1
|
Guo D, Sheng W, Cai Y, Shu J, Cai C. Genetic Association of Lipids and Lipid-Lowering Drug Target Genes With Attention Deficit Hyperactivity Disorder. J Atten Disord 2024; 28:1425-1436. [PMID: 38166458 DOI: 10.1177/10870547231222219] [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] [Indexed: 01/04/2024]
Abstract
BACKGROUND Lipid metabolism plays an essential role in nervous system development. Cholesterol deficiency leads to a variety of neurodevelopmental disorders, such as autism spectrum disorder and fragile X syndrome. There have been a lot of efforts to search for biological markers associated with and causal to ADHD, among which lipid is one possible etiological factor that is quite widely studied. We aimed to evaluate the causal relationship between lipids traits, lipid-lowering drugs, and attention deficit hyperactivity disorder (ADHD) outcomes using Mendelian randomization (MR) studies. METHODS We used summary data from genome-wide association studies to explore the causal relationships between circulating lipid-related traits and ADHD. Then, quantitative trait loci for the expression of lipid-lowering drug target genes and genetic variants associated with lipid traits were extracted. Summary-data-based MR and inverse-variance-weighted MR (IVW-MR) were used to investigate the correlation between the expression of these drug-target genes and ADHD. RESULTS After rigorous screening, 939 instrumental variables were finally included for univariable mendelian randomization analysis. However, there is no correlation between lipid profile and ADHD risk. Drug target analysis by IVW-MR method observed that APOB-mediated low-density lipoprotein cholesterol was associated with lower ADHD risk (odds ratio [OR] = 0.90, 95% confidence interval [CI] [0.84, 0.97]; p = .007), whereas LPL-mediated triglycerides levels were associated with a higher risk of ADHD (OR = 1.13, 95% CI [1.06, 1.21]; p < .001). CONCLUSION Our results suggest that APOB gene and LPL gene may be candidate drug target genes for the treatment of ADHD.
Collapse
Affiliation(s)
- Detong Guo
- Tianjin Children's Hospital (Tianjin University Children's Hospital), China
- Tianjin Medical University, China
| | - Wenchao Sheng
- Tianjin Children's Hospital (Tianjin University Children's Hospital), China
- Tianjin Medical University, China
| | | | - Jianbo Shu
- Tianjin Children's Hospital (Tianjin University Children's Hospital), China
- Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, China
| | - Chunquan Cai
- Tianjin Children's Hospital (Tianjin University Children's Hospital), China
- Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, China
| |
Collapse
|
2
|
Krieg S, Konrad M, Krieg A, Kostev K. What Is the Link between Attention-Deficit/Hyperactivity Disorder (ADHD) and Dyslipidemia in Adults? A German Retrospective Cohort Study. J Clin Med 2024; 13:4460. [PMID: 39124726 PMCID: PMC11312942 DOI: 10.3390/jcm13154460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Background: Alterations in the serum lipid profile have been suspected in many psychiatric disorders, such as schizophrenia and depression. However, studies on lipid status in attention-deficit/hyperactivity disorder (ADHD) are sparse and inconsistent. Methods: Using the nationwide, population-based IQVIA Disease Analyzer database, this retrospective cohort study included 5367 outpatients from general practices in Germany aged ≥18 years with a documented first diagnosis of ADHD between January 2005 and December 2021 and 26,835 propensity score-matched individuals without ADHD. Study outcomes were the first diagnosis of lipid metabolism disorders as a function of ADHD within up to 10 years of the index date. The cumulative 10-year incidence was analyzed using Kaplan-Meier curves and compared using the log-rank test. In addition, univariate Cox regression analyses were performed. Results: In the regression analysis, there was no significant association between ADHD and subsequent lipid metabolism disorders in the total population (HR: 0.94; 95% CI: 0.83-1.08), among women (HR: 1.04; 95% CI: 0.84-1.28), and among men (HR: 0.89; 95% CI: 0.74-1.06). In addition, no significant association was observed in the disease-stratified analyses. Conclusions: The findings of this study indicate that ADHD does not exert an influence on lipid metabolism. However, further investigation is warranted, particularly with respect to pharmacological interventions.
Collapse
Affiliation(s)
- Sarah Krieg
- Department of Inclusive Medicine, University Hospital Ostwestfalen-Lippe, Bielefeld University, 33617 Bielefeld, Germany
| | - Marcel Konrad
- Health & Social, FOM University of Applied Sciences for Economics and Management, 60486 Frankfurt am Main, Germany;
| | - Andreas Krieg
- Department of General and Visceral Surgery, Thoracic Surgery and Proctology, University Hospital Herford, Medical Campus OWL, Ruhr University Bochum, 32049 Herford, Germany;
| | | |
Collapse
|
3
|
Azzolino D, Bertoni C, De Cosmi V, Spolidoro GCI, Agostoni C, Lucchi T, Mazzocchi A. Omega-3 polyunsatured fatty acids and physical performance across the lifespan: a narrative review. Front Nutr 2024; 11:1414132. [PMID: 38966419 PMCID: PMC11223594 DOI: 10.3389/fnut.2024.1414132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/12/2024] [Indexed: 07/06/2024] Open
Abstract
Background and Aims Physical performance is a major contributor of mobility and independence during older life. Despite a progressive decline in musculoskeletal function starts from middle age, several factors acting during the life-course can negatively influence musculoskeletal functional capacities. Lifestyle interventions incorporating nutrition and physical exercise can help maximizing the muscle functional capacities in early life as well as preserving them later in life. Among various dietary compounds, omega-3 polyunsaturated fatty acids (PUFAs) are gaining growing attention for their potential effects on muscle membrane composition and muscle function. Indeed, several pathways are enhanced, such as an attenuation of pro-inflammatory oxidative stress, mitochondrial function, activation of the mammalian target of rapamycin (mTOR) signaling and reduction of insulin resistance. Methods We performed a narrative review to explore the existing literature on the relationship between omega-3 PUFAs and physical performance across the life-course. Results Growing evidence from randomized controlled trials (RCTs) suggests beneficial effects of omega-3 PUFAs on muscle function, including physical performance parameters in mid to later life. On the other hand, despite a direct association in early life is not available in literature, some mechanisms by which omega-3 PUFAs may contribute to improved adult physical performance could be hypothesized. Conclusion Omega-3 PUFAs are gaining growing attention for their positive effect on muscle function parameters. The integration of physical function measures in future studies would be of great interest to explore whether omega-3 PUFAs could contribute to improved muscle function, starting from early life and extending throughout the lifespan. However, larger and high-quality RCTs are needed to fully elucidate the beneficial effects of omega-3 PUFAs supplementation on muscle mass and function.
Collapse
Affiliation(s)
- Domenico Azzolino
- Geriatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Camilla Bertoni
- Department of Veterinary Sciences for Health, Animal Production and Food Safety, University of Milan, Milan, Italy
| | - Valentina De Cosmi
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità—Italian National Institute of Health, Rome, Italy
- Department of Clinical and Community Sciences, University of Milan, Milan, Italy
| | | | - Carlo Agostoni
- Department of Clinical and Community Sciences, University of Milan, Milan, Italy
- Pediatric Intermediate Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Tiziano Lucchi
- Geriatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Alessandra Mazzocchi
- Department of Clinical and Community Sciences, University of Milan, Milan, Italy
| |
Collapse
|
4
|
Camprodon-Boadas P, Gil-Dominguez A, De la Serna E, Sugranyes G, Lázaro I, Baeza I. Mediterranean Diet and Mental Health in Children and Adolescents: A Systematic Review. Nutr Rev 2024:nuae053. [PMID: 38758659 DOI: 10.1093/nutrit/nuae053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024] Open
Abstract
CONTEXT Childhood and adolescence are periods of critical importance in the development of mental health disorders. The Mediterranean diet (MD) has been linked to multiple positive health outcomes, including reduced incidence of mental health disorders and fewer psychiatric symptoms. OBJECTIVE This study aimed to investigate the association between adherence to an MD and mental health outcomes in children and adolescents. METHODS A systematic literature review was conducted of original research that explored the relationship between psychiatric symptoms or disorders and adherence to an MD. The literature search was conducted on PubMed, Scopus, Web of Science, MEDES, Dialnet, and Latindex from inception to November 2022, and the Newcastle-Ottawa Scale was used to evaluate the quality of studies. RESULTS A total of 13 studies (6 cross-sectional, 4 case-control, 2 randomized clinical trials, and 1 longitudinal cohort) out of 450 met the inclusion criteria. A total of 3058 children or adolescents with a mean age range from 8.6 to 16.2 years were included. Among the reviewed studies, 5 (71.42%) of those looking at attention-deficit/hyperactivity disorder, 4 (80%) examining depression, and 2 (50%) assessing anxiety found a significant protective association. Seven articles (53.84%) were found to be of high quality and 6 (46.15%) of moderate quality. CONCLUSION Adherence to an MD could be a protective factor for mental health in child and adolescent populations. This suggests that promoting an MD could help prevent the onset of clinical psychiatric symptoms, reduce symptom severity, and improve prognosis in young patients. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42021276316.
Collapse
Affiliation(s)
- Patricia Camprodon-Boadas
- Department of Child and Adolescent Psychiatry and Psychology, 2021SGR01319, Institut Clinic de Neurociències, Hospital Clínic Universitari, Barcelona, 08036, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, 08036, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Department of Medicine, Institute of Neurosciences, Universitat de Barcelona, Barcelona, 08036, Spain
| | - Aitana Gil-Dominguez
- Department of Medicine, Institute of Neurosciences, Universitat de Barcelona, Barcelona, 08036, Spain
| | - Elena De la Serna
- Department of Child and Adolescent Psychiatry and Psychology, 2021SGR01319, Institut Clinic de Neurociències, Hospital Clínic Universitari, Barcelona, 08036, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Gisela Sugranyes
- Department of Child and Adolescent Psychiatry and Psychology, 2021SGR01319, Institut Clinic de Neurociències, Hospital Clínic Universitari, Barcelona, 08036, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, 08036, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Iolanda Lázaro
- Cardiovascular Risk and Nutrition, Hospital del Mar Medical Research Institute (IMIM), Barcelona, 08003, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Immaculada Baeza
- Department of Child and Adolescent Psychiatry and Psychology, 2021SGR01319, Institut Clinic de Neurociències, Hospital Clínic Universitari, Barcelona, 08036, Spain
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, 08036, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Department of Medicine, Institute of Neurosciences, Universitat de Barcelona, Barcelona, 08036, Spain
| |
Collapse
|
5
|
Bozzatello P, Novelli R, Montemagni C, Rocca P, Bellino S. Nutraceuticals in Psychiatric Disorders: A Systematic Review. Int J Mol Sci 2024; 25:4824. [PMID: 38732043 PMCID: PMC11084672 DOI: 10.3390/ijms25094824] [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: 04/04/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Correct nutrition and diet are directly correlated with mental health, functions of the immune system, and gut microbiota composition. Diets with a high content of some nutrients, such as fibers, phytochemicals, and short-chain fatty acids (omega-3 fatty acids), seem to have an anti-inflammatory and protective action on the nervous system. Among nutraceuticals, supplementation of probiotics and omega-3 fatty acids plays a role in improving symptoms of several mental disorders. In this review, we collect data on the efficacy of nutraceuticals in patients with schizophrenia, autism spectrum disorders, major depression, bipolar disorder, and personality disorders. This narrative review aims to provide an overview of recent evidence obtained on this topic, pointing out the direction for future research.
Collapse
Affiliation(s)
- Paola Bozzatello
- Department of Neuroscience, University of Turin, Via Cherasco 15, 10126 Turin, Italy; (R.N.); (C.M.); (P.R.); (S.B.)
| | | | | | | | | |
Collapse
|
6
|
Vacy K, Thomson S, Moore A, Eisner A, Tanner S, Pham C, Saffery R, Mansell T, Burgner D, Collier F, Vuillermin P, O'Hely M, Boon WC, Meikle P, Burugupalli S, Ponsonby AL. Cord blood lipid correlation network profiles are associated with subsequent attention-deficit/hyperactivity disorder and autism spectrum disorder symptoms at 2 years: a prospective birth cohort study. EBioMedicine 2024; 100:104949. [PMID: 38199043 PMCID: PMC10825361 DOI: 10.1016/j.ebiom.2023.104949] [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: 07/05/2023] [Revised: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Attention-deficit/hyperactivity disorder (ADHD) and autism spectrum disorder (ASD) are neurodevelopmental conditions with early life origins. Alterations in blood lipids have been linked to ADHD and ASD; however, prospective early life data are limited. This study examined (i) associations between the cord blood lipidome and ADHD/ASD symptoms at 2 years of age, (ii) associations between prenatal and perinatal predictors of ADHD/ASD symptoms and cord blood lipidome, and (iii) mediation by the cord blood lipidome. METHODS From the Barwon Infant Study cohort (1074 mother-child pairs, 52.3% male children), child circulating lipid levels at birth were analysed using ultra-high-performance liquid chromatography-tandem mass spectrometry. These were clustered into lipid network modules via Weighted Gene Correlation Network Analysis. Associations between lipid modules and ADHD/ASD symptoms at 2 years, assessed with the Child Behavior Checklist, were explored via linear regression analyses. Mediation analysis identified indirect effects of prenatal and perinatal risk factors on ADHD/ASD symptoms through lipid modules. FINDINGS The acylcarnitine lipid module is associated with both ADHD and ASD symptoms at 2 years of age. Risk factors of these outcomes such as low income, Apgar score, and maternal inflammation were partly mediated by higher birth acylcarnitine levels. Other cord blood lipid profiles were also associated with ADHD and ASD symptoms. INTERPRETATION This study highlights that elevated cord blood birth acylcarnitine levels, either directly or as a possible marker of disrupted cell energy metabolism, are on the causal pathway of prenatal and perinatal risk factors for ADHD and ASD symptoms in early life. FUNDING The foundational work and infrastructure for the BIS was sponsored by the Murdoch Children's Research Institute, Deakin University, and Barwon Health. Subsequent funding was secured from the Minderoo Foundation, the European Union's Horizon 2020 research and innovation programme (ENDpoiNTs: No 825759), National Health and Medical Research Council of Australia (NHMRC) and Agency for Science, Technology and Research Singapore [APP1149047], The William and Vera Ellen Houston Memorial Trust Fund (via HOMER Hack), The Shepherd Foundation, The Jack Brockhoff Foundation, the Scobie & Claire McKinnon Trust, the Shane O'Brien Memorial Asthma Foundation, the Our Women Our Children's Fund Raising Committee Barwon Health, the Rotary Club of Geelong, the Ilhan Food Allergy Foundation, Geelong Medical and Hospital Benefits Association, Vanguard Investments Australia Ltd, the Percy Baxter Charitable Trust, and Perpetual Trustees.
Collapse
Affiliation(s)
- Kristina Vacy
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia; Melbourne School of Population and Global Health, University of Melbourne, Parkville 3010, Australia
| | - Sarah Thomson
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia
| | - Archer Moore
- Melbourne School of Mathematics and Statistics, University of Melbourne, Parkville 3010, Australia
| | - Alex Eisner
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia
| | - Sam Tanner
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia
| | - Cindy Pham
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville 3010, Australia; Department of Paediatrics, University of Melbourne, Parkville 3010, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville 3010, Australia; Department of Paediatrics, University of Melbourne, Parkville 3010, Australia
| | - Toby Mansell
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville 3010, Australia; Department of Paediatrics, University of Melbourne, Parkville 3010, Australia
| | - David Burgner
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville 3010, Australia; Department of Paediatrics, University of Melbourne, Parkville 3010, Australia; Department of Paediatrics, Monash University, Clayton 3168, Australia
| | - Fiona Collier
- Child Health Research Unit, Barwon Health, Geelong 3220, Australia; School of Medicine, Deakin University, Geelong 3220, Australia
| | - Peter Vuillermin
- Child Health Research Unit, Barwon Health, Geelong 3220, Australia
| | - Martin O'Hely
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville 3010, Australia; School of Medicine, Deakin University, Geelong 3220, Australia
| | - Wah Chin Boon
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia
| | - Peter Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne 3004, Australia; Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia
| | - Satvika Burugupalli
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne 3004, Australia
| | - Anne-Louise Ponsonby
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Parkville 3010, Australia; Department of Paediatrics, University of Melbourne, Parkville 3010, Australia.
| |
Collapse
|
7
|
Ma S, Zhu J, Xie S, Chen R, Li X, Wei W. Suboptimal dietary quality is associated with mental symptoms among adults aged 40 years and over in China: A population-based cross-sectional study. J Affect Disord 2023; 340:802-811. [PMID: 37597777 DOI: 10.1016/j.jad.2023.08.071] [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: 05/15/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND The previous studies an association between dietary patterns and psychiatric symptoms. However, few studies have examined the association of quality of dietary patterns and anxiety, depressive symptoms in the Chinese population. METHODS Between 2017 and 2019, a population-based, cross-sectional survey was carried out in China. Uniformed questionnaires collected the demographic characteristics and food data. The dietary quality of the adults was evaluated using the revised Diet Balance Index 2016 (DBI-16). We measured anxiety and depression symptoms using the the Generalized Anxiety Disorder (GAD)-7 and Patient Health Questionnaire (PHQ)-9. RESULTS A total of 73,737 participants were recruited during the survey period. 17.6 % and 13.7 % of residents suffer from anxiety and depression symptoms, respectively. The DBI-16 indicates that participants with anxiety or depression symptoms had higher scores of low bound score (LBS, refers to inadequate food intake) and dietary quality distance (DQD, refers to unbalanced food intake) than those without anxiety or depression. The logistic regression models showed that high levels of LBS and DQD problems were more strongly associated with anxiety (LBS:OR = 1.20, DQD:OR = 1.30) and depressive symptoms (LBS:OR = 1.21, DQD:OR = 1.44). On the contrary, higher bound score (HBS, refers to excessive food intake) was significantly negatively correlated with symptoms of anxiety and depression. Moreover, each increase in the food group was associated with 4 % lower odds of anxiety and 6 % lower odds of depression symptoms. LIMITATIONS Cross-sectional design and self-reporting of psychological symptoms and dietary information limit the generalizability of the results. CONCLUSION The dietary quality of adults aged 40 years and over in China is suboptimal, with excessive and inadequate food intake simultaneously. Dietary imbalance, and low dietary diversity may be related to anxiety and depressive symptoms.
Collapse
Affiliation(s)
- Shanrui Ma
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Juan Zhu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuanghua Xie
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ru Chen
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinqing Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenqiang Wei
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
8
|
Mastrorilli C, Arasi S, Barni S, Caimmi D, Chiera F, Comberiati P, Dinardo G, Giannetti A, Gismondi M, Gracci S, Paravati F, Pelosi U, Miraglia Del Giudice M, Bernardini R, Pecoraro L. IgE-Mediated and Non-IgE-Mediated Fish Allergy in Pediatric Age: A Holistic Approach-A Consensus by Diagnostic Commission of the Italian Society of Pediatric Allergy and Immunology. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1651. [PMID: 37763770 PMCID: PMC10537060 DOI: 10.3390/medicina59091651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/29/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023]
Abstract
Fish is one of the "big nine" foods triggering allergic reactions. For this reason, fish allergens must be accurately specified on food labels. Fish allergy affects less than 1% of the world population, but a higher prevalence is observed in pediatric cohorts, up to 7%. Parvalbumin is the main fish allergen found in the muscles. In childhood, sensitization to fish allergens occurs most frequently through the ingestion of fish, rarely transcutaneously or by inhalation. Fish allergy symptoms usually appear within two hours of the allergen contact. The diagnosis begins with the collection of the history. If it is suggestive of fish allergy, prick tests or the measurement of serum-specific IgE should be performed to confirm the suspicion. The oral food challenge is the gold standard for the diagnosis. It is not recommended in case of a severe allergic reaction. It is important to make a differential diagnosis with anisakiasis or scombroid poisoning, which have overlapping clinical features but differ in pathogenesis. Traditionally, managing fish allergy involves avoiding the triggering species (sometimes all bony fish species) and requires an action plan for accidental exposures. The present review will analyze IgE- and non-IgE-mediated fish allergy in children from epidemiology, pathogenesis to clinical features. Moreover, clinical management will be addressed with a particular focus on potential nutritional deficiencies.
Collapse
Affiliation(s)
- Carla Mastrorilli
- Admission and Emergency Pediatric Medicine and Surgery Unit, University Hospital Consortium Corporation Polyclinic of Bari, Pediatric Hospital Giovanni XXIII, 70124 Bari, Italy; (C.M.); (M.G.)
| | - Stefania Arasi
- Area of Translational Research in Pediatric Specialities, Allergy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Simona Barni
- Allergic Unit, Department of Pediatric, Meyer Children’s Hospital, 50139 Florence, Italy;
| | - Davide Caimmi
- Allergy Unit, CHU de Montpellier, Université de Montpellier, 34295 Montpellier, France;
- IDESP, UMR A11, Université de Montpellier, 34093 Montpellier, France
| | - Fernanda Chiera
- Department of Pediatrics, San Giovanni di Dio Hospital, 88900 Crotone, Italy; (F.C.); (F.P.)
| | - Pasquale Comberiati
- Section of Paediatrics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Giulio Dinardo
- Department of Woman, Child and of General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (G.D.); (M.M.D.G.)
| | - Arianna Giannetti
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Marco Gismondi
- Admission and Emergency Pediatric Medicine and Surgery Unit, University Hospital Consortium Corporation Polyclinic of Bari, Pediatric Hospital Giovanni XXIII, 70124 Bari, Italy; (C.M.); (M.G.)
| | - Serena Gracci
- Pediatrics and Neonatology Complex Unit, San Giuseppe Hospital, Azienda USL Toscana Centro, 50053 Empoli, Italy;
- Department of Pediatrics, University Hospital of Pisa, 56124 Pisa, Italy
| | - Francesco Paravati
- Department of Pediatrics, San Giovanni di Dio Hospital, 88900 Crotone, Italy; (F.C.); (F.P.)
| | - Umberto Pelosi
- Pediatric Unit, Santa Barbara Hospital, 09016 Iglesias, Italy;
| | - Michele Miraglia Del Giudice
- Department of Woman, Child and of General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (G.D.); (M.M.D.G.)
| | - Roberto Bernardini
- Pediatrics and Neonatology Complex Unit, San Giuseppe Hospital, Azienda USL Toscana Centro, 50053 Empoli, Italy;
| | - Luca Pecoraro
- Pediatric Unit, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37126 Verona, Italy;
| |
Collapse
|
9
|
Al-Beltagi M, Saeed NK, Bediwy AS, Elbeltagi R, Alhawamdeh R. Role of gastrointestinal health in managing children with autism spectrum disorder. World J Clin Pediatr 2023; 12:171-196. [PMID: 37753490 PMCID: PMC10518744 DOI: 10.5409/wjcp.v12.i4.171] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/08/2023] [Accepted: 08/17/2023] [Indexed: 09/06/2023] Open
Abstract
Children with autism spectrum disorders (ASD) or autism are more prone to gastrointestinal (GI) disorders than the general population. These disorders can significantly affect their health, learning, and development due to various factors such as genetics, environment, and behavior. The causes of GI disorders in children with ASD can include gut dysbiosis, immune dysfunction, food sensitivities, digestive enzyme deficiencies, and sensory processing differences. Many studies suggest that numerous children with ASD experience GI problems, and effective management is crucial. Diagnosing autism is typically done through genetic, neurological, functional, and behavioral assessments and observations, while GI tests are not consistently reliable. Some GI tests may increase the risk of developing ASD or exacerbating symptoms. Addressing GI issues in individuals with ASD can improve their overall well-being, leading to better behavior, cognitive function, and educational abilities. Proper management can improve digestion, nutrient absorption, and appetite by relieving physical discomfort and pain. Alleviating GI symptoms can improve sleep patterns, increase energy levels, and contribute to a general sense of well-being, ultimately leading to a better quality of life for the individual and improved family dynamics. The primary goal of GI interventions is to improve nutritional status, reduce symptom severity, promote a balanced mood, and increase patient independence.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Pediatric Department, Faculty of Medicine, Tanta University, Algharbia, Tanta 31511, Egypt
- Pediatrics, Univeristy Medical Center, King Abdulla Medical City, Arabian Gulf University, Dr. Sulaiman Al Habib Medical Group, Manama, Manama 26671, Bahrain
| | - Nermin Kamal Saeed
- Medical Microbiology Section, Pathology Department, Salmaniya Medical Complex, Ministry of Health, Manama, Manama 12, Bahrain
- Medical Microbiology Section, Pathology Department, Irish Royal College of Surgeon, Bahrain, Muharraq, Busaiteen 15503, Bahrain
| | - Adel Salah Bediwy
- Pulmonology Department, Faculty of Medicine, Tanta University, Algharbia, Tanta 31527, Egypt
- Pulmonology Department, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Dr. Sulaiman Al Habib Medical Group, Manama, Manama 26671, Bahrain
| | - Reem Elbeltagi
- Medicine, The Royal College of Surgeons in Ireland-Bahrain, Muharraq, Busiateen 15503, Bahrain
| | - Rawan Alhawamdeh
- Pediatrics Research, and Development Department, Genomics Creativity and Play Center, Manama, Manama 0000, Bahrain
- Pediatrics Research, and Development Department, SENSORYME Dubai 999041, United Arab Emirates
| |
Collapse
|
10
|
Derbyshire E, Maes M. The Role of Choline in Neurodevelopmental Disorders-A Narrative Review Focusing on ASC, ADHD and Dyslexia. Nutrients 2023; 15:2876. [PMID: 37447203 DOI: 10.3390/nu15132876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Neurodevelopmental disorders appear to be rising in prevalence, according to the recent Global Burden of Disease Study. This rise is likely to be multi-factorial, but the role of certain nutrients known to facilitate neurodevelopment should be considered. One possible contributing factor could be attributed to deficits in choline intake, particularly during key stages of neurodevelopment, which includes the first 1000 days of life and childhood. Choline, a key micronutrient, is crucial for optimal neurodevelopment and brain functioning of offspring. The present narrative review discusses the main research, describing the effect of choline in neurodevelopmental disorders, to better understand its role in the etiology and management of these disorders. In terms of findings, low choline intakes and reduced or altered choline status have been reported in relevant population subgroups: pregnancy (in utero), children with autism spectrum disorders, people with attention deficit hyperactivity disorder and those with dyslexia. In conclusion, an optimal choline provision may offer some neuronal protection in early life and help to mitigate some cognitive effects in later life attributed to neurodevelopmental conditions. Research indicates that choline may act as a modifiable risk factor for certain neurodevelopmental conditions. Ongoing research is needed to unravel the mechanisms and explanations.
Collapse
Affiliation(s)
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok 4002, Thailand
- Research Institute, Medical University of Plovdiv, 10330 Plovdiv, Bulgaria
| |
Collapse
|
11
|
Gillies D, Leach MJ, Perez Algorta G. Polyunsaturated fatty acids (PUFA) for attention deficit hyperactivity disorder (ADHD) in children and adolescents. Cochrane Database Syst Rev 2023; 4:CD007986. [PMID: 37058600 PMCID: PMC10103546 DOI: 10.1002/14651858.cd007986.pub3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Abstract
BACKGROUND Attention deficit hyperactivity disorder (ADHD) is a major problem in children and adolescents, characterised by age-inappropriate levels of inattention, hyperactivity, and impulsivity, and is associated with long-term social, academic, and mental health problems. The stimulant medications methylphenidate and amphetamine are the most frequently used treatments for ADHD, but these are not always effective and can be associated with side effects. Clinical and biochemical evidence suggests that deficiencies of polyunsaturated fatty acids (PUFA) could be related to ADHD. Research has shown that children and adolescents with ADHD have significantly lower plasma and blood concentrations of PUFA and, in particular, lower levels of omega-3 PUFA. These findings suggest that PUFA supplementation may reduce the attention and behaviour problems associated with ADHD. This review is an update of a previously published Cochrane Review. Overall, there was little evidence that PUFA supplementation improved symptoms of ADHD in children and adolescents. OBJECTIVES To compare the efficacy of PUFA to other forms of treatment or placebo in treating the symptoms of ADHD in children and adolescents. SEARCH METHODS We searched 13 databases and two trials registers up to October 2021. We also checked the reference lists of relevant studies and reviews for additional references. SELECTION CRITERIA We included randomised and quasi-randomised controlled trials that compared PUFA with placebo or PUFA plus alternative therapy (medication, behavioural therapy, or psychotherapy) with the same alternative therapy alone in children and adolescents (aged 18 years and under) diagnosed with ADHD. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. Our primary outcome was severity or improvement of ADHD symptoms. Our secondary outcomes were severity or incidence of behavioural problems; quality of life; severity or incidence of depressive symptoms; severity or incidence of anxiety symptoms; side effects; loss to follow-up; and cost. We used GRADE to assess the certainty of evidence for each outcome. MAIN RESULTS We included 37 trials with more than 2374 participants, of which 24 trials were new to this update. Five trials (seven reports) used a cross-over design, while the remaining 32 trials (52 reports) used a parallel design. Seven trials were conducted in Iran, four each in the USA and Israel, and two each in Australia, Canada, New Zealand, Sweden, and the UK. Single studies were conducted in Brazil, France, Germany, India, Italy, Japan, Mexico, the Netherlands, Singapore, Spain, Sri Lanka, and Taiwan. Of the 36 trials that compared a PUFA to placebo, 19 used an omega-3 PUFA, six used a combined omega-3/omega-6 supplement, and two used an omega-6 PUFA. The nine remaining trials were included in the comparison of PUFA to placebo, but also had the same co-intervention in the PUFA and placebo groups. Of these, four trials compared a combination of omega-3 PUFA plus methylphenidate to methylphenidate. One trial each compared omega-3 PUFA plus atomoxetine to atomoxetine; omega-3 PUFA plus physical training to physical training; and an omega-3 or omega-6 supplement plus methylphenidate to methylphenidate; and two trials compared omega-3 PUFA plus dietary supplement to dietary supplement. Supplements were given for a period of between two weeks and six months. Although we found low-certainty evidence that PUFA compared to placebo may improve ADHD symptoms in the medium term (risk ratio (RR) 1.95, 95% confidence interval (CI) 1.47 to 2.60; 3 studies, 191 participants), there was high-certainty evidence that PUFA had no effect on parent-rated total ADHD symptoms compared to placebo in the medium term (standardised mean difference (SMD) -0.08, 95% CI -0.24 to 0.07; 16 studies, 1166 participants). There was also high-certainty evidence that parent-rated inattention (medium-term: SMD -0.01, 95% CI -0.20 to 0.17; 12 studies, 960 participants) and hyperactivity/impulsivity (medium-term: SMD 0.09, 95% CI -0.04 to 0.23; 10 studies, 869 participants) scores were no different compared to placebo. There was moderate-certainty evidence that overall side effects likely did not differ between PUFA and placebo groups (RR 1.02, 95% CI 0.69 to 1.52; 8 studies, 591 participants). There was also moderate-certainty evidence that medium-term loss to follow-up was likely similar between groups (RR 1.03, 95% CI 0.77 to 1.37; 13 studies, 1121 participants). AUTHORS' CONCLUSIONS Although we found low-certainty evidence that children and adolescents receiving PUFA may be more likely to improve compared to those receiving placebo, there was high-certainty evidence that PUFA had no effect on total parent-rated ADHD symptoms. There was also high-certainty evidence that inattention and hyperactivity/impulsivity did not differ between PUFA and placebo groups. We found moderate-certainty evidence that overall side effects likely did not differ between PUFA and placebo groups. There was also moderate-certainty evidence that follow-up was similar between groups. It is important that future research addresses the current weaknesses in this area, which include small sample sizes, variability of selection criteria, variability of the type and dosage of supplementation, and short follow-up times.
Collapse
Affiliation(s)
- Donna Gillies
- Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Matthew J Leach
- School of Nursing & Midwifery, University of South Australia, Adelaide, Australia
| | | |
Collapse
|
12
|
Yap CX, Henders AK, Alvares GA, Giles C, Huynh K, Nguyen A, Wallace L, McLaren T, Yang Y, Hernandez LM, Gandal MJ, Hansell NK, Cleary D, Grove R, Hafekost C, Harun A, Holdsworth H, Jellett R, Khan F, Lawson LP, Leslie J, Levis Frenk M, Masi A, Mathew NE, Muniandy M, Nothard M, Miller JL, Nunn L, Strike LT, Cadby G, Moses EK, de Zubicaray GI, Thompson PM, McMahon KL, Wright MJ, Visscher PM, Dawson PA, Dissanayake C, Eapen V, Heussler HS, Whitehouse AJO, Meikle PJ, Wray NR, Gratten J. Interactions between the lipidome and genetic and environmental factors in autism. Nat Med 2023; 29:936-949. [PMID: 37076741 PMCID: PMC10115648 DOI: 10.1038/s41591-023-02271-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 02/22/2023] [Indexed: 04/21/2023]
Abstract
Autism omics research has historically been reductionist and diagnosis centric, with little attention paid to common co-occurring conditions (for example, sleep and feeding disorders) and the complex interplay between molecular profiles and neurodevelopment, genetics, environmental factors and health. Here we explored the plasma lipidome (783 lipid species) in 765 children (485 diagnosed with autism spectrum disorder (ASD)) within the Australian Autism Biobank. We identified lipids associated with ASD diagnosis (n = 8), sleep disturbances (n = 20) and cognitive function (n = 8) and found that long-chain polyunsaturated fatty acids may causally contribute to sleep disturbances mediated by the FADS gene cluster. We explored the interplay of environmental factors with neurodevelopment and the lipidome, finding that sleep disturbances and unhealthy diet have a convergent lipidome profile (with potential mediation by the microbiome) that is also independently associated with poorer adaptive function. In contrast, ASD lipidome differences were accounted for by dietary differences and sleep disturbances. We identified a large chr19p13.2 copy number variant genetic deletion spanning the LDLR gene and two high-confidence ASD genes (ELAVL3 and SMARCA4) in one child with an ASD diagnosis and widespread low-density lipoprotein-related lipidome derangements. Lipidomics captures the complexity of neurodevelopment, as well as the biological effects of conditions that commonly affect quality of life among autistic people.
Collapse
Affiliation(s)
- Chloe X Yap
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia.
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia.
| | - Anjali K Henders
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
| | - Gail A Alvares
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Anh Nguyen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Leanne Wallace
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
| | - Tiana McLaren
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
| | - Yuanhao Yang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
| | - Leanna M Hernandez
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Michael J Gandal
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Lifespan Brain Institute at Penn Medicine and The Children's Hospital of Philadelphia, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Narelle K Hansell
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Dominique Cleary
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Rachel Grove
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Faculty of Health, University of Technology Sydney, Sydney, New South Wales, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Claire Hafekost
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Alexis Harun
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Helen Holdsworth
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Rachel Jellett
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Olga Tennison Autism Research Centre, La Trobe University, Melbourne, Victoria, Australia
| | - Feroza Khan
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Lauren P Lawson
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Department of Psychology, Counselling and Therapy, La Trobe University, Melbourne, Victoria, Australia
| | - Jodie Leslie
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Mira Levis Frenk
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Anne Masi
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Nisha E Mathew
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Melanie Muniandy
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Olga Tennison Autism Research Centre, La Trobe University, Melbourne, Victoria, Australia
| | - Michaela Nothard
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Olga Tennison Autism Research Centre, La Trobe University, Melbourne, Victoria, Australia
| | - Jessica L Miller
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Lorelle Nunn
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Lachlan T Strike
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Gemma Cadby
- School of Population and Global Health, The University of Western Australia, Perth, Western Australia, Australia
| | - Eric K Moses
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Greig I de Zubicaray
- School of Psychology and Counselling, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Katie L McMahon
- School of Clinical Sciences, Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Margaret J Wright
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland, Australia
| | - Peter M Visscher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Paul A Dawson
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
| | - Cheryl Dissanayake
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Olga Tennison Autism Research Centre, La Trobe University, Melbourne, Victoria, Australia
| | - Valsamma Eapen
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Academic Unit of Child Psychiatry South West Sydney, Ingham Institute for Applied Medical Research, Liverpool Hospital, Sydney, New South Wales, Australia
| | - Helen S Heussler
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- Child Development Program, Children's Health Queensland, Brisbane, Queensland, Australia
| | - Andrew J O Whitehouse
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Naomi R Wray
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Jacob Gratten
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia.
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
- Cooperative Research Centre for Living with Autism, Long Pocket, Queensland, Australia.
| |
Collapse
|
13
|
Huber F, Schulz J, Schlack R, Hölling H, Ravens-Sieberer U, Meyer T, Rothenberger A, Wang B, Becker A. Long-term changes in serum levels of lipoproteins in children and adolescents with attention-deficit/hyperactivity disorder (ADHD). J Neural Transm (Vienna) 2023; 130:597-609. [PMID: 36826608 PMCID: PMC10050056 DOI: 10.1007/s00702-022-02583-5] [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: 07/21/2022] [Accepted: 12/26/2022] [Indexed: 02/25/2023]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder affecting approximately 5% of children worldwide. The causal mechanisms of ADHD remain unclear as the aetiology of this disorder seems to be multifactorial. One research field addresses the impact on lipid metabolism and particularly serum lipid fractions on the development of ADHD symptoms. This post hoc analysis aimed to investigate long-term changes in serum levels of lipoproteins in children and adolescents with ADHD and controls. Data of German children and adolescents from the nationwide and representative "Kinder- und Jugendgesundheitssurvey (KiGGS)" study were analysed at baseline and at a ten-year follow-up. At the two time points, participants in the control group were compared with those in the ADHD group, both before and after propensity score matching. Differences in total cholesterol, low-density lipoproteins (LDL), high-density lipoproteins (HDL) and triglycerides were assessed between matched children with and without ADHD. In addition, subgroups with versus without methylphenidate use were compared at both time points. At baseline before matching, there were no significant differences for lipid parameters between participants in the ADHD group (n = 1,219) and the control group (n = 9,741): total cholesterol (Exp(ß) = 0.999, 95%-CI 0.911-1.094, p = .979), LDL (Exp(ß) = 0.967, 95%-CI 0.872-1.071, p = .525), HDL (Exp(ß) = 1.095, 95%-CI 0.899-1.331, p = .366) and triglycerides (Exp(ß) = 1.038, 95%-CI 0.948-1.133, p = .412). Propensity score matching confirmed the non-significant differences between the ADHD and non-ADHD group at baseline. At the 10-year follow-up, n = 571 participants fulfilled complete inclusion criteria, among them 268 subjects were classified as ADHD. The two groups did not significantly differ in lipid fractions, neither cross-sectionally nor with regard to long-term changes. There was also no significant difference between methylphenidate subgroups. In this sample of children and adolescents we could not reveal any significant associations between serum lipid fractions and the diagnosis of ADHD, neither cross-sectionally nor longitudinally; even when methylphenidate use was considered. Thus, further studies using larger sample sizes are required to investigate putative long-term changes in serum lipid fractions related to ADHD.
Collapse
Affiliation(s)
- Franziska Huber
- Clinic for Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany
| | - Jan Schulz
- Clinic for Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany
| | - Robert Schlack
- Department of Epidemiology and Health Monitoring, Robert Koch Institute, Unit Mental Health, Berlin, Germany
| | - Heike Hölling
- Department of Epidemiology and Health Monitoring, Robert Koch Institute, Unit Mental Health, Berlin, Germany
| | - Ulrike Ravens-Sieberer
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Meyer
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Partner Site Göttingen, German Centre for Cardiovascular Research, Göttingen, Germany
| | - Aribert Rothenberger
- Clinic for Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany
| | - Biyao Wang
- Clinic for Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany
- Department of Clinical, Educational and Health Psychology, University College London, London, UK
| | - Andreas Becker
- Clinic for Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany
| |
Collapse
|
14
|
Herrera-Imbroda J, Flores-López M, Ruiz-Sastre P, Gómez-Sánchez-Lafuente C, Bordallo-Aragón A, Rodríguez de Fonseca F, Mayoral-Cleríes F. The Inflammatory Signals Associated with Psychosis: Impact of Comorbid Drug Abuse. Biomedicines 2023; 11:biomedicines11020454. [PMID: 36830990 PMCID: PMC9953424 DOI: 10.3390/biomedicines11020454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/27/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Psychosis and substance use disorders are two diagnostic categories whose association has been studied for decades. In addition, both psychosis spectrum disorders and drug abuse have recently been linked to multiple pro-inflammatory changes in the central nervous system. We have carried out a narrative review of the literature through a holistic approach. We used PubMed as our search engine. We included in the review all relevant studies looking at pro-inflammatory changes in psychotic disorders and substance use disorders. We found that there are multiple studies that relate various pro-inflammatory lipids and proteins with psychosis and substance use disorders, with an overlap between the two. The main findings involve inflammatory mediators such as cytokines, chemokines, endocannabinoids, eicosanoids, lysophospholipds and/or bacterial products. Many of these findings are present in different phases of psychosis and in substance use disorders such as cannabis, cocaine, methamphetamines, alcohol and nicotine. Psychosis and substance use disorders may have a common origin in an abnormal neurodevelopment caused, among other factors, by a neuroinflammatory process. A possible convergent pathway is that which interrelates the transcriptional factors NFκB and PPARγ. This may have future clinical implications.
Collapse
Affiliation(s)
- Jesús Herrera-Imbroda
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Facultad de Medicina, Universidad de Málaga, Andalucía Tech, Campus de Teatinos s/n, 29071 Málaga, Spain
- Departamento de Farmacología y Pediatría, Universidad de Málaga, Andalucía Tech, Campus de Teatinos s/n, 29071 Málaga, Spain
| | - María Flores-López
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Facultad de Psicología, Universidad de Málaga, Andalucía Tech, Campus de Teatinos s/n, 29071 Málaga, Spain
| | - Paloma Ruiz-Sastre
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Facultad de Medicina, Universidad de Málaga, Andalucía Tech, Campus de Teatinos s/n, 29071 Málaga, Spain
- Correspondence: (P.R.-S.); (C.G.-S.-L.)
| | - Carlos Gómez-Sánchez-Lafuente
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Facultad de Psicología, Universidad de Málaga, Andalucía Tech, Campus de Teatinos s/n, 29071 Málaga, Spain
- Correspondence: (P.R.-S.); (C.G.-S.-L.)
| | - Antonio Bordallo-Aragón
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Fermín Mayoral-Cleríes
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| |
Collapse
|
15
|
Wang L, Wang B, Wu C, Wang J, Sun M. Autism Spectrum Disorder: Neurodevelopmental Risk Factors, Biological Mechanism, and Precision Therapy. Int J Mol Sci 2023; 24:ijms24031819. [PMID: 36768153 PMCID: PMC9915249 DOI: 10.3390/ijms24031819] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous, behaviorally defined neurodevelopmental disorder. Over the past two decades, the prevalence of autism spectrum disorders has progressively increased, however, no clear diagnostic markers and specifically targeted medications for autism have emerged. As a result, neurobehavioral abnormalities, neurobiological alterations in ASD, and the development of novel ASD pharmacological therapy necessitate multidisciplinary collaboration. In this review, we discuss the development of multiple animal models of ASD to contribute to the disease mechanisms of ASD, as well as new studies from multiple disciplines to assess the behavioral pathology of ASD. In addition, we summarize and highlight the mechanistic advances regarding gene transcription, RNA and non-coding RNA translation, abnormal synaptic signaling pathways, epigenetic post-translational modifications, brain-gut axis, immune inflammation and neural loop abnormalities in autism to provide a theoretical basis for the next step of precision therapy. Furthermore, we review existing autism therapy tactics and limits and present challenges and opportunities for translating multidisciplinary knowledge of ASD into clinical practice.
Collapse
|
16
|
Matrisciano F. Functional Nutrition as Integrated Intervention for In- and Outpatient with Schizophrenia. Curr Neuropharmacol 2023; 21:2409-2423. [PMID: 36946488 PMCID: PMC10616917 DOI: 10.2174/1570159x21666230322160259] [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: 07/05/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 03/23/2023] Open
Abstract
Schizophrenia is a chronic and progressive disorder characterized by cognitive, emotional, and behavioral abnormalities associated with neuronal development and synaptic plasticity alterations. Genetic and epigenetic abnormalities in cortical parvalbumin-positive GABAergic interneurons and consequent alterations in glutamate-mediated excitatory neurotransmission during early neurodevelopment underlie schizophrenia manifestation and progression. Also, epigenetic alterations during pregnancy or early phases of postnatal life are associated with schizophrenia vulnerability and inflammatory processes, which are at the basis of brain pathology and a higher risk of comorbidities, including cardiovascular diseases and metabolic syndrome. In addition, schizophrenia patients adopt an unhealthy lifestyle and poor nutrition, leading to premature death. Here, I explored the role of functional nutrition as an integrated intervention for the long-term management of patients with schizophrenia. Several natural bioactive compounds in plant-based whole foods, including flavonoids, phytonutrients, vitamins, fatty acids, and minerals, modulate brain functioning by targeting neuroinflammation and improving cognitive decline. Although further clinical studies are needed, a functional diet rich in natural bioactive compounds might be effective in synergism with standard treatments to improve schizophrenia symptoms and reduce the risk of comorbidities.
Collapse
Affiliation(s)
- Francesco Matrisciano
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago (UIC), Chicago, IL, USA
| |
Collapse
|
17
|
Omega-3/6 supplementation for mild to moderate inattentive ADHD: a randomised, double-blind, placebo-controlled efficacy study in Italian children. Eur Arch Psychiatry Clin Neurosci 2022; 272:1453-1467. [PMID: 35672606 DOI: 10.1007/s00406-022-01428-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/09/2022] [Indexed: 11/03/2022]
Abstract
Recently there has been a growing interest in non-pharmacological treatments for ADHD. We evaluated the efficacy of a specific Omega-3/6 dietary supplement (two capsules containing 279 mg eicosapentaenoic acid [EPA], 87 mg Docosahexaenoic Acid [DHA], 30 mg gamma linolenic acid [GLA] each) in ameliorating inattentive symptoms in inattentive-ADHD children (6-12 years) with a baseline ADHD-RS-Inattention score ≥ 12. Secondary objectives included changes in global functioning, severity of illness, depression, and anxiety symptoms, learning disorders and in the fatty acids blood levels. The study was a randomised, double-blind, placebo-controlled efficacy and safety trial with a 6-month double-blind evaluation of Omega-3/6 vs placebo (Phase-I) and a further 6-month-open-label treatment with Omega-3/6 on all patients (Phase-II). In total 160 subjects were enrolled. No superiority of Omega-3/6 supplement to placebo was observed on the primary outcome (ADHD-RS-inattention score) after the first 6-months, with 46.3% of responders in the Omega-3/6 group and 45.6% in the placebo group; a slight (not statistically significant) reduction in Omega-6/3 ratio blood levels was measured in the active treatment group. Twelve months after enrolment, percentages of responders were similar between groups. A mild statistical, although not clinically significant, improvement was observed on the ADHD-RS-total score in the Omega-3/6 group but not on the ADHD-RS-Inattention score; a slight (not-statistically significant) reduction in Omega-6/3 ratio was observed in the group taking active treatment only during Phase II. In conclusion, no clinical beneficial effects of Omega-3/6 were detected on inattentive symptoms, suggesting a limited role of Omega-3/6 dietary products in children with mild ADHD-I.Trial registration: At the time of the Ethical submission, according to the clinical trial Italian law, registration was not mandatory for food additive as Omega 3/6 were then classified. The trial was approved by the Ethical Committee of the Cagliari University Hospital (resolution n. 662; September 22nd, 2011).
Collapse
|
18
|
Long-Term Effects of an Oligoantigenic Diet in Children with Attention-Deficit/Hyperactivity Disorder (ADHD) on Core Symptomatology. Nutrients 2022; 14:nu14235111. [PMID: 36501141 PMCID: PMC9737158 DOI: 10.3390/nu14235111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/24/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
In the early 1920s, it was discovered that nutrition is associated with what is known today as Attention-Deficit/Hyperactivity Disorder (ADHD) and that certain foods can worsen the symptoms. In previous studies, approximately 60% of the participants experience at least a 40% reduction in ADHD symptoms after an oligoantigenic diet (OD). The purpose of this study was to evaluate ADHD symptoms in children approximately 3.5 years after completing a 4-week oligoantigenic diet. Among 28 participants who completed the 4-week diet, 21 were re-assessed for this study after 3.5 years. The severity of ADHD symptoms was assessed with the ADHD-Rating-Scale-IV (ARS). Of 21 participants, 14 fulfilled the responder criterion, whereas 7 did not. At follow-up, 28% of the participants were taking medication. The mean ARS total score improved significantly from T1: M = 29.62 (SD = 9.80) to T2: M = 15.86 (SD = 8.56) between the time points before and after the diet (d = -1.91). There was also a lower ARS total score at the follow-up T5: M = 16.00 (SD = 10.52) compared to before the diet (d = -1.17). This study shows that individually adjusted nutrition significantly improved the ADHD symptomatology of the participants long-term. This suggests that an oligoantigenic diet with subsequent individual nutritional recommendations could become an additional treatment option for children with ADHD.
Collapse
|
19
|
Gallardo-Carrasco MC, Jiménez-Barbero JA, Bravo-Pastor MDM, Martin-Castillo D, Sánchez-Muñoz M. Serum Vitamin D, Folate and Fatty Acid Levels in Children with Autism Spectrum Disorders: A Systematic Review and Meta-Analysis. J Autism Dev Disord 2022; 52:4708-4721. [PMID: 34734376 PMCID: PMC9556366 DOI: 10.1007/s10803-021-05335-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2021] [Indexed: 10/30/2022]
Abstract
Vitamin and fatty acid deficiency in children diagnosed with autism has been linked to the etiology and course of the disease but the results have been inconsistent. In our work, we present a narrative review, which includes 20 observational studies that provide data on the blood levels of vitamin D, folate, or fatty acids of children diagnosed with ASD (Autism Spectrum Disorder-AG group), and of a control group (children without this disorder-CG group). The main characteristics and results are presented in a summary table. Of the 20 above-mentioned studies, a meta-analysis of vitamin D and folate levels was carried out in 14 of them, with a total of 2269 children (AG = 1159, CG = 1110). Vitamin D levels were lower in AG compared to CG: SMD, 95% CI = - 0.83 [- 1.15, - 0.50]. In terms of folate levels, a total of 299 children (AG = 148, CG = 151) were analyzed, finding no significant differences with the control group: SMD, 95% CI = - 0.16 [- 0.63, 0.32]. Only one study that provided data on fatty acids in children with ASD was included in the review although it was not possible to include it in the meta-analysis. We conclude that the nutritional status (vitamin and fatty acid levels) of patients diagnosed with ASD should be taken into account, as correct adjustment of these levels-may produce an improvement in the course of the disease and could also reduce the risk of its development.
Collapse
Affiliation(s)
| | | | - María del Mar Bravo-Pastor
- Nursing Department, University School of Nursing of Cartagena, University of Murcia, , Campus de Cartagena, Paseo Alfonso XIII, 61, PC: 30203 Cartagena, Murcia, Spain
| | - David Martin-Castillo
- Department of Nursing, University of Murcia, Espinardo Campus, Building 23, PC: 30100 Murcia, Spain
| | - María Sánchez-Muñoz
- “Los Arcos” Mental Health Center, Paraje Torre Octavio, 54, PC: 30739 Pozo Aledo, Murcia, Spain
| |
Collapse
|
20
|
Wei L, Zhang W, Li Y, Zhai J. The SIRT1-HMGB1 axis: Therapeutic potential to ameliorate inflammatory responses and tumor occurrence. Front Cell Dev Biol 2022; 10:986511. [PMID: 36081910 PMCID: PMC9448523 DOI: 10.3389/fcell.2022.986511] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammation is a common complication of many chronic diseases. It includes inflammation of the parenchyma and vascular systems. Sirtuin 1 (SIRT1) is a nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylase, which can directly participate in the suppression of inflammation. It can also regulate the activity of other proteins. Among them, high mobility group box 1 (HMGB1) signaling can be inhibited by deacetylating four lysine residues (55, 88, 90, and 177) in quiescent endothelial cells. HMGB1 is a ubiquitous nuclear protein, once translocated outside the cell, which can interact with various target cell receptors including the receptor for advanced glycation end-products (RAGE), toll-like receptor (TLR) 2, and TLR4 and stimulates the release of pro-inflammatory cyto-/chemokines. And SIRT1 has been reported to inhibit the activity of HMGB1. Both are related to the occurrence and development of inflammation and associated diseases but show an antagonistic relationship in controlling inflammation. Therefore, in this review, we introduce how this signaling axis regulates the emergence of inflammation-related responses and tumor occurrence, providing a new experimental perspective for future inflammation research. In addition, it explores diverse upstream regulators and some natural/synthetic activators of SIRT1 as a possible treatment for inflammatory responses and tumor occurrence which may encourage the development of new anti-inflammatory drugs. Meanwhile, this review also introduces the potential molecular mechanism of the SIRT1-HMGB1 pathway to improve inflammation, suggesting that SIRT1 and HMGB1 proteins may be potential targets for treating inflammation.
Collapse
Affiliation(s)
- Lanyi Wei
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, Jilin, China
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Wenrui Zhang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yueyang Li
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jinghui Zhai
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Jinghui Zhai,
| |
Collapse
|
21
|
Lawrence K, Myrissa K, Toribio-Mateas M, Minini L, Gregory AM. Trialling a microbiome-targeted dietary intervention in children with ADHD-the rationale and a non-randomised feasibility study. Pilot Feasibility Stud 2022; 8:108. [PMID: 35606889 PMCID: PMC9125862 DOI: 10.1186/s40814-022-01058-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 05/03/2022] [Indexed: 11/17/2022] Open
Abstract
Background Dietary interventions have been previously explored in children with ADHD. Elimination diets and supplementation can produce beneficial behaviour changes, but little is known about the mechanisms mediating change. We propose that these interventions may work, in part, by causing changes in the gut microbiota. A microbiome-targeted dietary intervention was developed, and its feasibility assessed. Methods A non-randomised feasibility study was conducted on nine non-medicated children with ADHD, aged 8–13 years (mean 10.39 years), using a prospective one-group pre-test/post-test design. Participants were recruited from ADHD support groups in London and took part in the 6-week microbiome-targeted dietary intervention, which was specifically designed to impact the composition of gut bacteria. Children were assessed pre- and post-intervention on measures of ADHD symptomatology, cognition, sleep, gut function and stool-sample microbiome analysis. The primary aim was to assess the study completion rate, with secondary aims assessing adherence, adverse events (aiming for no severe and minimal), acceptability and suitability of outcome measures. Results Recruitment proved to be challenging and despite targeting 230 participants directly through support groups, and many more through social media, nine families (of the planned 10) signed up for the trial. The completion rate for the study was excellent at 100%. Exploration of secondary aims revealed that (1) adherence to each aspect of the dietary protocol was very good; (2) two mild adverse events were reported; (3) parents rated the treatment as having good acceptability; (4) data collection and outcome measures were broadly feasible for use in an RCT with a few suggestions recommended; (5) descriptive data for outcome measures is presented and suggests that further exploration of gut microbiota, ADHD symptoms and sleep would be helpful in future research. Conclusions This study provides preliminary evidence for the feasibility of a microbiome-targeted dietary intervention in children with ADHD. Recruitment was challenging, but the diet itself was well-tolerated and adherence was very good. Families wishing to trial this diet may find it an acceptable intervention. However, recruitment, even for this small pilot study, was challenging. Because of the difficulty experienced recruiting participants, future randomised controlled trials may wish to adopt a simpler dietary approach which requires less parental time and engagement, in order to recruit the number of participants required to make meaningful statistical interpretations of efficacy. Trial registration ClinicalTrials.gov Identifier: NCT03737877. Registered 13 November 2018—retrospectively registered, within 2 days of the first participant being recruited. Supplementary Information The online version contains supplementary material available at 10.1186/s40814-022-01058-4.
Collapse
Affiliation(s)
- Kate Lawrence
- Department of Psychology & Pedagogic Science, Faculty of Sport, Allied Health and Performance Science, St Mary's University, Twickenham, London, UK.
| | - Kyriaki Myrissa
- Department of Health Sciences, Faculty of Sport, Allied Health and Performance Science, St Mary's University, Twickenham, London, UK
| | - Miguel Toribio-Mateas
- School of Health and Education, Middlesex University, London, UK.,School of Applied Science, London South Bank University, London, UK
| | - Lori Minini
- Department of Psychology & Pedagogic Science, Faculty of Sport, Allied Health and Performance Science, St Mary's University, Twickenham, London, UK
| | - Alice M Gregory
- Department of Psychology, Goldsmiths, University of London, London, UK
| |
Collapse
|
22
|
de la Torre-Aguilar MJ, Gomez-Fernandez A, Flores-Rojas K, Martin-Borreguero P, Mesa MD, Perez-Navero JL, Olivares M, Gil A, Gil-Campos M. Docosahexaenoic and Eicosapentaenoic Intervention Modifies Plasma and Erythrocyte Omega-3 Fatty Acid Profiles But Not the Clinical Course of Children With Autism Spectrum Disorder: A Randomized Control Trial. Front Nutr 2022; 9:790250. [PMID: 35425788 PMCID: PMC9002234 DOI: 10.3389/fnut.2022.790250] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 02/28/2022] [Indexed: 12/30/2022] Open
Abstract
BackgroundThe pathogenesis of autism spectrum disorder (ASD) is under investigation and one of the main alterations relates to the metabolic and inflammatory system dysfunctions. Indeed, based on a possible deficit of omega-3 fatty acids (FAs) of patients with ASD and looking for an anti-inflammatory effect, dietary supplements with omega-3 fatty acids have been proposed. We aimed to evaluate differences in plasma and erythrocyte FA profiles and plasma cytokines in patients with infantile ASD after supplementation with docosahexaenoic (DHA) and eicosapentaenoic (EPA) acids or placebo and both compared at baseline with a reference healthy group.MethodsA double-blind, randomized placebo-controlled intervention with DHA/EPA for 6 months was carried out in 54 children between 2 and 6 years diagnosed with ASD. They were selected and randomly assigned into two groups: 19 children received 800 mg/day of DHA and 25 mg/day of EPA, or placebo. In addition, another reference group of 59 healthy children of the same age was included. Plasma lipids and cytokines, and FA profiles in plasma and erythrocytes were measured at baseline and after 6 months of treatment in ASD children, and at baseline in the reference group.ResultsThere were no differences in demographic, anthropometric characteristics, and omega-3 intake between the healthy reference group and the ASD children at baseline. Children with ASD showed the higher plasma percentages of palmitic acid and total saturated FA and lower total omega-6 polyunsaturated FA (PUFA) compared with healthy children. An increased level of DHA and reduced EPA level in erythrocytes were detected in the ASD group vs. the reference group. After 6 months of treatment, the ASD group that received DHA enriched product significantly increased the plasma and erythrocyte percentages of DHA, but no differences were observed in the clinical test scores and other parameters as plasma cytokines between the two groups of ASD related to the intervention.ConclusionSpanish children with ASD exhibit an appropriate omega-3 FA status in plasma and erythrocytes. Neither a clinical improvement of ASD children nor a better anti-inflammatory or fatty acid state has been found after an intervention with DHA/EPA for 6 months. So, the prescription of n-3 LC-PUFA and other dietary supplements in ASD should be only indicated after a confirmed alteration of FA metabolism or omega-3 LC-PUFA deficiency evaluated by specific erythrocyte FA.Clinical Trial Registration[www.ClinicalTrials.gov], identifier [NCT03620097].
Collapse
Affiliation(s)
- Maria Jose de la Torre-Aguilar
- Pediatric Research and Metabolism Unit, Reina Sofia University Hospital, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba, Córdoba, Spain
| | - Antonio Gomez-Fernandez
- Pediatric Research and Metabolism Unit, Reina Sofia University Hospital, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba, Córdoba, Spain
| | - Katherine Flores-Rojas
- Pediatric Research and Metabolism Unit, Reina Sofia University Hospital, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba, Córdoba, Spain
| | - Pilar Martin-Borreguero
- Department of Child and Adolescent Clinical Psychiatry and Psychology, Reina Sofia University Hospital, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), Córdoba, Spain
| | - María Dolores Mesa
- Department of Biochemistry and Molecular Biology II University of Granada, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, Granada, Spain
- Biomedical Research Center, Institute of Nutrition and Food Technology “José Mataix,” University of Granada, Parque Tecnológico de la Salud, Granada, Spain
| | - Juan Luis Perez-Navero
- Pediatric Research and Metabolism Unit, Reina Sofia University Hospital, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba, Córdoba, Spain
- CIBERER (Ciber Rare Diseases), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- *Correspondence: Juan Luis Perez-Navero, ,
| | | | - Angel Gil
- Department of Biochemistry and Molecular Biology II University of Granada, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, Granada, Spain
- Biomedical Research Center, Institute of Nutrition and Food Technology “José Mataix,” University of Granada, Parque Tecnológico de la Salud, Granada, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Mercedes Gil-Campos
- Pediatric Research and Metabolism Unit, Reina Sofia University Hospital, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), University of Córdoba, Córdoba, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
23
|
van der Wurff I, Oenema A, de Ruijter D, Vingerhoets C, van Amelsvoort T, Rutten B, Mulkens S, Köhler S, Schols A, de Groot R. A Scoping Literature Review of the Relation between Nutrition and ASD Symptoms in Children. Nutrients 2022; 14:1389. [PMID: 35406004 PMCID: PMC9003544 DOI: 10.3390/nu14071389] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/18/2022] [Accepted: 03/24/2022] [Indexed: 02/01/2023] Open
Abstract
Autism spectrum disorder (ASD) is characterized by impairments in social interaction, communication skills, and repetitive and restrictive behaviors and interests. Even though there is a biological basis for an effect of specific nutrition factors on ASD symptoms and there is scientific literature available on this relationship, whether nutrition factors could play a role in ASD treatment is unclear. The goal of the current literature review was to summarize the available scientific literature on the relation between nutrition and autism spectrum disorder (ASD) symptoms in childhood, and to formulate practical dietary guidelines. A comprehensive search strategy including terms for ASD, nutrition factors (therapeutic diets, dietary patterns, specific food products, fatty acids and micronutrients) and childhood was developed and executed in six literature databases (Cinahl, Cochrane, Ovid Embase, PsycInfo, PubMed and Web of Science). Data from meta-analyses, systematic reviews and original studies were qualitatively summarized. A total of 5 meta-analyses, 29 systematic reviews and 27 original studies were retrieved that focused on therapeutic diets, specific food products, fatty acids and micronutrients and ASD symptoms during childhood. Results of the available studies were sparse and inconclusive, and hence, no firm conclusions could be drawn. There is currently insufficient evidence for a relation between nutrition and ASD symptoms in childhood, making it impossible to provide practical nutrition guidelines; more methodological sound research is needed.
Collapse
Affiliation(s)
- Inge van der Wurff
- Health Psychology, Faculty of Psychology, Open University of the Netherlands, 6419 AT Heerlen, The Netherlands
- Conditions for Lifelong Learning, Faculty of Educational Sciences, Open University of the Netherlands, 6419 AT Heerlen, The Netherlands;
| | - Anke Oenema
- Department of Health Promotion, Maastricht University, 6200 MD Maastricht, The Netherlands; (A.O.); (D.d.R.)
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD Maastricht, The Netherlands;
| | - Dennis de Ruijter
- Department of Health Promotion, Maastricht University, 6200 MD Maastricht, The Netherlands; (A.O.); (D.d.R.)
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD Maastricht, The Netherlands;
| | - Claudia Vingerhoets
- Department of Psychiatry and Neuropsychology, Maastricht University, 6200 MD Maastricht, The Netherlands; (C.V.); (T.v.A.); (B.R.); (S.M.); (S.K.)
| | - Thérèse van Amelsvoort
- Department of Psychiatry and Neuropsychology, Maastricht University, 6200 MD Maastricht, The Netherlands; (C.V.); (T.v.A.); (B.R.); (S.M.); (S.K.)
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Bart Rutten
- Department of Psychiatry and Neuropsychology, Maastricht University, 6200 MD Maastricht, The Netherlands; (C.V.); (T.v.A.); (B.R.); (S.M.); (S.K.)
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Sandra Mulkens
- Department of Psychiatry and Neuropsychology, Maastricht University, 6200 MD Maastricht, The Netherlands; (C.V.); (T.v.A.); (B.R.); (S.M.); (S.K.)
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6200 MD Maastricht, The Netherlands
- Department of Clinical Psychological Science, Maastricht University, 6200 MD Maastricht, The Netherlands
- SeysCentra, 6581 TE Malden, The Netherlands
| | - Sebastian Köhler
- Department of Psychiatry and Neuropsychology, Maastricht University, 6200 MD Maastricht, The Netherlands; (C.V.); (T.v.A.); (B.R.); (S.M.); (S.K.)
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Annemie Schols
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD Maastricht, The Netherlands;
- Department of Respiratory Medicine, Maastricht University, 6202 AZ Maastricht, The Netherlands
| | - Renate de Groot
- Conditions for Lifelong Learning, Faculty of Educational Sciences, Open University of the Netherlands, 6419 AT Heerlen, The Netherlands;
| |
Collapse
|
24
|
Morton SU, Leyshon BJ, Tamilia E, Vyas R, Sisitsky M, Ladha I, Lasekan JB, Kuchan MJ, Grant PE, Ou Y. A Role for Data Science in Precision Nutrition and Early Brain Development. Front Psychiatry 2022; 13:892259. [PMID: 35815018 PMCID: PMC9259898 DOI: 10.3389/fpsyt.2022.892259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Multimodal brain magnetic resonance imaging (MRI) can provide biomarkers of early influences on neurodevelopment such as nutrition, environmental and genetic factors. As the exposure to early influences can be separated from neurodevelopmental outcomes by many months or years, MRI markers can serve as an important intermediate outcome in multivariate analyses of neurodevelopmental determinants. Key to the success of such work are recent advances in data science as well as the growth of relevant data resources. Multimodal MRI assessment of neurodevelopment can be supplemented with other biomarkers of neurodevelopment such as electroencephalograms, magnetoencephalogram, and non-imaging biomarkers. This review focuses on how maternal nutrition impacts infant brain development, with three purposes: (1) to summarize the current knowledge about how nutrition in stages of pregnancy and breastfeeding impact infant brain development; (2) to discuss multimodal MRI and other measures of early neurodevelopment; and (3) to discuss potential opportunities for data science and artificial intelligence to advance precision nutrition. We hope this review can facilitate the collaborative march toward precision nutrition during pregnancy and the first year of life.
Collapse
Affiliation(s)
- Sarah U Morton
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, United States.,Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | | | - Eleonora Tamilia
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, United States.,Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Rutvi Vyas
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, United States.,Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital, Boston, MA, United States
| | - Michaela Sisitsky
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, United States.,Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital, Boston, MA, United States
| | - Imran Ladha
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, United States
| | | | | | - P Ellen Grant
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, United States.,Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States.,Department of Radiology, Boston Children's Hospital, Boston, MA, United States
| | - Yangming Ou
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, United States.,Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States.,Department of Radiology, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
25
|
Potential Role of Omega-3 Polyunsaturated Fatty Acids in Pediatric Food Allergy. Nutrients 2021; 14:nu14010152. [PMID: 35011028 PMCID: PMC8746967 DOI: 10.3390/nu14010152] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 02/07/2023] Open
Abstract
Polyunsaturated fatty acids (PUFAs) are involved both in immune system regulation and inflammation. In particular, within the PUFAs category, omega-3 (ω-3) may reduce inflammation, whereas omega-6 (ω-6) PUFAs are generally considered to have a proinflammatory effect. Recent evidence highlights an imbalance in the ω-3:ω-6 ratio with an increased intake of ω-6, as a consequence of the shift towards a westernized diet. In critical age groups such as infants, toddlers and young children, as well as pregnant and lactating women or fish allergic patients, ω-3 intake may be inadequate. This review aims to discuss the potential beneficial effects of PUFAs on pediatric food allergy prevention and treatment, both at prenatal and postnatal ages. Data from preclinical studies with PUFAs supplementation show encouraging effects in suppressing allergic response. Clinical studies results are still conflicting about the best timing and dosages of supplementation and which individuals are most likely to benefit; therefore, it is still not possible to draw firm conclusions. With regard to food-allergic children, it is still debated whether PUFAs could slow disease progression or not, since consistent data are lacking. In conclusion, more data on the effects of ω-3 PUFAs supplementation alone or in combination with other nutrients are warranted, both in the general and food allergic population.
Collapse
|
26
|
Increased PLA 2 activity in individuals at ultra-high risk for psychosis. Eur Arch Psychiatry Clin Neurosci 2021; 271:1593-1599. [PMID: 33677687 DOI: 10.1007/s00406-021-01246-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/24/2021] [Indexed: 10/22/2022]
Abstract
Phospholipase A2 is the main enzyme in the metabolism of membrane phospholipids. It comprises a family of enzymes divided into iPLA2, cPLA2 and sPLA2. Studies have reported increased PLA2 activity in psychotic patients, which suggests an accelerated breakdown of membrane phospholipids. In the present study we investigated whether increased PLA2 activity is also present in individuals at ultra-high risk (UHR) for psychosis. One-hundred fifty adults were included in this study (85 UHR and 65 controls). UHR was assessed using the "structured interview for prodromal syndromes". PLA2 activity was determined in platelets by a radio-enzymatic assay. We found in UHR individuals increased activities of iPLA2 (p < 0.001) and cPLA2 (p = 0.012) as compared to controls. No correlations were found between socio-demographic and clinical parameters and PLA2 activity. Our findings suggest that increased PLA2 activities may be useful as a biological risk-marker for psychotic disorders.
Collapse
|
27
|
The Role of Iron and Zinc in the Treatment of ADHD among Children and Adolescents: A Systematic Review of Randomized Clinical Trials. Nutrients 2021; 13:nu13114059. [PMID: 34836314 PMCID: PMC8618748 DOI: 10.3390/nu13114059] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/01/2021] [Accepted: 11/11/2021] [Indexed: 12/18/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder common from childhood to adulthood, affecting 5% to 12% among the general population in developed countries. Potential etiological factors have been identified, including genetic causes, environmental elements and epigenetic components. Nutrition is currently considered an influencing factor, and several studies have explored the contribution of restriction and dietary supplements in ADHD treatments. Iron is an essential cofactor required for a number of functions, such as transport of oxygen, immune function, cellular respiration, neurotransmitter metabolism (dopamine production), and DNA synthesis. Zinc is also an essential trace element, required for cellular functions related to the metabolism of neurotransmitters, melatonin, and prostaglandins. Epidemiological studies have found that iron and zinc deficiencies are common nutritional deficits worldwide, with important roles on neurologic functions (poor memory, inattentiveness, and impulsiveness), finicky appetite, and mood changes (sadness and irritability). Altered levels of iron and zinc have been related with the aggravation and progression of ADHD. Objective: This is a systematic review focused on the contribution of iron and zinc in the progression of ADHD among children and adolescents, and how therapies including these elements are tolerated along with its effectiveness (according to PRISMA guidelines). Method: The scientific literature was screened for randomized controlled trials published between January 2000 to July 2021. The databases consulted were Medline, PsycINFO, Web of Science, and Google Scholar. Two independent reviewers screened studies, extracted data, and assessed quality and risk of bias (CONSORT, NICE, and Cochrane checklists used). Conclusion: Nine studies met the eligibility criteria and were selected. Evidence was obtained regarding the contribution of iron-zinc supplementation in the treatment of ADHD among young individuals. The discussion was focused on how the deficits of these elements contribute to affectation on multiple ADHD correlates, and potential mechanisms explaining the mediational pathways. Evidence also suggested that treating ADHD with diet interventions might be particularly useful for specific subgroups of children and adolescents, but further investigations of the effects of these diet interventions are needed.
Collapse
|
28
|
Plaza-Diaz J, Flores-Rojas K, de la Torre-Aguilar MJ, Gomez-Fernández AR, Martín-Borreguero P, Perez-Navero JL, Gil A, Gil-Campos M. Dietary Patterns, Eating Behavior, and Nutrient Intakes of Spanish Preschool Children with Autism Spectrum Disorders. Nutrients 2021; 13:3551. [PMID: 34684552 PMCID: PMC8541028 DOI: 10.3390/nu13103551] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/11/2022] Open
Abstract
Eating behavior problems are characteristic of children with autism spectrum disorders (ASD) with a highly restricted range of food choices, which may pose an associated risk of nutritional problems. Hence, detailed knowledge of the dietary patterns (DPs) and nutrient intakes of ASD patients is necessary to carry out intervention strategies if required. The present study aimed to determine the DPs and macro-and micronutrient intakes in a sample of Spanish preschool children with ASD compared to typically developing control children. Fifty-four children with ASD (two to six years of age) diagnosed with ASD according to the Diagnostic Manual-5 criteria), and a control group of 57 typically developing children of similar ages were recruited. A validated food frequency questionnaire was used, and the intake of energy and nutrients was estimated through three non-consecutive 24-h dietary registrations. DPs were assessed using principal component analysis and hierarchical clustering analysis. Children with ASD exhibited a DP characterized by high energy and fat intakes and a low intake of vegetables and fruits. Likewise, meat intake of any type, both lean and fatty, was associated with higher consumption of fish and dietary fat. Furthermore, the increased consumption of dairy products was associated with increased consumption of cereals and pasta. In addition, they had frequent consumption of manufactured products with poor nutritional quality, e.g., beverages, sweets, snacks and bakery products. The percentages of children with ASD complying with the adequacy of nutrient intakes were higher for energy, saturated fat, calcium, and vitamin C, and lower for iron, iodine, and vitamins of group B when compared with control children. In conclusion, this study emphasizes the need to assess the DPs and nutrient intakes of children with ASD to correct their alterations and discard some potential nutritional diseases.
Collapse
Affiliation(s)
- Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain; (J.P.-D.); (A.G.)
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Katherine Flores-Rojas
- Pediatric Research and Metabolism Unit, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Av. Menéndez Pidal, s/n, 14010 Córdoba, Spain; (K.F.-R.); (A.R.G.-F.); (J.L.P.-N.); (M.G.-C.)
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - María José de la Torre-Aguilar
- Pediatric Research and Metabolism Unit, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Av. Menéndez Pidal, s/n, 14010 Córdoba, Spain; (K.F.-R.); (A.R.G.-F.); (J.L.P.-N.); (M.G.-C.)
| | - Antonio Rafael Gomez-Fernández
- Pediatric Research and Metabolism Unit, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Av. Menéndez Pidal, s/n, 14010 Córdoba, Spain; (K.F.-R.); (A.R.G.-F.); (J.L.P.-N.); (M.G.-C.)
| | - Pilar Martín-Borreguero
- Department of Child and Adolescent Clinical Psychiatry and Psychology, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), Reina Sofia University Hospital, Av. Menéndez Pidal, s/n, 14010 Córdoba, Spain;
| | - Juan Luis Perez-Navero
- Pediatric Research and Metabolism Unit, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Av. Menéndez Pidal, s/n, 14010 Córdoba, Spain; (K.F.-R.); (A.R.G.-F.); (J.L.P.-N.); (M.G.-C.)
- Centre for Biomedical Research on Rare Diseases (CIBERER), ISCIII, 28029 Madrid, Spain
| | - Angel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain; (J.P.-D.); (A.G.)
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Biomedical Research Center, Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Parque Tecnológico de la Salud, Avenida del Conocimiento, s/n, 18016 Granada, Spain
| | - Mercedes Gil-Campos
- Pediatric Research and Metabolism Unit, Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, Av. Menéndez Pidal, s/n, 14010 Córdoba, Spain; (K.F.-R.); (A.R.G.-F.); (J.L.P.-N.); (M.G.-C.)
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
29
|
Morandini HAE, Rao P, Hood SD, Griffiths K, Silk TJ, Zepf FD. Effects of dietary omega-3 intake on vigilant attention and resting-state functional connectivity in neurotypical children and adolescents. Nutr Neurosci 2021; 25:2269-2278. [PMID: 34369315 DOI: 10.1080/1028415x.2021.1955434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Vigilant Attention (VA) is a critical cognitive function allowing to maintain our attention, particularly in redundant or intellectually unchallenging situations. Evidence has shown that, as the brain develops, VA abilities rapidly improve throughout childhood and adolescence. Dietary omega-3 polyunsaturated fats (PUFA), playing a critical role for proper brain development and maturation of cortical regions, may contribute to variations in VA abilities. OBJECTIVE The present study investigated the effect of dietary omega-3 PUFA intake (docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA)) on resting-state functional connectivity (rsFC) of a meta-analytically defined VA network in 24 neurotypical children and adolescents (7.3-17.2 years) from the Healthy Brain Network databank. METHODS Functional MRI and phenotypical information were collected from the Healthy Brain Network databank. Intake of omega-3 DHA and EPA was assessed using a food frequency questionnaire and was adjusted for total calorie intake. Out of scanner VA-related performance was assessed using the VA condition of the Adaptive Cognitive Evaluation tool. RESULTS Overall, reported intake of omega-3 PUFA was not significantly associated with VA-related performance. Furthermore, energy-adjusted omega-3 intake was not significantly correlated with rsFC within the VA network. A complementary whole-brain analysis revealed that energy-adjusted omega-3 intake was correlated with decreased rsFC between parieto-occipital brain regions. CONCLUSION The present study was not able to detect a relationship between dietary omega-3 and rsFC or VA performance.
Collapse
Affiliation(s)
- Hugo A E Morandini
- Centre & Discipline of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia.,Division of Psychiatry, UWA Medical School, Faculty of Health & Medical Sciences, The University of Western Australia, Perth, Australia
| | - Pradeep Rao
- Centre & Discipline of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia.,Telethon Kids Institute, Perth, Australia.,Child and Adolescent Mental Health Service, Child and Adolescent Health Service, Perth, Australia
| | - Sean D Hood
- Division of Psychiatry, UWA Medical School, Faculty of Health & Medical Sciences, The University of Western Australia, Perth, Australia
| | - Kristi Griffiths
- The Brain Dynamics Centre, Westmead Institute for Medical Research, The University of Sydney, Westmead, Australia
| | - Timothy J Silk
- School of Psychology, Deakin University, Geelong, Australia.,Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Florian D Zepf
- Centre & Discipline of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Australia.,Telethon Kids Institute, Perth, Australia.,Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
30
|
Squarcina L, Nosari G, Marin R, Castellani U, Bellani M, Bonivento C, Fabbro F, Molteni M, Brambilla P. Automatic classification of autism spectrum disorder in children using cortical thickness and support vector machine. Brain Behav 2021; 11:e2238. [PMID: 34264004 PMCID: PMC8413814 DOI: 10.1002/brb3.2238] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 05/10/2021] [Accepted: 05/23/2021] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Autism spectrum disorder (ASD) is a neurodevelopmental condition with a heterogeneous phenotype. The role of biomarkers in ASD diagnosis has been highlighted; cortical thickness has proved to be involved in the etiopathogenesis of ASD core symptoms. We apply support vector machine, a supervised machine learning method, in order to identify specific cortical thickness alterations in ASD subjects. METHODS A sample of 76 subjects (9.5 ± 3.4 years old) has been selected, 40 diagnosed with ASD and 36 typically developed subjects. All children underwent a magnetic resonance imaging (MRI) examination; T1-MPRAGE sequences were analyzed to extract features for the characterization and parcellation of regions of interests (ROI); average cortical thickness (CT) has been measured for each ROI. For the classification process, the extracted features were used as input for a classifier to identify ASD subjects through a "learning by example" procedure; the features with best performance was then selected by "greedy forward-feature selection." Finally, this model underwent a leave-one-out cross-validation approach. RESULTS From the training set of 68 ROIs, five ROIs reached accuracies of over 70%. After this phase, we used a recursive feature selection process in order to identify the eight features with the best accuracy (84.2%). CT resulted higher in ASD compared to controls in all the ROIs identified at the end of the process. CONCLUSION We found increased CT in various brain regions in ASD subjects, confirming their role in the pathogenesis of this condition. Considering the brain development curve during ages, these changes in CT may normalize during development. Further validation on a larger sample is required.
Collapse
Affiliation(s)
- Letizia Squarcina
- Department of Pathophysiology and TransplantationUniversity of MilanVia Festa del Perdono, 7, 20122 MilanItaly
| | - Guido Nosari
- Department of Pathophysiology and TransplantationUniversity of MilanVia Festa del Perdono, 7, 20122 MilanItaly
| | - Riccardo Marin
- Department of InformaticsUniversity of VeronaVeronaItaly
| | | | - Marcella Bellani
- Department of NeurosciencesBiomedicine and Movement SciencesSection of PsychiatryUniversity of VeronaVeronaItaly
| | - Carolina Bonivento
- IRCCS “E. Medea”, Polo Friuli Venezia GiuliaSan Vito al Tagliamento (PN)Italy
| | | | - Massimo Molteni
- IRCCS “E. Medea”, Polo Friuli Venezia GiuliaSan Vito al Tagliamento (PN)Italy
| | - Paolo Brambilla
- Department of Pathophysiology and TransplantationUniversity of MilanVia Festa del Perdono, 7, 20122 MilanItaly
- Department of Neurosciences and Mental Health Department of Neurosciences and Mental HealthFondazione IRCCS Ca' Granda Ospedale Maggiore Policlinicovia Francesco Sforza 28, 20122 MilanItaly
| |
Collapse
|
31
|
Pecoraro L, Dalle Carbonare L, Castagnoli R, Marseglia GL, Piacentini G, Pietrobelli A. IgE-mediated fish allergy in children: is omega-3 supplementation useful? Int J Food Sci Nutr 2021; 73:154-157. [PMID: 34311650 DOI: 10.1080/09637486.2021.1957782] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The management of fish allergy relies on the elimination of all fish from the diet. Nevertheless, an exclusion diet can be problematic from a paediatric nutritional perspective. The issue of a substitute diet for children suffering from fish allergy seems to be not adequately addressed and the consequences of a fish exclusion diet in paediatric age are not known. Fish has an important nutritional value, it is rich in vitamins of group B, D and A, selenium, calcium and phosphorus, iron, zinc, magnesium, iodine and omega-3. While vitamins and iodine are normally present in the diet, omega-3 is present in few other foods, such as vegetable seed oils and nuts. Hence, the scientific research indicates a generic advice regarding a possible omega-3 supplementation in children with fish allergy. Given the knowledge about omega-3 supplementation having a potential good risk-benefit ratio and the absence of serious adverse events related to the omega-3 supplementation, this type of supplementation may seem advisable in children affected by fish allergy.
Collapse
Affiliation(s)
- Luca Pecoraro
- Department of Medicine, University of Verona, Verona, Italy.,Paediatric Clinic, ASST Mantua, Mantua, Italy
| | | | - Riccardo Castagnoli
- Department of Paediatrics, Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy.,Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy
| | - Gian Luigi Marseglia
- Department of Paediatrics, Foundation IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy.,Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy
| | - Giorgio Piacentini
- Department of Surgical Sciences, Dentistry, Gynaecology and Paediatrics, Paediatric Unit, University of Verona, Verona, Italy
| | - Angelo Pietrobelli
- Department of Surgical Sciences, Dentistry, Gynaecology and Paediatrics, Paediatric Unit, University of Verona, Verona, Italy.,Pennington Biomedical Research Centre, Baton Rouge, LA, USA
| |
Collapse
|
32
|
Bozzatello P, Blua C, Rocca P, Bellino S. Mental Health in Childhood and Adolescence: The Role of Polyunsaturated Fatty Acids. Biomedicines 2021; 9:850. [PMID: 34440053 PMCID: PMC8389598 DOI: 10.3390/biomedicines9080850] [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: 07/07/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/04/2023] Open
Abstract
There is increasing awareness of the importance of polyunsaturated fatty acids (PUFAs) for optimal brain development and function. In recent decades, researchers have confirmed the central role of PUFAs in a variety of patho-physiological processes. These agents modulate the mechanisms of brain cell signalling including the dopaminergic and serotonergic pathways. Therefore, nutritional insufficiencies of PUFAs may have adverse effects on brain development and developmental outcomes. The role of n-3 PUFAs has been studied in several psychiatric disorders in adulthood: schizophrenia, major depression, bipolar disorder, anxiety disorders, obsessive-compulsive disorder, post-traumatic stress disorder, attention deficit hyperactivity disorder (ADHD), autism spectrum disorders, eating disorders, substance use disorder, and borderline personality disorder. In contrast to the great number of studies conducted in adults, there are only limited data on the effects of n-3 PUFA supplementation in children and adolescents who suffer from mental disorders or show a high risk of developing psychiatric disorders. The aim of this review is to provide a complete and updated account of the available evidence of the impact of polyunsaturated fatty acids on developmental psychopathology in children and adolescents and the effect of fatty acid supplementation during developmental milestones, particularly in high-risk populations of children with minimal but detectable signs or symptoms of mental disorders.
Collapse
Affiliation(s)
| | | | | | - Silvio Bellino
- Department of Neuroscience, University of Turin, 10126 Turin, Italy; (P.B.); (C.B.); (P.R.)
| |
Collapse
|
33
|
Döpfner M, Dose C, Breuer D, Heintz S, Schiffhauer S, Banaschewski T. Efficacy of Omega-3/Omega-6 Fatty Acids in Preschool Children at Risk of ADHD: A Randomized Placebo-Controlled Trial. J Atten Disord 2021; 25:1096-1106. [PMID: 31680604 DOI: 10.1177/1087054719883023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: To evaluate the efficacy of an Omega-3/Omega-6 fatty acid supplement in preschool children at risk for ADHD. Method: Forty preschool children with elevated levels of ADHD symptoms were randomly assigned to either a verum or a placebo group. Children in the verum group received a 4-month treatment with Omega-3/Omega-6 fatty acids. Outcome measures comprised parent- and teacher-rated ADHD symptoms, which were the primary outcome variables, internalizing and externalizing problems, and intellectual abilities. Results: Intention-to-treat analyses of covariance, controlling for age and baseline data, revealed effects on parent- and teacher-rated ADHD symptoms (primary outcomes; parent ratings: F = 4.51, df = 1, p = .04, d = 0.63; teacher ratings: F = 4.67, df = 1, p = .04, d = 0.70), parent-rated internalizing symptoms (F = 8.47, df = 1, p < .01, d = 0.63), and parent- and teacher-rated externalizing symptoms (parent ratings: F = 4.58, df = 1, p = .04, d = 0.54; teacher ratings: F = 5.99, df = 1, p = .02, d = 0.79). Analyses involving only cases with available data yielded significant moderate effects on teacher-rated inattention symptoms (F = 4.60, df = 1, p = .04, d = 0.79) and parent-rated internalizing problems (F = 6.04, df = 1, p = .02, d = 0.57). Conclusion: The intention-to-treat analyses provide some evidence for positive effects of Omega-3/Omega-6 fatty acids. However, the results require replication in larger samples to allow for firm conclusions for practice.
Collapse
Affiliation(s)
- Manfred Döpfner
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Medical Faculty, University of Cologne, Germany.,School for Child and Adolescent Cognitive Behavior Therapy (AKiP), Medical Faculty, University of Cologne, Germany
| | - Christina Dose
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Medical Faculty, University of Cologne, Germany.,School for Child and Adolescent Cognitive Behavior Therapy (AKiP), Medical Faculty, University of Cologne, Germany
| | - Dieter Breuer
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Medical Faculty, University of Cologne, Germany.,School for Child and Adolescent Cognitive Behavior Therapy (AKiP), Medical Faculty, University of Cologne, Germany
| | - Stefan Heintz
- Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Stefanie Schiffhauer
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Medical Faculty, University of Cologne, Germany.,School for Child and Adolescent Cognitive Behavior Therapy (AKiP), Medical Faculty, University of Cologne, Germany
| | - Tobias Banaschewski
- Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
34
|
Poly-Unsaturated Fatty Acids in ADHD and in Other Neuropsychiatric Conditions: A Multiple Case Presentation. Pediatr Rep 2021; 13:234-240. [PMID: 34066347 PMCID: PMC8162565 DOI: 10.3390/pediatric13020032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/29/2021] [Accepted: 04/29/2021] [Indexed: 12/03/2022] Open
Abstract
Neurodevelopmental disorders are seen quite commonly by general pediatricians. They should be managed with a multi-professional approach. The potential beneficial effect of poly-unsaturated fatty acids (PUFAs) has been reported in recent literature, but guidelines describing their use in everyday practice are still lacking. We describe four cases as examples of the possible integration of a supplementation with PUFAs in the management of four relatively common clinical situations (i.e., children too young to receive pharmacological treatment for ADHD, children with nonspecific neurodevelopmental disorders, children whose parents refuse consent for pharmacological treatment of ADHD, and children for whom methylphenidate is not sufficient to achieve expected results).
Collapse
|
35
|
Watkins DJ, Meeker JD, Tamayo-Ortiz M, Sánchez BN, Schnaas L, Peterson KE, Téllez-Rojo MM. Gestational and peripubertal phthalate exposure in relation to attention performance in childhood and adolescence. ENVIRONMENTAL RESEARCH 2021; 196:110911. [PMID: 33640497 PMCID: PMC8119354 DOI: 10.1016/j.envres.2021.110911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 06/12/2023]
Abstract
The prevalence of Attention Deficit/Hyperactivity Disorder (ADHD) has been increasing. Research suggests that exposure to endocrine disrupting chemicals such as phthalates may play a role, but studies of in utero phthalate exposure and ADHD-related symptoms beyond early childhood are limited. We investigated associations between measures of in utero phthalate exposure and ADHD symptoms, such as inattention and impulsivity, in childhood (age 6-11 years, n = 221) and in adolescence (age 9-18 years, n = 200), as well as cross-sectional relationships between phthalate exposure and ADHD symptoms in adolescence (n = 491) among participants in the Early Life Exposure in Mexico to Environmental Toxicants (ELEMENT) cohort. Women provided urine samples up to three times during pregnancy and adolescents provided a urine sample at 9-18 years of age for phthalate metabolite measurement. We administered the Conners' Continuous Performance Test (CPT) when children were age 6-11 years and again at 9-18 years of age. We used multivariable linear regression to examine associations between the geometric mean of phthalate metabolite levels across pregnancy and CPT scores in childhood or adolescence separately, adjusting for age, years schooling (at 9-18 only), maternal education, and specific gravity. Although average in utero phthalate concentrations were not associated with CPT scores in childhood, interquartile range (IQR) increases of in utero MBzP, MCPP, and MBP were associated with 4.2%, 4.7%, and 4.5% (p < 0.05) higher Omissions scores in adolescence, respectively, indicating higher inattention. In utero MiBP levels were also associated with higher Inter-Stimulus Interval (ISI) and Variability scores (5.4% and 5.5% per IQR, p < 0.05) in adolescence. In addition, urinary DEHP metabolite levels during adolescence were cross-sectionally associated with poorer scores on several CPT indices indicating greater inattention. These findings suggest that in utero phthalate exposure may have adverse effects on attention, but these effects may not appear until adolescence, a period of extensive neurodevelopment. Future research investigating the long-term effects of in utero phthalate exposure on attention and ADHD in adolescence, as well as identification of potential mechanisms involved, is needed.
Collapse
Affiliation(s)
- Deborah J Watkins
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA.
| | - John D Meeker
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Marcela Tamayo-Ortiz
- National Council of Science and Technology, National Institute of Public Health, Mexico City, Mexico
| | - Brisa N Sánchez
- Department of Epidemiology and Biostatistics, Drexel Dornsife School of Public Health, Philadelphia, PA, USA
| | - Lourdes Schnaas
- National Institute of Perinatology, ABC Hospital, Mexico City, Mexico
| | - Karen E Peterson
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA; Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Martha María Téllez-Rojo
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Mexico
| |
Collapse
|
36
|
Casquero-Veiga M, Romero-Miguel D, MacDowell KS, Torres-Sanchez S, Garcia-Partida JA, Lamanna-Rama N, Gómez-Rangel V, Romero-Miranda A, Berrocoso E, Leza JC, Arango C, Desco M, Soto-Montenegro ML. Omega-3 fatty acids during adolescence prevent schizophrenia-related behavioural deficits: Neurophysiological evidences from the prenatal viral infection with PolyI:C. Eur Neuropsychopharmacol 2021; 46:14-27. [PMID: 33735708 DOI: 10.1016/j.euroneuro.2021.02.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 01/11/2021] [Accepted: 02/09/2021] [Indexed: 12/20/2022]
Abstract
The likely involvement of inflammation and oxidative stress (IOS) in mental disease has led to advocate anti-oxidant and anti-inflammatory drugs as therapeutic strategies in the treatment of schizophrenia. Since omega-3 fatty acids (ω-3) show anti-inflammatory/neuroprotective properties, we aim to evaluate whether ω-3 treatment during adolescence in the maternal immune stimulation (MIS) animal model of schizophrenia could prevent the brain and behavioural deficits described in adulthood. At gestational day 15, PolyI:C (4 mg/kg) or saline (VH) were injected to pregnant Wistar rats. Male offspring received ω-3 (800 mg/kg) or saline (Sal) daily from postnatal day (PND) 35-49, defining 4 groups: MIS-ω-3; MIS-Sal; VH-ω-3 and VH-Sal. At PND70, rats were submitted to prepulse inhibition test (PPI). FDG-PET and T2-weighted MRI brain studies were performed in adulthood and analyzed by means of SPM12. IOS markers were measured in selected brain areas. MIS-offspring showed a PPI deficit compared with VH-offspring and ω-3 treatment prevented this deficit. Also, ω-3 reduced the brain metabolism in the deep mesencephalic area and prevented the volumetric abnormalities in the hippocampus but not in the ventricles in MIS-offspring. Besides, ω-3 reduced the expression of iNOS and Keap1 and increased the activity/concentration of HO1, NQO1 and GPX. Our study demonstrates that administration of ω-3 during adolescence prevents PPI behavioural deficits and hippocampal volumetric abnormalities, and partially counteracts IOS deficits via iNOS and Nrf2-ARE pathways in the MIS model. This study highlights the need for novel strategies based on anti-inflammatory/anti-oxidant compounds to alter the disease course in high-risk populations at early stages.
Collapse
Affiliation(s)
- Marta Casquero-Veiga
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| | | | - Karina S MacDowell
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Department of Pharmacology & Toxicology, School of Medicine, University Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Sonia Torres-Sanchez
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Neuropsychopharmacology & Psychobiology Research Group, Psychobiology Area, Department of Psychology, University of Cádiz, Puerto Real (Cádiz), Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Jose Antonio Garcia-Partida
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain; Neuropsychopharmacology & Psychobiology Research Group, Department of Neuroscience, University of Cádiz, Cádiz, Spain
| | | | | | | | - Esther Berrocoso
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Neuropsychopharmacology & Psychobiology Research Group, Psychobiology Area, Department of Psychology, University of Cádiz, Puerto Real (Cádiz), Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Juan C Leza
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Department of Pharmacology & Toxicology, School of Medicine, University Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Celso Arango
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Departamento of Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain; Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain.
| | - María Luisa Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain.
| |
Collapse
|
37
|
de Andrade Wobido K, de Sá Barreto da Cunha M, Miranda SS, da Mota Santana J, da Silva DCG, Pereira M. Non-specific effect of omega-3 fatty acid supplementation on autistic spectrum disorder: systematic review and meta-analysis. Nutr Neurosci 2021; 25:1995-2007. [PMID: 33871323 DOI: 10.1080/1028415x.2021.1913950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
METHODS We searched seven databases and found 13 eligible controlled trials that use omega-3 supplementation in children and adolescents with ASD.Data extraction: We collected details on study design, intervention time, supplement dosage, and the autism assessment scale. Meta-analyses and subgroup analysis were conducted according to the autism symptoms. RESULTS Omega-3 and omega-6 supplementation improved ASD symptoms according to the Aberrant Behavior Checklist (standard mean difference - SMD = -0.13; CI 95% = -0.34, -0.02). However, using subgroup analysis, we observed no efficacy in terms of improvements in hyperactivity (SMD = -0.03; CI 95%: -0.43, 0.36), irritability (SMD = -0.18; CI 95%: -0.51, 0.15), stereotypy (SMD = -0.03; CI 95%: -0.43, 0.36), inappropriate speech (SMD = -0.68; CI 95%: -1.49, 0.14), lethargy (SMD = -0.22; CI 95%: -0.58, 0.14), and social function (SMD = -0.71; IC 95%: -1.56, 0.14). W-3 and w-6 supplementation also showed no efficacy according to the Social Responsiveness Scale (SMD = 0.08; CI 95%: -0.23, 0.39). The adverse effects were classified as mild and equally distributed between the placebo and intervention groups. CONCLUSIONS Despite w-3 and w-6 supplementation showing minimal beneficial effects in the treatment of autism, the subgroup analyses indicated that there is a lack of evidence on the beneficial role of w-3 and w-6 in treating ASD.Systematic Review Registration: PROSPERO number CRD42020146116.
Collapse
Affiliation(s)
- Kelma de Andrade Wobido
- Center for Biological and Health Sciences, Federal University of Western Bahia, Barreiras, Brazil
| | | | | | - Jerusa da Mota Santana
- Health Sciences Center, Federal University of Recôncavo da Bahia, Santo Antônio de Jesus, Bahia
| | | | - Marcos Pereira
- Institute of Collective Health, Federal University of Bahia, Salvador, Bahia
| |
Collapse
|
38
|
Esposito CM, Buoli M, Ciappolino V, Agostoni C, Brambilla P. The Role of Cholesterol and Fatty Acids in the Etiology and Diagnosis of Autism Spectrum Disorders. Int J Mol Sci 2021; 22:ijms22073550. [PMID: 33805572 PMCID: PMC8036564 DOI: 10.3390/ijms22073550] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/17/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Autism spectrum disorders (ASDs) are a group of neurodevelopmental disorders whose pathogenesis seems to be related to an imbalance of excitatory and inhibitory synapses, which leads to disrupted connectivity during brain development. Among the various biomarkers that have been evaluated in the last years, metabolic factors represent a bridge between genetic vulnerability and environmental aspects. In particular, cholesterol homeostasis and circulating fatty acids seem to be involved in the pathogenesis of ASDs, both through the contribute in the stabilization of cell membranes and the modulation of inflammatory factors. The purpose of the present review is to summarize the available data about the role of cholesterol and fatty acids, mainly long-chain ones, in the onset of ASDs. A bibliographic research on the main databases was performed and 36 studies were included in our review. Most of the studies document a correlation between ASDs and hypocholesterolemia, while the results concerning circulating fatty acids are less univocal. Even though further studies are necessary to confirm the available data, the metabolic biomarkers open to new treatment options such as the modulation of the lipid pattern through the diet.
Collapse
Affiliation(s)
- Cecilia Maria Esposito
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milan, Italy; (C.M.E.); (M.B.); (V.C.); (P.B.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Massimiliano Buoli
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milan, Italy; (C.M.E.); (M.B.); (V.C.); (P.B.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Valentina Ciappolino
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milan, Italy; (C.M.E.); (M.B.); (V.C.); (P.B.)
| | - Carlo Agostoni
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
- Pediatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Correspondence:
| | - Paolo Brambilla
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milan, Italy; (C.M.E.); (M.B.); (V.C.); (P.B.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| |
Collapse
|
39
|
Sun GY, Appenteng MK, Li R, Woo T, Yang B, Qin C, Pan M, Cieślik M, Cui J, Fritsche KL, Gu Z, Will M, Beversdorf D, Adamczyk A, Han X, Greenlief CM. Docosahexaenoic Acid (DHA) Supplementation Alters Phospholipid Species and Lipid Peroxidation Products in Adult Mouse Brain, Heart, and Plasma. Neuromolecular Med 2021; 23:118-129. [PMID: 32926329 PMCID: PMC9555299 DOI: 10.1007/s12017-020-08616-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/07/2020] [Indexed: 10/23/2022]
Abstract
The abundance of docosahexaenoic acid (DHA) in phospholipids in the brain and retina has generated interest to search for its role in mediating neurological functions. Besides the source of many oxylipins with pro-resolving properties, DHA also undergoes peroxidation, producing 4-hydroxyhexenal (4-HHE), although its function remains elusive. Despite wide dietary consumption, whether supplementation of DHA may alter the peroxidation products and their relationship to phospholipid species in brain and other body organs have not been explored sufficiently. In this study, adult mice were administered a control or DHA-enriched diet for 3 weeks, and phospholipid species and peroxidation products were examined in brain, heart, and plasma. Results demonstrated that this dietary regimen increased (n-3) and decreased (n-6) species to different extent in all major phospholipid classes (PC, dPE, PE-pl, PI and PS) examined. Besides changes in phospholipid species, DHA-enriched diet also showed substantial increases in 4-HHE in brain, heart, and plasma. Among different brain regions, the hippocampus responded to the DHA-enriched diet showing significant increase in 4-HHE. Considering the pro- and anti-inflammatory pathways mediated by the (n-6) and (n-3) polyunsaturated fatty acids, unveiling the ability for DHA-enriched diet to alter phospholipid species and lipid peroxidation products in the brain and in different body organs may be an important step forward towards understanding the mechanism(s) for this (n-3) fatty acid on health and diseases.
Collapse
Affiliation(s)
- Grace Y Sun
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Michael K Appenteng
- Department of Chemistry, University of Missouri, 125 Chemistry Bldg., Columbia, MO, 65211, USA
| | - Runting Li
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Taeseon Woo
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, 65211, USA
| | - Bo Yang
- Department of Chemistry, University of Missouri, 125 Chemistry Bldg., Columbia, MO, 65211, USA
| | - Chao Qin
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science & Center at San Antonio, San Antonio, TX, 78229, USA
| | - Meixia Pan
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science & Center at San Antonio, San Antonio, TX, 78229, USA
| | - Magdalena Cieślik
- Department of Cellular Signaling, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Jiankun Cui
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Kevin L Fritsche
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, 65211, USA
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Matthew Will
- Department of Psychological Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - David Beversdorf
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, 65211, USA
- Departments of Radiology, Neurology and Psychological Sciences, and the Thompson Center, University of Missouri, Columbia, MO, 65211, USA
| | - Agata Adamczyk
- Department of Cellular Signaling, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science & Center at San Antonio, San Antonio, TX, 78229, USA
| | - C Michael Greenlief
- Department of Chemistry, University of Missouri, 125 Chemistry Bldg., Columbia, MO, 65211, USA.
| |
Collapse
|
40
|
Lyall K, Windham GC, Snyder NW, Kuskovsky R, Xu P, Bostwick A, Robinson L, Newschaffer CJ. Association Between Midpregnancy Polyunsaturated Fatty Acid Levels and Offspring Autism Spectrum Disorder in a California Population-Based Case-Control Study. Am J Epidemiol 2021; 190:265-276. [PMID: 33524118 DOI: 10.1093/aje/kwaa171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022] Open
Abstract
Polyunsaturated fatty acids (PUFAs) are critical for brain development and have been linked with neurodevelopmental outcomes. We conducted a population-based case-control study in California to examine the association between PUFAs measured in midpregnancy serum samples and autism spectrum disorder (ASD) in offspring. ASD cases (n = 499) were identified through the California Department of Developmental Services and matched to live-birth population controls (n = 502) on birth month, year (2010 or 2011), and sex. Logistic regression models were used to examine crude and adjusted associations. In secondary analyses, we examined ASD with and without co-occurring intellectual disability (ID; n = 67 and n = 432, respectively) and effect modification by sex and ethnicity. No clear patterns emerged, though there was a modest inverse association with the top quartile of linoleic acid level (highest quartile vs. lowest: adjusted odds ratio = 0.74, 95% confidence interval: 0.49, 1.11; P for trend = 0.10). Lower levels of total and ω-3 PUFAs were associated with ASD with ID (lowest decile of total PUFAs vs. deciles 4-7: adjusted odds ratio = 2.78, 95% confidence interval: 1.13, 6.82) but not ASD without ID. We did not observe evidence of effect modification by the factors examined. These findings do not suggest a strong association between midpregnancy PUFA levels and ASD. In further work, researchers should consider associations with ASD with ID and in other time windows.
Collapse
|
41
|
El-Ansary A, Chirumbolo S, Bhat RS, Dadar M, Ibrahim EM, Bjørklund G. The Role of Lipidomics in Autism Spectrum Disorder. Mol Diagn Ther 2021; 24:31-48. [PMID: 31691195 DOI: 10.1007/s40291-019-00430-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental syndrome commonly diagnosed in early childhood; it is usually characterized by impairment in reciprocal communication and speech, repetitive behaviors, and social withdrawal with loss in communication skills. Its development may be affected by a variety of environmental and genetic factors. Trained physicians diagnose and evaluate the severity of ASD based on clinical evaluations of observed behaviors. As such, this approach is inevitably dependent on the expertise and subjective assessment of those administering the clinical evaluations. There is a need to identify objective biological markers associated with diagnosis or clinical severity of the disorder. Several important issues and concerns exist regarding the diagnostic competence of the many abnormal plasma metabolites produced in the different biochemical pathways evaluated in individuals with ASD. The search for high-performing bio-analytes to diagnose and follow-up ASD development is still a major target in medicine. Dysregulation in the oxidative stress response and proinflammatory processes are major etiological causes of ASD pathogenesis. Furthermore, dicarboxylic acid metabolites, cholesterol-related metabolites, phospholipid-related metabolites, and lipid transporters and mediators are impaired in different pathological conditions that have a role in the ASD etiology. A mechanism may exist by which pro-oxidant environmental stressors and abnormal metabolites regulate clinical manifestations and development of ASD.
Collapse
Affiliation(s)
- Afaf El-Ansary
- Central Laboratory, Female Centre for Scientific and Medical Studies, King Saud University, Riyadh, Saudi Arabia.,Autism Research and Treatment Center, Riyadh, Saudi Arabia.,CONEM Saudi Autism Research Group, King Saud University, Riyadh, Saudi Arabia.,Therapeutic Chemistry Department, National Research Centre, Giza, Egypt
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.,CONEM Scientific Secretary, Verona, Italy
| | - Ramesa Shafi Bhat
- Biochemistry Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Eiman M Ibrahim
- Central Laboratory, Female Centre for Scientific and Medical Studies, King Saud University, Riyadh, Saudi Arabia
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Toften 24, 8610, Mo i Rana, Norway.
| |
Collapse
|
42
|
Current Evidence on the Role of the Gut Microbiome in ADHD Pathophysiology and Therapeutic Implications. Nutrients 2021; 13:nu13010249. [PMID: 33467150 PMCID: PMC7830868 DOI: 10.3390/nu13010249] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/14/2022] Open
Abstract
Studies suggest that the bidirectional relationship existent between the gut microbiome (GM) and the central nervous system (CNS), or so-called the microbiome–gut–brain axis (MGBA), is involved in diverse neuropsychiatric diseases in children and adults. In pediatric age, most studies have focused on patients with autism. However, evidence of the role played by the MGBA in attention deficit/hyperactivity disorder (ADHD), the most common neurodevelopmental disorder in childhood, is still scanty and heterogeneous. This review aims to provide the current evidence on the functioning of the MGBA in pediatric patients with ADHD and the specific role of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) in this interaction, as well as the potential of the GM as a therapeutic target for ADHD. We will explore: (1) the diverse communication pathways between the GM and the CNS; (2) changes in the GM composition in children and adolescents with ADHD and association with ADHD pathophysiology; (3) influence of the GM on the ω-3 PUFA imbalance characteristically found in ADHD; (4) interaction between the GM and circadian rhythm regulation, as sleep disorders are frequently comorbid with ADHD; (5) finally, we will evaluate the most recent studies on the use of probiotics in pediatric patients with ADHD.
Collapse
|
43
|
Madireddy S, Madireddy S. Regulation of Reactive Oxygen Species-Mediated Damage in the Pathogenesis of Schizophrenia. Brain Sci 2020; 10:brainsci10100742. [PMID: 33081261 PMCID: PMC7603028 DOI: 10.3390/brainsci10100742] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/06/2020] [Accepted: 10/15/2020] [Indexed: 12/16/2022] Open
Abstract
The biochemical integrity of the brain is paramount to the function of the central nervous system, and oxidative stress is a key contributor to cerebral biochemical impairment. Oxidative stress, which occurs when an imbalance arises between the production of reactive oxygen species (ROS) and the efficacy of the antioxidant defense mechanism, is believed to play a role in the pathophysiology of various brain disorders. One such disorder, schizophrenia, not only causes lifelong disability but also induces severe emotional distress; however, because of its onset in early adolescence or adulthood and its progressive development, consuming natural antioxidant products may help regulate the pathogenesis of schizophrenia. Therefore, elucidating the functions of ROS and dietary antioxidants in the pathogenesis of schizophrenia could help formulate improved therapeutic strategies for its prevention and treatment. This review focuses specifically on the roles of ROS and oxidative damage in the pathophysiology of schizophrenia, as well as the effects of nutrition, antipsychotic use, cognitive therapies, and quality of life on patients with schizophrenia. By improving our understanding of the effects of various nutrients on schizophrenia, it may become possible to develop nutritional strategies and supplements to treat the disorder, alleviate its symptoms, and facilitate long-term recovery.
Collapse
Affiliation(s)
- Samskruthi Madireddy
- Independent Researcher, 1353 Tanaka Drive, San Jose, CA 95131, USA
- Correspondence: ; Tel.: +1-408-9214162
| | - Sahithi Madireddy
- Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA;
| |
Collapse
|
44
|
van der Wurff IS, Meyer BJ, de Groot RH. Effect of Omega-3 Long Chain Polyunsaturated Fatty Acids (n-3 LCPUFA) Supplementation on Cognition in Children and Adolescents: A Systematic Literature Review with a Focus on n-3 LCPUFA Blood Values and Dose of DHA and EPA. Nutrients 2020; 12:E3115. [PMID: 33053843 PMCID: PMC7599612 DOI: 10.3390/nu12103115] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
Omega-3 long chain polyunsaturated fatty acids (n-3 LCPUFA) supplementation in the cardiovascular field is effective if a certain Omega-3 index (O3I) is achieved or the daily n-3 LCPUFA dose is high enough. Whether this applies to studies on cognition in children and adolescents is unclear. The aims of the current review were to investigate whether: (1) a certain O3I level and (2) a minimum daily n-3 LCPUFA dose are required to improve cognition in 4-25 year olds. Web of Science and PubMed were searched. Inclusion criteria: placebo controlled randomized controlled trial; participants 4-25 years; supplementation with docosahexaenoic acid (DHA) and/or eicosapentaenoic acid (EPA); assessing cognition; in English and ≥10 participants per treatment arm. Thirty-three studies were included, 21 in typically developing participants, 12 in those with a disorder. A positive effect on cognitive measures was more likely in studies with an increase in O3I to >6%. Half of the studies in typically developing children with daily supplementation dose ≥450 mg DHA + EPA showed improved cognition. For children with a disorder no cut-off value was found. In conclusion, daily supplementation of ≥450 mg DHA + EPA per day and an increase in the O3I to >6% makes it more likely to show efficacy on cognition in children and adolescents.
Collapse
Affiliation(s)
- Inge S.M. van der Wurff
- Conditions for Lifelong Learning, Faculty of Educational Sciences, Open University of the Netherlands, 6419 Heerlen, The Netherlands;
| | - Barbara J. Meyer
- School of Medicine, Lipid Research Centre, Molecular Horizons, Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia;
| | - Renate H.M. de Groot
- Conditions for Lifelong Learning, Faculty of Educational Sciences, Open University of the Netherlands, 6419 Heerlen, The Netherlands;
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6200 Maastricht, The Netherlands
| |
Collapse
|
45
|
Recent Advances in the Pharmacological Management of Behavioral Disturbances Associated with Autism Spectrum Disorder in Children and Adolescents. Paediatr Drugs 2020; 22:473-483. [PMID: 32686015 DOI: 10.1007/s40272-020-00408-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neuropsychiatric condition affecting an estimated one in 36 children. Youth with ASD may have severe behavioral disturbances including irritability, aggression, and hyperactivity. Currently, there are only two medications (risperidone and aripiprazole) approved by the US Food and Drug Administration (FDA) for the treatment of irritability associated with ASD. Pharmacologic treatments are commonly used to target ASD-associated symptoms including irritability, mood lability, anxiety, and hyperactivity. However, evidence for the efficacy of many commonly used treatments is limited by the lack of large placebo-controlled trials of these medications in this population. Research into the pathophysiology of ASD has led to new targets for pharmacologic therapy including the neuroimmune system, the endocannabinoid system, and the glutamatergic neurotransmitter system. The goal of this review is to provide an overview of the current evidence base for commonly used treatments, as well as emerging treatment options for common behavioral disturbances seen in youth with ASD.
Collapse
|
46
|
Schiavi S, Carbone E, Melancia F, Buzzelli V, Manduca A, Campolongo P, Pallottini V, Trezza V. Perinatal supplementation with omega-3 fatty acids corrects the aberrant social and cognitive traits observed in a genetic model of autism based on FMR1 deletion in rats. Nutr Neurosci 2020; 25:898-911. [PMID: 32912100 DOI: 10.1080/1028415x.2020.1819107] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background and objective: Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder for which no treatments exist. Fragile X syndrome (FXS) is the most common form of inherited mental retardation and the most frequent monogenic cause of ASD. Given the lack of pharmacological treatments for ASD, increasing interest is devoted to non-pharmacological approaches, including dietary interventions. Omega-3 polyunsaturated fatty acids (PUFAs) are critical for neurobehavioraldevelopment. This study had two aims: 1. To validatethe recently developed Fmr1-Δexon 8 rat model of FXS; 2. To assess the impact of omega-3 PUFAs dietary supplementation during pregnancy and lactation on the altered behavior displayed by Fmr1-Δexon 8 rats.Methods: Female Fmr1-Δexon 8 and wild-type Sprague-Dawley rats were fed with either an omega-3 PUFAs enriched diet or with an isocaloric control diet during pregnancy and lactation. Behavioral experiments were carried out on the infant (Postnatal days (PNDs) 9 and 13), juvenile (PND 35) and adult (PND 90) male offspring.Results: Fmr1-Δexon 8 pups showed hypolocomotion, reduced ultrasonic vocalizations (USVs) emission and impaired social discrimination compared to wild-type controls. Juvenile and adult Fmr1-Δexon 8 rats showed deficits in the social and cognitive domains, that were counteracted by perinatal omega-3 PUFAs supplementation.Conclusion: Our results support the validity of the Fmr1-Δexon 8 rat model to mimic key autistic-like features and support an important role of omega-3 PUFAs during of neurodevelopment. Although the mechanisms underlying the beneficial effects of omega-3 PUFAs supplementation in ASD needs to be clarified, this dietary intervention holds promise to mitigate core and comorbid autistic features.
Collapse
Affiliation(s)
- Sara Schiavi
- Department of Science, University 'Roma Tre', Rome Italy
| | - Emilia Carbone
- Department of Science, University 'Roma Tre', Rome Italy
| | | | | | | | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, Santa Lucia Foundation, Rome, Italy
| | | | - Viviana Trezza
- Department of Science, University 'Roma Tre', Rome Italy
| |
Collapse
|
47
|
Bostwick A, Snyder NW, Windham GC, Whitman C, Pearl M, Robinson L, Newschaffer CJ, Lyall K. Polyunsaturated Fatty Acids in Newborn Bloodspots: Associations With Autism Spectrum Disorder and Correlation With Maternal Serum Levels. Autism Res 2020; 13:1601-1613. [PMID: 32897003 DOI: 10.1002/aur.2365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/11/2020] [Accepted: 06/26/2020] [Indexed: 11/11/2022]
Abstract
We conducted a population-based case-control study to examine newborn polyunsaturated fatty acid (PUFA) levels in association with autism spectrum disorder (ASD) and assess PUFA correlation across two time points. ASD cases (n = 200) were identified through the Department of Developmental Services and matched to live-birth population controls (n = 200) on birth month, year (2010-2011), and sex. Nonesterified PUFAs were measured by isotope dilution liquid chromatography-high resolution mass spectrometry from archived newborn dried blood spots and maternal mid-pregnancy serum samples. Crude and adjusted conditional logistic regression models were used to examine the association between neonatal PUFA levels, categorized in quartiles and according to distributional extremes, and ASD. Cubic splines were utilized to examine nonlinear relationships between continuous neonatal PUFAs and ASD. The correlation between neonatal and maternal levels was examined using Pearson correlation coefficients. In adjusted analyses of neonatal PUFA levels, no clear trends emerged, though there was an elevated odds ratio of ASD for the third quartile of linoleic acid, relative to the first (adjusted odds ratio = 2.49, 95% confidence interval: 1.31, 4.70). Cubic spline analysis suggested a nonlinear association between linoleic acid and ASD, though this was not robust to sensitivity analyses. While individual PUFAs were significantly correlated with one another within a given time point, aside from docohexaseanoic acid, PUFAs were not correlated across maternal and neonatal samples. Overall, our findings do not support an association between neonatal PUFA levels and ASD. Future work should confirm and expand these findings by examining associations with phenotypic subgroups and considering PUFAs in other time points. LAY SUMMARY: In this study, we examined whether levels of fats known as polyunsaturated fatty acids, measured in newborns, were related to later child diagnosis of autism spectrum disorder (ASD). Overall, we did not find strong evidence for hypothesized reduction in risk of ASD based on newborn levels of these fats. Future studies in larger samples and considering other time points may be useful to explain whether these fats are important in brain development related to ASD. Autism Res 2020, 13: 1601-1613. © 2020 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Anna Bostwick
- AJ Drexel Autism Institute, Drexel University, Philadelphia, Pennsylvania, USA
| | - Nathaniel W Snyder
- AJ Drexel Autism Institute, Drexel University, Philadelphia, Pennsylvania, USA
| | - Gayle C Windham
- Environmental Health Investigations Branch, California Department of Public Health, Richmond, California, USA
| | - Casey Whitman
- AJ Drexel Autism Institute, Drexel University, Philadelphia, Pennsylvania, USA
| | - Michelle Pearl
- Environmental Health Investigations Branch, California Department of Public Health, Richmond, California, USA
| | - Lucy Robinson
- Department of Epidemiology and Biostatistics, Dornsife School of Public Health, Drexel University, Philadelphia, Pennsylvania, USA
| | - Craig J Newschaffer
- AJ Drexel Autism Institute, Drexel University, Philadelphia, Pennsylvania, USA.,College of Health and Human Development, Pennsylvania State University, State College, Pennsylvania, USA
| | - Kristen Lyall
- AJ Drexel Autism Institute, Drexel University, Philadelphia, Pennsylvania, USA.,Department of Epidemiology and Biostatistics, Dornsife School of Public Health, Drexel University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
48
|
Bozzatello P, De Rosa ML, Rocca P, Bellino S. Effects of Omega 3 Fatty Acids on Main Dimensions of Psychopathology. Int J Mol Sci 2020; 21:ijms21176042. [PMID: 32839416 PMCID: PMC7504659 DOI: 10.3390/ijms21176042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/11/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
The usefulness of polyunsaturated fatty acids on inflammatory, cardiovascular, and the nervous system was studied in the last decades, but the mechanisms underlying their benefic properties are still partially unknown. These agents seem to express their action on the membrane phospholipid composition and permeability and modulation of second messenger cascades. In psychiatry, the efficacy and tolerability of omega-3 fatty acids were investigated in several psychiatric disorders, including major depression, bipolar disorder, personality disorders, high-risk conditions to develop psychosis, attention-deficit hyperactivity disorder, and autism spectrum disorders. Initial findings in this field are promising, and some relevant questions need to be addressed. In particular, the effects of these agents on the main symptom dimensions have to be investigated in a trans-diagnostic perspective. The present systematic review is aimed to examine the available data on the efficacy of omega-3 fatty acids on domains of psychotic symptoms, affective symptoms, impulsivity, and aggressiveness, and harmful behaviors, and suicide risk.
Collapse
Affiliation(s)
- Paola Bozzatello
- Department of Neuroscience, Faculty of Medicine, University of Turin, 10126 Turin, Italy; (P.B.); (M.L.D.R.); (P.R.)
- Center for Personality Disorders, Psychiatric Clinic, 10126 Turin, Italy
| | - Maria Laura De Rosa
- Department of Neuroscience, Faculty of Medicine, University of Turin, 10126 Turin, Italy; (P.B.); (M.L.D.R.); (P.R.)
- Center for Personality Disorders, Psychiatric Clinic, 10126 Turin, Italy
| | - Paola Rocca
- Department of Neuroscience, Faculty of Medicine, University of Turin, 10126 Turin, Italy; (P.B.); (M.L.D.R.); (P.R.)
| | - Silvio Bellino
- Department of Neuroscience, Faculty of Medicine, University of Turin, 10126 Turin, Italy; (P.B.); (M.L.D.R.); (P.R.)
- Center for Personality Disorders, Psychiatric Clinic, 10126 Turin, Italy
- Correspondence: ; Tel.: +39-011-6634848; Fax: +39-011-673473
| |
Collapse
|
49
|
Massari M, Novielli C, Mandò C, Di Francesco S, Della Porta M, Cazzola R, Panteghini M, Savasi V, Maggini S, Schaefer E, Cetin I. Multiple Micronutrients and Docosahexaenoic Acid Supplementation during Pregnancy: A Randomized Controlled Study. Nutrients 2020; 12:E2432. [PMID: 32823606 PMCID: PMC7468952 DOI: 10.3390/nu12082432] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/08/2020] [Accepted: 08/11/2020] [Indexed: 12/15/2022] Open
Abstract
Maternal dietary intake during pregnancy needs to meet increased nutritional demands to maintain metabolism and to support fetal development. Docosahexaenoic acid (DHA) is essential for fetal neuro-/visual development and in immunomodulation, accumulating rapidly within the developing brain and central nervous system. Levels available to the fetus are governed by the maternal diet. In this multicenter, parallel, randomized controlled trial, we evaluated once-daily supplementation with multiple micronutrients and DHA (i.e., multiple micronutrient supplementation, MMS) on maternal biomarkers and infant anthropometric parameters during the second and third trimesters of pregnancy compared with no supplementation. Primary efficacy endpoint: change in maternal red blood cell (RBC) DHA (wt% total fatty acids) during the study. Secondary variables: other biomarkers of fatty acid and oxidative status, vitamin D, and infant anthropometric parameters at delivery. Supplementation significantly increased RBC DHA levels, the omega-3 index, and vitamin D levels. Subscapular skinfold thickness was significantly greater with MMS in infants. Safety outcomes were comparable between groups. This first randomized controlled trial of supplementation with multiple micronutrients and DHA in pregnant women indicated that MMS significantly improved maternal DHA and vitamin D status in an industrialized setting-an important finding considering the essential roles of DHA and vitamin D.
Collapse
Affiliation(s)
- Maddalena Massari
- Department of Woman, Mother and Neonate, Buzzi Children Hospital, ASST Fatebenefratelli Sacco, 20154 Milan, Italy; (M.M.); (S.D.F.); (I.C.)
- Department of Woman, Mother and Neonate, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, 20157 Milan, Italy;
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | - Chiara Novielli
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | - Chiara Mandò
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | - Stefania Di Francesco
- Department of Woman, Mother and Neonate, Buzzi Children Hospital, ASST Fatebenefratelli Sacco, 20154 Milan, Italy; (M.M.); (S.D.F.); (I.C.)
| | - Matteo Della Porta
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | - Roberta Cazzola
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | - Mauro Panteghini
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | - Valeria Savasi
- Department of Woman, Mother and Neonate, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, 20157 Milan, Italy;
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| | | | | | - Irene Cetin
- Department of Woman, Mother and Neonate, Buzzi Children Hospital, ASST Fatebenefratelli Sacco, 20154 Milan, Italy; (M.M.); (S.D.F.); (I.C.)
- Department of Woman, Mother and Neonate, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, 20157 Milan, Italy;
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, 20157 Milan, Italy; (C.N.); (C.M.); (M.D.P.); (R.C.); (M.P.)
| |
Collapse
|
50
|
Pecoraro L, Tenero L, Pietrobelli A, Dalle Carbonare L, Czernin S, Widhalm K, Alvarez-Perea A, Piacentini G. Canned tuna tolerance in children with IgE-mediated fish allergy: an allergological and nutritional view. Minerva Pediatr 2020; 72:408-415. [PMID: 32686923 DOI: 10.23736/s0026-4946.20.05972-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Scientific research, diagnostic tools and clinical experience have shown that children suffering from IgE-mediated fish allergy do not need to follow a strict exclusion diet. In fact, they could tolerate some species of fish, which could be reintroduced in the diet by verifying their tolerance with an oral food challenge in a clinical setting. Consequently, it is possible to look a new insight on diagnosis and management of IgE-mediated fish allergy in children, considering the use of canned tuna in clinical settings. Authors performed a literature search through the Cochrane Library and Medline/PubMed databases. All quantitative and qualitative pediatric studies involving diagnosis and management of IgE-mediated fish allergy and the use of canned tuna in clinical settings were considered. Articles related to allergological and nutritional features of fish, and especially canned tuna, were selected. This research was conducted on May 2020. Canned tuna shows peculiar allergological and nutritional characteristics. Relating to allergy, canning process, characterized by cooking the fish under pressure for a time equal to about 7 hours, can lead a conformational change in parvalbumin, making it less allergenic. In terms of nutrition, canned tuna contains B, D and A vitamins and, above all, omega-3 fatty acids and shows a favourable and significantly sustainable nutritional profile. Lower allergenicity, adequate nutritional value and its rich availability in markets at reasonable costs, could make the use of canned tuna as a solution with an excellent risk/benefit ratio in the field of IgE-mediated fish allergy.
Collapse
Affiliation(s)
- Luca Pecoraro
- Department of Medicine, University of Verona, Verona, Italy - .,Clinic of Pediatric, ASST Mantova, Mantova, Italy -
| | - Laura Tenero
- , Division of Pediatric, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy
| | - Angelo Pietrobelli
- , Division of Pediatric, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy.,Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | - Sarah Czernin
- Division of Nutrition and Metabolism, Department of Pediatrics, Austrian Academic Institute for Clinical Nutrition, Vienna, Austria
| | - Kurt Widhalm
- Division of Nutrition and Metabolism, Department of Pediatrics, Austrian Academic Institute for Clinical Nutrition, Vienna, Austria
| | | | - Giorgio Piacentini
- , Division of Pediatric, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy
| |
Collapse
|