complementary roles in protection against psychopathology, which varied with environmental stress levels. CONCLUSIONS Our results underscore the importance of educational involvement and healthy nutrition in attenuating the neurodevelopmental sequelae of genetic risk factors. They also underscore the importance of characterizing early-life biomarkers associated with adult-onset pathologies.
Collapse
Affiliation(s)
- Raluca Petrican
- Institute of Population Health, Department of Psychology, University of Liverpool, Liverpool, United Kingdom.
| | - Alex Fornito
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| | - Emma Boyland
- Institute of Population Health, Department of Psychology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
6
|
Cunha-Rodrigues M, Rosário R, Duarte A, Silva MJ, Augusto C, Rodrigues M, Padrão P, Moreira P. Neurodevelopment and Dietary Intake in Toddlers-A Cross-Sectional Study from the Healthy Children 2021 Project. Nutrients 2023; 15:5105. [PMID: 38140364 PMCID: PMC10745499 DOI: 10.3390/nu15245105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Little is known about the potential associations between neurodevelopment, dietary diversity and food processing in the toddler period. This study aimed to estimate the association between these dietary quality dimensions and neurodevelopment in toddlers. Data for this cross-sectional analysis came from the Healthy Children 2021 project and included 212 toddlers (51.9% females, aged 12-36 months) from 15 Portuguese childcare centers. Neurodevelopment was assessed through Bayley Scales of Infant and Toddler Development. Dietary intake was gathered by a two-day non-consecutive dietary recall. The food items were categorised with NOVA classification. Dietary diversity was explored through Minimum Dietary Diversity (MDD). Logistic regression models adjusted for potential confounders were performed. Girls with a higher energy contribution of unprocessed/minimally processed foods and with an above median MDD score had higher odds of achieving a higher neurodevelopment score (aOR:1.04; 95%CI 1.01; 1.08 and aOR:2.26; 95%CI 1.01; 5.06, respectively); no significant association was observed in boys. Our findings suggest that these dietary dimensions are associated with a higher neurodevelopment in toddler girls. This should be further studied as a possible early link between dietary factors and neurodevelopment. Promotion of healthy eating can be promising in improving neurocognitive development and might help to introduce public health recommendations for toddlers' nutrition.
Collapse
Affiliation(s)
- Micaela Cunha-Rodrigues
- Faculty of Nutrition and Food Sciences, University of Porto, 4150-180 Porto, Portugal; (M.C.-R.); (M.R.); (P.P.)
| | - Rafaela Rosário
- School of Nursing, University of Minho, 4710-057 Braga, Portugal; (A.D.); (M.J.S.); (C.A.)
- Health Sciences Research Unit: Nursing (UICISA: E), Nursing School of Coimbra (ESEnfC), 3045-043 Coimbra, Portugal
- Nursing Research Centre, University of Minho, 4710-057 Braga, Portugal
| | - Ana Duarte
- School of Nursing, University of Minho, 4710-057 Braga, Portugal; (A.D.); (M.J.S.); (C.A.)
- Health Sciences Research Unit: Nursing (UICISA: E), Nursing School of Coimbra (ESEnfC), 3045-043 Coimbra, Portugal
- Research Centre on Child Studies (CIEC), Institute of Education, University do Minho, 4710-057 Braga, Portugal
| | - Maria José Silva
- School of Nursing, University of Minho, 4710-057 Braga, Portugal; (A.D.); (M.J.S.); (C.A.)
- Nursing Research Centre, University of Minho, 4710-057 Braga, Portugal
| | - Cláudia Augusto
- School of Nursing, University of Minho, 4710-057 Braga, Portugal; (A.D.); (M.J.S.); (C.A.)
- Health Sciences Research Unit: Nursing (UICISA: E), Nursing School of Coimbra (ESEnfC), 3045-043 Coimbra, Portugal
- Nursing Research Centre, University of Minho, 4710-057 Braga, Portugal
| | - Mónica Rodrigues
- Faculty of Nutrition and Food Sciences, University of Porto, 4150-180 Porto, Portugal; (M.C.-R.); (M.R.); (P.P.)
| | - Patrícia Padrão
- Faculty of Nutrition and Food Sciences, University of Porto, 4150-180 Porto, Portugal; (M.C.-R.); (M.R.); (P.P.)
- Epidemiology Research Unit and Laboratory for Integrative and Translational Research in Population Health, Institute of Public Health, University of Porto, 4050-600 Porto, Portugal
| | - Pedro Moreira
- Faculty of Nutrition and Food Sciences, University of Porto, 4150-180 Porto, Portugal; (M.C.-R.); (M.R.); (P.P.)
- Epidemiology Research Unit and Laboratory for Integrative and Translational Research in Population Health, Institute of Public Health, University of Porto, 4050-600 Porto, Portugal
| |
Collapse
|
7
|
Parolisi S, Montanari C, Borghi E, Cazzorla C, Zuvadelli J, Tosi M, Barone R, Bensi G, Bonfanti C, Dionisi Vici C, Biasucci G, Burlina A, Carbone MT, Verduci E. Possible role of tryptophan metabolism along the microbiota-gut-brain axis on cognitive & behavioral aspects in Phenylketonuria. Pharmacol Res 2023; 197:106952. [PMID: 37804926 DOI: 10.1016/j.phrs.2023.106952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Cognitive and psychiatric disorders are well documented across the lifetime of patients with inborn errors of metabolism (IEMs). Gut microbiota impacts behavior and cognitive functions through the gut-brain axis (GBA). According to recent research, a broad spectrum of GBA disorders may be influenced by a perturbed Tryptophan (Trp) metabolism and are associated with alterations in composition or function of the gut microbiota. Furthermore, early-life diets may influence children's neurodevelopment and cognitive deficits in adulthood. In Phenylketonuria (PKU), since the main therapeutic intervention is based on a life-long restrictive diet, important alterations of gut microbiota have been observed. Studies on PKU highlight the impact of alterations of gut microbiota on the central nervous system (CNS), also investigating the involvement of metabolic pathways, such as Trp and kynurenine (KYN) metabolisms, involved in numerous neurodegenerative disorders. An alteration of Trp metabolism with an imbalance of the KYN pathway towards the production of neurotoxic metabolites implicated in numerous neurodegenerative and inflammatory diseases has been observed in PKU patients supplemented with Phe-free amino acid medical foods (AA-MF). The present review investigates the possible link between gut microbiota and the brain in IEMs, focusing on Trp metabolism in PKU. Considering the evidence collected, cognitive and behavioral well-being should always be monitored in routine IEMs clinical management. Further studies are required to evaluate the possible impact of Trp metabolism, through gut microbiota, on cognitive and behavioral functions in IEMs, to identify innovative dietetic strategies and improve quality of life and mental health of these patients.
Collapse
Affiliation(s)
- Sara Parolisi
- UOSD Metabolic Diseases, AORN Santobono-Pausilipon, Naples, Italy
| | - Chiara Montanari
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Elisa Borghi
- Department of Health Science, University of Milan, Milan, Italy
| | - Chiara Cazzorla
- Division of Inherited Metabolic Diseases, DIDAS Department of Diagnostic Services, University Hospital, Padua, Italy
| | - Juri Zuvadelli
- Clinical Department of Pediatrics, San Paolo Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Martina Tosi
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy; Department of Health Science, University of Milan, Milan, Italy
| | - Rita Barone
- Child Neuropsychiatry Unit, Department of Clinical and Experimental Medicine, AOU Policlinico "G.Rodolico-San Marco", University of Catania, Catania, Italy
| | - Giulia Bensi
- Paediatrics & Neonatology Unit, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Cristina Bonfanti
- Rare metabolic disease unit, Pediatric Department, San Gerardo Hospital, Monza, Italy
| | | | - Giacomo Biasucci
- Paediatrics & Neonatology Unit, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Alberto Burlina
- Division of Inherited Metabolic Diseases, DIDAS Department of Diagnostic Services, University Hospital, Padua, Italy
| | - Maria T Carbone
- UOSD Metabolic Diseases, AORN Santobono-Pausilipon, Naples, Italy
| | - Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy; Department of Health Science, University of Milan, Milan, Italy.
| |
Collapse
|
8
|
Colombo J, Harris CL, Wampler JL, Zhuang W, Shaddy DJ, Liu BY, Wu SS. Improved Neurodevelopmental Outcomes at 5.5 Years of Age in Children Who Received Bovine Milk Fat Globule Membrane and Lactoferrin in Infant Formula Through 12 Months: A Randomized Controlled Trial. J Pediatr 2023; 261:113483. [PMID: 37192722 DOI: 10.1016/j.jpeds.2023.113483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/18/2023]
Abstract
OBJECTIVE To evaluate the neurodevelopmental outcomes at 5.5 years of age in children who were previously randomized to cow milk-based infant formula (control) or similar formula (milk fat globule membrane + lactoferrin) with added sources of bovine milk fat globule membrane and bovine lactoferrin through 12 months of age. DESIGN Children who completed study feeding were invited to participate in follow-up assessments: cognitive development across multiple domains (primary outcome; Wechsler Preschool & Primary Scale of Intelligence, 4th Edition), inhibitory control/rule learning (Stroop Task), flexibility/rule learning (Dimensional Change Card Sort), and behavior/emotion (Child Behavior Checklist). RESULTS Of 292 eligible participants (control: 148, milk fat globule membrane + lactoferrin: 144), 116 enrolled and completed assessments (control: 59, milk fat globule membrane + LF: 57). There were no group demographic differences except family income (milk fat globule membrane + lactoferrin significantly higher). Wechsler Preschool & Primary Scale of Intelligence, 4th Edition composite scores (mean ± standard error) for Visual Spatial (100.6 ± 1.7 vs 95.3 ± 1.7; P = .027), Processing Speed (107.1 ± 1.4 vs 100.0 ± 1.4; P < .001), and Full-Scale IQ (98.7 ± 1.4 vs 93.5 ± 1.5; P = .012) were significantly higher for milk fat globule membrane + lactoferrin versus control, even after controlling for demographic/socioeconomic factors. Stroop Task scores were significantly higher in milk fat globule membrane + lactoferrin versus control (P < .001). Higher Dimensional Change Card Sort scores (P = .013) in the border phase (most complex/challenging) were detected, and more children passed the border phase (32% vs 12%; P = .039) for milk fat globule membrane versus control. No group differences in Child Behavior Checklist score were detected. CONCLUSIONS Children who received infant formula to 12 months of age with added bovine milk fat globule membrane and bovine lactoferrin versus standard formula demonstrated improved cognitive outcomes in multiple domains at 5.5 years of age, including measures of intelligence and executive function. TRIAL REGISTRATION Clinicaltrials.gov: https://clinicaltrials.gov/ct2/show/NCT04442477.
Collapse
Affiliation(s)
- John Colombo
- Schiefelbusch Institute for Life Span Studies, University of Kansas, Lawrence, KS.
| | - Cheryl L Harris
- Medical Affairs, ReckittǀMead Johnson Nutrition Institute, Evansville, IN
| | - Jennifer L Wampler
- Medical Affairs, ReckittǀMead Johnson Nutrition Institute, Evansville, IN
| | - Weihong Zhuang
- Medical Affairs, ReckittǀMead Johnson Nutrition Institute, Evansville, IN
| | - D Jill Shaddy
- Department of Dietetics & Nutrition, University of Kansas Medical Center, Kansas City, KS
| | - Bryan Y Liu
- School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Steven S Wu
- Medical Affairs, ReckittǀMead Johnson Nutrition Institute, Evansville, IN; Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
9
|
Lapidot Y, Maya M, Reshef L, Cohen D, Ornoy A, Gophna U, Muhsen K. Relationships of the gut microbiome with cognitive development among healthy school-age children. Front Pediatr 2023; 11:1198792. [PMID: 37274812 PMCID: PMC10235814 DOI: 10.3389/fped.2023.1198792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/05/2023] [Indexed: 06/07/2023] Open
Abstract
Background The gut microbiome might play a role in neurodevelopment, however, evidence remains elusive. We aimed to examine the relationship between the intestinal microbiome and cognitive development of school-age children. Methods This cross-sectional study included healthy Israeli Arab children from different socioeconomic status (SES). The microbiome was characterized in fecal samples by implementing 16S rRNA gene sequencing. Cognitive function was measured using Stanford-Binet test, yielding full-scale Intelligence Quotient (FSIQ) score. Sociodemographics and anthropometric and hemoglobin measurements were obtained. Multivariate models were implemented to assess adjusted associations between the gut microbiome and FSIQ score, while controlling for age, sex, SES, physical growth, and hemoglobin levels. Results Overall, 165 children (41.2% females) aged 6-9 years were enrolled. SES score was strongly related to both FSIQ score and the gut microbiome. Measures of α-diversity were significantly associated with FSIQ score, demonstrating a more diverse, even, and rich microbiome with increased FSIQ score. Significant differences in fecal bacterial composition were found; FSIQ score explained the highest variance in bacterial β-diversity, followed by SES score. Several taxonomic differences were significantly associated with FSIQ score, including Prevotella, Dialister, Sutterella, Ruminococcus callidus, and Bacteroides uniformis. Conclusions We demonstrated significant independent associations between the gut microbiome and cognitive development in school-age children.
Collapse
Affiliation(s)
- Yelena Lapidot
- Department of Epidemiology and Preventive Medicine, School of Public Health, the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Maayan Maya
- Department of Epidemiology and Preventive Medicine, School of Public Health, the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Leah Reshef
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Dani Cohen
- Department of Epidemiology and Preventive Medicine, School of Public Health, the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Asher Ornoy
- Adelson School of Medicine, Ariel University, Ariel, Israel
- Department of Medical Neurobiology, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Uri Gophna
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Khitam Muhsen
- Department of Epidemiology and Preventive Medicine, School of Public Health, the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
10
|
Borasio F, De Cosmi V, D’Oria V, Scaglioni S, Syren MLE, Turolo S, Agostoni C, Coniglio M, Molteni M, Antonietti A, Lorusso ML. Associations between Dietary Intake, Blood Levels of Omega-3 and Omega-6 Fatty Acids and Reading Abilities in Children. Biomolecules 2023; 13:biom13020368. [PMID: 36830737 PMCID: PMC9952928 DOI: 10.3390/biom13020368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Lower levels of omega-3 polyunsaturated fatty acids (PUFAs) have been described in individuals with reading difficulties, but the degree and the nature of such deficiencies as well as the role of nutrition are a matter of debate. The aim of the present study was to investigate the associations between PUFA blood levels, nutritional status, and reading/writing/phonological awareness performances in 42 school-age children with varying levels of reading ability. Significant correlations were found between PUFA levels (specific omega-6/omega-3 ratios), the ratio of omega-6-derived calories to the total amount of calories and reading scores. Mediation analysis showed a mediating effect of fatty acids on the association between reading speed scores and nutritional status. Moderation analysis, moreover, showed that the associations of omega-6/omega-3 ratios in the blood and Kcal omega-6/Kcal total in dietary intake were moderated by reading speed performances. Results of the mediation and moderation models confirm that the associations of dietary intake with PUFA levels in the blood vary depending on learning abilities. Reading skills appear to be sensitive to the effects of a complex set of favorable conditions related to the presence of higher omega-3 blood levels. These conditions may reflect the action of dietary as well as genetic and epigenetic mechanisms.
Collapse
Affiliation(s)
- Francesca Borasio
- Scientific Institute IRCSS E. Medea, Unit of Child Psychopathology, 23842 Bosisio Parini, Italy
- Department of Psychology, Catholic University of the Sacred Heart, 20123 Milan, Italy
| | - Valentina De Cosmi
- Department of Clinical and Community Sciences, University of Milan, 20122 Milan, Italy
| | - Veronica D’Oria
- Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Anestesia e Terapia Intensiva Donna-Bambino, 20122 Milan, Italy
| | - Silvia Scaglioni
- Fondazione De Marchi, Department of Pediatrics, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | | | - Stefano Turolo
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, SC Nephrology Dialysis and Pediatric Transplantation, 20122 Milan, Italy
| | - Carlo Agostoni
- Department of Clinical and Community Sciences, University of Milan, 20122 Milan, Italy
- SC Pediatria-Immunoreumatologia, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Marilena Coniglio
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, SC Child and Adolescent Neuropsychiatry, 20122 Milan, Italy
| | - Massimo Molteni
- Scientific Institute IRCSS E. Medea, Unit of Child Psychopathology, 23842 Bosisio Parini, Italy
| | - Alessandro Antonietti
- Department of Psychology, Catholic University of the Sacred Heart, 20123 Milan, Italy
| | - Maria Luisa Lorusso
- Scientific Institute IRCSS E. Medea, Unit of Child Psychopathology, 23842 Bosisio Parini, Italy
- Correspondence:
| |
Collapse
|
11
|
Dou L, Gu J, Pan Y, Huang D, Huang Z, Bao H, Wu W, Zhu P, Tao F, Hao J. Prenatal Healthy Dietary Patterns Are Associated with Reduced Behavioral Problems of Preschool Children in China: A Latent Class Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2214. [PMID: 36767579 PMCID: PMC9916231 DOI: 10.3390/ijerph20032214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 06/18/2023]
Abstract
The relation between maternal dietary patterns during pregnancy and offspring behavioral problems is less verified. Therefore, we have aimed to assess the relationship between them and have hypothesized that children of mothers with healthy dietary patterns during pregnancy have better behavior. The 1612 mother-child pairs of the China-Anhui Birth Cohort Study (C-ABCS) have been enrolled as the study population. The dietary behaviors of mothers during early and mid-pregnancy have been investigated using a semi-quantitative food frequency questionnaire. Preschool child behavioral problems have been assessed. Clusters of maternal food groups intakes have been identified using latent class analysis, and the association between maternal dietary patterns and child behavioral problems has been subsequently analyzed using logistic regression. Maternal age at inclusion is 26.56 ± 3.51 years. There has been a preponderance of boys (53.3%). Maternal food groups intakes have been classified into four groups: "High-consumed pattern (HCP)", "Southern dietary pattern (SDP)", "Northern dietary pattern (NDP)", and "Low-consumed pattern (LCP)". The offspring with maternal SDP and NDP have lower emotional symptoms compared to the offspring with maternal LCP in the first trimester (p < 0.05). It has been reported to lower conduct problems in children with maternal SDP than the children with maternal LCP in the second trimester (p < 0.05). In boys, we have detected associations between first-trimester SDP and lower emotional symptoms (p < 0.05) and between second-trimester SDP with decreased peer relationship problems (p < 0.05). In girls, total difficulty scores are lower with second-trimester SDP (p < 0.05). Maternal SDP in early and mid-pregnancy predicts reduced behavioral problems in preschool children, while maternal HCP and NDP during pregnancy may result in fewer developmental benefits.
Collapse
Affiliation(s)
- Lianjie Dou
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Jijun Gu
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230032, China
| | - Ying Pan
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230032, China
| | - Dan Huang
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230032, China
| | - Zhaohui Huang
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230032, China
- Anhui Provincial Center for Women and Child Health, Hefei 230001, China
| | - Huihui Bao
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230032, China
| | - Wanke Wu
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230032, China
| | - Peng Zhu
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230032, China
| | - Fangbiao Tao
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230032, China
| | - Jiahu Hao
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei 230032, China
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei 230032, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
12
|
Lima RA, Soares FC, van Poppel M, Savinainen S, Mäntyselkä A, Haapala EA, Lakka T. Determinants of Cognitive Performance in Children and Adolescents: A Populational Longitudinal Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:8955. [PMID: 35897325 PMCID: PMC9331797 DOI: 10.3390/ijerph19158955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 12/07/2022]
Abstract
We evaluated the determinants of cognitive performance in children and adolescents. This is a longitudinal study, secondary analysis of the Physical Activity and Nutrition in Children (PANIC) study. We assessed 502 children (51.6% girls) at middle childhood (range: 6.6 to 9.0 years), at late childhood, 437 children (51.0% girls, range: 8.8 to 11.2 years), and in 277 adolescents (54.5% girls, range: 15.0 to 17.4 years). Raven's progressive matrices tests estimated the participants' cognitive performance (outcome variable) at all time points. In total, we evaluated 29 factors from various dimensions (prenatal, neonatal, child fitness, lifestyle and anthropometrics). None of the neonatal and anthropometric parameters were associated with cognitive performance. Preeclampsia (prenatal) and listening to music, writing, arts and craft and watching TV (lifestyle) were negatively associated with cognitive performance. Shuttle run and box and block tests (fitness), and playing music, reading and time at the computer (lifestyle) were positive determinants of cognitive performance in children and adolescents. Fitness and lifestyle factors during childhood and adolescence diminished the importance of prenatal factors on cognitive performance and lifestyle factors were especially relevant in regard to cognitive performance. Reading was positively associated with cognitive performance, regardless of age and time dedicated, and should be promoted.
Collapse
Affiliation(s)
- Rodrigo Antunes Lima
- Research, Innovation and Teaching Unit, Parc Sanitari Sant Joan de Déu, CIBERSAM, 08830 Sant Boi de Llobregat, Spain
| | - Fernanda Cunha Soares
- Division of Orthodontics and Pediatric Dentistry, Department of Dental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden;
| | | | - Saija Savinainen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, 70211 Kuopio, Finland; (S.S.); (E.A.H.); (T.L.)
- Department of Pediatrics, Kuopio University Hospital, 70211 Kuopio, Finland;
| | - Aino Mäntyselkä
- Department of Pediatrics, Kuopio University Hospital, 70211 Kuopio, Finland;
| | - Eero A. Haapala
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, 70211 Kuopio, Finland; (S.S.); (E.A.H.); (T.L.)
- Faculty of Sport and Health Sciences, University of Jyväskylä, 40014 Jyväskylä, Finland
| | - Timo Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, 70211 Kuopio, Finland; (S.S.); (E.A.H.); (T.L.)
- Department of Clinical Physiology and Nuclear Medicine, School of Medicine, Kuopio University Hospital, University of Eastern Finland, 70211 Kuopio, Finland
- Kuopio Research Institute of Exercise Medicine, 70100 Kuopio, Finland
| |
Collapse
|
13
|
AKIN S, GÜLTEKİN F, GÜLER EM. Dikkat Eksikliği ve Hiperaktivite Bozukluğunda Yağ Asitlerinin Rolü. ACTA MEDICA ALANYA 2022. [DOI: 10.30565/medalanya.1059552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is a childhood-onset disorder that affects 5% to 12% of children worldwide. Etiological factors, including nutrition, are involved in this disease, which is characterized by inattention, impulsivity, and hyperactivity symptoms. Fats, which form an important part of the daily diet, can have effects on ADHD and its symptoms. In the literature, it is stated that omega-3 fatty acids are low in children with ADHD, and supplementation studies may be effective in improving symptoms. In addition, high omega-6/omega-3 fatty acids ratio in the diet and diets rich in saturated and trans fatty acids are associated with ADHD. In this review, the relationship between ADHD and dietary fatty acids will be evaluated.
Collapse
Affiliation(s)
- Sümeyye AKIN
- SAĞLIK BİLİMLERİ ÜNİVERSİTESİ, HAMİDİYE SAĞLIK BİLİMLERİ ENSTİTÜSÜ, TIBBİ BİYOKİMYA (DR)
| | - Fatih GÜLTEKİN
- SAĞLIK BİLİMLERİ ÜNİVERSİTESİ, HAMİDİYE TIP FAKÜLTESİ, TEMEL TIP BİLİMLERİ BÖLÜMÜ, TIBBİ BİYOKİMYA ANABİLİM DALI
| | - Eray Metin GÜLER
- SAĞLIK BİLİMLERİ ÜNİVERSİTESİ, HAMİDİYE TIP FAKÜLTESİ, TEMEL TIP BİLİMLERİ BÖLÜMÜ, TIBBİ BİYOKİMYA ANABİLİM DALI
| |
Collapse
|
14
|
Jiang CB, Kao CS, Chien LC, Chen YJ, Liao KW. Associations among prenatal and postnatal arsenic, lead, and cadmium exposures and motor development in 3-year-old children: a longitudinal birth cohort study in Taiwan. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:43191-43200. [PMID: 35091938 DOI: 10.1007/s11356-021-18321-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/21/2021] [Indexed: 06/14/2023]
Abstract
Prenatal and postnatal exposures to heavy metals have been suggested to interfere with neurodevelopment, but the neurotoxicity of lead (Pb), arsenic (As), and cadmium (Cd) is still unclear. In this study, we aimed to assess the associations between the levels of As, Cd, and Pb and children's neurodevelopment. A total of 299 mother-infant pairs were recruited in this study and their meconium were collected. After three years, 53 children underwent the Bayley Scales of Infant and Toddler Development (Bayley-III) examinations and provided hair and fingernail specimens. The levels of As, Cd, and Pb in the meconium, hair, and fingernail were measured by inductively coupled plasma mass spectrometry; the median levels were the following: meconium, 42.7, 5.57, and 25.6 ng/g, respectively; hair, 0.19, 0.05, and 3.61 μg/g, respectively; and fingernail, 0.29, 0.04, and 0.84 μg/g, respectively. After adjusting for potential confounding factors, we found that the log-transformed levels of As in the hair samples was negatively associated with gross motor development (β = - 0.032; 95% confidence interval: - 0.061 to - 0.004). We conclude that postnatal exposure to As is a crucial period for gross motor development in children, while the effects of Cd and Pb on neurodevelopment are less clear.
Collapse
Affiliation(s)
- Chuen-Bin Jiang
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, MacKay Children's Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Chi-Sian Kao
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Ling-Chu Chien
- School of Public Health, Taipei Medical University, Taipei, Taiwan
- Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yi-Jhen Chen
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Kai-Wei Liao
- School of Food Safety, College of Nutrition, Taipei Medical University, 250 Wu-Xing Street, Taipei, Taiwan.
| |
Collapse
|
15
|
Lertxundi N, Molinuevo A, Valvi D, Gorostiaga A, Balluerka N, Shivappa N, Hebert J, Navarrete-Muñoz EM, Vioque J, Tardón A, Vrijheid M, Roumeliotaki T, Koutra K, Chatzi L, Ibarluzea J. Dietary inflammatory index of mothers during pregnancy and Attention Deficit-Hyperactivity Disorder symptoms in the child at preschool age: a prospective investigation in the INMA and RHEA cohorts. Eur Child Adolesc Psychiatry 2022; 31:615-624. [PMID: 33398651 PMCID: PMC8713648 DOI: 10.1007/s00787-020-01705-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Inflammation provides a substrate for mechanisms that underlie the association of maternal diet during pregnancy with Attention Deficit-Hyperactivity Disorder (ADHD) symptoms in childhood. However, no previous study has quantified the proinflammatory potential of maternal diet as a risk factor for ADHD. Thus, we evaluated the association of maternal dietary inflammatory index (DII®) scores during pregnancy with ADHD symptoms in 4-year-old children born in two Mediterranean regions. We analyzed data from two population-based birth cohort studies-INMA (Environment and Childhood) four subcohorts in Spain (N = 2097), and RHEA study in Crete (Greece) (N = 444). The DII score of maternal diet was calculated based on validated food frequency questionnaires completed during pregnancy (12th and/or 32nd week of gestation). ADHD symptoms were assessed by ADHD-DSM-IV in INMA cohort and by ADHDT test in RHEA cohort, with questionnaires filled-out by teachers and parents, respectively. The associations between maternal DII and ADHD symptoms were analysed using multivariable-adjusted zero-inflated negative binomial regression models in each cohort study separately. Meta-analysis was conducted to combine data across the cohorts for fitting within one model. The DII was significantly higher in RHEA (RHEA = 2.09 [1.94, 2.24]) in comparison to INMA subcohorts (Asturias = - 1.52 [- 1.67, - 1.38]; Gipuzkoa = - 1.48 [- 1.64, - 1.33]; Sabadell = - 0.95 [- 1.07, - 0.83]; Valencia = - 0.76 [- 0.90, - 0.62]). Statistically significant reduced risk of inattention symptomatology (OR = 0.86; CI 95% = 0.77-0.96), hyperactivity symptomatology (OR = 0.82; CI 95% = 0.72-0.92) and total ADHD symptomatology (OR = 0.82; CI 95% = - 0.72 to 0.93) were observed with increased maternal DII in boys. No statistically significant associations were observed in girls between maternal DII and inattention, hyperactivity and total ADHD symptomatology. We found reduced risk of ADHD symptomatology with increased DII only in boys. This relationship requires further exploration in other settings.
Collapse
Grants
- R21 ES029328, R21 ES028903 NIEHS NIH HHS
- P30 ES007048 NIEHS NIH HHS
- R21 ES028903 NIEHS NIH HHS
- R21 ES029328 NIEHS NIH HHS
- CIRIT 1999SGR 00241, Departament de Salut, Generalitat de Catalunya
- G03/176, CB06/02/0041, FIS-PI041436, FIS- PI081151, FIS-PI042018, FISPI09/02311, FIS-PI06/0867, FIS-PS09/00090, FIS-FEDER PI11/1007 FIS-FEDER 03/1615, 04/1509, 04/1112, 04/1931 , 05/1079, 05/1052, 06/1213, 07/0314, and 09/02647 Instituto de Salud Carlos III
- R21 ES029328, R21 ES028903 NIEHS NIH HHS
Collapse
Affiliation(s)
- Nerea Lertxundi
- University of the Basque Country UPV/EHU, Donostia-San Sebastián, Spain.
- Health Research Institute BIODONOSTIA, Donostia-San Sebastián, Spain.
| | - Amaia Molinuevo
- Public Health Division of Gipuzkoa, Donostia-San Sebastian, Spain
| | - Dania Valvi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine At Mount Sinai, New York, NY, USA
| | - Arantxa Gorostiaga
- University of the Basque Country UPV/EHU, Donostia-San Sebastián, Spain
- Health Research Institute BIODONOSTIA, Donostia-San Sebastián, Spain
| | - Nekane Balluerka
- University of the Basque Country UPV/EHU, Donostia-San Sebastián, Spain
- Health Research Institute BIODONOSTIA, Donostia-San Sebastián, Spain
| | - Nitin Shivappa
- Cancer Prevention and Control Program and Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Connecting Health Innovations LLC, Columbia, SC, USA
| | - James Hebert
- Cancer Prevention and Control Program and Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- Connecting Health Innovations LLC, Columbia, SC, USA
| | - Eva María Navarrete-Muñoz
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Institute for Health and Biomedical Research, ISABIAL-UMH, Alicante, Spain
- Department of Surgery and Pathology, Miguel Hernández University, Alicante, Spain
| | - Jesus Vioque
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Institute for Health and Biomedical Research, ISABIAL-UMH, Alicante, Spain
| | | | - Martine Vrijheid
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
- ISGlobal Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Theano Roumeliotaki
- Department of Psychology, School of Social Sciences, University of Crete, Heraklion, Greece
| | - Katerina Koutra
- Department of Psychology, School of Social Sciences, University of Crete, Heraklion, Greece
| | - Leda Chatzi
- Department of Preventive Medicine, University of Southern California, Los Angeles, USA
| | - Jesus Ibarluzea
- University of the Basque Country UPV/EHU, Donostia-San Sebastián, Spain
- Health Research Institute BIODONOSTIA, Donostia-San Sebastián, Spain
- Public Health Division of Gipuzkoa, Donostia-San Sebastian, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|
16
|
Nieto-Ruiz A, García-Santos JA, Verdejo-Román J, Diéguez E, Sepúlveda-Valbuena N, Herrmann F, Cerdó T, De-Castellar R, Jiménez J, Bermúdez MG, Pérez-García M, Miranda MT, López-Sabater MC, Catena A, Campoy C. Infant Formula Supplemented With Milk Fat Globule Membrane, Long-Chain Polyunsaturated Fatty Acids, and Synbiotics Is Associated With Neurocognitive Function and Brain Structure of Healthy Children Aged 6 Years: The COGNIS Study. Front Nutr 2022; 9:820224. [PMID: 35356726 PMCID: PMC8959863 DOI: 10.3389/fnut.2022.820224] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/01/2022] [Indexed: 12/25/2022] Open
Abstract
Background Adequate nutrient intake during the first few months of life plays a critical role on brain structure and function development. Objectives To analyze the long-term effects of an experimental infant formula (EF) on neurocognitive function and brain structure in healthy children aged 6 years compared to those fed with a standard infant formula or breastfed. Methods The current study involved 108 healthy children aged 6 years and participating in the COGNIS Study. At 0-2 months, infants were randomized to receive up to 18 months of life a standard infant formula (SF) or EF enriched with milk fat globule membrane (MFGM), long-chain polyunsaturated fatty acids (LC-PUFAs) and synbiotics. Furthermore, a reference group of breastfed (BF) infants were also recruited. Children were assessed using neurocognitive tests and structural Magnetic Resonance Imaging (MRI) at 6 years old. Results Experimental infant formula (EF) children showed greater volumes in the left orbital cortex, higher vocabulary scores and IQ, and better performance in an attention task than BF children. EF children also presented greater volumes in parietal regions than SF kids. Additionally, greater cortical thickness in the insular, parietal, and temporal areas were found in children from the EF group than those fed with SF or BF groups. Further correlation analyses suggest that higher volumes and cortical thickness of different parietal and frontal regions are associated with better cognitive development in terms of language (verbal comprehension) and executive function (working memory). Finally, arachidonic acid (ARA), adrenic acid (AdA), docosahexaenoic acid (DHA) levels in cheek cell glycerophospholipids, ARA/DHA ratio, and protein, fatty acid, and mineral intake during the first 18 months of life seem to be associated with changes in the brain structures at 6 years old. Conclusions Supplemented infant formula with MFGM components, LC-PUFAs, and synbiotics seems to be associated to long-term effects on neurocognitive development and brain structure in children at 6 years old. Clinical Trial Registration https://www.clinicaltrials.gov/, identifier: NCT02094547.
Collapse
Affiliation(s)
- Ana Nieto-Ruiz
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Health Sciences Technological Park, Granada, Spain
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
| | - José A. García-Santos
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Health Sciences Technological Park, Granada, Spain
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
| | - Juan Verdejo-Román
- Department of Personality, Assessment & Psychological Treatment, School of Psychology, University of Granada, Granada, Spain
| | - Estefanía Diéguez
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Health Sciences Technological Park, Granada, Spain
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
| | - Natalia Sepúlveda-Valbuena
- Nutrition and Biochemistry Department, Faculty of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Florian Herrmann
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
| | - Tomás Cerdó
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
- Carlos III Health Institute, Madrid, Spain
| | | | | | - Mercedes G. Bermúdez
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Health Sciences Technological Park, Granada, Spain
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
| | - Miguel Pérez-García
- Department of Personality, Assessment & Psychological Treatment, School of Psychology, University of Granada, Granada, Spain
- Mind, Brain and Behavior Research Centre—CIMCYC, University of Granada, Granada, Spain
| | - M. Teresa Miranda
- Department of Biostatistics, School of Medicine, University of Granada, Granada, Spain
| | - M. Carmen López-Sabater
- Department of Nutrition, Food Sciences and Gastronomy, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institut de Recerca en Nutrició i Seguretat Alimentària de la UB (INSA-UB), Barcelona, Spain
- National Network of Research in Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III (Barcelona's Node), Madrid, Spain
| | - Andrés Catena
- Mind, Brain and Behavior Research Centre—CIMCYC, University of Granada, Granada, Spain
- Department of Experimental Psychology, School of Psychology, University of Granada, Granada, Spain
| | - Cristina Campoy
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Health Sciences Technological Park, Granada, Spain
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
- National Network of Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III (Granada's Node), Madrid, Spain
| |
Collapse
|
17
|
Vlug LE, Verloop MW, Dierckx B, Bosman L, de Graaff JC, Rings EH, Wijnen RM, de Koning BA, Legerstee JS. Cognitive Outcomes in Children With Conditions Affecting the Small Intestine: A Systematic Review and Meta-analysis. J Pediatr Gastroenterol Nutr 2022; 74:368-376. [PMID: 35226646 PMCID: PMC8860224 DOI: 10.1097/mpg.0000000000003368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/09/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The aim of the study was to assess cognitive outcomes in children with intestinal failure (IF) and children at high risk of IF with conditions affecting the small intestine requiring parenteral nutrition. METHODS EMBASE, Cochrane, Web of Science, Google Scholar, MEDLINE, and PsycINFO were searched from inception to October 2020. Studies were included constituting original data on developmental quotient (DQ), intelligence quotient (IQ) and/or severe developmental delay/disability (SDD) rates assessed with standardized tests. We used appropriate standardized tools to extract data and assess study quality. We performed random effects meta-analyses to estimate pooled means of DQ/IQ and pooled SDD rates (general population mean for DQ/IQ: 100, for percentage with SDD: 1.8%) for 4 groups: IF, surgical necrotizing enterocolitis (NEC), abdominal wall defects (AWD), and midgut malformations (MM). Associations of patient characteristics with DQ/IQ were evaluated with meta-regressions. RESULTS Thirty studies met the inclusion criteria. The pooled mean DQ/IQ for IF, NEC, AWD, and MM were 86.8, 83.3, 96.6, and 99.5, respectively. The pooled SDD rates for IF, NEC, AWD and MM were 28.6%, 32.8%, 8.5%, and 3.7%, respectively. Meta-regressions indicated that lower gestational age, longer hospital stay, and higher number of surgeries but not parenteral nutrition duration, were associated with lower DQ/IQ. CONCLUSIONS Adverse developmental outcomes are common in children with IF and NEC, and to a much lesser extent in children with AWD and MM. It is important to monitor cognitive development in children with conditions affecting the small intestine and to explore avenues for prevention and remediation.
Collapse
Affiliation(s)
- Lotte E. Vlug
- Division of Gastroenterology, Department of Pediatrics
| | | | - Bram Dierckx
- Department of Child and Adolescent Psychiatry/Psychology
| | - Lotte Bosman
- Department of Child and Adolescent Psychiatry/Psychology
| | - Jurgen C. de Graaff
- Department of Anesthesiology, Erasmus MC Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam
| | - Edmond H.H.M. Rings
- Division of Gastroenterology, Department of Pediatrics
- Division of Gastroenterology, Department of Pediatrics, Willem Alexander Children's Hospital, Leiden University Medical Center, Leiden
| | - René M.H. Wijnen
- Department of Pediatric Surgery, Erasmus MC Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | | |
Collapse
|
18
|
Silveira JS, Ramires Júnior OV, Schmitz F, Ferreira FS, Rodrigues FC, Silva RC, Savio LEB, Wyse ATS. Folic acid supplementation during pregnancy alters behavior in male rat offspring: nitrative stress and neuroinflammatory implications. Mol Neurobiol 2022; 59:2150-2170. [PMID: 35044624 DOI: 10.1007/s12035-022-02724-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/30/2021] [Indexed: 12/27/2022]
Abstract
Pregnancy diet can impact offspring's neurodevelopment, metabolism, redox homeostasis, and inflammatory status. In pregnancy, folate demand is increased due to the requirement for one-carbon transfer reactions. The present study was proposed to investigate the effect of folic acid supplementation throughout pregnancy on a battery of behavior tests (olfactory preference, motor activity, exploratory capacity, habituation, memory, anxiety- and depression-like behavior). Redox homeostasis and neuroinflammatory status in cerebral cortex were also investigated. After pregnancy confirmation, the pregnant rats were randomly divided into two groups, according to the diet: group 1, (control) standard diet (2 mg/kg diet of folic acid) and group 2, supplemented diet with 4 mg/kg diet of folic acid. Throughout the gestational period, the pregnant rats received experimental diets. Results show that the supplemented diet with 4 mg/kg diet of folic acid throughout pregnancy impaired memory and motricity of the offspring when compared with control (standard diet). It was also observed an increase in anxiety- and depression-like behavior in this group. Nitrite levels increased in cerebral cortex of the offspring, when compared to control group. In contrast, iNOS expression and immunocontent were not altered. Moreover, we identify an increase in TNF-α, IL-1β, IL-6, IL-10, and MCP-1 gene expression in the cerebral cortex. In conclusion, our study showed that the supplemented diet with 4 mg/kg diet of folic acid throughout pregnancy may cause behavioral and biochemical changes in the male offspringGraphical abstract After pregnancy confirmation, the pregnant rats were randomly divided into two groups, according to the diet: group 1, (control) standard diet (2 mg/kg diet of folic acid) and group 2, supplemented diet with 4 mg/kg diet of folic acid. Throughout the gestational period, the pregnant rats received experimental diets. Results show that folic acid supplementation did not impair the mother-pup relationship. We showed that supplemented diet with 4 mg/kg diet of folic acid during pregnancy impairs memory and motricity of the offspring when compared with standard diet. It was also observed an increase in anxiety- and depression-like behavior in this group. Nitrative stress and neuroinflammation parameters were increased in the cerebral cortex of the offspring. ROS, reactive oxygen species.
Collapse
Affiliation(s)
- Josiane Silva Silveira
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Laboratório de Neuroproteção E Doenças Metabólicas (Wyse's Lab), Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, Rio Grande do Sul, 90035003, Brazil
| | - Osmar Vieira Ramires Júnior
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Laboratório de Neuroproteção E Doenças Metabólicas (Wyse's Lab), Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, Rio Grande do Sul, 90035003, Brazil
| | - Felipe Schmitz
- Laboratório de Neuroproteção E Doenças Metabólicas (Wyse's Lab), Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, Rio Grande do Sul, 90035003, Brazil
| | - Fernanda Silva Ferreira
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Rio Grande do Sul, Brazil.,Laboratório de Neuroproteção E Doenças Metabólicas (Wyse's Lab), Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, Rio Grande do Sul, 90035003, Brazil
| | - Fabiana Cristina Rodrigues
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Bloco G. Av. Carlos Chagas Filho, 373. Cidade Universitária, Ilha Do Fundão, Rio de Janeiro, 21941-902, Brazil
| | - Robson Coutinho Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Bloco G. Av. Carlos Chagas Filho, 373. Cidade Universitária, Ilha Do Fundão, Rio de Janeiro, 21941-902, Brazil
| | - Luiz Eduardo Baggio Savio
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Bloco G. Av. Carlos Chagas Filho, 373. Cidade Universitária, Ilha Do Fundão, Rio de Janeiro, 21941-902, Brazil
| | - Angela T S Wyse
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Rio Grande do Sul, Brazil. .,Laboratório de Neuroproteção E Doenças Metabólicas (Wyse's Lab), Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, Rio Grande do Sul, 90035003, Brazil.
| |
Collapse
|
19
|
The Role of Vitamin D Supplementation in Children with Autism Spectrum Disorder: A Narrative Review. Nutrients 2021; 14:nu14010026. [PMID: 35010901 PMCID: PMC8746934 DOI: 10.3390/nu14010026] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022] Open
Abstract
Children with autism spectrum disorder (ASD) present with persistent deficits in both social communication and interactions, along with the presence of restricted and repetitive behaviors, resulting in significant impairment in significant areas of functioning. Children with ASD consistently reported significantly lower vitamin D levels than typically developing children. Moreover, vitamin D deficiency was found to be strongly correlated with ASD severity. Theoretically, vitamin D can affect neurodevelopment in children with ASD through its anti-inflammatory properties, stimulating the production of neurotrophins, decreasing the risk of seizures, and regulating glutathione and serotonin levels. A Title/Abstract specific search for publications on Vitamin D supplementation trials up to June 2021 was performed using two databases: PubMed and Cochrane Library. Twelve experimental studies were included in the synthesis of this review. Children with ASD reported a high prevalence of vitamin D deficiency or insufficiency. In general, it was observed that improved vitamin D status significantly reduced the ASD severity, however, this effect was not consistently different between the treatment and control groups. The variations in vitamin D dose protocols and the presence of concurrent interventions might provide an explanation for the variability of results. The age of the child for introducing vitamin D intervention was identified as a possible factor determining the effectiveness of the treatment. Common limitations included a small number of participants and a short duration of follow-ups in the selected studies. Long-term, well-designed randomized controlled trials are warranted to confirm the effect of vitamin D on severity in children with ASD.
Collapse
|
20
|
Lee P, Tse LA, László KD, Wei D, Yu Y, Li J. Association of maternal gestational weight gain with intellectual developmental disorder in the offspring: a nationwide follow-up study in Sweden. BJOG 2021; 129:540-549. [PMID: 34455681 DOI: 10.1111/1471-0528.16887] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2021] [Indexed: 12/01/2022]
Abstract
OBJECTIVES To investigate the association between maternal gestational weight gain (GWG) and offspring's intellectual developmental disorders (IDD); how this association is modified by maternal early-pregnancy BMI. DESIGN Population-based cohort study. SETTING AND POPULATION All liveborn singletons with information on maternal GWG in the Swedish Medical Register during 1992-2006 (n = 467 485). METHODS We used three GWG classifications, (1) Institute of Medicine (IOM) guidelines ('ideal' GWG: maternal underweight = 12.7-18.1 kg; normal = 11.3-15.9 kg; overweight = 6.8-11.3 kg; obesity = 5.0-9.1 kg), (2) LifeCycle project recommendation ('ideal' GWG: maternal underweight = 14.0-16.0 kg; normal = 10.0-18.0 kg; overweight = 2.0-16.0 kg; obesity class I = 2.0-6.0 kg; obesity class II ≤0.0-4.0 kg; obesity class III ≤0.0-6.0 kg) and (3) GWG centiles. Hazard ratio (HR) and 95% CI for offspring's IDD risk using Cox regression. MAIN OUTCOME MEASURES IDD was extracted from Swedish National Patient Register (code ICD-9:317-319/ICD-10:F70-F79). RESULTS Forty-one per cent of children were born to mothers with excessive GWG, 32.8% with ideal GWG and 26.2% with inadequate GWG according to IOM guidelines. Inadequate GWG was associated with 21% higher risk of offspring's IDD (95% CI 1.11-1.31) relative to ideal GWG. In contrast, when using the LifeCycle classification, children of mothers with inadequate GWG (HR 1.14, 95% CI 1.05-1.24) or excessive GWG (HR 1.09, 95% CI 1.01-1.17) had higher risks of IDD than those of mothers with ideal GWG. When using GWG centiles, extremely low GWG (<20th centile) and low GWG (20th-40th centile) were associated with elevated offspring's IDD risk. Further stratified analysis by maternal early-pregnancy body mass index (BMI) showed that overweight/obese mothers (BMI ≥25 kg/m2 ) with extremely excessive GWG (>25 kg) was associated with an increased offspring's IDD. CONCLUSION Our findings suggest that inadequate maternal GWG may increase offspring's IDD risk, irrespective of maternal early-pregnancy BMI. Extremely excessive GWG (>25 kg) may increase offspring's IDD risk, but only among mothers with an early-pregnancy BMI ≥25 kg/m2 . TWEETABLE ABSTRACT Inadequate maternal weight gain during pregnancy may increase the risk of offspring's intellectual disability, regardless of maternal BMI.
Collapse
Affiliation(s)
- Pmy Lee
- JC School of Public Health and Primary Care, the Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - L A Tse
- JC School of Public Health and Primary Care, the Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - K D László
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - D Wei
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Y Yu
- Department of Clinical Medicine-Department of Clinical Epidemiology, Aarhus University, Aarhus, Denmark
| | - J Li
- Department of Clinical Medicine-Department of Clinical Epidemiology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
21
|
Sanefuji M, Senju A, Shimono M, Ogawa M, Sonoda Y, Torio M, Ichimiya Y, Suga R, Sakai Y, Honjo S, Kusuhara K, Ohga S. Breast feeding and infant development in a cohort with sibling pair analysis: the Japan Environment and Children's Study. BMJ Open 2021; 11:e043202. [PMID: 34380712 PMCID: PMC8359520 DOI: 10.1136/bmjopen-2020-043202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVES To investigate the association between breast feeding and infant development during the first year of life using sibling comparison. DESIGN Nationwide prospective birth cohort study with sibling pair analysis. SETTING 15 regional centres that participated in the Japan Environment and Children's Study. PARTICIPANTS This study included 77 119 children (singleton, term birth and no malformation/severe diseases) whose mothers were registered between January 2011 and March 2014, including 3521 duos or trios of siblings. PRIMARY OUTCOME MEASURES The primary outcome was developmental delay at 6 and 12 months of age, assessed using the Japanese translation of the Ages and Stages Questionnaires, third edition. Multivariable regression analyses adjusted for confounders were performed to estimate the risk ratios of delay associated with any or exclusive breast feeding. Pairs of siblings discordant for statuses were selected, and conditional regression analyses were conducted with a matched cohort design. RESULTS Developmental delay was identified in 6162 (8.4%) and 10 442 (14.6%) children at 6 and 12 months of age, respectively. Any breast feeding continued until 6 months or 12 months old was associated with reduced developmental delay at 12 months of age (adjusted risk ratio (95% CI): 0.81 (0.77 to 0.85) and 0.81 (0.78 to 0.84), respectively). Furthermore, exclusive breast feeding until 3 months was associated with reduced developmental delay at 12 months of age (adjusted risk ratio, 0.86 (95% CI 0.83 to 0.90)). In sibling pair analysis, the association between any breast feeding until 12 months and reduced developmental delay at 12 months of age persisted (adjusted risk ratio, 0.64 (95% CI 0.43 to 0.93)). CONCLUSIONS The present study demonstrated the association of continuous breast feeding with reduced developmental delay at 1 year of age using sibling pair analysis, in which unmeasured confounding factors are still present but less included. This may provide an argument to promote breastfeeding continuation.
Collapse
Affiliation(s)
- Masafumi Sanefuji
- Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ayako Senju
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu, Japan
- Regional Center for Japan Environment and Children's Study, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Masayuki Shimono
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu, Japan
- Regional Center for Japan Environment and Children's Study, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Masanobu Ogawa
- Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuri Sonoda
- Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Michiko Torio
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuko Ichimiya
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Reiko Suga
- Regional Center for Japan Environment and Children's Study, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoshi Honjo
- Department of Pediatrics, National Hospital Organization Fukuoka National Hospital, Fukuoka, Japan
| | - Koichi Kusuhara
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu, Japan
- Regional Center for Japan Environment and Children's Study, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shouichi Ohga
- Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
22
|
Chiu Y, Fadadu RP, Gaskins AJ, Rifas‐Shiman SL, Laue HE, Moley KH, Hivert M, Baccarelli A, Oken E, Chavarro JE, Cardenas A. Dietary fat intake during early pregnancy is associated with cord blood DNA methylation at IGF2 and H19 genes in newborns. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2021; 62:388-398. [PMID: 34288135 PMCID: PMC8364885 DOI: 10.1002/em.22452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/11/2021] [Accepted: 07/16/2021] [Indexed: 05/03/2023]
Abstract
Maternal fat intake during pregnancy affects fetal growth, but mechanisms underlying this relationship are unclear. We performed an exploratory study of the associations of fat consumption during pregnancy with cord blood DNA methylation of the insulin-like growth factor 2 (IGF2) and H19 genes. We used data from 96 uncomplicated full-term pregnancies of mothers of whom majority had normal body mass index (BMI) (66%) in Project Viva, a prospective pre-birth cohort. We assessed maternal diet with validated food frequency questionnaires during the first and second trimesters and measured DNA methylation in segments of the IGF2- and H19-differentially methylated regions (DMRs) by pyrosequencing DNA extracted from umbilical cord blood samples. Mean (SD) age was 32.8 (4.1) years and prepregnancy BMI was 24.0 (4.4) kg/m2 . Mean DNA methylation was 56.3% (3.9%) for IGF2-DMR and 44.6% (1.9%) for H19-DMR. Greater first trimester intake of omega-6 polyunsaturated fat (effect per 1% of calories at the expense of carbohydrates) was associated with lower DNA methylation of IGF2-DMR (-1.2%; 95% confidence interval [CI]: -2.2%, -0.2%) and higher DNA methylation at H19-DMR (0.8%; 95% CI: 0.3%, 1.3%). On the other hand, greater first trimester intake of omega-3 polyunsaturated fat was associated with lower DNA methylation of the H19-DMR (-4.3%; 95% CI: -7.9%, -0.8%). We did not find significant associations of IGF2 and H19 methylation with IGF2 cord blood levels. Our findings suggest that early prenatal fat intake (omega-3, omega-6, and saturated fatty acids) may influence DNA methylation at the IGF2 and H19 locus, which could impact fetal development and long-term health.
Collapse
Affiliation(s)
- Yu‐Han Chiu
- Department of EpidemiologyHarvard TH Chan School of Public HealthBostonMassachusettsUSA
| | - Raj P. Fadadu
- School of MedicineUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Division of Environmental Health SciencesUniversity of California, Berkeley School of Public Health, BerkeleyBerkeleyCaliforniaUSA
| | - Audrey J. Gaskins
- Department of EpidemiologyRollins School of Public Health, Emory UniversityAtlantaGeorgiaUSA
| | - Sheryl L. Rifas‐Shiman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
| | - Hannah E. Laue
- Department of EpidemiologyGeisel School of Medicine at Dartmouth CollegeHanoverNew HampshireUSA
| | - Kelle H. Moley
- Department of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Marie‐France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
- Diabetes Unit, Massachusetts General HospitalBostonMassachusettsUSA
| | - Andrea Baccarelli
- Department of Environmental Health SciencesMailman School of Public Health, Columbia UniversityNew York CityNew YorkUSA
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population MedicineHarvard Medical School and Harvard Pilgrim Health Care InstituteBostonMassachusettsUSA
| | - Jorge E. Chavarro
- Department of EpidemiologyHarvard TH Chan School of Public HealthBostonMassachusettsUSA
- Department of NutritionHarvard TH Chan School of Public HealthBostonMassachusettsUSA
- Channing Division of Network Medicine, Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Andres Cardenas
- Division of Environmental Health SciencesUniversity of California, Berkeley School of Public Health, BerkeleyBerkeleyCaliforniaUSA
| |
Collapse
|
23
|
Mantey AA, Annan RA, Lutterodt HE, Twumasi P. Iron status predicts cognitive test performance of primary school children from Kumasi, Ghana. PLoS One 2021; 16:e0251335. [PMID: 34010354 PMCID: PMC8133497 DOI: 10.1371/journal.pone.0251335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 04/25/2021] [Indexed: 01/01/2023] Open
Abstract
Background Good nutritional status of school-aged children is crucial in achieving improved cognition. The objective of this study was to assess the relationship between nutritional status and cognition of school-aged children in the Kumasi metropolis, Ghana. Methods 389 children were selected from ten government-owned schools. Socio-demographic and anthropometric data were collected. Blood samples were collected and analyzed for nutrients levels. Dietary intakes were assessed using food frequency questionnaire and previous day’s nutrients intake. Cognition test was performed using the Ravens Colored Progressive Matrix (RCPM). Results Mean age of participants was 8.9±1.4 years, mean RCPM score was 17.9±5.4. More girls scored RCPM below the 40th percentile (45.5%) than boys (33.7%), while mother’s level of education significantly associated with RCPM percentiles of the children (p = 0.037). Four dietary patterns were generated from food frequency data, and scores of the second pattern, depicting vegetables, non-fruits, bread and cereals, showed a weak negative correlation (r = -0.132, p = 0.026) with previous day’s dietary zinc intake. Cognitive status did not vary by anthropometric and dietary patterns. More anemic (54.4%) than non-anemic (33.3%) children were below the 40th RCPM percentile. Mean previous day’s intake for folate (p<0.001), vitamin B6 (p = 0.018), iron (p<0.001), and zinc (p = 0.001) differed significantly between the cognitive test score percentiles of the children. Spearman rank correlation showed weak positive associations between RCPM score and hemoglobin (r = 0.246, p = 0.003) and serum ferritin (r = 0.176, p = 0.036). Binary regression analysis showed anemic children (aOR = 0.4; 95%CI = 0.2–0.8, p = 0.014), compared with non-anemic had decreased odds, while boys, compared with girls had increased odds (aOR = 2.0 95%CI = 1.0–4.0, p = 0.035) for scoring above the 50th RCPM percentile. Conclusions Iron status, especially hemoglobin levels, correlated with the cognitive performance of school-aged children in the metropolis. Thus nutritional strategies aimed at reducing iron deficiency anemia are needed.
Collapse
Affiliation(s)
- Afua Afreh Mantey
- Department of Laboratory Technology, Kumasi Technical University, Kumasi, Ghana
- Department of Biochemistry and Biotechnology, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- * E-mail:
| | - Reginald Adjetey Annan
- Department of Biochemistry and Biotechnology, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Herman Erick Lutterodt
- Department of Food Science and Technology, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | | |
Collapse
|
24
|
Tsuji JS, Lennox KP, Watson HN, Chang ET. Essential concepts for interpreting the dose-response of low-level arsenic exposure in epidemiological studies. Toxicology 2021; 457:152801. [PMID: 33905760 DOI: 10.1016/j.tox.2021.152801] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 11/25/2022]
Abstract
Scientifically robust selections of epidemiological studies and assessments of the dose-response of inorganic arsenic in the low-dose range must consider key issues specific to arsenic in order to reduce risk of bias. The abundance of toxicological, mechanistic, and epidemiological evidence on arsenic enables a nuanced assessment of risk of bias in epidemiological studies of low-level arsenic, as opposed to a generic evaluation based only on standard principles. Important concepts in this context include 1) arsenic metabolism and mode of action for toxicity and carcinogenicity; 2) effects of confounding factors such as diet, health status including nutritional deficiencies, use of tobacco and other substances, and body composition; 3) strengths and limitations of various metrics for assessing relevant exposures consistent with the mode of action; and 4) the potential for bias in the positive direction for the observed dose-response relationship as exposure increases in the low-dose range. As an example, evaluation of a recent dose-response modeling using eight epidemiological studies of inorganic arsenic and bladder cancer demonstrated that the pooled risk estimate was markedly affected by the single study that was ranked as having a high risk of bias, based on the above factors. The other seven studies were also affected by these factors to varying, albeit lesser, degrees that can influence the apparent dose-response in the low-dose range (i.e., drinking water concentration of 65 µg/L or dose of approximately ≤1 µg/kg-day). These issues are relevant considerations for assessing health risks of oral exposures to inorganic arsenic in the U.S. population, and setting evidence-based regulatory limits to protect human health.
Collapse
|
25
|
Bordeleau M, Fernández de Cossío L, Chakravarty MM, Tremblay MÈ. From Maternal Diet to Neurodevelopmental Disorders: A Story of Neuroinflammation. Front Cell Neurosci 2021; 14:612705. [PMID: 33536875 PMCID: PMC7849357 DOI: 10.3389/fncel.2020.612705] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Providing the appropriate quantity and quality of food needed for both the mother's well-being and the healthy development of the offspring is crucial during pregnancy. However, the macro- and micronutrient intake also impacts the body's regulatory supersystems of the mother, such as the immune, endocrine, and nervous systems, which ultimately influence the overall development of the offspring. Of particular importance is the association between unhealthy maternal diet and neurodevelopmental disorders in the offspring. Epidemiological studies have linked neurodevelopmental disorders like autism spectrum disorders, attention-deficit-hyperactivity disorder, and schizophrenia, to maternal immune activation (MIA) during gestation. While the deleterious consequences of diet-induced MIA on offspring neurodevelopment are increasingly revealed, neuroinflammation is emerging as a key underlying mechanism. In this review, we compile the evidence available on how the mother and offspring are both impacted by maternal dietary imbalance. We specifically explore the various inflammatory and anti-inflammatory effects of dietary components and discuss how changes in inflammatory status can prime the offspring brain development toward neurodevelopmental disorders. Lastly, we discuss research evidence on the mechanisms that sustain the relationship between maternal dietary imbalance and offspring brain development, involving altered neuroinflammatory status in the offspring, as well as genetic to cellular programming notably of microglia, and the evidence that the gut microbiome may act as a key mediator.
Collapse
Affiliation(s)
- Maude Bordeleau
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | | | - M. Mallar Chakravarty
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Cerebral Imaging Centre, Douglas Mental Health University, McGill University, Montréal, QC, Canada
- Department of Psychiatry, McGill University, Montréal, QC, Canada
- Department of Biological and Biomedical Engineering, McGill University, Montréal, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
26
|
Campoy C, Azaryah H, Torres-Espínola FJ, Martínez-Zaldívar C, García-Santos JA, Demmelmair H, Haile G, Rzehak P, Koletzko B, Györei E, Décsi T, Ramírez-Tortosa MDC, Reischl E, Molloy AM, Luna JDD, Pérez-García M. Long-Chain Polyunsaturated Fatty Acids, Homocysteine at Birth and Fatty Acid Desaturase Gene Cluster Polymorphisms are Associated with Children's Processing Speed up to Age 9 Years. Nutrients 2020; 13:E131. [PMID: 33396458 PMCID: PMC7824114 DOI: 10.3390/nu13010131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/18/2020] [Accepted: 12/29/2020] [Indexed: 01/26/2023] Open
Abstract
Both pre- and early postnatal supplementation with docosahexaenoic acid (DHA), arachidonic acid (AA) and folate have been related to neural development, but their long-term effects on later neural function remain unclear. We evaluated the long-term effects of maternal prenatal supplementation with fish-oil (FO), 5-methyltetrahydrofolate (5-MTHF), placebo or FO + 5-MTHF, as well as the role of fatty acid desaturase (FADS) gene cluster polymorphisms, on their offspring's processing speed at later school age. This study was conducted in NUHEAL children at 7.5 (n = 143) and 9 years of age (n = 127). Processing speed tasks were assessed using Symbol Digit Modalities Test (SDMT), Children Color Trails Test (CCTT) and Stroop Color and Word Test (SCWT). Long-chain polyunsaturated fatty acids, folate and total homocysteine (tHcy) levels were determined at delivery from maternal and cord blood samples. FADS and methylenetetrahydrofolate reductase (MTHFR) 677 C > T genetic polymorphisms were analyzed. Mixed models (linear and logistic) were performed. There were significant differences in processing speed performance among children at different ages (p < 0.001). The type of prenatal supplementation had no effect on processing speed in children up to 9 years. Secondary exploratory analyses indicated that children born to mothers with higher AA/DHA ratio at delivery (p < 0.001) and heterozygotes for FADS1 rs174556 (p < 0.05) showed better performance in processing speed at 9 years. Negative associations between processing speed scores and maternal tHcy levels at delivery were found. Our findings suggest speed processing development in children up to 9 years could be related to maternal factors, including AA/DHA and tHcy levels, and their genetic background, mainly FADS polymorphism. These considerations support that maternal prenatal supplementation should be quantitatively adequate and individualized to obtain better brain development and mental performance in the offspring.
Collapse
Affiliation(s)
- Cristina Campoy
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 180016 Granada, Spain; (H.A.); (F.J.T.-E.); (C.M.-Z.); (J.A.G.-S.)
- Department of Paediatrics, School of Medicine, University of Granada, Avda, Investigación 11, 180016 Granada, Spain
- Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada’s Node, Institute of Health Carlos III, 28029 Madrid, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs-GRANADA), Health Sciences Technological Park, 18012 Granada, Spain
- Instituto de Neurociencias “Doctor Olóriz”, Health Sciences Technological Park, 18012 Granada, Spain
| | - Hatim Azaryah
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 180016 Granada, Spain; (H.A.); (F.J.T.-E.); (C.M.-Z.); (J.A.G.-S.)
- Department of Paediatrics, School of Medicine, University of Granada, Avda, Investigación 11, 180016 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs-GRANADA), Health Sciences Technological Park, 18012 Granada, Spain
| | - Francisco J. Torres-Espínola
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 180016 Granada, Spain; (H.A.); (F.J.T.-E.); (C.M.-Z.); (J.A.G.-S.)
- Department of Paediatrics, School of Medicine, University of Granada, Avda, Investigación 11, 180016 Granada, Spain
- Instituto de Neurociencias “Doctor Olóriz”, Health Sciences Technological Park, 18012 Granada, Spain
| | - Cristina Martínez-Zaldívar
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 180016 Granada, Spain; (H.A.); (F.J.T.-E.); (C.M.-Z.); (J.A.G.-S.)
- Department of Paediatrics, School of Medicine, University of Granada, Avda, Investigación 11, 180016 Granada, Spain
| | - José Antonio García-Santos
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 180016 Granada, Spain; (H.A.); (F.J.T.-E.); (C.M.-Z.); (J.A.G.-S.)
- Department of Paediatrics, School of Medicine, University of Granada, Avda, Investigación 11, 180016 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs-GRANADA), Health Sciences Technological Park, 18012 Granada, Spain
| | - Hans Demmelmair
- Ludwig-Maximilians-Universität München, Department of Paediatrics, Paediatrics, Dr. von Hauner Children’s Hospital, University of Munich Hospitals, 80337 Munich, Germany; (H.D.); (G.H.); (P.R.); (B.K.)
| | - Gudrun Haile
- Ludwig-Maximilians-Universität München, Department of Paediatrics, Paediatrics, Dr. von Hauner Children’s Hospital, University of Munich Hospitals, 80337 Munich, Germany; (H.D.); (G.H.); (P.R.); (B.K.)
| | - Peter Rzehak
- Ludwig-Maximilians-Universität München, Department of Paediatrics, Paediatrics, Dr. von Hauner Children’s Hospital, University of Munich Hospitals, 80337 Munich, Germany; (H.D.); (G.H.); (P.R.); (B.K.)
| | - Berthold Koletzko
- Ludwig-Maximilians-Universität München, Department of Paediatrics, Paediatrics, Dr. von Hauner Children’s Hospital, University of Munich Hospitals, 80337 Munich, Germany; (H.D.); (G.H.); (P.R.); (B.K.)
| | - Eszter Györei
- Department of Paediatrics, University of Pécs, 7623 Pécs, József Attila u. 7, 7623 Pécs, Hungary; (E.G.); (T.D.)
| | - Tamas Décsi
- Department of Paediatrics, University of Pécs, 7623 Pécs, József Attila u. 7, 7623 Pécs, Hungary; (E.G.); (T.D.)
| | - María del Carmen Ramírez-Tortosa
- Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology “José Mataix Verdú”, Biomedical Research Center, University of Granada, 18100 Granada, Spain;
| | - Eva Reischl
- Helmholtz Zentrum Munchen, Research Unit of Molecular Epidemiology, D-85764 Neuherberg, Germany;
| | - Anne M. Molloy
- School of Medicine, Trinity College, 152–160 Pearse Street, D02 Dublin 2, Ireland;
| | - Juan de Dios Luna
- Department of Biostatistics, School of Medicine, University of Granada, 18016 Granada, Spain;
| | - Miguel Pérez-García
- Mind, Brain and Behaviour International Research Centre (CIMCYC), University of Granada, 18011 Granada, Spain;
- Spanish Network of Biomedical Research Centre on Mental Health (CIBERSAM), Granada’s Node, Institute of Health Carlos III, 28029 Madrid, Spain
| |
Collapse
|
27
|
The Effects of an Infant Formula Enriched with Milk Fat Globule Membrane, Long-Chain Polyunsaturated Fatty Acids and Synbiotics on Child Behavior up to 2.5 Years Old: The COGNIS Study. Nutrients 2020; 12:nu12123825. [PMID: 33333767 PMCID: PMC7765166 DOI: 10.3390/nu12123825] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 01/10/2023] Open
Abstract
Although early life nutrition influences brain development and mental health, the long-term effects of supplemented infant formula on children´s behavior remain unclear. We analyzed the effects of a bioactive nutrients-enriched-infant formula on children’s behavior up to 2.5 years, compared to a standard infant formula or breastfeeding. Current analysis involved 70 children who were fed a standard infant formula (SF, n = 29) or a bioactive compounds enriched-infant formula (EF, n = 41), during their first 18 months of life, and 33 breastfed (BF) children (reference group) participating in the COGNIS study. Behavioral problems were evaluated using the Child Behavior Checklist at 18 months and 2.5 years. Different statistical analyses were performed using SPSS. EF children aged 2.5 years presented fewer pathological affective problems than SF children. Besides, SF children were classified more frequently as bordering on internalizing problems than BF children. Rates of externalizing problems were increased in SF infants compared to EF and BF infants. Higher maternal IQ was found to have beneficial effects on internalizing and total problem rate in their offspring at 18 months of life; finally, higher maternal educational level was related with fewer ADHD problems in children at 18 months, as well as internalizing, externalizing, total and anxiety problems in children aged 2.5 years. Our analysis suggests that enriched infant formula fed infants seem to show fewer behavioral problems up to 2.5 years compared to a standard infant formula-fed infants. In addition to type of early feeding, maternal IQ and educational level seem to play a key role on children behavioral development.
Collapse
|
28
|
Abstract
OBJECTIVE To analyse nutritional and packaging characteristics of toddler-specific foods and milks in the Australian retail food environment to identify how such products fit within the Australian Dietary Guidelines (ADG) and the NOVA classification. DESIGN Cross-sectional retail audit of toddler foods and milks. On-pack product attributes were recorded. Products were categorised as (1) food or milk; (2) snack food or meal and (3) snacks sub-categorised depending on main ingredients. Products were classified as a discretionary or core food as per the ADG and level of processing according to NOVA classification. SETTING Supermarkets and pharmacies in Australia. RESULTS A total of 154 foods and thirty-two milks were identified. Eighty percentage of foods were snacks, and 60 % of foods were classified as core foods, while 85 % were ultraprocessed (UP). Per 100 g, discretionary foods provided significantly more energy, protein, total and saturated fat, carbohydrate, total sugar and Na (P < 0·001) than core foods. Total sugars were significantly higher (P < 0·001) and Na significantly lower (P < 0·001) in minimally processed foods than in UP foods. All toddler milks (n 32) were found to have higher energy, carbohydrate and total sugar levels than full-fat cow's milk per 100 ml. Claims and messages were present on 99 % of foods and all milks. CONCLUSIONS The majority of toddler foods available in Australia are UP snack foods and do not align with the ADG. Toddler milks, despite being UP, do align with the ADG. A strengthened regulatory approach may address this issue.
Collapse
|
29
|
Azaryah H, Verdejo-Román J, Martin-Pérez C, García-Santos JA, Martínez-Zaldívar C, Torres-Espínola FJ, Campos D, Koletzko B, Pérez-García M, Catena A, Campoy C. Effects of Maternal Fish Oil and/or 5-Methyl-Tetrahydrofolate Supplementation during Pregnancy on Offspring Brain Resting-State at 10 Years Old: A Follow-Up Study from the NUHEAL Randomized Controlled Trial. Nutrients 2020; 12:E2701. [PMID: 32899673 PMCID: PMC7551257 DOI: 10.3390/nu12092701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 01/10/2023] Open
Abstract
Recent studies have shown that maternal supplementation with folate and long-chain polyunsaturated fatty acids (LC-PUFAs) during pregnancy may affect children's brain development. We aimed at examining the potential long-term effect of maternal supplementation with fish oil (FO) and/or 5-methyl-tetrahydrofolate (5-MTHF) on the brain functionality of offspring at the age of 9.5-10 years. The current study was conducted as a follow-up of the Spanish participants belonging to the Nutraceuticals for a Healthier Life (NUHEAL) project; 57 children were divided into groups according to mother's supplementation and assessed through functional magnetic resonance imaging (fMRI) scanning and neurodevelopment testing. Independent component analysis and double regression methods were implemented to investigate plausible associations. Children born to mothers supplemented with FO (FO and FO + 5-MTHF groups, n = 33) showed weaker functional connectivity in the default mode (DM) (angular gyrus), the sensorimotor (SM) (motor and somatosensory cortices) and the fronto-parietal (FP) (angular gyrus) networks compared to the No-FO group (placebo and 5-MTHF groups, n = 24) (PFWE < 0.05). Furthermore, no differences were found regarding the neuropsychological tests, except for a trend of better results in an object recall (memory) test. Considering the No-FO group, the aforementioned networks were associated negatively with attention and speed-processing functions. Mother's FO supplementation during pregnancy seems to be able to shape resting-state network functioning in their children at school age and appears to produce long-term effects on children´s cognitive processing.
Collapse
Affiliation(s)
- Hatim Azaryah
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain; (H.A.); (J.A.G.-S.); (C.M.-Z.); (F.J.T.-E.); (D.C.)
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
| | - Juan Verdejo-Román
- Mind, Brain and Behaviour International Research Centre (CIMCYC), University of Granada, 18011 Granada, Spain; (J.V.-R.); (C.M.-P.); (M.P.-G.); (A.C.)
| | - Cristina Martin-Pérez
- Mind, Brain and Behaviour International Research Centre (CIMCYC), University of Granada, 18011 Granada, Spain; (J.V.-R.); (C.M.-P.); (M.P.-G.); (A.C.)
| | - José Antonio García-Santos
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain; (H.A.); (J.A.G.-S.); (C.M.-Z.); (F.J.T.-E.); (D.C.)
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs-GRANADA), Health Sciences Technological Park, 18012 Granada, Spain
| | - Cristina Martínez-Zaldívar
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain; (H.A.); (J.A.G.-S.); (C.M.-Z.); (F.J.T.-E.); (D.C.)
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
| | - Francisco J. Torres-Espínola
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain; (H.A.); (J.A.G.-S.); (C.M.-Z.); (F.J.T.-E.); (D.C.)
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
| | - Daniel Campos
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain; (H.A.); (J.A.G.-S.); (C.M.-Z.); (F.J.T.-E.); (D.C.)
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
| | - Berthold Koletzko
- Ludwig-Maximiliams-Universität München, Dr. von Hauner Children’s Hospital, University of Munich Hospitals, 80337 Munich, Germany;
| | - Miguel Pérez-García
- Mind, Brain and Behaviour International Research Centre (CIMCYC), University of Granada, 18011 Granada, Spain; (J.V.-R.); (C.M.-P.); (M.P.-G.); (A.C.)
| | - Andrés Catena
- Mind, Brain and Behaviour International Research Centre (CIMCYC), University of Granada, 18011 Granada, Spain; (J.V.-R.); (C.M.-P.); (M.P.-G.); (A.C.)
| | - Cristina Campoy
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain; (H.A.); (J.A.G.-S.); (C.M.-Z.); (F.J.T.-E.); (D.C.)
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (Ibs-GRANADA), Health Sciences Technological Park, 18012 Granada, Spain
- Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada’s Node, Institute of Health Carlos III, 28029 Madrid, Spain
| |
Collapse
|
30
|
Tinkov AA, Mazaletskaya AL, Ajsuvakova OP, Bjørklund G, Huang PT, Chernova LN, Skalny AA, Skalny AV. ICP-MS Assessment of Hair Essential Trace Elements and Minerals in Russian Preschool and Primary School Children with Attention-Deficit/Hyperactivity Disorder (ADHD). Biol Trace Elem Res 2020; 196:400-409. [PMID: 31691190 DOI: 10.1007/s12011-019-01947-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/18/2019] [Indexed: 12/15/2022]
Abstract
The objective of the present study was to investigate the relationship between hair essential trace element and mineral content and ADHD in preschool (4-6 years old) and primary school children (6-10 years old) in relation to age and gender. Hair essential trace element and mineral content in 90 Russian children with ADHD and 90 age- and gender-matched neurotypical controls were assessed using inductively coupled plasma mass-spectrometry after microwave digestion. The obtained data demonstrate that hair Co, Cu, Mn, Si, and Zn contents in ADHD children was significantly reduced by 18%, 10%, 27%, 16%, and 19% as compared to the control values, respectively. The most significant decrease in children with ADHD was observed for hair Mg levels, being 29% lower than those in neurotypical children. After adjustment for age and gender, the observed difference in hair element content was more characteristic for preschool children and girls, respectively. Multiple linear regression analysis demonstrated that in a crude model (hair element levels as predictors), only hair Zn content was significantly inversely associated with ADHD (β = - 0.169; p = 0.025). Adjustment for anthropometric parameters (model 2) did not increase the predictive ability of the model, although it improved the association between hair Zn and ADHD in children (β = - 0.194; p = 0.014). Hypothetically, the observed alterations may at least partially contribute to neurobehavioral disturbances in children with ADHD. Moreover, the results of the present study raise the question about the potential benefits of Zn and Mg supplementation in children with ADHD. However, further detailed studies are required to investigate micronutrient deficiencies in ADHD.
Collapse
Affiliation(s)
- Alexey A Tinkov
- Yaroslavl State University, Yaroslavl, Russia, 150003.
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia, 119146.
- RUDN University, Moscow, Russia.
| | | | - Olga P Ajsuvakova
- Yaroslavl State University, Yaroslavl, Russia, 150003
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia, 119146
- RUDN University, Moscow, Russia
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| | | | | | - Andrey A Skalny
- Yaroslavl State University, Yaroslavl, Russia, 150003
- RUDN University, Moscow, Russia
| | - Anatoly V Skalny
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia, 119146
- RUDN University, Moscow, Russia
- Federal Scientific Center of Biological Systems and Agrotechnologies of the Russian Academy of Sciences, Orenburg, Russia, 460000
| |
Collapse
|
31
|
Jaddoe VWV, Felix JF, Andersen AMN, Charles MA, Chatzi L, Corpeleijn E, Donner N, Elhakeem A, Eriksson JG, Foong R, Grote V, Haakma S, Hanson M, Harris JR, Heude B, Huang RC, Inskip H, Järvelin MR, Koletzko B, Lawlor DA, Lindeboom M, McEachan RRC, Mikkola TM, Nader JLT, de Moira AP, Pizzi C, Richiardi L, Sebert S, Schwalber A, Sunyer J, Swertz MA, Vafeiadi M, Vrijheid M, Wright J, Duijts L. The LifeCycle Project-EU Child Cohort Network: a federated analysis infrastructure and harmonized data of more than 250,000 children and parents. Eur J Epidemiol 2020; 35:709-724. [PMID: 32705500 PMCID: PMC7387322 DOI: 10.1007/s10654-020-00662-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 07/04/2020] [Indexed: 12/15/2022]
Abstract
Early life is an important window of opportunity to improve health across the full lifecycle. An accumulating body of evidence suggests that exposure to adverse stressors during early life leads to developmental adaptations, which subsequently affect disease risk in later life. Also, geographical, socio-economic, and ethnic differences are related to health inequalities from early life onwards. To address these important public health challenges, many European pregnancy and childhood cohorts have been established over the last 30 years. The enormous wealth of data of these cohorts has led to important new biological insights and important impact for health from early life onwards. The impact of these cohorts and their data could be further increased by combining data from different cohorts. Combining data will lead to the possibility of identifying smaller effect estimates, and the opportunity to better identify risk groups and risk factors leading to disease across the lifecycle across countries. Also, it enables research on better causal understanding and modelling of life course health trajectories. The EU Child Cohort Network, established by the Horizon2020-funded LifeCycle Project, brings together nineteen pregnancy and childhood cohorts, together including more than 250,000 children and their parents. A large set of variables has been harmonised and standardized across these cohorts. The harmonized data are kept within each institution and can be accessed by external researchers through a shared federated data analysis platform using the R-based platform DataSHIELD, which takes relevant national and international data regulations into account. The EU Child Cohort Network has an open character. All protocols for data harmonization and setting up the data analysis platform are available online. The EU Child Cohort Network creates great opportunities for researchers to use data from different cohorts, during and beyond the LifeCycle Project duration. It also provides a novel model for collaborative research in large research infrastructures with individual-level data. The LifeCycle Project will translate results from research using the EU Child Cohort Network into recommendations for targeted prevention strategies to improve health trajectories for current and future generations by optimizing their earliest phases of life.
Collapse
Affiliation(s)
- Vincent W V Jaddoe
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, The Generation R Study Group, (Na 29-18), PO Box 2040, 3000 CA, Rotterdam, The Netherlands. .,Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Janine F Felix
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, The Generation R Study Group, (Na 29-18), PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Anne-Marie Nybo Andersen
- Section of Epidemiology, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Marie-Aline Charles
- Université de Paris, Centre for Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Paris, France.,ELFE Joint Unit, French Institute for Demographic Studies (Ined), French Institute for Medical Research and Health (INSERM), French Blood Agency, Aubervilliers, France
| | - Leda Chatzi
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Eva Corpeleijn
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Nina Donner
- Concentris Research Management GmbH, Fürstenfeldbruck, Germany
| | - Ahmed Elhakeem
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Folkhälsan Research Center, Helsinki, Finland.,Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore.,Singapore Institute for Clinical Sciences (SICS), Agency for Science and Technology (A*STAR), Singapore, Singapore
| | - Rachel Foong
- Telethon Kids Institute, Perth, WA, Australia.,School of Physiotherapy and Exercise Science, Curtin University, Perth, WA, Australia
| | - Veit Grote
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU, Munich, Germany
| | - Sido Haakma
- University of Groningen, University Medical Center Groningen, Genomics Coordination Center, Groningen, The Netherlands
| | - Mark Hanson
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jennifer R Harris
- Centre for Fertility and Health, The Norwegian Institute of Public Health, Oslo, Norway.,Division of Health Data and Digitalization, Norwegian Institute of Public Health, Oslo, Norway
| | - Barbara Heude
- Université de Paris, Centre for Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Paris, France
| | | | - Hazel Inskip
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK.,MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Marjo-Riitta Järvelin
- Center for Life-Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland.,Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK.,Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK.,Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
| | - Berthold Koletzko
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU, Munich, Germany
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.,NIHR Bristol Biomedical Research Centre, Bristol, UK
| | - Maarten Lindeboom
- Department of Economics, VU University Amsterdam, Amsterdam, The Netherlands
| | - Rosemary R C McEachan
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | | | - Johanna L T Nader
- Department of Genetics and Bioinformatics, Division of Health Data and Digitalisation, Norwegian Institute of Public Health, Oslo, Norway
| | - Angela Pinot de Moira
- Section of Epidemiology, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Costanza Pizzi
- Cancer Epidemiology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Lorenzo Richiardi
- Cancer Epidemiology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Sylvain Sebert
- Center for Life-Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Ameli Schwalber
- Concentris Research Management GmbH, Fürstenfeldbruck, Germany
| | - Jordi Sunyer
- ISGlobal, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Morris A Swertz
- University of Groningen, University Medical Center Groningen, Genomics Coordination Center, Groningen, The Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marina Vafeiadi
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Martine Vrijheid
- ISGlobal, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - John Wright
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Liesbeth Duijts
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, The Generation R Study Group, (Na 29-18), PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | |
Collapse
|
32
|
Skalny AV, Mazaletskaya AL, Ajsuvakova OP, Bjørklund G, Skalnaya MG, Notova SV, Chernova LN, Skalny AA, Burtseva TI, Tinkov AA. Hair trace element concentrations in autism spectrum disorder (ASD) and attention deficit/hyperactivity disorder (ADHD). J Trace Elem Med Biol 2020; 61:126539. [PMID: 32438295 DOI: 10.1016/j.jtemb.2020.126539] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 04/09/2020] [Accepted: 04/21/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND The existing data demonstrate that alteration of trace element and mineral status in children with neurodevelopmental disorders including ASD and ADHD. However, comparative analysis of the specific patterns of trace element and mineral metabolism in children with ASD and ADHD was not performed. Therefore, the primary objective of the present study was to assess hair trace element and mineral levels in boys with ADHD, ASD, as well as ADHD with ASD. METHODS Boys with ADHD (n = 52), ASD (n = 53), both ADHD and ASD (n = 52), as well as neurotypical controls (n = 52) were examined. Hair analysis was performed using inductively-coupled plasma mass-spectrometry. RESULTS The obtained data demonstrate that hair Co, Mg, Mn, and V levels were significantly reduced in children with ADHD and ASD, and especially in boys with ADHD + ASD. Hair Zn was found to be reduced by 20% (p = 0.009) only in children with ADHD + ASD as compared to healthy controls. Factor analysis demonstrated that ASD was associated with significant alteration of hair Co, Fe, Mg, Mn, and V levels, whereas impaired hair Mg, Mn, and Zn content was also significantly associated with ADHD. In regression models hair Zn and Mg were negatively associated with severity of neurodevelopmental disorders. The revealed similarity of trace element and mineral disturbances in ASD and ADHD may be indicative of certain similar pathogenetic features. CONCLUSION The obtained data support the hypothesis that trace elements and minerals, namely Mg, Mn, and Zn, may play a significant role in development of both ADHD and ASD. Improvement of Mg, Mn, and Zn status in children with ASD and ADHD may be considered as a nutritional strategy for improvement of neurodevelopmental disturbances, although clinical trials and experimental studies are highly required to support this hypothesis.
Collapse
Affiliation(s)
- Anatoly V Skalny
- Yaroslavl State University, Sovetskaya St., 14, 150003, Yaroslavl, Russia; IM Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya St., 9, 119146, Moscow, Russia; RUDN University, Miklukho-Maklaya St., 6, 117198, Moscow, Russia
| | | | - Olga P Ajsuvakova
- Yaroslavl State University, Sovetskaya St., 14, 150003, Yaroslavl, Russia; IM Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya St., 9, 119146, Moscow, Russia; RUDN University, Miklukho-Maklaya St., 6, 117198, Moscow, Russia
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, 8602, Mo i Rana, Norway
| | - Margarita G Skalnaya
- IM Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya St., 9, 119146, Moscow, Russia; RUDN University, Miklukho-Maklaya St., 6, 117198, Moscow, Russia
| | - Svetlana V Notova
- IM Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya St., 9, 119146, Moscow, Russia; Federal Research Centre of Biological Systems and Agro-technologies of the Russian Academy of Sciences, 9 Yanvarya St., 29, 460000, Orenburg, Russia
| | | | - Andrey A Skalny
- Yaroslavl State University, Sovetskaya St., 14, 150003, Yaroslavl, Russia; RUDN University, Miklukho-Maklaya St., 6, 117198, Moscow, Russia
| | - Tatiana I Burtseva
- IM Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya St., 9, 119146, Moscow, Russia; Orenburg State University, Pobedy Ave., 46, 460000, Orenburg, Russia
| | - Alexey A Tinkov
- Yaroslavl State University, Sovetskaya St., 14, 150003, Yaroslavl, Russia; IM Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya St., 9, 119146, Moscow, Russia; RUDN University, Miklukho-Maklaya St., 6, 117198, Moscow, Russia.
| |
Collapse
|
33
|
Morton SU, Vyas R, Gagoski B, Vu C, Litt J, Larsen RJ, Kuchan MJ, Lasekan JB, Sutton BP, Grant PE, Ou Y. Maternal Dietary Intake of Omega-3 Fatty Acids Correlates Positively with Regional Brain Volumes in 1-Month-Old Term Infants. Cereb Cortex 2020; 30:2057-2069. [PMID: 31711132 PMCID: PMC8355466 DOI: 10.1093/cercor/bhz222] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/31/2019] [Accepted: 08/22/2019] [Indexed: 01/05/2023] Open
Abstract
Maternal nutrition is an important factor for infant neurodevelopment. However, prior magnetic resonance imaging (MRI) studies on maternal nutrients and infant brain have focused mostly on preterm infants or on few specific nutrients and few specific brain regions. We present a first study in term-born infants, comprehensively correlating 73 maternal nutrients with infant brain morphometry at the regional (61 regions) and voxel (over 300 000 voxel) levels. Both maternal nutrition intake diaries and infant MRI were collected at 1 month of life (0.9 ± 0.5 months) for 92 term-born infants (among them, 54 infants were purely breastfed and 19 were breastfed most of the time). Intake of nutrients was assessed via standardized food frequency questionnaire. No nutrient was significantly correlated with any of the volumes of the 61 autosegmented brain regions. However, increased volumes within subregions of the frontal cortex and corpus callosum at the voxel level were positively correlated with maternal intake of omega-3 fatty acids, retinol (vitamin A) and vitamin B12, both with and without correction for postmenstrual age and sex (P < 0.05, q < 0.05 after false discovery rate correction). Omega-3 fatty acids remained significantly correlated with infant brain volumes after subsetting to the 54 infants who were exclusively breastfed, but retinol and vitamin B12 did not. This provides an impetus for future larger studies to better characterize the effect size of dietary variation and correlation with neurodevelopmental outcomes, which can lead to improved nutritional guidance during pregnancy and lactation.
Collapse
Affiliation(s)
- Sarah U Morton
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Rutvi Vyas
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Borjan Gagoski
- Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Radiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Catherine Vu
- Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Jonathan Litt
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Ryan J Larsen
- Beckman Institute, University of Illinois at Urbana—Champaign, Urbana, IL 61801, USA
| | | | | | - Brad P Sutton
- Beckman Institute, University of Illinois at Urbana—Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana—Champaign, Urbana, IL 61801, USA
| | - P Ellen Grant
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Radiology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Yangming Ou
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Radiology, Boston Children’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
34
|
Nieto-Ruiz A, García-Santos JA, Bermúdez MG, Herrmann F, Diéguez E, Sepúlveda-Valbuena N, García S, Miranda MT, De-Castellar R, Rodríguez-Palmero M, Catena A, Campoy C. Cortical Visual Evoked Potentials and Growth in Infants Fed with Bioactive Compounds-Enriched Infant Formula: Results from COGNIS Randomized Clinical Trial. Nutrients 2019; 11:nu11102456. [PMID: 31615134 PMCID: PMC6835488 DOI: 10.3390/nu11102456] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/09/2019] [Accepted: 10/12/2019] [Indexed: 12/18/2022] Open
Abstract
Postnatal nutrition is essential for growth and neurodevelopment. We analyzed the influence of a new enriched-infant formula with bioactive compounds on growth, neurodevelopment, and visual function (VF) in healthy infants during their first 18 months of life. A total of 170 infants were randomized in the COGNIS randomized clinical trial (RCT) to receive a standard infant formula (SF = 85) or a new experimental infant formula supplemented with functional nutrients (EF = 85). As a control, 50 breastfed infants (BF) were enrolled. Growth patterns were evaluated up to 18 months of life; neurodevelopment was assessed by general movements at 2, 3, and 4 months; VF was measured by cortical visual evoked potentials at 3 and 12 months. No differences in growth and neurodevelopment were found between groups. Regarding VF, SF and EF infants presented prolonged latencies and lower amplitudes in the P100 wave than BF infants. In the EF group, a higher percentage of infants presented response at 7½′ of arc at 12 months compared to 3 months of age; a similar proportion of BF and EF infants presented responses at 7½′ of arc at 12 months of age. Early nutritional intervention with bioactive compounds could narrow the gap in growth and neurodevelopment between breastfed and formula-fed infants.
Collapse
Affiliation(s)
- Ana Nieto-Ruiz
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain.
- Mind, Brain and Behaviour Research Centre-CIMCYC, University of Granada, 18011 Granada, Spain.
| | - José Antonio García-Santos
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain.
| | - Mercedes G Bermúdez
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain.
| | - Florian Herrmann
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain.
| | - Estefanía Diéguez
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain.
| | - Natalia Sepúlveda-Valbuena
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
- Nutrition and Biochemistry Department, Faculty of Sciences, Pontificia Universidad Javeriana, Bogotá 110231, Colombia.
| | - Salomé García
- Clinical Service of Neurophysiology, Clinical University Hospital San Cecilio, 18016 Granada, Spain.
| | - M Teresa Miranda
- Department of Biostatistics, School of Medicine, University of Granada,18016 Granada, Spain.
| | | | | | - Andrés Catena
- Mind, Brain and Behaviour Research Centre-CIMCYC, University of Granada, 18011 Granada, Spain.
| | - Cristina Campoy
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, 18016 Granada, Spain.
- Department of Paediatrics, School of Medicine, University of Granada, Avda. Investigación 11, 18016 Granada, Spain.
- Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada´s Node, Institute of Health Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
35
|
Cerdó T, Diéguez E, Campoy C. Early nutrition and gut microbiome: interrelationship between bacterial metabolism, immune system, brain structure, and neurodevelopment. Am J Physiol Endocrinol Metab 2019; 317:E617-E630. [PMID: 31361544 DOI: 10.1152/ajpendo.00188.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Disturbances of diet during pregnancy and early postnatal life may impact colonization of gut microbiota during early life, which could influence infant health, leading to potential long-lasting consequences later in life. This is a nonsystematic review that explores the recent scientific literature to provide a general perspective of this broad topic. Several studies have shown that gut microbiota composition is related to changes in metabolism, energy balance, and immune system disturbances through interaction between microbiota metabolites and host receptors by the gut-brain axis. Moreover, recent clinical studies suggest that an intestinal dysbiosis in gut microbiota may result in cognitive disorders and behavioral problems. Furthermore, recent research in the field of brain imaging focused on the study of the relationship between gut microbial ecology and large-scale brain networks, which will help to decipher the influence of the microbiome on brain function and potentially will serve to identify multiple mediators of the gut-brain axis. Thus, knowledge about optimal nutrition by modulating gut microbiota-brain axis activity will allow a better understanding of the molecular mechanisms involved in the crosstalk between gut microbiota and the developing brain during critical windows. In addition, this knowledge will open new avenues for developing novel microbiota-modulating based diet interventions during pregnancy and early life to prevent metabolic disorders, as well as neurodevelopmental deficits and brain functional disorders.
Collapse
Affiliation(s)
- Tomás Cerdó
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
- BioHealth Research Institute (Ibs-Granada), Health Sciences Technological Park, Granada, Spain
- Neurosciences Institute, Biomedical Research Centre, University of Granada, Granada, Spain
| | - Estefanía Diéguez
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
| | - Cristina Campoy
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain
- EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
- BioHealth Research Institute (Ibs-Granada), Health Sciences Technological Park, Granada, Spain
- Neurosciences Institute, Biomedical Research Centre, University of Granada, Granada, Spain
- Spanish Network of Biomedical Research in Epidemiology and Public Health (CIBERESP), Granada's node, Carlos III Health Institute of Health Carlos III, Madrid, Spain
- Brain, Behavior and Health Excellence Research Unit (SC2). University of Granada, Granada, Spain
| |
Collapse
|
36
|
Caparros-Gonzalez RA, Giménez-Asensio MJ, González-Alzaga B, Aguilar-Garduño C, Lorca-Marín JA, Alguacil J, Gómez-Becerra I, Gómez-Ariza JL, García-Barrera T, Hernandez AF, López-Flores I, Rohlman DS, Romero-Molina D, Ruiz-Pérez I, Lacasaña M. Childhood chromium exposure and neuropsychological development in children living in two polluted areas in southern Spain. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 252:1550-1560. [PMID: 31277024 DOI: 10.1016/j.envpol.2019.06.084] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 06/19/2019] [Accepted: 06/21/2019] [Indexed: 05/28/2023]
Abstract
This study aimed to assess the association between exposure to chromium and neuropsychological development among children. A cross-sectional study was conducted with 393 children aged 6-11 years old randomly selected from State-funded schools in two provinces in Southern Spain (Almeria and Huelva), in 2010 and 2012. Chromium levels in urine and hair samples were analyzed by inductively coupled plasma mass spectrometry with an octopole reaction system. Neuropsychological development was evaluated using the Wechsler Intelligence Scale for Children-Fourth Edition (WISC-IV) and three computerized tests from the Behavioural Assessment and Research System (BARS): Reaction Time Test (RTT), Continuous Performance Test (CPT) and Selective Attention Test (SAT). Multivariable linear regression models adjusted for potential confounders, including heavy metals, were applied to examine the association between chromium levels and neuropsychological outcomes. A 10-fold increase in urine chromium levels was associated with a decrease of 5.99 points on the WISC-IV Full-Scale IQ (95% CI: 11.98 to -0.02). Likewise, a 10-fold increase in urine chromium levels in boys was associated with a decrease of 0.03 points in the percentage of omissions (95% CI: 0.0 to 0.05) in the SAT, with an increase of 68.35 points in latency (95% CI: 6.60 to 130.12) in the RTT, and with an increase in the number of trials with latencies > 1000 ms (β = 37.92; 95% CI: 2.73 to 73.12) in the RTT. An inverse significant association was detected between chromium levels in hair and latency in the SAT in boys (β = -50.53; 95% CI: 86.86 to -14.22) and girls (β = -55.95; 95% CI: 78.93 to -32.97). Excluding trials with latencies >1000 ms in the RTT increased latency scores by 29.36 points in boys (95% CI: 0.17 to 58.57), and 39.91 points in girls (95% CI: 21.25 to 58.59). This study is the first to show the detrimental effects of postnatal chromium exposure on neuropsychological development in school-aged children.
Collapse
Affiliation(s)
- Rafael A Caparros-Gonzalez
- Mind, Brain and Behaviour Research Center (CIMCYC), Faculty of Psychology, University of Granada, Granada, Spain; Department of Nursing, Faculty of Health Sciences, University of Jaen, Jaen, Spain
| | | | - Beatriz González-Alzaga
- Andalusian School of Public Health (EASP), Granada, Spain; Instituto de Investigación Biosanitaria (ibs. GRANADA), Granada, Spain
| | | | - J Andrés Lorca-Marín
- Department of Clinical, Experimental and Social Psychology, University of Huelva, Huelva, Spain; Natural Resources, Health, and Environment Research Centre (RENSMA), University of Huelva, Huelva, Spain
| | - Juan Alguacil
- Natural Resources, Health, and Environment Research Centre (RENSMA), University of Huelva, Huelva, Spain; CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | | | - José L Gómez-Ariza
- Professor José Carlos Vílchez Martín, Department of Chemistry, University of Huelva, Huelva, Spain; Natural Resources, Health, and Environment Research Centre (RENSMA), University of Huelva, Huelva, Spain
| | - Tamara García-Barrera
- Professor José Carlos Vílchez Martín, Department of Chemistry, University of Huelva, Huelva, Spain; Natural Resources, Health, and Environment Research Centre (RENSMA), University of Huelva, Huelva, Spain
| | - Antonio F Hernandez
- Department of Legal Medicine, Toxicology and Physical Anthropology, University of Granada, Granada, Spain
| | - Inmaculada López-Flores
- Department of Genetics, Faculty of Sciences, University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria (ibs. GRANADA), Granada, Spain
| | - Diane S Rohlman
- Department of Occupational and Environmental Health, College of Public Health, University of Iowa, USA
| | - Desiree Romero-Molina
- Department of Statistics and Operational Research, Faculty of Sciences, University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria (ibs. GRANADA), Granada, Spain
| | - Isabel Ruiz-Pérez
- Andalusian School of Public Health (EASP), Granada, Spain; Instituto de Investigación Biosanitaria (ibs. GRANADA), Granada, Spain; CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Marina Lacasaña
- Andalusian School of Public Health (EASP), Granada, Spain; Instituto de Investigación Biosanitaria (ibs. GRANADA), Granada, Spain; CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain.
| |
Collapse
|
37
|
López-Vicente M, Ribas Fitó N, Vilor-Tejedor N, Garcia-Esteban R, Fernández-Barrés S, Dadvand P, Murcia M, Rebagliato M, Ibarluzea J, Lertxundi A, Fernández-Somoano A, Tardón A, López-Sabater MC, Romaguera D, Vrijheid M, Sunyer J, Julvez J. Prenatal Omega-6:Omega-3 Ratio and Attention Deficit and Hyperactivity Disorder Symptoms. J Pediatr 2019; 209:204-211.e4. [PMID: 30929929 DOI: 10.1016/j.jpeds.2019.02.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/06/2019] [Accepted: 02/12/2019] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To evaluate whether higher omega-6:omega-3 (n-6:n-3) long-chain polyunsaturated fatty acid ratio in cord plasma is associated with more symptoms of attention deficit and hyperactivity disorder (ADHD) at 4 and 7 years of age. STUDY DESIGN This study was based on a population-based birth cohort in Spain. N-6 arachidonic acid and n-3 eicosapentaenoic and docosahexaenoic acid concentrations were measured in cord plasma. At 4 years old, ADHD symptoms were reported by teachers through the ADHD Diagnostic and Statistical Manual of Mental Disorders, 4th ed checklist (n = 580). At 7 years old, ADHD symptoms were reported by parents through the Conners' Rating Scale-Revised (short form; n = 642). The ADHD variable was treated as continuous (score) and as dichotomous (symptom diagnostic criteria). Child and family general characteristics were prospectively collected through questionnaires. We applied pooled zero-inflated negative binomial and logistic regressions adjusted for covariates. RESULTS A higher omega-6:omega-3 long-chain polyunsaturated fatty acid ratio in cord plasma was associated with a higher ADHD index (incidence rate ratio, 1.13; 95% CI, 1.03, 1.23) at 7 years old. The association was not observed at 4 years old (incidence rate ratio, 1.04; 95% CI, 0.92-1.18). No associations were found using ADHD symptom diagnostic criteria. CONCLUSIONS High prenatal omega-6:omega-3 long-chain polyunsaturated fatty acid ratio preceded the appearance of subclinical ADHD symptoms during mid-childhood. Our findings suggest that maternal diet during pregnancy may modulate the risk to develop long-term ADHD symptoms in the offspring.
Collapse
Affiliation(s)
- Mónica López-Vicente
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain; Hospital del Mar Medical Research Institute, Barcelona, Spain.
| | | | - Natalia Vilor-Tejedor
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Raquel Garcia-Esteban
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Sílvia Fernández-Barrés
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Payam Dadvand
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Mario Murcia
- Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain; Epidemiology and Environmental Health Joint Research Unit, FISABIO-Jaume I University-University of València, València, Spain
| | - Marisa Rebagliato
- Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain; Epidemiology and Environmental Health Joint Research Unit, FISABIO-Jaume I University-University of València, València, Spain; Medicine Department, Jaume I University, Castelló de la Plana, Spain
| | - Jesús Ibarluzea
- Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain; University of Basque Country, San Sebastián, Spain; Health Research Institute, BIODONOSTIA, San Sebastián, Spain; Sub-Directorate of Public Health of Gipuzkoa, Department of Health, Goverment of Basque Country, San Sebastián, Spain
| | - Aitana Lertxundi
- Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain; University of Basque Country, San Sebastián, Spain; Health Research Institute, BIODONOSTIA, San Sebastián, Spain
| | - Ana Fernández-Somoano
- Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain; IUOPA-Preventive Medicine and Public Health Area, University of Oviedo, Asturias, Spain
| | - Adonina Tardón
- Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain; IUOPA-Preventive Medicine and Public Health Area, University of Oviedo, Asturias, Spain
| | - M Carmen López-Sabater
- Department of Nutrition, Food Sciences and Gastronomy, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain; Biomedical Research Networking Centres Obesity and Nutrition (CIBEROBN), Madrid, Spain
| | - Dora Romaguera
- Biomedical Research Networking Centres Obesity and Nutrition (CIBEROBN), Madrid, Spain; Balearic Islands Health Research Institute, Son Espases University Hospital, Palma de Mallorca, Spain
| | - Martine Vrijheid
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Jordi Sunyer
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain; Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Jordi Julvez
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Biomedical Research Networking Centres Epidemiology and Public Health (CIBERESP), Madrid, Spain; Hospital del Mar Medical Research Institute, Barcelona, Spain
| |
Collapse
|
38
|
Maternal nut intake in pregnancy and child neuropsychological development up to 8 years old: a population-based cohort study in Spain. Eur J Epidemiol 2019; 34:661-673. [DOI: 10.1007/s10654-019-00521-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/17/2019] [Indexed: 02/03/2023]
|
39
|
Joosten KFM, Eveleens RD, Verbruggen SCAT. Nutritional support in the recovery phase of critically ill children. Curr Opin Clin Nutr Metab Care 2019; 22:152-158. [PMID: 30585805 DOI: 10.1097/mco.0000000000000549] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW The metabolic stress response of a critically ill child evolves over time and thus it seems reasonable that nutritional requirements change during their course of illness as well. This review proposes strategies and considerations for nutritional support during the recovery phase to gain optimal (catch-up) growth with preservation of lean body mass. RECENT FINDINGS Critical illness impairs nutritional status, muscle mass and function, and neurocognition, but early and high intakes of artificial nutrition during the acute phase cannot resolve this. Although (parenteral) nutrient restriction during the acute phase appears to be beneficial, persistent nutrient restriction, when the metabolic stress response resolves, has short-term and long-term detrimental consequences. Requirements increase markedly during the recovery phase to enable recovery and catch-up growth. Such large amounts of intake demand for alternate approach, especially when intestinal problems constitute a barrier for full enteral feeding. As part of the nutritional recovery, mobilization and exercise are essential to achieve catch-up growth with an optimal body composition. SUMMARY During the recovery phase of paediatric critical illness (catch-up) growth and muscle recovery require nutritional intakes at least two times the resting energy expenditure.
Collapse
Affiliation(s)
- Koen F M Joosten
- Paediatric Intensive Care, Department of Paediatrics and Paediatric Surgery, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands
| | | | | |
Collapse
|
40
|
Moderately elevated preconception fasting plasma total homocysteine is a risk factor for psychological problems in childhood. Public Health Nutr 2019; 22:1615-1623. [PMID: 30636652 DOI: 10.1017/s1368980018003610] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE We investigated the effect of maternal preconception fasting plasma total homocysteine (tHcy) on psychological problems in children aged 6 years from normal pregnancies. DESIGN A longitudinal study was carried out from preconception, throughout each trimester of pregnancy, until 6 years of age in the offspring. Fasting blood samples at 2-10 weeks preconception and non-fasting samples at birth were collected. Parents completed the Child Behaviour Checklist (CBCL) and teachers the Inattention-Overactivity with Aggression (IOWA) scale for the 6-year-old children. SETTING Elevated tHcy during pregnancy has been associated with several adverse outcomes and with neurodevelopmental impairment in the offspring.ParticipantsThe initial sample consisted of 139 healthy non-pregnant women who were planning on becoming pregnant. Eighty-one mother-child dyads were followed from preconception until 6 years of age. RESULTS After adjusting for covariables, multiple linear regression models showed that higher preconception tHcy was associated with higher scores in internalizing dimension (β=0·289; P=0.028), specifically in withdrawn behaviour (β=0·349; P=0·009), anxiety/depression (β=0·303; P=0·019) and social problems (β=0·372; P=0·009). Aggressive behaviour in the school setting was higher in children whose mothers had higher preconception tHcy (β=0·351; P=0·014). CONCLUSIONS Moderately elevated preconception tHcy may increase the risk of psychological problems in offspring during childhood. These findings add to the evidence that maternal nutritional status, even before being pregnant, can affect later offspring health and may be important to consider when developing future public health policy.
Collapse
|
41
|
Perinatal nutrition impacts on the functional development of the visual tract in infants. Pediatr Res 2019; 85:72-78. [PMID: 30237571 DOI: 10.1038/s41390-018-0161-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 07/18/2018] [Accepted: 07/23/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND OBJECTIVE We investigated the associations of maternal diet and serum fatty acids during pregnancy and in early infancy on infantile neurodevelopment. METHODS Pattern-reversal visual evoked potentials (pVEP) as depictors of central nervous system maturation were recorded from 56 children when they were 2 years old. Maternal nutrient intakes were calculated from food diaries and fish consumption from questionnaires collected during pregnancy. Serum phospholipid fatty acids were determined by gas chromatography in late pregnancy and from infants at 1 month of age. RESULTS The children of the women who consumed fish three or more times per week during the last trimester of pregnancy had a higher pVEP component P100 amplitude for 60' (mean 23.4, SD 8.1) and 30' (mean 20.4, SD 6.7) of arcminute check sizes compared to those who consumed fish 0-2 times per week (mean 15.0, SD 4.8, p = 0.023, adjusted for birth weight and gender p = 0.058 and mean 13.4, SD 2.0, respectively, p = 0.028, adjusted p = 0.072). Maternal and child serum phospholipid fatty acids correlated with child pVEP measurements. CONCLUSION The results of this small-scale study suggest that fish consumption during pregnancy and perinatal serum fatty acid status may associate with neurodevelopment within visual system during infancy.
Collapse
|
42
|
Oxidative Stress, Maternal Diabetes, and Autism Spectrum Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3717215. [PMID: 30524654 PMCID: PMC6247386 DOI: 10.1155/2018/3717215] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/17/2018] [Indexed: 12/25/2022]
Abstract
Autism spectrum disorders (ASD) are a group of early-onset neurodevelopmental conditions characterized by alterations in brain connectivity with cascading effects on neuropsychological functions. To date, in the framework of an increasing interest about environmental conditions which could interact with genetic factors in ASD pathogenesis, many authors have stressed that changes in the intrauterine environment at different stages of pregnancy, such as those linked to maternal metabolic pathologies, may lead to long-term conditions in the newborn. In particular, a growing number of epidemiological studies have highlighted the role of obesity and maternal diabetes as a risk factor for developing both somatic and psychiatric disorders in humans, including ASD. While literature still fails in identifying specific etiopathological mechanisms, a growing body of evidence is available about the presence of a relationship between maternal immune dysregulation, inflammation, oxidative stress, and the development of ASD in the offspring. In this framework, results from high-fat diet animal models about the role played by oxidative stress in shaping offspring neurodevelopment may help in clarifying the pathways through which maternal metabolic conditions are linked with ASD. The aim of this review is to provide an overview of literature about the effects of early life insults linked to oxidative stress which may be involved in ASD etiopathogenesis and how this relationship can be explained in biological terms.
Collapse
|
43
|
Oliveros E, Vázquez E, Barranco A, Ramírez M, Gruart A, Delgado-García JM, Buck R, Rueda R, Martín MJ. Sialic Acid and Sialylated Oligosaccharide Supplementation during Lactation Improves Learning and Memory in Rats. Nutrients 2018; 10:E1519. [PMID: 30332832 PMCID: PMC6212975 DOI: 10.3390/nu10101519] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/08/2018] [Accepted: 10/11/2018] [Indexed: 12/21/2022] Open
Abstract
Sialic acids (Sia) are postulated to improve cognitive abilities. This study evaluated Sia effects on rat behavior when administered in a free form as N-acetylneuraminic acid (Neu5Ac) or conjugated as 6'-sialyllactose (6'-SL). Rat milk contains Sia, which peaks at Postnatal Day 9 and drops to a minimum by Day 15. To bypass this Sia peak, a cohort of foster mothers was used to raise the experimental pups. A group of pups received a daily oral supplementation of Neu5Ac to mimic the amount naturally present in rat milk, and another group received the same molar amount of Sia as 6'-SL. The control group received water. After weaning, rats were submitted to behavioral evaluation. One year later, behavior was re-evaluated, and in vivo long-term potentiation (LTP) was performed. Brain samples were collected and analyzed at both ages. Adult rats who received Sia performed significantly better in the behavioral assessment and showed an enhanced LTP compared to controls. Within Sia groups, 6'-SL rats showed better scores in some cognitive outcomes compared to Neu5Ac rats. At weaning, an effect on polysialylated-neural cell adhesion molecule (PSA-NCAM) levels in the frontal cortex was only observed in 6'-SL fed rats. Providing Sia during lactation, especially as 6'-SL, improves memory and LTP in adult rats.
Collapse
Affiliation(s)
- Elena Oliveros
- R&D Abbott Nutrition, 18004 Granada, Spain.
- Doctoral programme in Biomedicine, School of Health Sciences, University of Granada, 18071 Granada, Spain.
| | | | | | | | - Agnes Gruart
- Division of Neurosciences, Pablo de Olavide University, 41013 Seville, Spain.
| | | | | | | | | |
Collapse
|
44
|
House JS, Mendez M, Maguire RL, Gonzalez-Nahm S, Huang Z, Daniels J, Murphy SK, Fuemmeler BF, Wright FA, Hoyo C. Periconceptional Maternal Mediterranean Diet Is Associated With Favorable Offspring Behaviors and Altered CpG Methylation of Imprinted Genes. Front Cell Dev Biol 2018; 6:107. [PMID: 30246009 PMCID: PMC6137242 DOI: 10.3389/fcell.2018.00107] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 08/20/2018] [Indexed: 12/28/2022] Open
Abstract
Background: Maternal diet during pregnancy has been shown to influence the child neuro-developmental outcomes. Studies examining effects of dietary patterns on offspring behavior are sparse. Objective: Determine if maternal adherence to a Mediterranean diet is associated with child behavioral outcomes assessed early in life, and to evaluate the role of differentially methylated regions (DMRs) regulating genomically imprinted genes in these associations. Methods: Among 325 mother/infant pairs, we used regression models to evaluate the association between tertiles of maternal periconceptional Mediterranean diet adherence (MDA) scores derived from a Food Frequency Questionnaire, and social and emotional scores derived from the Infant Toddler Social and Emotional Assessment (ITSEA) questionnaire in the second year of life. Methylation of nine genomically imprinted genes was measured to determine if MDA was associated with CpG methylation. Results: Child depression was inversely associated with maternal MDA (Bonferroni-corrected p = 0.041). While controlling for false-discovery, compared to offspring of women with the lowest MDA tertile, those with MDA scores in middle and high MDA tertiles had decreased odds for atypical behaviors [OR (95% CI) = 0.40 (0.20, 0.78) for middle and 0.40 (0.17, 0.92) for highest tertile], for maladaptive behaviors [0.37 (0.18, 0.72) for middle tertile and 0.42 (0.18, 0.95) for highest tertile] and for an index of autism spectrum disorder behaviors [0.46 (0.23, 0.90) for middle and 0.35 (0.15, 0.80) for highest tertile]. Offspring of women with the highest MDA tertile were less likely to exhibit depressive [OR = 0.28 (0.12, 0.64)] and anxiety [0.42 (0.18, 0.97)] behaviors and increased odds of social relatedness [2.31 (1.04, 5.19)] behaviors when compared to low MDA mothers. Some associations varied by sex. Perinatal MDA score was associated with methylation differences for imprinted control regions of PEG10/SGCE [females: Beta (95% CI) = 1.66 (0.52, 2.80) - Bonferroni-corrected p = 0.048; males: -0.56 (-1.13, -0.00)], as well as both MEG3 and IGF2 in males [0.97 (0.00, 1.94)] and -0.92 (-1.65, -0.19) respectively. Conclusion: In this ethnically diverse cohort, maternal adherence to a Mediterranean diet in early pregnancy was associated with favorable neurobehavioral outcomes in early childhood and with sex-dependent methylation differences of MEG3, IGF2, and SGCE/PEG10 DMRs.
Collapse
Affiliation(s)
- John S House
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States.,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States
| | - Michelle Mendez
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rachel L Maguire
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Sarah Gonzalez-Nahm
- Department of Health, Behavior and Society, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Zhiqing Huang
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, United States
| | - Julie Daniels
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Susan K Murphy
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, United States
| | - Bernard F Fuemmeler
- Department of Health Behavior and Policy, Virginia Commonwealth University, Richmond, VA, United States
| | - Fred A Wright
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States.,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States.,Department of Statistics, North Carolina State University, Raleigh, NC, United States
| | - Cathrine Hoyo
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
45
|
Canever L, Freire TG, Mastella GA, Damázio L, Gomes S, Fachim I, Michels C, Carvalho G, Godói AK, Peterle BR, Gava FF, Valvassori SS, Budni J, Quevedo J, Zugno AI. Changes in behavioural parameters, oxidative stress and neurotrophins in the brain of adult offspring induced to an animal model of schizophrenia: The effects of FA deficient or FA supplemented diet during the neurodevelopmental phase. Prog Neuropsychopharmacol Biol Psychiatry 2018; 86:52-64. [PMID: 29782958 DOI: 10.1016/j.pnpbp.2018.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 05/16/2018] [Accepted: 05/17/2018] [Indexed: 01/28/2023]
Abstract
A deficiency of maternal folic acid (FA) can compromise the function and development of the brain, and may produce a susceptibility to diseases such as schizophrenia (SZ) in the later life of offspring. The aim of this study was to evaluate the effects of both FA deficient and FA supplemented diets during gestation and lactation on behavioural parameters, the markers of oxidative stress and neurotrophic factors in adult offspring which had been subjected to an animal model of SZ. Female mother rats (Dam's) were separated into experimental maternal groups, which began receiving a special diet (food) consisting of the AIN-93 diet, a control diet, or an FA deficient diet during the periods of pregnancy and lactation. Dam's receiving the control diet were further subdivided into four groups: one group received only control diet, while three groups to receive supplementation with FA at different doses (5, 10 and 50 mg/kg). Adult offspring bred from the Dam's were divided into ten groups for induction of the animal model of SZ through the administration of ketamine (Ket) (25 mg/kg). After the last administration of the drug, the animals were subjected to the behavioural tests and were then euthanized. The frontal cortex (FC) and hippocampus (Hip) were then dissected for later biochemical analysis. Our data demonstrates that Ket induced the model of SZ by altering the behavioural parameters (e.g. hyperlocomotion, social impairment, deficits in the sensory-motor profile and memory damage in the adult animals); and also caused changes in the parameters of oxidative stress (lipid hydroperoxide - LPO; 8-isoprostane - 8-ISO; 4-hydroxynonenal - 4-HNE; protein carbonyl content; superoxide dismutase - SOD and catalase - CAT) as well as in the levels of neurotrophic factors (brain-derived neurotrophic factor - BDNF and nerve growth factor - NGF) particularly within the FC of adult offspring. A deficiency in maternal FA, alone or in combination with ket, was able to induce hyperlocomotion and social impairment in the offspring with increased levels of lipid and protein damage (LPO, 8-ISO, 4-HNE, carbonylation of protein) within the FC, increased activity of antioxidant enzymes (SOD and CAT) in both of the brain structures studied, and also reduced the levels of neurotrophins (BDNF and NGF), particularly within the Hip of the adult offspring. Supplementation of FA (5, 10 and 50 mg/kg) to the Dam's was mostly able to prevent the cognitive damage which was induced by Ket in the adult animals. FA (10 and 50 mg/kg) attenuated the action of Ket in the animals in relation to the biochemical parameters, proving the possible neuroprotective effect of FA in the adulthood of offspring that were subjected to the animal model of SZ. Our study indicates that the intake of maternal FA during pregnancy and lactation plays an important role, particularly in the regulation of markers of oxidative stress and neurotrophins.
Collapse
Affiliation(s)
- L Canever
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
| | - T G Freire
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
| | - G A Mastella
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
| | - L Damázio
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
| | - S Gomes
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
| | - I Fachim
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
| | - C Michels
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
| | - G Carvalho
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
| | - A K Godói
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
| | - B R Peterle
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
| | - F F Gava
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
| | - S S Valvassori
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
| | - J Budni
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
| | - J Quevedo
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil; Center for Experimental Models in Psychiatry, Department of Psychiatry and Behavioral Sciences, Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - A I Zugno
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil.
| |
Collapse
|
46
|
Veras LV, Chotai PN, Tumen AZ, Gosain A. Impaired growth outcomes in children with congenital colorectal diseases. J Surg Res 2018; 229:102-107. [PMID: 29936975 DOI: 10.1016/j.jss.2018.03.069] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/02/2018] [Accepted: 03/29/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Cloaca, Hirschsprung disease, and anorectal malformations (CHARM) are congenital anomalies of the hindgut. Small series have suggested that children suffering from one of these anomalies may be at risk for growth impairment. We sought to expand on these findings in a comprehensive cohort, hypothesizing that patients with Medicaid insurance or African-American (AA) race would be at higher risk for poor growth. METHODS Following Institutional Review Board (IRB) approval, single-institution retrospective review of children with CHARM anomalies was performed (2009-2016). Body mass index (BMI) value Z-scores were obtained using the 2006 World Health Organization (age 0-24 mo) and 2000 Centers for Disease Control (CDC) (age >2 y) growth charts and calculators (statistical analysis system). Patient factors and BMI Z-scores were analyzed with descriptive statistics and Fisher's exact test. RESULTS One hundred sixty-six patients (Cloaca n = 16, Hirschsprung disease [HD] n = 71, anorectal malformation [ARM] n = 79) were identified. The BMI Z-score distribution for the entire CHARM cohort was lower than controls (P < 0.0001). HD and ARM BMI Z-scores were also lower versus controls (P < 0.0007, P < 0.0037). Requiring more or less than the average number of surgeries did not impact BMI Z-score [P = non-significant (NS)]. Patients with Medicaid had lower Z-scores versus private or commercial insurance (P < 0.0001). AA race BMI Z-score distribution was lower than controls (P < 0.0002), but there was no statistical difference in BMI Z-scores when comparing AA versus non-AA CHARM patients (P = NS). CONCLUSIONS Patients born with CHARM anomalies are at risk for impaired growth. Furthermore study is warranted to identify modifiable risk factors contributing to this impairment. Longitudinal follow-up should include interventions to mitigate these risks.
Collapse
Affiliation(s)
- Laura V Veras
- Division of Pediatric Surgery, Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Pranit N Chotai
- Division of Pediatric Surgery, Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Andrew Z Tumen
- Division of Pediatric Surgery, Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Ankush Gosain
- Division of Pediatric Surgery, Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee; Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee.
| |
Collapse
|
47
|
Statistical analysis of human microarray data shows that dietary intervention with n-3 fatty acids, flavonoids and resveratrol enriches for immune response and disease pathways. Br J Nutr 2018; 119:239-249. [PMID: 29345217 DOI: 10.1017/s0007114517003506] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
n-3 Fatty acids, flavonoids and resveratrol are well publicised for their beneficial effects on human health and wellbeing. Identifying common, underlying biological mechanisms targeted by these functional foods would therefore be informative for the public health sector for advising on nutritional health and disease, food and drug product development and consumer interest. The aim of this study was to explore the potential effects of gene expression changes associated with n-3 fatty acids EPA and DHA, flavonoids and resveratrol on modifying biological systems and disease pathways. To test this, publicly available human microarray data for significant gene expression changes associated with dietary intervention with EPA/DHA, flavonoids and resveratrol was subjected to pathway analysis and significance testing for overlap with signals from genome-wide association studies (GWAS) for common non-communicable diseases and biological functions. There was an enrichment of genes implicated in immune responses and disease pathways which was common to all of the treatment conditions tested. Analysis of biological functions and disease pathways indicated anti-tumorigenic properties for EPA/DHA. In line with this, significance testing of the intersection of genes associated with these functional foods and GWAS hits for common biological functions (ageing and cognition) and non-communicable diseases (breast cancer, CVD, diabesity, neurodegeneration and psychiatric disorders) identified significant overlap between the EPA/DHA and breast cancer gene sets. Dietary intervention with EPA/DHA, flavonoids and resveratrol can target important biological and disease pathways suggesting a potentially important role for these bioactive compounds in the prevention and treatment of dietary-related diseases.
Collapse
|
48
|
Taylor RM, Fealy SM, Bisquera A, Smith R, Collins CE, Evans TJ, Hure AJ. Effects of Nutritional Interventions during Pregnancy on Infant and Child Cognitive Outcomes: A Systematic Review and Meta-Analysis. Nutrients 2017; 9:E1265. [PMID: 29156647 PMCID: PMC5707737 DOI: 10.3390/nu9111265] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/06/2017] [Accepted: 11/13/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Epidemiological studies have demonstrated that folate, iodine and iron intake during pregnancy impacts on foetal brain development and cognitive function. However, in human studies, the relationship with other dietary nutrients is less clear. OBJECTIVE This systematic review aims to critically appraise the current literature and meta-analyses results from nutritional interventions during pregnancy that aimed to optimise infant and child cognitive outcomes. DESIGN Ten electronic databases were searched for articles published up to August 2017. The search was limited to articles published in English. Randomised controlled trials (RCTs) testing the impact of any nutritional intervention (dietary counselling, education, nutrient supplementation, fortified foods and/or foods) during pregnancy on cognitive outcomes of children (<10 years old). Two independent reviewers assessed study eligibility and quality using the American Dietetic Association quality criteria checklist for primary research. Standardised mean differences were used for nine cognitive domains to measure effects for meta-analyses. RESULTS A total of 34 RCTs were included (21 studies included children aged less than 35 months, 10 studies included children aged 36-60 months and 3 studies included children aged 61-119 months). The types of nutritional interventions included nutrient supplements, whole foods, fortified foods and nutrition education. The following nine cognition outcomes: attention, behaviour, crystallised intelligence, fluid intelligence, global cognition, memory, motor skills, visual processing, and problem solving were not significantly impacted by nutritional interventions, although 65% of studies conducted post-hoc data analyses and were likely to be underpowered. Although, long chain polyunsaturated fatty acids (LCPUFA) supplementation was associated with a marginal increase in crystallised intelligence (Effect size (ES): 0.25; 95% confidence interval (95% CI): -0.04, 0.53), the effect was not statistically significant (p = 0.09), with significant study heterogeneity (p = 0.00). CONCLUSIONS LCPUFA supplementation may be associated with an improvement in child crystallised intelligence, however further research is warranted. The remaining eight cognition domains were not significantly impacted by maternal nutritional interventions.
Collapse
Affiliation(s)
- Rachael M Taylor
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia.
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia.
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia.
| | - Shanna M Fealy
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia.
- Faculty of Health and Medicine, School of Nursing & Midwifery, University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Alessandra Bisquera
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia.
- Clinical Research Design IT and Statistical Support (CReDITSS) Unit, Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia.
| | - Roger Smith
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia.
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia.
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia.
| | - Clare E Collins
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia.
- Faculty of Health and Medicine, School of Health Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.
- Priority Research Centre in Physical Activity and Nutrition, University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Tiffany-Jane Evans
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia.
- Clinical Research Design IT and Statistical Support (CReDITSS) Unit, Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia.
| | - Alexis J Hure
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW 2308, Australia.
- Hunter Medical Research Institute, 1 Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia.
- Priority Research Centre for Gender, Health and Ageing, University of Newcastle, Callaghan, NSW 2308, Australia.
| |
Collapse
|
49
|
Mitra-Ganguli T, Kalita S, Bhushan S, Stough C, Kean J, Wang N, Sethi V, Khadilkar A. A Randomized, Double-Blind Study Assessing Changes in Cognitive Function in Indian School Children Receiving a Combination of Bacopa monnieri and Micronutrient Supplementation vs. Placebo. Front Pharmacol 2017; 8:678. [PMID: 29204115 PMCID: PMC5699204 DOI: 10.3389/fphar.2017.00678] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/11/2017] [Indexed: 12/04/2022] Open
Abstract
Several studies have indicated a chronic cognitive enhancing effect of Bacopa monnieri across different ages and cognitive impairment associated with vitamin and mineral deficiencies in children. Therefore, we investigated the effects of 4-month supplementation with a combination of B. monnieri extract and multiple micronutrients on cognitive functions in Indian school children aged 7–12 years. This was a randomized, double-blind, parallel design, single-center study in which 300 children were randomized to receive a beverage either fortified with B. monnieri and multiple micronutrients (“fortified”) or a non-fortified isocaloric equivalent (“control”) twice-daily for 4 months. Cognitive function was assessed by the Cambridge Neuropsychological Automated Test Battery (CANTAB) administered at baseline, Day 60 and Day 121. The primary endpoint was change in short-term memory (working memory) from baseline in subjects receiving “fortified” vs. “control” beverages after 4 months. Secondary endpoints included sustained attention, episodic memory, and executive function. The “fortified” beverage did not significantly improve short-term memory or any of the secondary outcomes tested relative to the “control” beverage. However, the spatial working memory “strategy” score showed significant improvement on Day 60 (difference between groups in change from baseline: −0.55; p < 0.05), but not on Day 121 due to the active intervention. Study products were well-tolerated. Reasons for these unexpected findings are discussed.
Collapse
Affiliation(s)
| | | | | | - Con Stough
- Swinburne Centre for Human Psychopharmacology, Swinburne University, Victoria, BC, Australia
| | - James Kean
- Swinburne Centre for Human Psychopharmacology, Swinburne University, Victoria, BC, Australia
| | - Nan Wang
- GSK Research and Development Centre, Haryana, India
| | - Vidhu Sethi
- GSK Research and Development Centre, Haryana, India
| | - Anuradha Khadilkar
- Jehangir Clinical Development Centre Pvt. Ltd., Jehangir Hospital Premises, Maharashtra, India
| |
Collapse
|
50
|
Hibbeln JR, SanGiovanni JP, Golding J, Emmett PM, Northstone K, Davis JM, Schuckit M, Heron J. Meat Consumption During Pregnancy and Substance Misuse Among Adolescent Offspring: Stratification of TCN2 Genetic Variants. Alcohol Clin Exp Res 2017; 41:1928-1937. [PMID: 28975627 DOI: 10.1111/acer.13494] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/29/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Reducing meat consumption is often advised; however, inadvertent nutritional deficiencies during pregnancy may result in residual neurodevelopmental harms to offspring. This study assessed possible effects of maternal diets in pregnancy on adverse substance use among adolescent offspring. METHODS Pregnant women and their 13-year-old offspring taking part in a prospective birth cohort study, the Avon Longitudinal Study of Parents and Children (ALSPAC), provided Food Frequency Questionnaire data from which dietary patterns were derived using principal components analysis. Multivariable logistic regression models including potential confounders evaluated adverse alcohol, cannabis, and tobacco use of the children at 15 years of age. RESULTS Lower maternal meat consumption was associated with greater problematic substance use among 15-year-old offspring in dose-response patterns. Comparing never to daily meat consumption after adjustment, risks were greater for all categories of problem substance use: alcohol, odds ratio OR = 1.75, 95% CI = (1.23, 2.56), p < 0.001; tobacco use OR = 1.85, 95% CI = (1.28, 2.63), p < 0.001; and cannabis OR = 2.70, 95% CI = (1.89, 4.00), p < 0.001. Given the likelihood of residual confounding, potential causality was evaluated using stratification for maternal allelic variants that impact biological activity of cobalamin (vitamin B12) and iron. Lower meat consumption disproportionally increased the risks of offspring substance misuse among mothers with optimally functional (homozygous) variants (rs1801198) of the gene transcobalamin 2 gene (TCN2) which encodes the vitamin B12 transport protein transcobalamin 2 implicating a causal role for cobalamin deficits. Functional maternal variants in iron metabolism were unrelated to the adverse substance use. Risks potentially attributable to cobalamin deficits during pregnancy include adverse adolescent alcohol, cannabis, and tobacco use (14, 37, and 23, respectively). CONCLUSIONS Lower prenatal meat consumption was associated with increased risks of adolescent substance misuse. Interactions between TCN2 variant status and meat intake implicate cobalamin deficiencies.
Collapse
Affiliation(s)
- Joseph R Hibbeln
- Section on Nutritional Neurosciences, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland
| | - John Paul SanGiovanni
- Section on Nutritional Neurosciences, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland.,Georgetown University School of Medicine, Washington, District of Columbia
| | - Jean Golding
- School of Social and Community Based Medicine, University of Bristol, Bristol, United Kingdom
| | - Pauline M Emmett
- School of Social and Community Based Medicine, University of Bristol, Bristol, United Kingdom
| | - Kate Northstone
- School of Social and Community Based Medicine, University of Bristol, Bristol, United Kingdom
| | - John M Davis
- University of Illinois at Chicago, Chicago, Illinois
| | - Marc Schuckit
- University of California, San Diego, San Diego, California
| | - Jon Heron
- School of Social and Community Based Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|