1
|
Luo L, Tao FB. Impact of age on cardiometabolic health in children at adiposity rebound: the role of genetic mechanisms. World J Pediatr 2025; 21:252-265. [PMID: 40097891 DOI: 10.1007/s12519-025-00893-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Identifying effective predictors early in life is crucial to enable timely prevention and intervention to improve cardiometabolic health outcomes. Adiposity rebound (AR) is an important period in early life, with earlier AR increasing the risk of cardiometabolic abnormalities. However, the role and mechanism of genetic factors in this association are unclear. Therefore, this study reviews the potential genetic mechanisms influencing the age at AR, as well as the genetic mechanisms linking earlier AR with cardiometabolic abnormalities. DATA SOURCES A comprehensive literature search was conducted in PubMed and China National Knowledge Infrastructure databases using a combination of medical subject headings terms and related keywords, including "adiposity rebound", "cardiometabolic", "obesity", "BMI trajectory", "diabetes mellitus", "dyslipidemias", "hypertension", "metabolic syndrome", "genetics", and "epigenetic". Citation tracking was performed as a supplementary search strategy. All potentially relevant articles were subsequently subjected to full-text evaluation for eligibility assessment. RESULTS Polymorphisms in the DMRT1, FTO, LEPR, and TFAP2B genes, along with obesity susceptibility, can influence the age at AR. Single-nucleotide polymorphisms associated with the age at AR are enriched in the insulin-like growth factor 1 (IGF-1) signaling pathway, which can be modulated by the LEPR and TFAP2B genes. Shared genetic mechanisms between cardiometabolic abnormalities and the age at AR are influenced by obesity-related genetic variants. These variants regulate the growth hormone (GH)/IGF-1 axis, advancing AR and leading to cardiometabolic abnormalities. Earlier AR alters adiponectin and leptin levels, further activating the GH/IGF-1 axis and creating a vicious cycle. Long-term breastfeeding can counteract the adverse effects of obesity-related genetic susceptibility on AR timing, thereby reducing the genetic risk of cardiometabolic abnormalities. CONCLUSIONS Our results support earlier AR as a marker for identifying cardiometabolic risk and screening high-risk populations at the genetic level.
Collapse
Affiliation(s)
- Ling Luo
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, China
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Anhui Medical University, Hefei, 230032, China
| | - Fang-Biao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, China.
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, 230032, China.
- Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
2
|
Kim JH, Kim SJ, Chung IH, Lim JS. Adiposity Rebound Timing in Small for Gestational Age Children Treated With Growth Hormone: Results From LG Growth Study Data. J Korean Med Sci 2025; 40:e12. [PMID: 39901524 PMCID: PMC11790395 DOI: 10.3346/jkms.2025.40.e12] [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: 04/19/2024] [Accepted: 09/30/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Adiposity rebound (AR) refers to the period during growth when the body mass index reaches its lowest point before increasing again. The timing of AR is associated with the development of obesity and puberty onset. Although studies have evaluated AR timing in Korean children, none has focused on children born small for gestational age (SGA). METHODS This study analyzed data from a multicenter observational clinical trial (LG Growth Study) to determine AR timing in children born SGA without catch-up growth (CUG) who were treated with growth hormone (GH) therapy. The study also aimed to identify factors associated with AR timing, examine the influence of AR timing on puberty onset, and assess the effectiveness of GH therapy. RESULTS A total of 151 children born SGA without CUG were included. Of them, 15% experienced AR between 4 and 5 years of age, 42% between 5 and 6 years, 27% between 6 and 7 years, and 16% after 7 years of age. A significant positive correlation was noted between the height standard deviation score at the start of treatment and AR timing. However, no significant correlation was observed between AR timing and puberty onset or the effectiveness of GH therapy. CONCLUSION This study provides insights into AR timing in prepubertal children who meet the specific SGA criteria and its relationship with growth outcomes. The findings suggest that AR in children born SGA who do not experience CUG occurs later than in the general population, with no significant relationship between AR timing and puberty onset or growth outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01604395.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Department of Pediatrics, Dongguk University Ilsan Hospital, Goyang, Korea
| | - Su Jin Kim
- Department of Pediatrics, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| | - In Hyuk Chung
- Department of Pediatrics, National Health Insurance Service Ilsan Hospital, Goyang, Korea
| | - Jung Sub Lim
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul, Korea.
| |
Collapse
|
3
|
Leroy A, Gupta V, Tint MT, Ooi DSQ, Yap F, Lek N, Godfrey KM, Chong YS, Lee YS, Eriksson JG, Álvarez MA, Michael N, Wang D. Prospective prediction of childhood body mass index trajectories using multi-task Gaussian processes. Int J Obes (Lond) 2025; 49:340-347. [PMID: 39548218 PMCID: PMC11805709 DOI: 10.1038/s41366-024-01679-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Body mass index (BMI) trajectories have been used to assess the growth of children with respect to their peers, and to anticipate future obesity and disease risk. While retrospective BMI trajectories have been actively studied, models to prospectively predict continuous BMI trajectories have not been investigated. MATERIALS AND METHODS Using longitudinal BMI measurements between birth and age 10 y from a mother-offspring cohort, we leveraged a multi-task Gaussian process approach to develop and evaluate a unified framework for modeling, clustering, and prospective prediction of BMI trajectories. We compared its sensitivity to missing values in the longitudinal follow-up of children, compared its prediction performance to cubic B-spline and multilevel Jenss-Bayley models, and used prospectively predicted BMI trajectories to assess the probability of future BMIs crossing the clinical cutoffs for obesity. RESULTS MagmaClust identified 5 distinct patterns of BMI trajectories between 0 to 10 y. The method outperformed both cubic B-spline and multilevel Jenss-Bayley models in the accuracy of retrospective BMI trajectories while being more robust to missing data (up to 90%). It was also better at prospectively forecasting BMI trajectories of children for periods ranging from 2 to 8 years into the future, using historic BMI data. Given BMI data between birth and age 2 years, prediction of overweight/obesity status at age 10 years, as computed from MagmaClust's predictions exhibited high specificity (0.94), negative predictive value (0.89), and accuracy (0.86). The accuracy, sensitivity, and positive predictive value of predictions increased as BMI data from additional time points were utilized for prediction. CONCLUSION MagmaClust provides a unified, probabilistic, non-parametric framework to model, cluster, and prospectively predict childhood BMI trajectories and overweight/obesity risk. The proposed method offers a convenient tool for clinicians to monitor BMI growth in children, allowing them to prospectively identify children with high predicted overweight/obesity risk and implement timely interventions.
Collapse
Affiliation(s)
- Arthur Leroy
- Department of Computer Science, The University of Manchester, Manchester, UK
| | - Varsha Gupta
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
- Bioinformatics Institute, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Mya Thway Tint
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Delicia Shu Qin Ooi
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Fabian Yap
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Republic of Singapore
- Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Ngee Lek
- Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Republic of Singapore
- Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Centre and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Yap Seng Chong
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Yung Seng Lee
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Division of Paediatric Endocrinology, Department of Paediatrics, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, Republic of Singapore
| | - Johan G Eriksson
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Mauricio A Álvarez
- Department of Computer Science, The University of Manchester, Manchester, UK
- Department of Computer Science, University of Sheffield, Sheffield, UK
| | - Navin Michael
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Dennis Wang
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore.
- Bioinformatics Institute, Agency for Science Technology and Research (A*STAR), Singapore, Republic of Singapore.
- Department of Computer Science, University of Sheffield, Sheffield, UK.
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
4
|
Adam-Raileanu A, Miron I, Lupu A, Bozomitu L, Sasaran MO, Russu R, Rosu ST, Nedelcu AH, Salaru DL, Baciu G, Mihai CM, Chisnoiu T, Beser OF, Lupu VV. Fetal Growth Restriction and Its Metabolism-Related Long-Term Outcomes-Underlying Mechanisms and Clinical Implications. Nutrients 2025; 17:555. [PMID: 39940412 PMCID: PMC11819745 DOI: 10.3390/nu17030555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
The developmental origins of adult disease theory support the concept that undernourished fetuses are at risk of developing metabolic syndrome due to the energy-saving 'Thrifty Phenotype'. This metabolic plasticity represents an evolutionary adaptation that allows individuals to resist the intense pressure caused by cyclically recurring periods of nutritional deprivation. A comprehensive review was conducted following an extensive literature search in the PubMed/Medline and EMBASE databases concerning reports on fetal/intrauterine growth restriction and its metabolic-related long-term outcomes. We only included articles written in English that were published before 1 July 2024. There are several underlying mechanisms and metabolic and endocrine adjustments shaped by the perinatal environment, and they all contribute to progression towards adult disease. From in utero malnutrition or other insults during the fetal period to fetal programing and postnatal catch-up growth, it is difficult to identify the exact moment when this adaptative phenomenon meant to assure fetal survival and to set children on their own physiological growth curves lose its beneficial effect, establishing the trajectory to obesity, insulin resistance, and other hallmarks of metabolic syndrome. With clinical correspondence to an altered body mass, composition, and eating behaviors, it is evident that the metabolic complications linked to FGR are intricate and arise from disturbances in several pathways and organs, but the underlying processes responsible for the long-term consequences are just starting to be understood. The lack of continuity in perinatal-to-pediatric FGR research sets the challenge of exploring new directions in future scientific opportunities. These will hopefully represent a cornerstone in the management of FGR-related metabolic disorders in children, preventing these disorders from evolving into adult disease.
Collapse
Affiliation(s)
- Anca Adam-Raileanu
- Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.A.-R.); (I.M.); (V.V.L.)
| | - Ingrith Miron
- Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.A.-R.); (I.M.); (V.V.L.)
| | - Ancuta Lupu
- Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.A.-R.); (I.M.); (V.V.L.)
| | - Laura Bozomitu
- Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.A.-R.); (I.M.); (V.V.L.)
| | - Maria Oana Sasaran
- Pediatrics, Faculty of Medicine, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
| | - Ruxandra Russu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (R.R.); (S.T.R.); (A.H.N.); (D.L.S.)
| | - Solange Tamara Rosu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (R.R.); (S.T.R.); (A.H.N.); (D.L.S.)
| | - Alin Horatiu Nedelcu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (R.R.); (S.T.R.); (A.H.N.); (D.L.S.)
| | - Delia Lidia Salaru
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (R.R.); (S.T.R.); (A.H.N.); (D.L.S.)
| | - Ginel Baciu
- Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania;
| | - Cristina Maria Mihai
- Pediatrics, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania; (C.M.M.); (T.C.)
| | - Tatiana Chisnoiu
- Pediatrics, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania; (C.M.M.); (T.C.)
| | - Omer Faruk Beser
- Department of Pediatric Gastroenterology, Hepatology & Nutrition, Cerrahpasa Medical Faculty, Istanbul University Cerrahpasa, 34776 Istanbul, Turkey;
| | - Vasile Valeriu Lupu
- Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.A.-R.); (I.M.); (V.V.L.)
| |
Collapse
|
5
|
Yang X, Eckel SP, Maldonado LE, Yang T, Chen X, Vigil M, Toledo-Corral CM, Dunton GF, Grubbs BH, Al-Marayati L, Lerner D, Lurvey N, Habre R, Farzan SF, Bastain TM, Breton C. Perceived Stress During Late Pregnancy and Infant Body Composition at 1 Month. J Endocr Soc 2025; 9:bvae222. [PMID: 39876876 PMCID: PMC11772522 DOI: 10.1210/jendso/bvae222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Indexed: 01/31/2025] Open
Abstract
Context Worldwide, obesity remains one of the most challenging crises with children being one of the most susceptible populations. The effect of maternal stress during pregnancy on newborn body composition, measured by fat mass and lean mass has, not been extensively studied. Objectives We evaluated the association between perceived stress during late pregnancy and infant adiposity at 1 month and assessed effect modification by infant sex and preterm birth. Methods Mother-infant dyads (N = 138) were included from the ongoing MADRES cohort. Maternal perceived stress during late pregnancy was measured by the 10-item Perceived Stress Scale (PSS), as a cumulative score, during the third trimester. Infant adiposity measures, collected at 1 month by EchoMRI, included weight, fat mass (FM), and lean mass with FM-related ratios derived. Multivariable linear regression models with interaction terms were performed. Results Most mothers reported low to moderate stress (mean ± SD PSS: 13.2 ± 5.6) during late pregnancy. A 1-SD higher PSS was associated with higher FM% (FM (g)/weight (g): β = 0.78%; 95% CI, 0.13-1.44) but we did not find significant associations for the other adiposity measures. Statistically significant effects of perceived stress on FM-related measures were observed in male infants and preterm infants (both P for interaction <.05) but were null among female infants or term infants. Conclusion In this predominately low-income Hispanic population, perceived stress during late pregnancy was associated with higher FM-related body composition measures during early infancy; this association was stronger among male and preterm infants compared to the overall population and other subgroups.
Collapse
Affiliation(s)
- Xiaoran Yang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Sandrah P Eckel
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Luis E Maldonado
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Tingyu Yang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Xinci Chen
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Mario Vigil
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Claudia M Toledo-Corral
- Department of Health Sciences, California State University, Northridge, Northridge, CA 91330, USA
| | - Genevieve F Dunton
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Brendan H Grubbs
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Laila Al-Marayati
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | | | | | - Rima Habre
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Shohreh F Farzan
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Theresa M Bastain
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Carrie Breton
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| |
Collapse
|
6
|
Mahboub N, Ayoub E, Mounzer C, Baltagi TK, Papandreou D, de Vries N, Rizk R. Metabolic Syndrome as a Risk Factor Among Lebanese Patients with Substance Use Disorder Undergoing Treatment for Recovery Through Rehabilitation or Opioid Substitution Treatment. Clin Pract 2024; 14:2661-2680. [PMID: 39727798 DOI: 10.3390/clinpract14060210] [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: 10/21/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: Data about metabolic syndrome (MS) in people who use drugs (PWUD) undergoing treatment for recovery are limited. We aimed to explore the extent of the MS and its predominant components and determinants in a sample of PWUD undergoing treatment for recovery through rehabilitation or opioid substitution treatment (OST) in Lebanon. Furthermore, we investigated the effect of each treatment modality on the MS; Methods: This was a cross-sectional study, in which demographics and treatment-related, nutritional, and biochemical data of the participants were collected. MS was defined according to the American Heart Association and the National Heart, Lung, and Blood Institute (AHA/NHLBI) criteria. Descriptive statistics were presented, and bivariate and multivariate analyses were conducted; Results: A total of 155 male subjects with the following characteristics were included: OST: n = 80; rehabilitation: n = 75; mean age: 32.53 ± 8.39 years; mean body mass index (BMI): 27.41 ± 4.99 Kg/m2; mean duration of treatment: 18 months. More than half of the sample had low HDL-C (56.8%) and/or elevated blood pressure (51.6%), 42.9% had elevated WC, 21.9% had elevated TG, and 12.3% had elevated FBS. Furthermore, 7.2% of the sample had no components of the MS, 29.2% had one component, 40.9% had two components, 16.9% had three components, and 5.8% had four components. MS was identified in 22.7% of the sample. Higher age was associated with higher odds of being diagnosed with MS (OR = 1.072; 95% CI: 1.021-1.126), whereas higher duration of current treatment was associated with lower odds (OR = 0.969; 95% CI: 0.944-0.995); Conclusions: MS and its components are prevalent in PWUD undergoing treatment for recovery. Routine screening and preventive measures are essential to prevent metabolic syndrome, particularly among older people and treatment newcomers.
Collapse
Affiliation(s)
- Nadine Mahboub
- Department of Nutrition and Food Sciences, Faculty of Arts and Sciences, Lebanese International University, Beirut P.O. Box 146404, Lebanon
| | - Elissa Ayoub
- Department of Nutrition and Dietetics, Faculty of Public Health Branch 2, Lebanese University, Beirut P.O. Box 6573, Lebanon
| | - Carine Mounzer
- Department of Nutrition and Dietetics, Faculty of Public Health Branch 2, Lebanese University, Beirut P.O. Box 6573, Lebanon
| | - Tatiana Kate Baltagi
- Department of Biochemistry, College of Arts and Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Dimitrios Papandreou
- Department of Clinical Nutrition & Dietetics, College of Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Nanne de Vries
- Department of Health Promotion, CAPHRI School for Public Health and Primary Care, Maastricht University, P.O. Box 616, 6200 LK Maastricht, The Netherlands
| | - Rana Rizk
- Department of Nutrition and Food Science, Faculty of Arts and Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
- Institut National de Santé Publique, d'Epidémiologie Clinique Et de Toxicologie (INSPECT-LB), Beirut P.O. Box 14404, Lebanon
| |
Collapse
|
7
|
Bagot S, Pereira B, Miles-Chan J, Gryson C, Chanséaume Bussière E, Duclos M, Thivel D, Isacco L. The habitual degree of weight loss might be associated with specific fat and protein intakes during a period of weight maintenance in athletes used to weight variations: preliminary results from the WAVE study. Nutr Res 2024; 129:14-27. [PMID: 39178640 DOI: 10.1016/j.nutres.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/26/2024]
Abstract
Weight variations are common in sporting life, with important inter-individual variability in the degree of an athlete's habitual weight loss. As a part of the WAVE study (NCT04107545), the main objective of this preliminary study was to determine whether the habitual degree of weight loss was associated with anthropometric, body composition, nutritional or psychometric profiles during a period of weight maintenance in athletes accustomed to weight variations. We hypothesized that athletes accustomed to a higher habitual degree of weight loss may have a higher body weight and body fat mass, and may present a more controlled diet regimen and cognitive restriction than athletes with a lower habitual degree of weight loss. During a period of weight maintenance, 62 athletes (24.0 ± 5.3 years; 26 women) completed anthropometry and body composition measurements, a 48-hours food diary and self-reported questionnaires to determine their weight variation practice, nutritional profile and mood state. Athletes were stratified within inter- and intra-quartile groups according to their habitual degree of weight loss. Athletes with a higher habitual degree of weight loss were those who consumed more protein (P < .001) and less fat (P = .01) as a proportion of total energy compared with those losing less weight, without any difference in body composition between the groups. The rapid weight loss score was significantly higher in individuals losing more weight (P < .001) and no difference was observed for the mood state profile. The present results suggest a potential control of nutritional regulation during a period of weight maintenance in order to spare fat-free mass and favor fat mass loss in athletes who are routinely losing more weight. Fat-free mass may be the main nutritional driver due to low body fat mass in athletes, which may limit the "catch-up fat" phenomenon commonly observed in nonathletic population.
Collapse
Affiliation(s)
- Sarah Bagot
- Clermont Auvergne University, UPR 3533, Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), CRNH Auvergne, Clermont-Ferrand, France; Nutrifizz, Clermont-Ferrand, France.
| | - Bruno Pereira
- Clermont-Ferrand University Hospital, Biostatistics Unit (DRCI), Clermont-Ferrand, France
| | - Jennifer Miles-Chan
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | | | | | - Martine Duclos
- Department of Sport Medicine and Functional Explorations, CHU Clermont-Ferrand, Clermont-Ferrand, France; International Research Chair Health in Motion, Clermont Auvergne University Foundation, Clermont-Ferrand, France
| | - David Thivel
- Clermont Auvergne University, UPR 3533, Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), CRNH Auvergne, Clermont-Ferrand, France; International Research Chair Health in Motion, Clermont Auvergne University Foundation, Clermont-Ferrand, France
| | - Laurie Isacco
- Clermont Auvergne University, UPR 3533, Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), CRNH Auvergne, Clermont-Ferrand, France; International Research Chair Health in Motion, Clermont Auvergne University Foundation, Clermont-Ferrand, France
| |
Collapse
|
8
|
Vasconcelos I, von Hafe M, Adão R, Leite-Moreira A, Brás-Silva C. Corticotropin-releasing hormone and obesity: From fetal life to adulthood. Obes Rev 2024; 25:e13763. [PMID: 38699883 DOI: 10.1111/obr.13763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/02/2024] [Accepted: 03/19/2024] [Indexed: 05/05/2024]
Abstract
Obesity is among the most common chronic disorders, worldwide. It is a complex disease that reflects the interactions between environmental influences, multiple genetic allelic variants, and behavioral factors. Recent developments have also shown that biological conditions in utero play an important role in the programming of energy homeostasis systems and might have an impact on obesity and metabolic disease risk. The corticotropin-releasing hormone (CRH) family of neuropeptides, as a central element of energy homeostasis, has been evaluated for its role in the pathophysiology of obesity. This review aims to summarize the relevance and effects of the CRH family of peptides in the pathophysiology of obesity spanning from fetal life to adulthood.
Collapse
Affiliation(s)
- Inês Vasconcelos
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Madalena von Hafe
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rui Adão
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Adelino Leite-Moreira
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Carmen Brás-Silva
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal
| |
Collapse
|
9
|
Yoshida A, Takata Y, Tabara Y, Maruyama K, Inoue S, Osawa H, Sugiyama T. Interaction effect between low birthweight and resistin gene rs1862513 variant on insulin resistance and type 2 diabetes mellitus in adulthood: Toon Genome Study. J Diabetes Investig 2024; 15:725-735. [PMID: 38421160 PMCID: PMC11143422 DOI: 10.1111/jdi.14163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/11/2024] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
AIMS/INTRODUCTION Gene-environment interactions are considered to critically influence type 2 diabetes mellitus development; however, the underlying mechanisms and specific interactions remain unclear. Given the increasing prevalence of low birthweight (LBW) influenced by the intrauterine environment, we sought to investigate genetic factors related to type 2 diabetes development in individuals with LBW. MATERIALS AND METHODS The interaction between 20 reported type 2 diabetes susceptibility genes and the development of type 2 diabetes in LBW (<2,500 g) individuals in a population-based Japanese cohort (n = 1,021) was examined by logistic regression and stratified analyses. RESULTS Logistic regression analyses showed that only the G/G genotype at the rs1862513 locus of the resistin gene (RETN), an established initiator of insulin resistance, was closely related to the prevalence of type 2 diabetes in individuals with LBW. Age, sex and current body mass index-adjusted stratified analyses showed a significant interaction effect of LBW and the RETN G/G genotype on fasting insulin, homeostatic model assessment 2-insulin resistance, Matsuda index and the prevalence of type 2 diabetes (all P-values for interaction <0.05). The adjusted odds ratio for type 2 diabetes in the LBW + G/G genotype group was 7.33 (95% confidence interval 2.43-22.11; P = 0.002) compared with the non-LBW + non-G/G genotype group. Similar results were obtained after excluding the influence of malnutrition due to World War II. CONCLUSIONS Simultaneous assessment of LBW and the RETN G/G genotype can more accurately predict the risk of future type 2 diabetes than assessing each of these factors alone, and provide management strategies, including early lifestyle intervention in LBW population.
Collapse
Affiliation(s)
- Ayaka Yoshida
- Department of Obstetrics and GynecologyEhime University Graduate School of MedicineToonEhimeJapan
| | - Yasunori Takata
- Department of Diabetes and Molecular GeneticsEhime University Graduate School of MedicineToonEhimeJapan
| | - Yasuharu Tabara
- Center for Genomic MedicineKyoto University Graduate School of MedicineKyotoJapan
- Graduate School of Public HealthShizuoka Graduate University of Public HealthShizuokaJapan
| | - Koutatsu Maruyama
- Department of Bioscience, Graduate School of AgricultureEhime UniversityToonEhimeJapan
| | - Shota Inoue
- Department of Obstetrics and GynecologyEhime University Graduate School of MedicineToonEhimeJapan
| | - Haruhiko Osawa
- Department of Diabetes and Molecular GeneticsEhime University Graduate School of MedicineToonEhimeJapan
| | - Takashi Sugiyama
- Department of Obstetrics and GynecologyEhime University Graduate School of MedicineToonEhimeJapan
| |
Collapse
|
10
|
Ngema M, Xulu ND, Ngubane PS, Khathi A. Pregestational Prediabetes Induces Maternal Hypothalamic-Pituitary-Adrenal (HPA) Axis Dysregulation and Results in Adverse Foetal Outcomes. Int J Mol Sci 2024; 25:5431. [PMID: 38791468 PMCID: PMC11122116 DOI: 10.3390/ijms25105431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Maternal type 2 diabetes mellitus (T2DM) has been shown to result in foetal programming of the hypothalamic-pituitary-adrenal (HPA) axis, leading to adverse foetal outcomes. T2DM is preceded by prediabetes and shares similar pathophysiological complications. However, no studies have investigated the effects of maternal prediabetes on foetal HPA axis function and postnatal offspring development. Hence, this study investigated the effects of pregestational prediabetes on maternal HPA axis function and postnatal offspring development. Pre-diabetic (PD) and non-pre-diabetic (NPD) female Sprague Dawley rats were mated with non-prediabetic males. After gestation, male pups born from the PD and NPD groups were collected. Markers of HPA axis function, adrenocorticotropin hormone (ACTH) and corticosterone, were measured in all dams and pups. Glucose tolerance, insulin and gene expressions of mineralocorticoid (MR) and glucocorticoid (GR) receptors were further measured in all pups at birth and their developmental milestones. The results demonstrated increased basal concentrations of ACTH and corticosterone in the dams from the PD group by comparison to NPD. Furthermore, the results show an increase basal ACTH and corticosterone concentrations, disturbed MR and GR gene expression, glucose intolerance and insulin resistance assessed via the Homeostasis Model Assessment (HOMA) indices in the pups born from the PD group compared to NPD group at all developmental milestones. These observations reveal that pregestational prediabetes is associated with maternal dysregulation of the HPA axis, impacting offspring HPA axis development along with impaired glucose handling.
Collapse
Affiliation(s)
| | | | | | - Andile Khathi
- School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Westville, Private Bag X54001, Durban 4041, KwaZulu Natal, South Africa; (M.N.); (N.D.X.); (P.S.N.)
| |
Collapse
|
11
|
Magodoro IM, Castle AC, Tshuma N, Goedecke JH, Sewpaul R, Manasa J, Manne-Goehler J, Ntusi N, Nyirenda MJ, Siedner MJ. Associations of HIV and prevalent type 2 diabetes mellitus in the context of obesity in South Africa. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.10.24304033. [PMID: 38559082 PMCID: PMC10980116 DOI: 10.1101/2024.03.10.24304033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
It is unclear how rising obesity among people with HIV (PWH) in sub-Saharan Africa (SSA) impacts their risk of type 2 diabetes mellitus (diabetes). Using a South African national cross-sectional sample of adult PWH and their peers without HIV (PWOH), we examined the associations between HIV and prevalent diabetes across the spectrum of body mass index (BMI), waist circumference (WC) and waist-to-height ratio (WtHR). Analyses were sex stratified, and adjusted for age, sociodemographic and behavioral factors. The prevalence of diabetes among males was similar between PWH and PWOH, overall and at all levels of adiposity. In contrast, overall diabetes prevalence was higher among female PWOH than female PWH. However, there were differences according to adiposity such that, compared to female PWOH, relative diabetes prevalence in female PWH was reduced with obesity but accentuated with leanness. These differences in the relationship between adiposity and diabetes by HIV serostatus call for better mechanistic understanding of sex-specific adipose tissue biology in HIV in South Africa, and possibly in other HIV endemic settings in SSA.
Collapse
|
12
|
Cerasani J, Consales A, Gangi S, Macchi M, Morniroli D, Vizzari G, Tiraferri V, Petrelli A, Mosca F, Giannì ML. Exclusive human milk feeding and prevalence of early adiposity rebound in ELBW infants: a retrospective cohort study. Eur J Pediatr 2024; 183:1295-1303. [PMID: 38112801 PMCID: PMC10950974 DOI: 10.1007/s00431-023-05374-6] [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: 05/27/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
The purpose of the present study was to evaluate the prevalence of early adiposity rebound (EAR) and factors associated with its occurrence in a cohort of extremely low birth weight infants (ELBW). We conducted a retrospective longitudinal study including ELBW infants followed-up for up to 10 years after discharge. EAR was defined as occurring before 5 years of age. A multivariate binary logistic regression analysis was performed to evaluate maternal and perinatal variables independently associated with EAR. Out of 212 ELBW infants included in the analysis, 40.6% developed EAR and 21.5% showed it before 4 years of age. Only formula milk feeding at discharge was independently associated with a higher risk of EAR. The mean BMI of children with EAR was higher than that of children without EAR. Furthermore, the prevalence of overweight and obesity was higher in the EAR group than in the timely AR group. Conclusions: ELBW infants in our cohort developed EAR in a relatively high percentage of cases. In this already at-risk population, EAR may represent a further risk factor for an adverse metabolic outcome. Monitoring preterm infants' growth within a long-term follow-up program and promoting and supporting human milk feeding is advisable. What is Known: • Preterm-born infants are at high risk for long-term adverse health outcomes, especially cardiovascular and metabolic. • The occurrence of early adiposity rebound (EAR) is associated with the risk of later obesity and metabolic syndrome. What is New: • The occurrence of EAR in ELBW infants may represent an additional risk factor for later adverse metabolic outcomes in an already vulnerable population. • Future preventive strategies should include a long-term follow-up and the promotion of exclusive breastfeeding.
Collapse
Affiliation(s)
- Jacopo Cerasani
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Alessandra Consales
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| | - Silvana Gangi
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marta Macchi
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Morniroli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giulia Vizzari
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Tiraferri
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Angelo Petrelli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Fabio Mosca
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Lorella Giannì
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
13
|
Bochantin-Winders KA, Slavick KR, Jurgens IM, Hurlbert JL, Menezes ACB, Kirsch JD, Borowicz PP, Schauer CS, Dahlen CR. Influence of sire plane of nutrition and targeted body weight gain on ewe lamb growth, glucose metabolism, and ovarian reserve. J Anim Sci 2024; 102:skae301. [PMID: 39367596 PMCID: PMC11600961 DOI: 10.1093/jas/skae301] [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: 07/18/2024] [Accepted: 10/03/2024] [Indexed: 10/06/2024] Open
Abstract
Rambouillet rams were managed on either a positive (POS; gain 12% body weight [BW]; n = 8), maintenance (MAINT; maintain BW; n = 8), or negative (NEG; lose 12% BW; n = 8) plane of nutrition before breeding. Rams were bred to ewes (n = 10 per ram) that were managed similarly throughout gestation, and lambs were fed a common diet postnatally. Two ewe lambs (7.6 ± 0.02 mo of age, BW = 47.1 ± 1.17 kg) from each sire were selected and within-pair, randomly assigned to be managed for a moderate (MOD, 0.11 kg/d; n = 23) or accelerated (ACC, 0.20 kg/d; n = 22) rate of gain for 56 d. Ewe lamb BW was recorded on a weekly basis and blood was collected on days 0, 28, and 56 for analysis of insulin-like growth factor 1 (IGF-1), triiodothyronine (T3), thyroxine (T4), glucose, blood urea nitrogen (BUN), and non-esterified fatty acids (NEFA). Intravenous glucose tolerance tests (IVGTT) were conducted from days -7 to -4 and days 57 to 64. A unilateral ovariectomy was performed and ovarian follicles were staged and counted macro and microscopically. Sire treatment × day and ewe treatment × day interactions were present for BW (P ≤ 0.05), where POS had slower growth than MAINT and NEG, and tended (P = 0.10) to have reduced average daily gain (ADG) when managed at an accelerated rate of gain. By design, ACC had greater BW and ADG than MOD (P < 0.05). Concentrations of IGF-1 and T4 were greater in ACC than MOD (P ≤ 0.05), and NEG tended to have greater concentrations of IGF-1 than POS and MAINT (P = 0.08). At the first IVGTT, the concentration of insulin was influenced by a sire treatment × time interaction (P ≤ 0.05), suggesting impaired secretion in NEG-sires ewes, but no differences in area under the curve (AUC) for glucose, insulin, or their ratio (P ≥ 0.11). No interactive effects of sire and ewe treatment (P ≥ 0.52) were observed at the second IVGTT, but insulin and insulin:glucose ratio were influenced by sire treatment × time (P ≤ 0.02), as NEG had greater insulin concentration at 60 min than MAINT (P = 0.03) and greater AUC than POS and MAINT (P ≤ 0.04). No differences in ovary size, weight, or total counts of macro and microscopic follicles were observed (P ≥ 0.23). Ewes-fed ACC had a greater number of small surface follicles (P = 0.02), whereas MOD tended to have a greater number of large surface follicles and tertiary follicles (P < 0.06). These findings suggest that the paternal plane of nutrition influences female offspring physiology, particularly at varying growth rates.
Collapse
Affiliation(s)
- Kerri A Bochantin-Winders
- Department of Animal Sciences and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58105, USA
| | - Kathryn R Slavick
- Department of Animal Sciences and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58105, USA
| | - Isabella M Jurgens
- Department of Animal Sciences and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58105, USA
| | - Jennifer L Hurlbert
- Department of Animal Sciences and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58105, USA
| | - Ana Clara B Menezes
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, USA
| | - James D Kirsch
- Department of Animal Sciences and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58105, USA
| | - Pawel P Borowicz
- Department of Animal Sciences and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58105, USA
| | - Christopher S Schauer
- Hettinger Research Extension Center, North Dakota State University, Hettinger, ND 58639, USA
| | - Carl R Dahlen
- Department of Animal Sciences and Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 58105, USA
| |
Collapse
|
14
|
Bochantin-Winders KA, Baumgaertner F, Hurlbert JL, Menezes ACB, Kirsch JD, Dorsam ST, Schauer CS, Dahlen CR. Divergent planes of nutrition in mature rams influences body composition, hormone and metabolite concentrations, and offspring birth measurements, but not semen characteristics or offspring growth. J Anim Sci 2024; 102:skae207. [PMID: 39044680 PMCID: PMC11347781 DOI: 10.1093/jas/skae207] [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: 01/30/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024] Open
Abstract
Objectives of this experiment were to characterize the effects of ram plane of nutrition on body composition, concentrations of hormones and metabolites, sperm characteristics, and offspring outcomes. Mature Rambouillet rams (n = 24, BW = 82.9 ± 2.63 kg) were individually housed and randomly assigned to either a positive (POS; n = 8), maintenance (MAINT; n = 8), or negative (NEG; n = 8) plane of nutrition for an 84-day feeding period. Rams were fed a common diet, with daily feed allocations adjusted weekly based on body weight (BW) to achieve the targeted weight gain or loss (approximately 12% of initial BW). On 0, 28, 56, and 84-d, body condition score (BCS) and scrotal circumference (SC) were recorded, and blood and semen were collected. Following the feeding period, rams were placed in pens with 10 ewes each for a 28-d breeding period. Ewes were managed similarly throughout gestation and body weight and measurements were recorded at birth and weaning. Data were analyzed as repeated measures in time where appropriate with the mixed procedure of SAS, and individual ram was the experimental unit for all analysis. Ram BW was influenced by a treatment × day interaction (P < 0.001), with POS (0.12 ± 0.01 kg) having greater daily weight change than MAINT (0.1 ± 0.01 kg), which was greater than NEG (-0.12 ± 0.01 kg). Ram BCS and SC were influenced by treatment × day interactions (P ≤ 0.01), being similar on day 0 but POS being greater than NEG by day 56. Concentrations of triiodothyronine (T3) and T3:T4 ratio exhibited treatment × day interactions (P ≤ 0.02), as POS had greater values than NEG by day 84 (P ≤ 0.02). Concentration of insulin-like growth factor-1 was greater in POS than MAINT and NEG (P ≤ 0.02), and non-esterified fatty acids and thyroxine (T4) were influenced by a day effect (P ≤ 0.01), but testosterone was unaffected (P ≥ 0.09). Minimal differences in semen volume, sperm concentration, motility, or morphology were observed among treatments (P ≥ 0.31). A similar proportion of ewes bred by rams in the respective treatments lambed and weaned lambs (P ≥ 0.54). Birth weight, chest circumference, and shoulder-hip length were greater (P ≤ 0.05) in NEG lambs compared with POS and MAINT; however, no differences were detected in weaning weight and weaning body measurements (P ≥ 0.40). Findings suggest paternal nutrition during the period of sperm development may influence offspring outcomes, potentially as a result of in-utero programming of paternal origin.
Collapse
Affiliation(s)
| | | | - Jennifer L Hurlbert
- Department of Animal Science, North Dakota State University, Fargo, ND, 58105, USA
| | - Ana Clara B Menezes
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, USA
| | - James D Kirsch
- Department of Animal Science, North Dakota State University, Fargo, ND, 58105, USA
| | - Sheri T Dorsam
- Department of Animal Science, North Dakota State University, Fargo, ND, 58105, USA
| | - Christopher S Schauer
- Hettinger Research Extension Center, North Dakota State University, Hettinger, ND 58639, USA
| | - Carl R Dahlen
- Department of Animal Science, North Dakota State University, Fargo, ND, 58105, USA
| |
Collapse
|
15
|
Magodoro IM, Castle AC, Tshuma N, Goedecke JH, Sewpaul R, Manasa J, Manne-Goehler J, Ntusi NAB, Nyirenda MJ, Siedner MJ. Associations of HIV and prevalent type 2 diabetes mellitus in the context of obesity in South Africa. JOURNAL OF MULTIMORBIDITY AND COMORBIDITY 2024; 14:26335565241293691. [PMID: 39492946 PMCID: PMC11528680 DOI: 10.1177/26335565241293691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024]
Abstract
Background It is unclear how rising obesity among people with HIV (PWH) impacts their risk of type 2 diabetes mellitus (diabetes). We examined associations between HIV, prevalent diabetes and adiposity among South African PWH and their peers without HIV (PWOH). Methods HIV status was ascertained by antibody testing. Diabetes was defined as current use of oral hypoglycemics, insulin, and/or HbA1c ≥6.5%. Adiposity was measured by body mass index (BMI), waist circumference and waist-to-height ratio. Their associations were examined using sex-stratified multivariable fractional polynomial generalized linear models, reporting adjusted prevalence and prevalence ratios (adjPR). Results The mean age among 1,254 PWH and 4,381 PWOH was 41 years (95%CI 28, 56). The prevalence of diabetes among males was similar between PWH [11.3% (7.1, 15.5)] and PWOH [9.8% (8.5, 11.1); p=0.740]. By contrast, diabetes prevalence was higher among female PWOH [15.7% (14.4, 17.0)] than female PWH [10.5 (8.3, 12.8)%; adjPR: 0.67 (0.51, 0.82); p<0.001]. This difference was accentuated with obesity but reversed with leanness. At BMI ≥25 kg/m2, female PWH had lower diabetes prevalence [adjPR: 0.58 (0.41, 0.76); p<0.001] than female PHIV. In contrast, at BMI <18 kg/m2, female PWH had higher prevalence [adjPR: 1.72 (-1.53, 4.96); p=0.756] than female PWOH. Conclusion We found sex-specific differences in the relationship between adiposity and diabetes prevalence by HIV serostatus in South Africa. Notably, females living with obesity and HIV had lower prevalence of diabetes than females living with obesity and without HIV, which may have particular implications for diabetes prevention programs in the region.
Collapse
Affiliation(s)
- Itai M Magodoro
- Department of Medicine, University of Cape Town, Cape Town, Republic of South Africa
| | - Alison C Castle
- Africa Health Research Institute, Mtubatuba, Republic of South Africa
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
- Medical Practice Evaluation Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ndumiso Tshuma
- The Best Health Solutions, Johannesburg, Republic of South Africa
| | - Julia H Goedecke
- Biomedical Research and Innovation Platform, South African Medical Research Council, Cape Town, Republic of South Africa
- Health through Physical Activity, Lifestyle and Sport Research Centre (HPALS), Division of Physiological Sciences, Department of Human Biology, University of Cape Town, Cape Town, Republic of South Africa
| | - Ronel Sewpaul
- Public Health, Societies and Belonging (PHSB) Division, Human Sciences Research Council, Cape Town, Republic of South Africa
| | - Justen Manasa
- Biomedical Research and Training Institute, Harare, Zimbabwe
- College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Jennifer Manne-Goehler
- Medical Practice Evaluation Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ntobeko AB Ntusi
- Department of Medicine, University of Cape Town, Cape Town, Republic of South Africa
- South African Medical Research Council Extramural Unit on Noncommunicable and Infectious Diseases, Cape Town, Republic of South Africa
- ARUA/Guild Cluster of Research Excellence on Noncommunicable Diseases and Associated Multimorbidity
| | | | - Mark J Siedner
- Africa Health Research Institute, Mtubatuba, Republic of South Africa
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
- Medical Practice Evaluation Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Faculty of Medicine, University of KwaZulu-Natal, Durban, Republic of South Africa
| |
Collapse
|
16
|
Zheng W, Zhang KX, Yuan XX, Luo JY, Wang J, Song W, Liang SN, Wang XX, Guo CM, Li GH. Maternal weight, blood lipids, and the offspring weight trajectories during infancy and early childhood in twin pregnancies. World J Pediatr 2023; 19:961-971. [PMID: 36877432 DOI: 10.1007/s12519-023-00703-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/07/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND The intrauterine environment has a profound and long-lasting influence on the health of the offspring. However, its impact on the postnatal catch-up growth of twin children remains unclarified. Therefore, this study aimed to explore the maternal factors in pregnancy associated with twin offspring growth. METHODS This study included 3142 live twin children born to 1571 mothers from the Beijing Birth Cohort Study conducted from 2016 to 2021 in Beijing, China. Original and corrected weight-for-age standard deviation scores of the twin offspring from birth to 36 months of age were calculated according to the World Health Organization Child Growth Standards. The corresponding weight trajectories were identified by the latent trajectory model. Maternal factors in pregnancy associated with the weight trajectories of the twin offspring were examined after adjustment for potential confounders. RESULTS Five weight trajectories of the twin children were identified, with 4.9% (154/3142) exhibiting insufficient catch-up growth, 30.6% (961/3142), and 46.8% (1469/3142) showing adequate catch-up growth from different birth weights, and 15.0% (472/3142) and 2.7% (86/3142) showing various degrees of excessive catch-up growth. Maternal short stature [adjusted odds ratio (OR) = 0.691, 95% confidence interval (CI) = 0.563-0.848, P = 0.0004] and lower total gestational weight gain (GWG) (adjusted OR = 0.774, 95% CI = 0.616-0.972, P = 0.03) were associated with insufficient catch-up growth of the offspring. Maternal stature (adjusted OR = 1.331, 95% CI = 1.168-1.518, P < 0.001), higher pre-pregnancy body mass index (BMI) (adjusted OR = 1.230, 95% CI = 1.090-1.387, P < 0.001), total GWG (adjusted OR = 1.207, 95% CI = 1.068-1.364, P = 0.002), GWG rate (adjusted OR = 1.165, 95% CI = 1.027-1.321, P = 0.02), total cholesterol (TC) (adjusted OR = 1.150, 95% CI = 1.018-1.300, P = 0.03) and low-density lipoprotein-cholesterol (LDL-C) (adjusted OR = 1.177, 95% CI = 1.041-1.330) in early pregnancy were associated with excessive growth of the offspring. The pattern of weight trajectories was similar between monochorionic and dichorionic twins. Maternal height, pre-pregnancy BMI, GWG, TC and LDL-C in early pregnancy were positively associated with excess growth in dichorionic twins, yet a similar association was observed only between maternal height and postnatal growth in monochorionic twins. CONCLUSION This study identified the effect of maternal stature, weight status, and blood lipid profiles during pregnancy on postnatal weight trajectories of the twin offspring, thereby providing a basis for twin pregnancy management to improve the long-term health of the offspring.
Collapse
Affiliation(s)
- Wei Zheng
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Ke-Xin Zhang
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Xian-Xian Yuan
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Jin-Ying Luo
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
- Obstetrics and Gynecology Department, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, China
| | - Jia Wang
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Wei Song
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Sheng-Nan Liang
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Xiao-Xin Wang
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Cui-Mei Guo
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Guang-Hui Li
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China.
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China.
| |
Collapse
|
17
|
Yonke JA, Seymour KA, El-Kadi SW. Branched-chain amino acid supplementation does not enhance lean tissue accretion in low birth weight neonatal pigs, despite lower Sestrin2 expression in skeletal muscle. Amino Acids 2023; 55:1389-1404. [PMID: 37743429 DOI: 10.1007/s00726-023-03319-9] [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: 01/31/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023]
Abstract
Postnatal muscle growth is impaired in low birth weight (L) neonatal pigs. Leucine supplementation has been established as a dietary intervention to enhance muscle growth in growing animals. The aim of this study was to investigate the efficacy of supplementing L neonatal pig formulas with branched-chain amino acids (B) to enhance the rate of protein accretion. Twenty-four 3-day old pigs were divided into two groups low (L) and normal birth weight (N) based on weight at birth. Pigs were assigned to a control (C) or 1% branched-chain amino acids (B) formulas, and fed at 250 mL·kg body weight -1·d-1 for 28 d. Body weight of pigs in the L group was less than those in the N group (P < 0.01). However, fractional body weight was greater for L pigs compared with their N siblings from day 24 to 28 of feeding regardless of formula (P < 0.01). In addition, feed efficiency (P < 0.0001) and efficiently of protein accretion (P < 0.0001) were greater for L than N pigs regardless of supplementation. Pigs fed the B formula had greater plasma leucine, isoleucine, and valine concentrations compared with those fed the C formula (P < 0.05). Longissimus dorsi Sestrin2 protein expression was less for pigs in the L group compared with those in the N group (P < 0.01), but did not result in a corresponding increase in translation initiation signaling. Longissimus dorsi mRNA expression of BCAT2 was less for LB pigs compared with those in the LC group, and was intermediate for NC and NB pigs (P < 0.05). Hepatic mRNA expression of BCKDHA was greater for pigs in the L compared with those in the N groups (P < 0.05). However, plasma branched-chain keto-acid concentration was reduced for C compared with those in the B group (P < 0.05). These data suggest that branched-chain amino acid supplementation does not improve lean tissue accretion of low and normal birth weight pigs, despite a reduction in Sestrin2 expression in skeletal muscle of low birth weight pigs. The modest improvement in fractional growth rate of low birth weight pigs compared with their normal birth weight siblings was likely due to a more efficient dietary protein utilization.
Collapse
Affiliation(s)
- Joseph A Yonke
- School of Animal Sciences, Virginia Tech, 175 West Campus Drive, Blacksburg, VA, 24061, USA
| | - Kacie A Seymour
- School of Animal Sciences, Virginia Tech, 175 West Campus Drive, Blacksburg, VA, 24061, USA
| | - Samer W El-Kadi
- School of Animal Sciences, Virginia Tech, 175 West Campus Drive, Blacksburg, VA, 24061, USA.
| |
Collapse
|
18
|
White MR, Yates DT. Dousing the flame: reviewing the mechanisms of inflammatory programming during stress-induced intrauterine growth restriction and the potential for ω-3 polyunsaturated fatty acid intervention. Front Physiol 2023; 14:1250134. [PMID: 37727657 PMCID: PMC10505810 DOI: 10.3389/fphys.2023.1250134] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/14/2023] [Indexed: 09/21/2023] Open
Abstract
Intrauterine growth restriction (IUGR) arises when maternal stressors coincide with peak placental development, leading to placental insufficiency. When the expanding nutrient demands of the growing fetus subsequently exceed the capacity of the stunted placenta, fetal hypoxemia and hypoglycemia result. Poor fetal nutrient status stimulates greater release of inflammatory cytokines and catecholamines, which in turn lead to thrifty growth and metabolic programming that benefits fetal survival but is maladaptive after birth. Specifically, some IUGR fetal tissues develop enriched expression of inflammatory cytokine receptors and other signaling cascade components, which increases inflammatory sensitivity even when circulating inflammatory cytokines are no longer elevated after birth. Recent evidence indicates that greater inflammatory tone contributes to deficits in skeletal muscle growth and metabolism that are characteristic of IUGR offspring. These deficits underlie the metabolic dysfunction that markedly increases risk for metabolic diseases in IUGR-born individuals. The same programming mechanisms yield reduced metabolic efficiency, poor body composition, and inferior carcass quality in IUGR-born livestock. The ω-3 polyunsaturated fatty acids (PUFA) are diet-derived nutraceuticals with anti-inflammatory effects that have been used to improve conditions of chronic systemic inflammation, including intrauterine stress. In this review, we highlight the role of sustained systemic inflammation in the development of IUGR pathologies. We then discuss the potential for ω-3 PUFA supplementation to improve inflammation-mediated growth and metabolic deficits in IUGR offspring, along with potential barriers that must be considered when developing a supplementation strategy.
Collapse
Affiliation(s)
| | - Dustin T. Yates
- Stress Physiology Laboratory, Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
19
|
Tamarelle J, Creze MM, Savathdy V, Phonekeo S, Wallenborn J, Siengsounthone L, Fink G, Odermatt P, Kounnavong S, Sayasone S, Vonaesch P. Dynamics and consequences of nutrition-related microbial dysbiosis in early life: study protocol of the VITERBI GUT project. Front Nutr 2023; 10:1111478. [PMID: 37275646 PMCID: PMC10232750 DOI: 10.3389/fnut.2023.1111478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Introduction Early life under- and overnutrition (jointly termed malnutrition) is increasingly recognized as an important risk factor for adult obesity and metabolic syndrome, a diet-related cluster of conditions including high blood sugar, fat and cholesterol. Nevertheless, the exact factors linking early life malnutrition with metabolic syndrome remain poorly characterized. We hypothesize that the microbiota plays a crucial role in this trajectory and that the pathophysiological mechanisms underlying under- and overnutrition are, to some extent, shared. We further hypothesize that a "dysbiotic seed microbiota" is transmitted to children during the birth process, altering the children's microbiota composition and metabolic health. The overall objective of this project is to understand the precise causes and biological mechanisms linking prenatal or early life under- or overnutrition with the predisposition to develop overnutrition and/or metabolic disease in later life, as well as to investigate the possibility of a dysbiotic seed microbiota inheritance in the context of maternal malnutrition. Methods/design VITERBI GUT is a prospective birth cohort allowing to study the link between early life malnutrition, the microbiota and metabolic health. VITERBI GUT will include 100 undernourished, 100 normally nourished and 100 overnourished pregnant women living in Vientiane, Lao People's Democratic Republic (PDR). Women will be recruited during their third trimester of pregnancy and followed with their child until its second birthday. Anthropometric, clinical, metabolic and nutritional data are collected from both the mother and the child. The microbiota composition of maternal and child's fecal and oral samples as well as maternal vaginal and breast milk samples will be determined using amplicon and shotgun metagenomic sequencing. Epigenetic modifications and lipid profiles will be assessed in the child's blood at 2 years of age. We will investigate for possible associations between metabolic health, epigenetics, and microbial changes. Discussion We expect the VITERBI GUT project to contribute to the emerging literature linking the early life microbiota, epigenetic changes and growth/metabolic health. We also expect this project to give new (molecular) insights into the mechanisms linking malnutrition-induced early life dysbiosis and metabolic health in later life, opening new avenues for microbiota-engineering using microbiota-targeted interventions.
Collapse
Affiliation(s)
- Jeanne Tamarelle
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Margaux M. Creze
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Vanthanom Savathdy
- Lao Tropical and Public Health Institute, Ministry of Health, Vientiane, Lao People’s Democratic Republic (PDR)
| | - Sengrloun Phonekeo
- Lao Tropical and Public Health Institute, Ministry of Health, Vientiane, Lao People’s Democratic Republic (PDR)
| | - Jordyn Wallenborn
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Latsamy Siengsounthone
- Lao Tropical and Public Health Institute, Ministry of Health, Vientiane, Lao People’s Democratic Republic (PDR)
| | - Günther Fink
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Peter Odermatt
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Sengchanh Kounnavong
- Lao Tropical and Public Health Institute, Ministry of Health, Vientiane, Lao People’s Democratic Republic (PDR)
| | - Somphou Sayasone
- Lao Tropical and Public Health Institute, Ministry of Health, Vientiane, Lao People’s Democratic Republic (PDR)
| | - Pascale Vonaesch
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
20
|
Reidy PT, Smith AD, Jevnikar BE, Doctor AK, Williams RW, Kachulkin AA, Monnig JM, Fix DK, Petrocelli JJ, Mahmassani ZS, McKenzie AI, de Hart NMMP, Drummond MJ. Muscle disuse as hindlimb unloading in early postnatal mice negatively impacts grip strength in adult mice: a pilot study. J Appl Physiol (1985) 2023; 134:787-798. [PMID: 36759163 PMCID: PMC10042595 DOI: 10.1152/japplphysiol.00681.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/24/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Physical inactivity has many detrimental effects on health, yet the impact of physical inactivity in early life on muscle health in adulthood remains unknown. Early postnatal malnutrition has prolonged effects into adulthood and we propose that early postnatal (P) physical inactivity would have similar negative effects. To test this hypothesis, we exposed postnatal mice (∼P28, C57BL/6J) to 14 days of physical inactivity (shortly after weaning, from ∼P28 to P42 days of age) in the form of muscle disuse with hindlimb unloading (HU). After this early-life physical inactivity, they were allowed to normally ambulate until 5 mo of age (P140, adulthood) when they underwent 14 days of HU with and without 7-day recovery. They were then tested for physical function (grip strength) and muscles were extracted and weighed. Immunofluorescence was carried out on these muscle cross sections for analysis of myofiber cross-sectional area (fCSA), macrophage density (CD68+ cells), and extracellular matrix (ECM) area. Muscle weights and fCSA and myofiber diameter were used to quantify changes in muscle and fiber size. Compared with age-matched controls, no notable effects of early-life physical inactivity (HU) on skeletal muscle and myofiber size were observed. However, a significant reduction in adult grip strength was observed in those exposed to HU early in life. This was associated with reduced muscle macrophages and increased ECM area. Exposure to a short period of early life disuse has negative enduring effects into adulthood impacting grip strength, muscle macrophages, and muscle composition as low muscle quality.NEW & NOTEWORTHY We demonstrate that early life disuse resulted in less grip strength in adulthood. Analysis of muscle composition demonstrated no loss of whole muscle or myofiber size indicating lower muscle quality akin to premature aging. This poor muscle quality was characterized by altered muscle macrophages and extracellular matrix area. We demonstrate intriguing correlations between this loss of grip strength and muscle macrophages and also area of noncontractile tissue in the muscle.
Collapse
Affiliation(s)
- Paul T Reidy
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Austin D Smith
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Benjamin E Jevnikar
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Abbas K Doctor
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Ryan W Williams
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Anthony A Kachulkin
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Jackie M Monnig
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, United States
| | - Dennis K Fix
- Department of Physical Therapy & Athletic Training, University of Utah, Salt Lake City, Utah, United States
| | - Jonathan J Petrocelli
- Department of Physical Therapy & Athletic Training, University of Utah, Salt Lake City, Utah, United States
| | - Ziad S Mahmassani
- Department of Physical Therapy & Athletic Training, University of Utah, Salt Lake City, Utah, United States
| | - Alec I McKenzie
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Naomi M M P de Hart
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Micah J Drummond
- Department of Physical Therapy & Athletic Training, University of Utah, Salt Lake City, Utah, United States
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
21
|
Alfano R, Zugna D, Barros H, Bustamante M, Chatzi L, Ghantous A, Herceg Z, Keski-Rahkonen P, de Kok TM, Nawrot TS, Relton CL, Robinson O, Roumeliotaki T, Scalbert A, Vrijheid M, Vineis P, Richiardi L, Plusquin M. Cord blood epigenome-wide meta-analysis in six European-based child cohorts identifies signatures linked to rapid weight growth. BMC Med 2023; 21:17. [PMID: 36627699 PMCID: PMC9831885 DOI: 10.1186/s12916-022-02685-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/29/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Rapid postnatal growth may result from exposure in utero or early life to adverse conditions and has been associated with diseases later in life and, in particular, with childhood obesity. DNA methylation, interfacing early-life exposures and subsequent diseases, is a possible mechanism underlying early-life programming. METHODS Here, a meta-analysis of Illumina HumanMethylation 450K/EPIC-array associations of cord blood DNA methylation at single CpG sites and CpG genomic regions with rapid weight growth at 1 year of age (defined with reference to WHO growth charts) was conducted in six European-based child cohorts (ALSPAC, ENVIRONAGE, Generation XXI, INMA, Piccolipiù, and RHEA, N = 2003). The association of gestational age acceleration (calculated using the Bohlin epigenetic clock) with rapid weight growth was also explored via meta-analysis. Follow-up analyses of identified DNA methylation signals included prediction of rapid weight growth, mediation of the effect of conventional risk factors on rapid weight growth, integration with transcriptomics and metabolomics, association with overweight in childhood (between 4 and 8 years), and comparison with previous findings. RESULTS Forty-seven CpGs were associated with rapid weight growth at suggestive p-value <1e-05 and, among them, three CpGs (cg14459032, cg25953130 annotated to ARID5B, and cg00049440 annotated to KLF9) passed the genome-wide significance level (p-value <1.25e-07). Sixteen differentially methylated regions (DMRs) were identified as associated with rapid weight growth at false discovery rate (FDR)-adjusted/Siddak p-values < 0.01. Gestational age acceleration was associated with decreasing risk of rapid weight growth (p-value = 9.75e-04). Identified DNA methylation signals slightly increased the prediction of rapid weight growth in addition to conventional risk factors. Among the identified signals, three CpGs partially mediated the effect of gestational age on rapid weight growth. Both CpGs (N=3) and DMRs (N=3) were associated with differential expression of transcripts (N=10 and 7, respectively), including long non-coding RNAs. An AURKC DMR was associated with childhood overweight. We observed enrichment of CpGs previously reported associated with birthweight. CONCLUSIONS Our findings provide evidence of the association between cord blood DNA methylation and rapid weight growth and suggest links with prenatal exposures and association with childhood obesity providing opportunities for early prevention.
Collapse
Affiliation(s)
- Rossella Alfano
- Medical Research Council Centre for Environment and Health, Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, UK
- Centre for Environmental Sciences, Hasselt University, Agoralaan Building D, 3590, Diepenbeek, Belgium
| | - Daniela Zugna
- Department of Medical Sciences, University of Turin and CPO-Piemonte, Turin, Italy
| | - Henrique Barros
- Institute of Public Health, University of Porto, Porto, Portugal
| | - Mariona Bustamante
- ISGlobal, Institute of Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Leda Chatzi
- Department of Preventive Medicine, University of Southern California, Los Angeles, USA
| | - Akram Ghantous
- International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, 69008, Lyon, France
| | - Zdenko Herceg
- International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, 69008, Lyon, France
| | - Pekka Keski-Rahkonen
- International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, 69008, Lyon, France
| | - Theo M de Kok
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, Agoralaan Building D, 3590, Diepenbeek, Belgium
| | - Caroline L Relton
- Μedical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Oliver Robinson
- Medical Research Council Centre for Environment and Health, Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, UK
- Mohn Centre for Children's Health and Well-being, The School of Public Health, Imperial College London, London, UK
| | - Theano Roumeliotaki
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Greece
| | - Augustin Scalbert
- International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, 69008, Lyon, France
| | - Martine Vrijheid
- International Agency for Research on Cancer (IARC), 150 Cours Albert Thomas, 69008, Lyon, France
| | - Paolo Vineis
- Medical Research Council Centre for Environment and Health, Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, UK
| | - Lorenzo Richiardi
- Department of Medical Sciences, University of Turin and CPO-Piemonte, Turin, Italy
| | - Michelle Plusquin
- Centre for Environmental Sciences, Hasselt University, Agoralaan Building D, 3590, Diepenbeek, Belgium.
| |
Collapse
|
22
|
Robertson OC, Marceau K, Moding KJ, Knopik VS. Developmental pathways linking obesity risk and early puberty: The thrifty phenotype and fetal overnutrition hypotheses. DEVELOPMENTAL REVIEW 2022. [DOI: 10.1016/j.dr.2022.101048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
23
|
Baldassarre ME, Panza R, Cresi F, Salvatori G, Corvaglia L, Aceti A, Giannì ML, Liotto N, Ilardi L, Laforgia N, Maggio L, Lionetti P, Agostoni C, Orfeo L, Di Mauro A, Staiano A, Mosca F. Complementary feeding in preterm infants: a position paper by Italian neonatal, paediatric and paediatric gastroenterology joint societies. Ital J Pediatr 2022; 48:143. [PMID: 35932061 PMCID: PMC9354266 DOI: 10.1186/s13052-022-01275-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/04/2022] [Indexed: 11/23/2022] Open
Abstract
Nutrition in the first 1000 days of life is essential to ensure appropriate growth rates, prevent adverse short- and long-term outcomes, and allow physiologic neurocognitive development. Appropriate management of early nutritional needs is particularly crucial for preterm infants. Although the impact of early nutrition on health outcomes in preterm infants is well established, evidence-based recommendations on complementary feeding for preterm neonates and especially extremely low birth weight and extremely low gestational age neonates are still lacking. In the present position paper we performed a narrative review to summarize current evidence regarding complementary feeding in preterm neonates and draw recommendation shared by joint societies (SIP, SIN and SIGENP) for paediatricians, healthcare providers and families with the final aim to reduce the variability of attitude and timing among professionals.
Collapse
Affiliation(s)
- Maria Elisabetta Baldassarre
- Department of Biomedical Science and Human Oncology, Section of Neonatology and Neonatal Intensive Care Unit, "Aldo Moro" University of Bari, Policlinico Hospital - Piazza Giulio Cesare n. 11, 70124, Bari, Italy
| | - Raffaella Panza
- Department of Biomedical Science and Human Oncology, Section of Neonatology and Neonatal Intensive Care Unit, "Aldo Moro" University of Bari, Policlinico Hospital - Piazza Giulio Cesare n. 11, 70124, Bari, Italy. .,Neonatology and Neonatal Intensive Care Unit, "A. Perrino" Hospital, Brindisi, Italy.
| | - Francesco Cresi
- Neonatology and Neonatal Intensive Care Unit, Department of Public Health and Paediatrics, University of Turin, Torino, Italy
| | - Guglielmo Salvatori
- Neonatal Intensive Care Unit and Human Milk Bank, Department of Neonatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Luigi Corvaglia
- Department of Medical and Surgical Sciences - University of Bologna, Neonatal Intensive Care Unit - IRCCS AOUBO, Bologna, Italy
| | - Arianna Aceti
- Department of Medical and Surgical Sciences - University of Bologna, Neonatal Intensive Care Unit - IRCCS AOUBO, Bologna, Italy
| | - Maria Lorella Giannì
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, 20122, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy
| | - Nadia Liotto
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, 20122, Milan, Italy
| | - Laura Ilardi
- Neonatology and Neonatal Intensive Care Unit ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Nicola Laforgia
- Department of Interdisciplinary Medicine - Section of Neonatology and Neonatal Intensive Care Unit, "Aldo Moro" University of Bari, 70124, Bari, Italy
| | - Luca Maggio
- UOC Neonatology and Neonatal Intensive Care Unit, AO San Camillo Forlanini, Rome, Italy
| | - Paolo Lionetti
- Gastroenterology Unit, NEUROFARBA Department, University of Florence, Meyer Children's Hospital, Florence, Italy
| | - Carlo Agostoni
- Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Paediatric Intermediate Care Unit, Milan, Italy
| | - Luigi Orfeo
- Neonatal Intensive Care Unit, "San Giovanni Calibita Fatebenefratelli" Hospital, Isola Tiberina, Rome, Italy
| | - Antonio Di Mauro
- Paediatric Primary Care, National Paediatric Health Care System, Via Conversa 12, Margherita di Savoia, BT, Italy
| | - Annamaria Staiano
- Department of Translational Medical Science, Section of Paediatrics, University of Naples "Federico II", Naples, Italy
| | - Fabio Mosca
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, 20122, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy
| | | |
Collapse
|
24
|
Alfano R, Plusquin M, Robinson O, Brescianini S, Chatzi L, Keski-Rahkonen P, Handakas E, Maitre L, Nawrot T, Robinot N, Roumeliotaki T, Sassi F, Scalbert A, Vrijheid M, Vineis P, Richiardi L, Zugna D. Cord blood metabolites and rapid postnatal growth as multiple mediators in the prenatal propensity to childhood overweight. Int J Obes (Lond) 2022; 46:1384-1393. [PMID: 35508813 PMCID: PMC9239910 DOI: 10.1038/s41366-022-01108-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND The mechanisms underlying childhood overweight and obesity are poorly known. Here, we investigated the direct and indirect effects of different prenatal exposures on offspring rapid postnatal growth and overweight in childhood, mediated through cord blood metabolites. Additionally, rapid postnatal growth was considered a potential mediator on childhood overweight, alone and sequentially to each metabolite. METHODS Within four European birth-cohorts (N = 375 mother-child dyads), information on seven prenatal exposures (maternal education, pre-pregnancy BMI, weight gain and tobacco smoke during pregnancy, age at delivery, parity, and child gestational age), selected as obesogenic according to a-priori knowledge, was collected. Cord blood levels of 31 metabolites, associated with rapid postnatal growth and/or childhood overweight in a previous study, were measured via liquid-chromatography-quadrupole-time-of-flight-mass-spectrometry. Rapid growth at 12 months and childhood overweight (including obesity) between four and eight years were defined with reference to WHO growth charts. Single mediation analysis was performed using the imputation approach and multiple mediation analysis using the extended-imputation approach. RESULTS Single mediation suggested that the effect of maternal education, pregnancy weight gain, parity, and gestational age on rapid postnatal growth but not on childhood overweight was partly mediated by seven metabolites, including cholestenone, decenoylcarnitine(C10:1), phosphatidylcholine(C34:3), progesterone and three unidentified metabolites; and the effect of gestational age on childhood overweight was mainly mediated by rapid postnatal growth. Multiple mediation suggested that the effect of gestational age on childhood overweight was mainly mediated by rapid postnatal growth and that the mediating role of the metabolites was marginal. CONCLUSION Our findings provide evidence of the involvement of in utero metabolism in the propensity to rapid postnatal growth and of rapid postnatal growth in the propensity to childhood overweight. We did not find evidence supporting a mediating role of the studied metabolites alone between the studied prenatal exposures and the propensity to childhood overweight.
Collapse
Affiliation(s)
- Rossella Alfano
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium.
- Μedical Research Council-Health Protection Agency Centre for Environment and Health, Imperial College London, London, UK.
| | - Michelle Plusquin
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Oliver Robinson
- Μedical Research Council-Health Protection Agency Centre for Environment and Health, Imperial College London, London, UK
| | - Sonia Brescianini
- Centre for Behavioural Science and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Lida Chatzi
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Pekka Keski-Rahkonen
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Evangelos Handakas
- Μedical Research Council-Health Protection Agency Centre for Environment and Health, Imperial College London, London, UK
| | - Lea Maitre
- Barcelona Institute of Global Health (ISGlobal), Barcelona, Spain
| | - Tim Nawrot
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Nivonirina Robinot
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Theano Roumeliotaki
- Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Greece
| | - Franco Sassi
- Centre for Health Economics & Policy Innovation, Department of Economics & Public Policy, Imperial College Business School, South Kensington Campus, London, UK
| | - Augustin Scalbert
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Martine Vrijheid
- Barcelona Institute of Global Health (ISGlobal), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Paolo Vineis
- Μedical Research Council-Health Protection Agency Centre for Environment and Health, Imperial College London, London, UK
| | - Lorenzo Richiardi
- Cancer Epidemiology Unit, Department of Medical Sciences, University of Turin and CPO-Piemonte, Torino, Italy
| | - Daniela Zugna
- Cancer Epidemiology Unit, Department of Medical Sciences, University of Turin and CPO-Piemonte, Torino, Italy
| |
Collapse
|
25
|
Tan KML, Tint MT, Kothandaraman N, Michael N, Sadananthan SA, Velan SS, Fortier MV, Yap F, Tan KH, Gluckman PD, Chong YS, Chong MFF, Lee YS, Godfrey KM, Eriksson JG, Cameron-Smith D. The Kynurenine Pathway Metabolites in Cord Blood Positively Correlate With Early Childhood Adiposity. J Clin Endocrinol Metab 2022; 107:e2464-e2473. [PMID: 35150259 PMCID: PMC9113811 DOI: 10.1210/clinem/dgac078] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT The kynurenine pathway generates metabolites integral to energy metabolism, neurotransmission, and immune function. Circulating kynurenine metabolites positively correlate with adiposity in children and adults, yet it is not known whether this relationship is present already at birth. OBJECTIVE In this prospective longitudinal study, we investigate the relationship between cord blood kynurenine metabolites and measures of adiposity from birth to 4.5 years. METHODS Liquid chromatography-tandem mass spectrometry was used to quantify cord blood kynurenine metabolites in 812 neonates from the Growing Up in Singapore Towards healthy Outcomes (GUSTO) study. Fat percentage was measured by air displacement plethysmography and abdominal adipose tissue compartment volumes; superficial (sSAT) and deep subcutaneous (dSAT) and internal adipose tissue were quantified by magnetic resonance imaging at early infancy in a smaller subset of neonates, and again at 4 to 4.5 years of age. RESULTS Cord blood kynurenine metabolites appeared to be higher in female newborns, higher in Indian newborns compared with Chinese newborns, and higher in infants born by cesarean section compared with vaginal delivery. Kynurenine, xanthurenic acid, and quinolinic acid were positively associated with birthweight, but not with subsequent weight during infancy and childhood. Quinolinic acid was positively associated with sSAT at birth. Kynurenic acid and quinolinic acid were positively associated with fat percentage at 4 years. CONCLUSION Several cord blood kynurenine metabolite concentrations were positively associated with birthweight, with higher kynurenic acid and quinolinic acid correlating to higher percentage body fat in childhood, suggesting these cord blood metabolites as biomarkers of early childhood adiposity.
Collapse
Affiliation(s)
- Karen Mei-Ling Tan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 117609, Singapore
- Department of Laboratory Medicine, National University Hospital, 119074, Singapore
| | - Mya-Thway Tint
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 117609, Singapore
- Department of Obstetrics and Gynaecology, Human Potential Translational Research Programme, Yong Loo Lin School of Medicine (YLLSOM), National University of Singapore, 119228, Singapore
| | - Narasimhan Kothandaraman
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 117609, Singapore
| | - Navin Michael
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 117609, Singapore
| | - Suresh Anand Sadananthan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 117609, Singapore
| | - S Sendhil Velan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 117609, Singapore
- Institute of Bioengineering and Bioimaging (IBB), Agency for Science Technology and Research, 138669, Singapore
| | - Marielle V Fortier
- Department of Diagnostic and Interventional Imaging, KK Women’s and Children’s Hospital, 229899, Singapore
| | - Fabian Yap
- Duke-National University of Singapore (NUS) Medical School, 169857, Singapore
- Department of Pediatric Endocrinology, KK Women’s and Children’s Hospital, 229899, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore
| | - Kok Hian Tan
- Duke-National University of Singapore (NUS) Medical School, 169857, Singapore
- Perinatal Audit and Epidemiology, Department of Maternal Fetal Medicine, KK Women’s and Children’s Hospital, 119228, Singapore
| | - Peter D Gluckman
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 117609, Singapore
- Liggins Institute, University of Auckland, Auckland 1023, New Zealand
| | - Yap-Seng Chong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 117609, Singapore
- Department of Obstetrics and Gynaecology, Human Potential Translational Research Programme, Yong Loo Lin School of Medicine (YLLSOM), National University of Singapore, 119228, Singapore
- Yong Loo Lin School of Medicine (YLLSOM), National University of Singapore, 117597, Singapore
| | - Mary F F Chong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 117609, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, 117549, Singapore
| | - Yung Seng Lee
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 117609, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore
- Khoo Teck Puat – National University Children’s Medical Institute, National University Health System, 119074, Singapore
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton SO16 6YD, United Kingdom
- NIHR Southampton Biomedical Research Centre, University of Southampton Hospital, Southampton SO16 6YD, United Kingdom
| | - Johan G Eriksson
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 117609, Singapore
- Department of Obstetrics and Gynaecology, Human Potential Translational Research Programme, Yong Loo Lin School of Medicine (YLLSOM), National University of Singapore, 119228, Singapore
- Folkhälsan Research Center, 00250 Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, 00290 Helsinki, Finland
| | - David Cameron-Smith
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), 117609, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117596, Singapore
- Correspondence: Professor David Cameron Smith, Singapore Institute for Clinical Sciences, Brenner Centre for Molecular Medicine, Agency for Science, Technology and Research, 30 Medical Drive 117609, Singapore.
| |
Collapse
|
26
|
Billah MM, Khatiwada S, Morris MJ, Maloney CA. Effects of paternal overnutrition and interventions on future generations. Int J Obes (Lond) 2022; 46:901-917. [PMID: 35022547 PMCID: PMC9050512 DOI: 10.1038/s41366-021-01042-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023]
Abstract
In the last two decades, evidence from human and animal studies suggests that paternal obesity around the time of conception can have adverse effects on offspring health through developmental programming. This may make significant contributions to the current epidemic of obesity and related metabolic and reproductive complications like diabetes, cardiovascular disease, and subfertility/infertility. To date, changes in seminal fluid composition, sperm DNA methylation, histone composition, small non-coding RNAs, and sperm DNA damage have been proposed as potential underpinning mechanism to program offspring health. In this review, we discuss current human and rodent evidence on the impact of paternal obesity/overnutrition on offspring health, followed by the proposed mechanisms, with a focus on sperm DNA damage underpinning paternal programming. We also summarize the different intervention strategies implemented to minimize effects of paternal obesity. Upon critical review of literature, we find that obesity-induced altered sperm quality in father is linked with compromised offspring health. Paternal exercise intervention before conception has been shown to improve metabolic health. Further work to explore the mechanisms underlying benefits of paternal exercise on offspring are warranted. Conversion to healthy diets and micronutrient supplementation during pre-conception have shown some positive impacts towards minimizing the impact of paternal obesity on offspring. Pharmacological approaches e.g., metformin are also being applied. Thus, interventions in the obese father may ameliorate the potential detrimental impacts of paternal obesity on offspring.
Collapse
Affiliation(s)
| | - Saroj Khatiwada
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Margaret J Morris
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | | |
Collapse
|
27
|
Xiao P, Goodarzi P, Pezeshki A, Hagen DE. RNA-seq reveals insights into molecular mechanisms of metabolic restoration via tryptophan supplementation in low birth weight piglet model. J Anim Sci 2022; 100:skac156. [PMID: 35552417 PMCID: PMC9155244 DOI: 10.1093/jas/skac156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/26/2022] [Indexed: 11/23/2022] Open
Abstract
Low birth weight (LBW) is associated with metabolic disorders in early life. While dietary l-tryptophan (Trp) can ameliorate postprandial plasma triglycerides (TG) disposal in LBW piglets, the genetic and biological basis underlying Trp-caused alterations in lipid metabolism is poorly understood. In this study, we collected 24 liver samples from 1-mo-old LBW and normal birth weight (NBW) piglets supplemented with different concentrations of dietary Trp (NBW with 0% Trp, N0; LBW with 0% Trp, L0; LBW with 0.4% Trp, L4; LBW with 0.8% Trp, L8; N = 6 in each group.) and conducted systematic, transcriptome-wide analysis using RNA sequencing (RNA-seq). We identified 39 differentially expressed genes (DEG) between N0 and L0, and genes within "increased dose effect" clusters based on dose-series expression profile analysis, enriched in fatty acid response of gene ontology (GO) biological process (BP). We then identified RNA-binding proteins including SRSF1, DAZAP1, PUM2, PCBP3, IGF2BP2, and IGF2BP3 significantly (P < 0.05) enriched in alternative splicing events (ASE) in comparison with L0 as control. There were significant positive and negative relationships between candidate genes from co-expression networks (including PID1, ANKRD44, RUSC1, and CYP2J34) and postprandial plasma TG concentration. Further, we determined whether these candidate hub genes were also significantly associated with metabolic and cardiovascular traits in humans via human phenome-wide association study (Phe-WAS), and analysis of mammalian orthologs suggests a functional conservation between human and pig. Our work demonstrates that transcriptomic changes during dietary Trp supplementation in LBW piglets. We detected candidate genes and related BP that may play roles on lipid metabolism restoration. These findings will help to better understand the amino acid support in LBW metabolic complications.
Collapse
Affiliation(s)
- Ping Xiao
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Parniyan Goodarzi
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Adel Pezeshki
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Darren E Hagen
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
28
|
Córdoba-Rodríguez DP, Iglesia I, Gómez-Bruton A, Álvarez Sauras ML, Miguel-Berges ML, Flores-Barrantes P, Casajús JA, Moreno LA, Rodríguez G. Early Life Factors Associated with Lean Body Mass in Spanish Children: CALINA Study. CHILDREN 2022; 9:children9050585. [PMID: 35626762 PMCID: PMC9139173 DOI: 10.3390/children9050585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 11/16/2022]
Abstract
Early life is critical for the programming of body composition. The literature links perinatal factors with fat mass development and its future effects (e.g., obesity); however, little evidence exists between early life factors and lean body mass (LBM). This study follows up on a cohort of 416 Spanish children at ages six to eight, previously evaluated at birth in the CALINA study. Here, we studied the association between early life factors, LBM, and limb strength. Parental origin/nutritional status, maternal smoking during pregnancy, gestational diabetes/weight gain/age, birth weight (BW), early feeding, and rapid weight gain (RWG) were collected from primary care records. Bioimpedance analysis, dual-energy X-ray absorptiometry, peripheral quantitative computed tomography, and a handgrip/standing long jump test were used to assess fat-free mass index (FFMI), total lean soft tissue mass index (TLSTMI), muscle cross-sectional area index (MCSAI), and limb strength, respectively. In girls, maternal smoking, gestational age, and BW were positively associated with FFM/LSTM. In boys, the parents’ BMI, BW, and RWG were positively associated with FFM/LSTM. BW was associated with handgrip strength in both. Maternal BMI in girls and RWG in boys were negatively associated with the standing long jump. Early life programming plays a key role in determining LBM in children.
Collapse
Affiliation(s)
- Diana Paola Córdoba-Rodríguez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia;
| | - Iris Iglesia
- Growth, Exercise, Nutrition and Development (GENUD) Research Group, Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza, 50009 Zaragoza, Spain; (A.G.-B.); (M.L.M.-B.); (P.F.-B.); (J.A.C.); (L.A.M.); (G.R.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Red de Salud Materno Infantil y del Desarrollo (SAMID), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS), RD21/0012/0012, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-876843756
| | - Alejandro Gómez-Bruton
- Growth, Exercise, Nutrition and Development (GENUD) Research Group, Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza, 50009 Zaragoza, Spain; (A.G.-B.); (M.L.M.-B.); (P.F.-B.); (J.A.C.); (L.A.M.); (G.R.)
- Departamento de Fisiatría y Enfermería, Facultad de Ciencias de la Salud y del Deporte (FCSD), Universidad de Zaragoza, 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III Madrid, 28029 Madrid, Spain
| | | | - María L. Miguel-Berges
- Growth, Exercise, Nutrition and Development (GENUD) Research Group, Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza, 50009 Zaragoza, Spain; (A.G.-B.); (M.L.M.-B.); (P.F.-B.); (J.A.C.); (L.A.M.); (G.R.)
| | - Paloma Flores-Barrantes
- Growth, Exercise, Nutrition and Development (GENUD) Research Group, Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza, 50009 Zaragoza, Spain; (A.G.-B.); (M.L.M.-B.); (P.F.-B.); (J.A.C.); (L.A.M.); (G.R.)
| | - José Antonio Casajús
- Growth, Exercise, Nutrition and Development (GENUD) Research Group, Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza, 50009 Zaragoza, Spain; (A.G.-B.); (M.L.M.-B.); (P.F.-B.); (J.A.C.); (L.A.M.); (G.R.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Departamento de Fisiatría y Enfermería, Facultad de Ciencias de la Salud y del Deporte (FCSD), Universidad de Zaragoza, 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III Madrid, 28029 Madrid, Spain
| | - Luis A. Moreno
- Growth, Exercise, Nutrition and Development (GENUD) Research Group, Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza, 50009 Zaragoza, Spain; (A.G.-B.); (M.L.M.-B.); (P.F.-B.); (J.A.C.); (L.A.M.); (G.R.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III Madrid, 28029 Madrid, Spain
| | - Gerardo Rodríguez
- Growth, Exercise, Nutrition and Development (GENUD) Research Group, Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza, 50009 Zaragoza, Spain; (A.G.-B.); (M.L.M.-B.); (P.F.-B.); (J.A.C.); (L.A.M.); (G.R.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Red de Salud Materno Infantil y del Desarrollo (SAMID), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Área de Pediatría, Universidad de Zaragoza, 50009 Zaragoza, Spain
| |
Collapse
|
29
|
Infant feeding practices associated with adiposity peak and rebound in the EDEN mother-child cohort. Int J Obes (Lond) 2022; 46:809-816. [PMID: 34980907 DOI: 10.1038/s41366-021-01059-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 12/01/2021] [Accepted: 12/21/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND/OBJECTIVE High magnitude of adiposity peak and early adiposity rebound are early risk markers of later obesity. Infant diet represents one of the main modifiable determinants of early growth. This study aimed to investigate the association between infant feeding practices and age and magnitude of adiposity peak and rebound. SUBJECTS/METHODS Analyses were based on data from the French EDEN mother-child cohort. Data on breastfeeding and complementary feeding were collected at birth and 4, 8, and 12 months. From clinical examinations and measurements collected in the child's health booklet up to 12 years, individual growth curves were modeled, and ages and magnitudes of adiposity peak and rebound were estimated. Associations between infant feeding practices and growth were investigated by multivariable linear regression in children after testing a child-sex interaction. RESULTS In the studied population (n = 1225), adiposity peak occurred at a mean of 9.9 ± 2 months and adiposity rebound at 5.5 ± 1.4 years. Associations between infant feeding practices and adiposity peak or rebound were moderated by child sex. For girls, each additional month of breastfeeding was related to a 2-day increase in the age at adiposity peak (p < 0.001), and an 18-day increase in the age at adiposity peak (p = 0.004). Whereas for boys, each additional month for the age at complementary food introduction was associated with a 29-day increase in the age at adiposity rebound (p = 0.02). For boys, long breastfeeding duration was only related to reduced body mass index at adiposity peak. CONCLUSIONS Child sex has a moderating effect on the association between infant feeding practices and adiposity peak or rebound. The well-known association between breastfeeding duration and early growth seems stronger in girls than boys. The association found for complementary feeding in boys may give new insights into preventing obesity.
Collapse
|
30
|
Chen M, Ma Y, Ma T, Li Y, Gao D, Chen L, Liu J, Zhang Y, Jiang J, Wang X, Dong Y, Ma J. The association between growth patterns and blood pressure in children and adolescents: A cross-sectional study of seven provinces in China. J Clin Hypertens (Greenwich) 2021; 23:2053-2064. [PMID: 34847290 PMCID: PMC8696227 DOI: 10.1111/jch.14393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/12/2021] [Accepted: 11/02/2021] [Indexed: 01/22/2023]
Abstract
Aimed to investigate the associations between different growth patterns with high blood pressure, and further examine the mediation effect of BMI between growth patterns and high blood pressure among children and adolescents. A total of 31581 children and adolescents aged 7-18 years were selected based on the stratified cluster sampling method. Logistics regression models were used to calculate the odds rations (ORs) and 95% confidence interval (95%CI) of the association between different growth patterns and high blood pressure. Mediation effect analyses were applied to estimate the effect of BMI on the increase of blood pressure levels in different growth patterns. In different sex and ages, compared to reference group of normal growth, blood pressure levels and prevalence of high blood pressure of the catch-up growth were higher, but that of the catch-down growth were lower. The prevalence of high blood pressure was 11.69%, 16.06%, and 9.68% in normal growth, catch-up growth, and catch-down growth, respectively. In total, compared with the normal growth pattern, the ORs (95%CI) of high blood pressure, high systolic blood pressure and high diastolic blood pressure in the catch-up growth were 1.171(1.073,1.280), 1.110(1.001,1.230) and 1.141(1.025,1.270) (p < .05), respectively. Additionally, the mediation effect of current BMI existed in the association between blood pressure levels and different growth patterns, particularly in boys. Our findings suggested that different growth patterns after birth could modify blood pressure, and the potential risks of high blood pressure could be increased by catch-up growth at childhood and adolescence.
Collapse
Affiliation(s)
- Manman Chen
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Ying Ma
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Tao Ma
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Yanhui Li
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Di Gao
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Li Chen
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Jieyu Liu
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Yi Zhang
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Jun Jiang
- Department of Plant Science and Landscape ArchitectureUniversity of MarylandCollege ParkMarylandUSA
| | - Xinxin Wang
- School of Public Health and ManagementKey Laboratory of Environmental Factors and Chronic Disease ControlNingxia Medical UniversityNingxiaChina
| | - Yanhui Dong
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| | - Jun Ma
- School of Public HealthNational Health Commission Key Laboratory of Reproductive HealthInstitute of Child and Adolescent HealthPeking UniversityBeijingChina
| |
Collapse
|
31
|
Gibbs RL, Yates DT. The Price of Surviving on Adrenaline: Developmental Programming Responses to Chronic Fetal Hypercatecholaminemia Contribute to Poor Muscle Growth Capacity and Metabolic Dysfunction in IUGR-Born Offspring. FRONTIERS IN ANIMAL SCIENCE 2021; 2:769334. [PMID: 34966907 PMCID: PMC8713512 DOI: 10.3389/fanim.2021.769334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Maternofetal stress induces fetal programming that restricts skeletal muscle growth capacity and metabolic function, resulting in intrauterine growth restriction (IUGR) of the fetus. This thrifty phenotype aids fetal survival but also yields reduced muscle mass and metabolic dysfunction after birth. Consequently, IUGR-born individuals are at greater lifelong risk for metabolic disorders that reduce quality of life. In livestock, IUGR-born animals exhibit poor growth efficiency and body composition, making these animals more costly and less valuable. Specifically, IUGR-associated programming causes a greater propensity for fat deposition and a reduced capacity for muscle accretion. This, combined with metabolic inefficiency, means that these animals produce less lean meat from greater feed input, require more time on feed to reach market weight, and produce carcasses that are of less quality. Despite the health and economic implications of IUGR pathologies in humans and food animals, knowledge regarding their specific underlying mechanisms is lacking. However, recent data indicate that adaptive programing of adrenergic sensitivity in multiple tissues is a contributing factor in a number of IUGR pathologies including reduced muscle mass, peripheral insulin resistance, and impaired glucose metabolism. This review highlights the findings that support the role for adrenergic programming and how it relates to the lifelong consequences of IUGR, as well as how dysfunctional adrenergic signaling pathways might be effective targets for improving outcomes in IUGR-born offspring.
Collapse
Affiliation(s)
- Rachel L. Gibbs
- Stress Physiology Laboratory, Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Dustin T. Yates
- Stress Physiology Laboratory, Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
32
|
Abstract
The observation that 64% of English adults are overweight or obese despite a rising prevalence in weight-loss attempts suggests our understanding of energy balance is fundamentally flawed. Weight-loss is induced through a negative energy balance; however, we typically view weight change as a static function, in that energy intake and energy expenditure are independent variables, resulting in a fixed rate of weight-loss assuming a constant energy deficit. Such static modelling provides the basis for the clinical assumption that a 14644 kJ (3500 kcal) deficit translates to a 1 lb weight-loss. However, this '3500 kcal (14644 kJ) rule' is consistently shown to significantly overestimate weight-loss. Static modelling disregards obligatory changes in energy expenditure associated with the loss of metabolically active tissue, i.e. skeletal muscle. Additionally, it disregards the presence of adaptive thermogenesis, the underfeeding-associated fall in resting energy expenditure beyond that caused by loss of fat-free mass. This metabolic manipulation of energy expenditure is observed from the onset of energy restriction to maintain weight at a genetically pre-determined set point. As a result, the observed magnitude of weight-loss is disproportionally less, followed by earlier weight plateau, despite strict compliance to a dietary intervention. By simulating dynamic changes in energy expenditure associated with underfeeding, mathematical modelling may provide a more accurate method of weight-loss prediction. However, accuracy at an individual level is limited due to difficulty estimating energy requirements, physical activity and dietary intake in free-living individuals. In the present paper, we aim to outline the contribution of dynamic changes in energy expenditure to weight-loss resistance and weight plateau.
Collapse
|
33
|
Singer D, Thiede LP, Perez A. Adults Born Preterm: Long-Term Health Risks of Former Very Low Birth Weight Infants. DEUTSCHES ARZTEBLATT INTERNATIONAL 2021; 118:521-527. [PMID: 33734986 DOI: 10.3238/arztebl.m2021.0164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/02/2020] [Accepted: 02/22/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Advances in neonatology now enable increasing numbers of very low birth weight neonates (<1500 g) to survive into early adulthood and beyond. What are the implications for their long-term care? METHODS Selective literature search on the outcome of very low birth weight neonates in adulthood ("adults born preterm"). RESULTS Robust data are available on the pulmonary, metabolic, cardiovascular, renal, neurocognitive, sensory-visual, social-emotional, mental, reproductive, and musculoskeletal long-term risks. On the somatic level, elevated rates have been documented for asthma (odds Ratio [OR] 2.37), diabetes mellitus (OR 1.54), and chronic renal disease (hazard ratio [HR] 3.01), along with the cardiovascular and cerebrovascular sequelae of a tendency toward arterial hypertension. On the psychosocial level, the main findings are deficits in romantic partnerships (OR 0.72) and a lower reproduction rate (relative risk [RR] male/female 0.24/0.33). The affected women also have an elevated risk of preterm delivery. CONCLUSION A risk profile with both somatic and psychosocial aspects can be discerned for adults who were born prematurely, even if some of these risks are present in low absolute numbers. As the ability to compensate for latent deficits declines with age, such adults may suffer from "premature aging as the late price of premature birth." A holistic approach to care with personalized prevention strategies-which for most of them was discontinued at discharge from pediatric follow-up-therefore seems appropriate in adulthood as well.
Collapse
|
34
|
Gillen ZM, Mustad VA, Shoemaker ME, Mckay BD, Leutzinger TJ, Lopez-Pedrosa JM, Rueda R, Cramer JT. Impact of slow versus rapid digesting carbohydrates on substrate oxidation in pre-pubertal children: A randomized crossover trial. Clin Nutr 2021; 40:3718-3728. [PMID: 34130017 DOI: 10.1016/j.clnu.2021.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/21/2021] [Accepted: 05/04/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND & AIMS Consumption of rapid digesting sugars by children are under increased scrutiny because of their contribution to unhealthy weight gain. Previous studies in adults and children have suggested that altering the blend of carbohydrates (CHOs) consumed may cause shifts in substrate utilization. The purpose of this study was to examine the effects of consuming a slow digesting carbohydrate (SDC) and rapid digesting carbohydrate (RDC) on CHO and fat oxidation, glucose, and insulin responses at rest, during exercise, and post-exercise rest in pre-pubescent children. METHODS A randomized, double-blind, crossover design was used. Nineteen pre-pubescent children (n = 10 boys, n = 9 girls, mean ± standard error, age = 9.84 ± 0.37-yrs) participated. Visits to the laboratory began with a 30-min measurement of resting metabolism followed by consumption of either an RDC or SDC drink. Postprandial resting metabolism was recorded for 60-min, immediately followed by 60-min of submaximal cycling exercise while metabolism was recorded, which was immediately followed by another 60-min recording of post-exercise metabolism. Total CHO and fat oxidation, endogenous and exogenous CHO oxidation, blood glucose, and insulin were assessed. RESULTS Total CHO oxidation rate (g∙min-1) was greater after the RDC drink at 60 min (p = 0.032). Endogenous CHO oxidation rate (g∙min-1) was greater after the SDC drink at 15 min (p ≤ 0.010). Cumulative endogenous CHO oxidation (g) was greater after the SDC drink at 45 min (p = 0.009). Endogenous CHO oxidation accounted for a greater proportion of substrate oxidation after the first 60-min rest period (p = 0.028), while exogenous CHO oxidation accounted for a greater proportion of substrate oxidation for the RDC at all time points (p ≤ 0.019). CONCLUSIONS The present study provides novel data suggesting that an SDC promotes greater endogenous substrate utilization in pre-pubertal children, which may have beneficial health impacts on energy intake and carbohydrate regulation/metabolism during growth and development. CLINICAL TRIALS REGISTRY NUMBER NCT03185884, clinicaltrials.gov.
Collapse
Affiliation(s)
- Zachary M Gillen
- Department of Kinesiology, Mississippi State University, Mississippi State, MS 39762, USA
| | - Vikkie A Mustad
- Abbott Nutrition, 2900 Easton Square Place, Columbus, OH, 43219, USA
| | - Marni E Shoemaker
- College of Health Sciences, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Brianna D Mckay
- Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Todd J Leutzinger
- Department of Biomechanics, University of Nebraska Omaha, Omaha, NE, 68182, USA
| | | | | | - Joel T Cramer
- College of Health Sciences, The University of Texas at El Paso, El Paso, TX, 79968, USA.
| |
Collapse
|
35
|
Zhou S, Lin L, Bao Z, Meng T, Wang S, Chen G, Li Q, Liu Z, Bao H, Han N, Wang H, Guo Y. The association of prenatal exposure to particulate matter with infant growth: A birth cohort study in Beijing, China. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 277:116792. [PMID: 33721799 DOI: 10.1016/j.envpol.2021.116792] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 06/12/2023]
Abstract
Limited studies examined the associations of prenatal exposure to particulate matter (PM) and children's growth with inconsistent results, and no study focused on PM1. We matched a birth cohort (10,547 children) with daily PM1 and PM2.5 concentrations by maternal home addresses. Air pollution concentrations were predicted by satellite remote sensing data, meteorological factors, and land use information. The weight and length of children in the birth cohort were measured at approximately one year old. We calculated the Z-score of weight for length (WFL) and body mass index (BMI) and then defined overweight and obesity (OWOB) based on WHO Standards. Generalized linear regression and modified Poisson regression were used to identify the association of prenatal exposure to PM1 or PM2.5 with anthropometric measurements and risk of OWOB. We also determined the mediation effect of preterm birth on the associations. Results showed that a 10 μg/m3 increase in prenatal exposure to PM1 and PM2.5 was significantly associated with a 0.105 [95% confidence interval (CI): 0.067, 0.144] and 0.063 (95% CI: 0.029, 0.097) increase in WFL Z-score for one-year-old children. Similar associations were found for BMI Z-score. A 10 μg/m3 increase in prenatal PM1 and PM2.5 exposure was significantly associated with 1.012 (95%CI: 1.003, 1.021) and 1.010 (95%CI: 1.002, 1.018) times higher risk of OWOB. . Preterm birth mediated 7.5% [direct effect (DE) = 0.106, P < 0.001; indirect effect (IE) = 0.009, P < 0.001)] and 9.9% (DE = 0.064, P < 0.001; IE = 0.007, P < 0.001) of the association between prenatal PM1 and PM2.5 exposure and WFL Z-score of the children. The association of prenatal PM1 and PM2.5 exposure with BMI Z-score of children was also mediated by preterm birth by 6.6% (DE = 0.111, P < 0.001; IE = 0.008, P < 0.001) and 9.1% (DE = 0.064, P < 0.001; IE = 0.006, P < 0.001). These results remained robust in the sensitivity analyses. In conclusion, prenatal exposure to PM1 and PM2.5 increased WFL, BMI Z-scores and higher risk of OWOB for one-year-old children. The associations were partially mediated by preterm birth. These findings call for the urgent action on air pollution regulation to protect early-life health among children.
Collapse
Affiliation(s)
- Shuang Zhou
- Department of Maternal and Child Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Lizi Lin
- Department of Maternal and Child Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China; Guangdong Provincial Engineering Technology Research Center of Environmental and Health Risk Assessment; Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zheng Bao
- Tongzhou Maternal and Child Health Hospital, Beijing, 101101, China
| | - Tong Meng
- Tongzhou Maternal and Child Health Hospital, Beijing, 101101, China
| | - Shanshan Wang
- Tongzhou Maternal and Child Health Hospital, Beijing, 101101, China
| | - Gongbo Chen
- Guangdong Provincial Engineering Technology Research Center of Environmental and Health Risk Assessment; Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qin Li
- Department of Maternal and Child Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China; Reproductive Medical Centre, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China
| | - Zheng Liu
- Department of Maternal and Child Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Heling Bao
- Department of Maternal and Child Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Na Han
- Tongzhou Maternal and Child Health Hospital, Beijing, 101101, China
| | - Haijun Wang
- Department of Maternal and Child Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China.
| | - Yuming Guo
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| |
Collapse
|
36
|
Mazzoli A, Gatto C, Crescenzo R, Cigliano L, Iossa S. Prolonged Changes in Hepatic Mitochondrial Activity and Insulin Sensitivity by High Fructose Intake in Adolescent Rats. Nutrients 2021; 13:nu13041370. [PMID: 33921866 PMCID: PMC8073121 DOI: 10.3390/nu13041370] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 12/16/2022] Open
Abstract
Persistence of damage induced by unhealthy diets during youth has been little addressed. Therefore, we investigated the impact of a short-term fructose-rich diet on liver metabolic activity in adolescent rats and the putative persistence of alterations after removing fructose from the diet. Adolescent rats were fed a fructose-rich diet for three weeks and then switched to a control diet for further three weeks. Body composition and energy balance were not affected by fructose-rich diet, while increased body lipids and lipid gain were found after the rescue period. Switching to a control diet reversed the upregulation of plasma fructose, uric acid, lipocalin, and haptoglobin, while plasma triglycerides, alanine aminotransferase, lipopolysaccharide, and tumor necrosis factor alpha remained higher. Hepatic steatosis and ceramide were increased by fructose-rich diet, but reversed by returning to a control diet, while altered hepatic response to insulin persisted. Liver fatty acid synthase and stearoyl-CoA desaturase (SCD) activities were upregulated by fructose-rich diet, and SCD activity remained higher after returning to the control diet. Fructose-induced upregulation of complex II-driven mitochondrial respiration, peroxisome proliferator-activated receptor-gamma coactivator 1 alpha, and peroxisome proliferator activated receptor α also persisted after switching to control diet. In conclusion, our results show prolonged fructose-induced dysregulation of liver metabolic activity.
Collapse
|
37
|
Resende CB, Luft VC, Duncan B, Griep RH, Molina MDCB, Barreto SM, Schmidt MI. Birth weight and body fat mass in adults assessed by bioimpedance in the ELSA-Brasil study. CAD SAUDE PUBLICA 2021; 37:e00061619. [PMID: 33729303 DOI: 10.1590/0102-311x00061619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 07/06/2020] [Indexed: 01/13/2023] Open
Abstract
Intrauterine life is a critical period for the development of body fat and metabolic risk. This study investigated associations between birth weight and total and truncal body fat in adults. To do so, we analyzed data on 10,011 adults participating in the Brazilian Longitudinal Study of Adult Health (ELSA-Brasil) who self-reported birth weight as < 2.5kg, 2.5-4.0kg, or > 4.0kg at baseline (2008-2010) and underwent bioimpedance in the next follow-up visit (2012-2014). Greater mean total and truncal fat mass were seen in those with high birth weight compared with adequate birth weight (p < 0.001) in both sexes (total fat: 25.2 vs. 23.1kg in men and 31.4 vs. 27.7kg in women, and truncal fat: 13.5 vs. 12.4kg in men and 15.9 vs. 14.2kg in women). U-shaped patterns were observed in restricted cubic-spline analyses in the subset of 5,212 individuals reporting exact birth weights, although statistically significant only for those with high birth weight. In the whole sample, in comparing high to adequate birth weight, the latter predicted having a large (> 85 percentile) total and truncal fat mass, respectively: OR = 1.76, 95%CI: 1.37-2.25 (men) and OR = 1.86, 95%CI: 1.42-2.44 (women); OR = 1.68, 95% CI: 1.31-2.16 (men) and OR = 1.73, 95%CI: 1.31-2.28 (women). However, low birth weight predicted having a large (> 85 percentile) % truncal fat only in women (OR = 1.40, 95%CI: 1.03-1.91). In conclusion, in these men and women born in a period in which fetal malnutrition was prevalent, birth weight showed complex, frequently non-linear associations with adult body fat, highlighting the need for interventions to prevent low and high birth weight during pregnancy.
Collapse
Affiliation(s)
| | - Vivian Cristine Luft
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brasil.,Hospital das Clínicas de Porto Alegre, Porto Alegre, Brasil
| | - Bruce Duncan
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brasil
| | | | | | - Sandhi Maria Barreto
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | - Maria Ines Schmidt
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brasil
| |
Collapse
|
38
|
Dulloo AG. Physiology of weight regain: Lessons from the classic Minnesota Starvation Experiment on human body composition regulation. Obes Rev 2021; 22 Suppl 2:e13189. [PMID: 33543573 DOI: 10.1111/obr.13189] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/23/2022]
Abstract
Since its publication in 1950, the Biology of Human Starvation, which describes the classic longitudinal Minnesota Experiment of semistarvation and refeeding in healthy young men, has been the undisputed source of scientific reference about the impact of long-term food deprivation on human physiology and behavior. It has been a guide in developing famine and refugee relief programs for international agencies, in exploring the effects of food deprivation on the cognitive and social functioning of those with anorexia nervosa and bulimia nervosa, and in gaining insights into metabolic adaptations that undermine obesity therapy and cachexia rehabilitation. In more recent decades, the application of a systems approach to the analysis of its data on longitudinal changes in body composition, basal metabolic rate, and food intake during the 24 weeks of semistarvation and 20 weeks of refeeding has provided rare insights into the multitude of control systems that govern the regulation of body composition during weight regain. These have underscored an internal (autoregulatory) control of lean-fat partitioning (highly sensitive to initial adiposity), which operates during weight loss and weight regain and revealed the existence of feedback loops between changes in body composition and the control of food intake and adaptive thermogenesis for the purpose of accelerating the recovery of fat mass and fat-free mass. This paper highlights the general features and design of this grueling experiment of simulated famine that has allowed the unmasking of fundamental control systems in human body composition autoregulation. The integration of its outcomes constitutes the "famine reactions" that drive the normal physiology of weight regain and obesity relapse and provides a mechanistic "autoregulation-based" explanation of how dieting and weight cycling, transition to sedentarity, or developmental programming may predispose to obesity. It also provides a system physiology framework for research toward elucidating proteinstatic and adipostatic mechanisms that control hunger-appetite and adaptive thermogenesis, with major implications for a better understanding (and management) of cachexia, obesity, and cardiometabolic diseases.
Collapse
Affiliation(s)
- Abdul G Dulloo
- Faculty of Science and Medicine, Department of Endocrinology, Metabolism and Cardiovascular System, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
39
|
Monnard CR, Dulloo AG. Polyunsaturated fatty acids as modulators of fat mass and lean mass in human body composition regulation and cardiometabolic health. Obes Rev 2021; 22 Suppl 2:e13197. [PMID: 33471425 DOI: 10.1111/obr.13197] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/22/2022]
Abstract
It is now recognized that the amount and type of dietary fat consumed play an important role in metabolic health. In humans, high intake of polyunsaturated fatty acids (PUFAs) has been associated with reductions in cardiovascular disease risk, improvements in glucose homeostasis, and changes in body composition that involve reductions in central adiposity and, more recently, increases in lean body mass. There is also emerging evidence, which suggests that high intakes of the plant-based essential fatty acids (ePUFAs)-n-6 linoleic acid (LA) and n-3 α-linolenic acid (ALA)-have a greater impact on body composition (fat mass and lean mass) and on glucose homeostasis than the marine-derived long-chain n-3 PUFA-eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). In addition, high intake of both ePUFAs (LA and ALA) may also have anti-inflammatory effects in humans. The purpose of this review is to highlight the emerging evidence, from both epidemiological prospective studies and clinical intervention trials, of a role for PUFA, in particular ePUFA, in the long-term regulation of body weight and body composition, and their impact on cardiometabolic health. It also discusses current notions about the mechanisms by which PUFAs modulate fat mass and lean mass through altered control of energy intake, thermogenesis, or lean-fat partitioning.
Collapse
Affiliation(s)
- Cathriona R Monnard
- Faculty of Science and Medicine, Department of Endocrinology, Metabolism and Cardiovascular System, University of Fribourg, Fribourg, Switzerland
| | - Abdul G Dulloo
- Faculty of Science and Medicine, Department of Endocrinology, Metabolism and Cardiovascular System, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
40
|
Countering impaired glucose homeostasis during catch-up growth with essential polyunsaturated fatty acids: is there a major role for improved insulin sensitivity? Nutr Diabetes 2021; 11:4. [PMID: 33414371 PMCID: PMC7791023 DOI: 10.1038/s41387-020-00143-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 11/08/2022] Open
Abstract
Background/Objectives Catch-up growth, an important risk factor for later obesity and type 2 diabetes, is often characterized by a high rate of fat deposition associated with hyperinsulinemia and glucose intolerance. We tested here the hypothesis that refeeding on a high-fat diet rich in essential polyunsaturated fatty acids (ePUFA) improves glucose homeostasis primarily by enhancing insulin sensitivity in skeletal muscles and adipose tissues. Methods Rats were caloric restricted for 2 weeks followed by 1–2 weeks of isocaloric refeeding on either a low-fat (LF) diet, a high-fat (HF) diet based on animal fat and high in saturated and monounsaturated fatty acids (HF SMFA diet), or a HF diet based on vegetable oils (1:1 mixture of safflower and linseed oils) and rich in the essential fatty acids linoleic and α-linolenic acids (HF ePUFA diet). In addition to measuring body composition and a test of glucose tolerance, insulin sensitivity was assessed during hyperinsulinemic-euglycemic clamps at the whole-body level and in individual skeletal muscles and adipose tissue depots. Results Compared to animals refed the LF diet, those refed the HF-SMFA diet showed a higher rate of fat deposition, higher plasma insulin and glucose responses during the test of glucose tolerance, and markedly lower insulin-stimulated glucose utilization at the whole body level (by a-third to a-half) and in adipose tissue depots (by 2–5 folds) during insulin clamps. While refeeding on the ePUFA diet prevented the increases in fat mass and in plasma insulin and glucose, the results of insulin clamps revealed that insulin-stimulated glucose utilization was not increased in skeletal muscles and only marginally higher in adipose tissues and at the whole-body level. Conclusions These results suggest only a minor role for enhanced insulin sensitivity in the mechanisms by which diets high in ePUFA improves glucose homeostasis during catch-up growth.
Collapse
|
41
|
Lin D, Chen D, Huang J, Li Y, Wen X, Wang L, Shi H. Pre-Birth and Early-Life Factors Associated With the Timing of Adiposity Peak and Rebound: A Large Population-Based Longitudinal Study. Front Pediatr 2021; 9:742551. [PMID: 35004537 PMCID: PMC8727998 DOI: 10.3389/fped.2021.742551] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
Background: The late occurrence of adiposity peak (AP) and the early occurrence of adiposity rebound (AR) are considered the earliest indicators for obesity and its related health conditions later in life. However, there is still limited information for their upstream factors. Therefore, in this study, we aimed to identify the parental and child factors associated with the timing of AP and AR in the early stage of life. Methods: This is a population-based longitudinal study conducted in Shanghai, China. The BMI data of children born between September 2010 and October 2013 were followed from birth to 80 months. Subject-specific body mass index trajectories were fitted by non-linear mixed-effect models with natural cubic spline functions, and the individual's age at AP and AR was estimated. The generalized linear regression models were applied to identify the upstream factors of late occurrence of AP and early occurrence AR. Results: For 7,292 children with estimated AP, boys were less likely to have a late AP [adjusted risk ratio (RR) = 0.83, 95% confidence interval (CI): 0.77-0.90, p < 0.001], but preterm born children had a higher risk of a late AP (adjusted RR = 1.25, 95% CI: 1.07-1.47, p < 0.01). For 10,985 children with estimated AR, children with breastfeeding longer than 4 months were less likely to have an early AR (adjusted RR = 0.80, 95% CI: 0.73-0.87, p < 0.001), but children who were born to advanced-age mothers and who were born small for gestational age had a higher risk of having an early AR (adjusted RR = 1.21, 95% CI: 1.07-1.36, p < 0.01; adjusted RR = 1.20, 95% CI: 1.04-1.39, p = 0.01). Conclusions: Modifiable pre-birth or early-life factors associated with the timing of AP or AR were found. Our findings may help develop prevention and intervention strategies at the earliest stage of life to control later obesity and the health conditions and diseases linked to it.
Collapse
Affiliation(s)
- Dan Lin
- Department of Maternal, Child and Adolescent Health, School of Public Health, Fudan University, Shanghai, China
| | - Didi Chen
- Department of School Health, Minhang District Center of Disease Control and Prevention, Shanghai, China.,Minhang Branch, School of Public Health, Fudan University, Shanghai, China
| | - Jun Huang
- Department of Child Care, Minhang Maternal and Child Health Center, Shanghai, China
| | - Yun Li
- Department of Child Care, Minhang Maternal and Child Health Center, Shanghai, China
| | - Xiaosa Wen
- Department of School Health, Minhang District Center of Disease Control and Prevention, Shanghai, China.,Minhang Branch, School of Public Health, Fudan University, Shanghai, China
| | - Ling Wang
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Huijing Shi
- Department of Maternal, Child and Adolescent Health, School of Public Health, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Di Munno C, Busiello RA, Calonne J, Salzano AM, Miles-Chan J, Scaloni A, Ceccarelli M, de Lange P, Lombardi A, Senese R, Cioffi F, Visser TJ, Peeters RP, Dulloo AG, Silvestri E. Adaptive Thermogenesis Driving Catch-Up Fat Is Associated With Increased Muscle Type 3 and Decreased Hepatic Type 1 Iodothyronine Deiodinase Activities: A Functional and Proteomic Study. Front Endocrinol (Lausanne) 2021; 12:631176. [PMID: 33746903 PMCID: PMC7971177 DOI: 10.3389/fendo.2021.631176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
Refeeding after caloric restriction induces weight regain and a disproportionate recovering of fat mass rather than lean mass (catch-up fat) that, in humans, associates with higher risks to develop chronic dysmetabolism. Studies in a well-established rat model of semistarvation-refeeding have reported that catch-up fat associates with hyperinsulinemia, glucose redistribution from skeletal muscle to white adipose tissue and suppressed adaptive thermogenesis sustaining a high efficiency for fat deposition. The skeletal muscle of catch-up fat animals exhibits reduced insulin-stimulated glucose utilization, mitochondrial dysfunction, delayed in vivo contraction-relaxation kinetics, increased proportion of slow fibers and altered local thyroid hormone metabolism, with suggestions of a role for iodothyronine deiodinases. To obtain novel insights into the skeletal muscle response during catch-up fat in this rat model, the functional proteomes of tibialis anterior and soleus muscles, harvested after 2 weeks of caloric restriction and 1 week of refeeding, were studied. Furthermore, to assess the implication of thyroid hormone metabolism in catch-up fat, circulatory thyroid hormones as well as liver type 1 (D1) and liver and skeletal muscle type 3 (D3) iodothyronine deiodinase activities were evaluated. The proteomic profiling of both skeletal muscles indicated catch-up fat-induced alterations, reflecting metabolic and contractile adjustments in soleus muscle and changes in glucose utilization and oxidative stress in tibialis anterior muscle. In response to caloric restriction, D3 activity increased in both liver and skeletal muscle, and persisted only in skeletal muscle upon refeeding. In parallel, liver D1 activity decreased during caloric restriction, and persisted during catch-up fat at a time-point when circulating levels of T4, T3 and rT3 were all restored to those of controls. Thus, during catch-up fat, a local hypothyroidism may occur in liver and skeletal muscle despite systemic euthyroidism. The resulting reduced tissue thyroid hormone bioavailability, likely D1- and D3-dependent in liver and skeletal muscle, respectively, may be part of the adaptive thermogenesis sustaining catch-up fat. These results open new perspectives in understanding the metabolic processes associated with the high efficiency of body fat recovery after caloric restriction, revealing new implications for iodothyronine deiodinases as putative biological brakes contributing in suppressed thermogenesis driving catch-up fat during weight regain.
Collapse
Affiliation(s)
- Celia Di Munno
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | | | - Julie Calonne
- Department of Medicine, Physiology, University of Fribourg, Fribourg, Switzerland
| | - Anna Maria Salzano
- Institute for the Animal Production System in the Mediterranean Environment, National Research Council, Naples, Italy
| | - Jennifer Miles-Chan
- Department of Medicine, Physiology, University of Fribourg, Fribourg, Switzerland
| | - Andrea Scaloni
- Institute for the Animal Production System in the Mediterranean Environment, National Research Council, Naples, Italy
| | - Michele Ceccarelli
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Pieter de Lange
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | | | - Rosalba Senese
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Federica Cioffi
- Department of Science and Technologies, University of Sannio, Benevento, Italy
| | - Theo J. Visser
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus MC, Rotterdam, Netherlands
| | - Robin P. Peeters
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus MC, Rotterdam, Netherlands
| | - Abdul G. Dulloo
- Department of Medicine, Physiology, University of Fribourg, Fribourg, Switzerland
| | - Elena Silvestri
- Department of Science and Technologies, University of Sannio, Benevento, Italy
- *Correspondence: Elena Silvestri,
| |
Collapse
|
43
|
Moreno-Fernandez J, Ochoa JJ, Lopez-Frias M, Diaz-Castro J. Impact of Early Nutrition, Physical Activity and Sleep on the Fetal Programming of Disease in the Pregnancy: A Narrative Review. Nutrients 2020; 12:nu12123900. [PMID: 33419354 PMCID: PMC7766505 DOI: 10.3390/nu12123900] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/13/2022] Open
Abstract
Early programming is the adaptation process by which nutrition and environmental factors alter development pathways during prenatal growth, inducing changes in postnatal metabolism and diseases. The aim of this narrative review, is evaluating the current knowledge in the scientific literature on the effects of nutrition, environmental factors, physical activity and sleep on development pathways. If in utero adaptations were incorrect, this would cause a mismatch between prenatal programming and adulthood. Adequate caloric intake, protein, mineral, vitamin, and long-chain fatty acids, have been noted for their relevance in the offspring brain functions and behavior. Fetus undernutrition/malnutrition causes a delay in growth and have detrimental effects on the development and subsequent functioning of the organs. Pregnancy is a particularly vulnerable period for the development of food preferences and for modifications in the emotional response. Maternal obesity increases the risk of developing perinatal complications and delivery by cesarean section and has long-term implications in the development of metabolic diseases. Physical exercise during pregnancy contributes to overall improved health post-partum. It is also interesting to highlight the relevance of sleep problems during pregnancy, which influence adequate growth and fetal development. Taking into account these considerations, we conclude that nutrition and metabolic factors during early life play a key role of health promotion and public health nutrition programs worldwide to improve the health of the offspring and the health costs of hospitalization.
Collapse
Affiliation(s)
- Jorge Moreno-Fernandez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, E-18071 Granada, Spain; (J.M.-F.); (M.L.-F.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, E-18071 Granada, Spain
| | - Julio J. Ochoa
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, E-18071 Granada, Spain; (J.M.-F.); (M.L.-F.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, E-18071 Granada, Spain
- Correspondence: ; Tel.: +34-958-241-000 (ext. 20317)
| | - Magdalena Lopez-Frias
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, E-18071 Granada, Spain; (J.M.-F.); (M.L.-F.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, E-18071 Granada, Spain
| | - Javier Diaz-Castro
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, E-18071 Granada, Spain; (J.M.-F.); (M.L.-F.); (J.D.-C.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, E-18071 Granada, Spain
| |
Collapse
|
44
|
Lu X, Hu S, Liao Y, Zheng J, Zeng T, Zhong X, Liu G, Gou L, Chen L. Vascular endothelial growth factor B promotes transendothelial fatty acid transport into skeletal muscle via histone modifications during catch-up growth. Am J Physiol Endocrinol Metab 2020; 319:E1031-E1043. [PMID: 32954823 DOI: 10.1152/ajpendo.00090.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Caloric restriction (CR) followed by refeeding, a phenomenon known as catch-up growth (CUG), results in excessive lipid deposition and insulin resistance in skeletal muscle, but the underlying mechanisms remain elusive. Recent reports have suggested that vascular endothelial growth factor B (VEGF-B) controls muscle lipid accumulation by regulating endothelial fatty acid transport. Here, we found continuous activation of VEGF-B signaling and increased lipid uptake in skeletal muscle from CR to refeeding, as well as increased lipid deposition and impaired insulin sensitivity after refeeding in the skeletal muscle of CUG rodents. Inhibiting VEGF-B signaling reduced fatty acid uptake in and transport across endothelial cells. Knockdown of Vegfb in the tibialis anterior (TA) muscle of CUG mice significantly attenuated muscle lipid accumulation and ameliorated muscle insulin sensitivity by decreasing lipid uptake. Furthermore, we showed that aberrant histone methylation (H3K9me1) and acetylation (H3K14ac and H3K18ac) at the Vegfb promoter might be the main cause of persistent VEGF-B upregulation in skeletal muscle during CUG. Modifying these aberrant loci using their related enzymes [PHD finger protein 2 (PHF2) or E1A binding protein p300 (p300)] could regulate VEGF-B expression in vitro. Collectively, our findings indicate that VEGF-B can promote transendothelial lipid transport and lead to lipid overaccumulation and insulin resistance in skeletal muscle during CUG, which might be mediated by histone methylation and acetylation.
Collapse
Affiliation(s)
- Xiaodan Lu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Shengqing Hu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Yunfei Liao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Juan Zheng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Xueyu Zhong
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Geng Liu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Luoning Gou
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| |
Collapse
|
45
|
Wu Y, Yin G, Wang P, Huang Z, Lin S. Effects of different diet-induced postnatal catch-up growth on glycolipid metabolism in intrauterine growth retardation male rats. Exp Ther Med 2020; 20:134. [PMID: 33082866 PMCID: PMC7560533 DOI: 10.3892/etm.2020.9263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 06/24/2020] [Indexed: 02/05/2023] Open
Abstract
A number of studies have reported the occurrence of long-term metabolic disorders in mammals following intrauterine growth retardation (IUGR). However, the effects of dietary patterns during IUGR have not been fully elucidated. The present study aimed to evaluate the effects of different dietary patterns during critical growth windows on metabolic outcomes in the offspring of rats with IUGR. Male offspring rats from mothers fed either a normal or low-protein diet were randomly assigned to one of the following groups: Normal diet throughout pregnancy, lactation and after weaning (CON); normal diet throughout pregnancy and high-fat diet throughout lactation and after weaning (N + H + H); low-protein diet throughout pregnancy and high-fat diet throughout lactation and after weaning (IUGR + H + H); low-protein diet throughout pregnancy and lactation and high-fat diet after weaning (IUGR + L + H); and low-protein diet throughout pregnancy and normal diet throughout lactation and after weaning. During lactation, the male offspring in the N + H + H group exhibited the fastest growth rate, whereas the slowest rate was in the IUGR + L + H group. Following weaning, all IUGR groups demonstrated significant catch-up growth. Abnormal insulin tolerance were observed in the N + H + H, IUGR + H + H and IUGR + L + H groups and insulin sensitivity was decreased in IUGR + L + H group. The triglycerides/high-density lipoprotein ratio in the IUGR + L + H group was significantly higher compared with in the other groups. The abdominal circumference, Lee's index and adipocyte diameter of IUGR groups were significantly increased compared with the CON group. High levels of leptin and interleukin-6 in adipose tissues, and low adiponectin were observed in the IUGR + L + H group. Different dietary patterns during specific growth windows showed numerous impacts on glycolipid metabolism in IUGR offspring. The present study elucidated the mechanisms and potential options for IUGR treatment and prevention.
Collapse
Affiliation(s)
- Yixi Wu
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Guoshu Yin
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Ping Wang
- Center of Reproductive Medicine, Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Zhihua Huang
- Center of Reproductive Medicine, Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Shaoda Lin
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
- Correspondence to: Dr Shaoda Lin, Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, 57 Changping Road, Shantou, Guangdong 515000, P.R. China
| |
Collapse
|
46
|
Baldassarre ME, Di Mauro A, Caroli M, Schettini F, Rizzo V, Panza R, De Giorgi A, Capozza M, Fanelli M, Laforgia N. Premature Birth is an Independent Risk Factor for Early Adiposity Rebound: Longitudinal Analysis of BMI Data from Birth to 7 Years. Nutrients 2020; 12:nu12123654. [PMID: 33261215 PMCID: PMC7760942 DOI: 10.3390/nu12123654] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/17/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Adiposity rebound (AR) refers to the second rise of the body mass index (BMI) curve that usually occurs physiologically between five and seven years of age. AR timing has a great impact on patients’ health, since early adiposity rebound (EAR) is associated with the development of metabolic syndrome later in life. We aimed to investigate the prevalence of EAR in a cohort of inborn preterm infants admitted to the Neonatal Intensive Care Section of the Policlinico University Hospital of Bari, Italy. Secondarily, we assessed whether some determinants such as (1) gender; (2) delivery mode; (3) birth weight and classification into small, normal, or large for gestational age; (4) type of feeding; (5) breastfeeding duration; (6) timing of introduction of solid food; (7) parental education; and (8) parental pre-pregnancy body mass index (BMI) influenced EAR in this cohort. The tertiary aim was to evaluate the prevalence of obesity or being overweight at seven years of age in children according to early versus timely AR. This is a prospective, population-based longitudinal study conducted at the Neonatal Intensive Care Section of the Policlinico University Hospital of Bari, Italy. Inborn preterm infants admitted to the neonatal ward between 2009 and 2011 were eligible. Enrolled preterm infants were evaluated at birth and at 1, 3, 6, 9, 12, 15, 18, and 24 months and 3, 4, 5, 6, and 7 years of age. Weight and height data were analyzed, and BMI was calculated. AR was assessed in the growth trajectory in a body mass index (BMI) plot. Of the 250 preterm newborns included, 100 completed the seven-year follow-up and entered the final analysis, 138 were lost during the seven-year follow-up, and in 12 cases parents withdrew over the course of the study. The prevalence of EAR in our cohort of preterm newborns was 54% at seven years of age. Early adiposity rebound was associated with being large for gestational age (LGA) at birth. No other factors were associated with EAR. Early adiposity rebounders had a significantly higher BMI at seven years compared to children with timely AR (17.2 ± 2.7 vs. 15.6 ± 2.05, p = 0.021). No significant differences were found in the prevalence of obesity or being overweight at seven years of age in children with early or timely AR (29% vs. 14%, p = 0.202). Ex-preterm infants have an increased risk of EAR. Since EAR may lead to long-term detrimental health effects with the onset of various chronic diseases (e.g., obesity, metabolic syndrome, etc.), healthcare providers should be prepared to counteract its occurrence, especially in delicate sub-populations of infants.
Collapse
Affiliation(s)
- Maria Elisabetta Baldassarre
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, “Aldo Moro” University of Bari, 70100 Bari, Italy; (A.D.M.); (F.S.); (V.R.); (R.P.); (A.D.G.); (M.C.); (N.L.)
- Correspondence:
| | - Antonio Di Mauro
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, “Aldo Moro” University of Bari, 70100 Bari, Italy; (A.D.M.); (F.S.); (V.R.); (R.P.); (A.D.G.); (M.C.); (N.L.)
| | | | - Federico Schettini
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, “Aldo Moro” University of Bari, 70100 Bari, Italy; (A.D.M.); (F.S.); (V.R.); (R.P.); (A.D.G.); (M.C.); (N.L.)
| | - Valentina Rizzo
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, “Aldo Moro” University of Bari, 70100 Bari, Italy; (A.D.M.); (F.S.); (V.R.); (R.P.); (A.D.G.); (M.C.); (N.L.)
| | - Raffaella Panza
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, “Aldo Moro” University of Bari, 70100 Bari, Italy; (A.D.M.); (F.S.); (V.R.); (R.P.); (A.D.G.); (M.C.); (N.L.)
| | - Alessia De Giorgi
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, “Aldo Moro” University of Bari, 70100 Bari, Italy; (A.D.M.); (F.S.); (V.R.); (R.P.); (A.D.G.); (M.C.); (N.L.)
| | - Manuela Capozza
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, “Aldo Moro” University of Bari, 70100 Bari, Italy; (A.D.M.); (F.S.); (V.R.); (R.P.); (A.D.G.); (M.C.); (N.L.)
| | - Margherita Fanelli
- Department of Interdisciplinary Medicine, “Aldo Moro” University of Bari, 70100 Bari, Italy;
| | - Nicola Laforgia
- Department of Biomedical Science and Human Oncology, Neonatology and Neonatal Intensive Care Unit, “Aldo Moro” University of Bari, 70100 Bari, Italy; (A.D.M.); (F.S.); (V.R.); (R.P.); (A.D.G.); (M.C.); (N.L.)
| |
Collapse
|
47
|
Crossin R, Arunogiri S. Harms associated with inhalant misuse in adolescent females - a review of the pre-clinical and clinical evidence. Drug Alcohol Depend 2020; 216:108232. [PMID: 32862119 DOI: 10.1016/j.drugalcdep.2020.108232] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 01/14/2023]
Abstract
BACKGROUND Inhalant misuse, or the misuse of products containing toluene is common in adolescents, and is associated with diverse physiological and psychological harms. Females comprise over half those who misuse inhalants in adolescence, however, the majority of the evidence has been derived from male-only or mixed-sex studies without exploration of sex differences. Female adolescence is a critical maturational period with potential for growth, reproductive, cognitive and psychological harms that may lead to long-term health consequences. We therefore summarise evidence of female-specific harms arising from inhalant misuse. METHODS We synthesised pre-clinical and clinical studies of inhalant misuse which were conducted in females, or where sex-differences were reported, into a narrative literature review. RESULTS Females experience growth impairments and metabolic dysfunction arising from inhalant misuse, but data on sex-differences are inconclusive. Inhalant misuse in early adolescence may impact menarche and subsequent reproductive capacity, but studies have predominantly focused on the effects of inhalants on offspring rather than on the exposed female. There is limited evidence of sex-differences in relation to cognitive outcomes following exposure to inhalants in pre-clinical models. Females are at an increased risk of psychological harms associated with inhalant misuse, particularly depression and suicidal behaviour. CONCLUSIONS The type and magnitude of harms associated with inhalant misuse are sex-specific, but data are limited. We recommend that both pre-clinical and clinical studies of inhalant misuse include both males and females, and should specifically test for and report sex-differences. This can be used to build an evidence base for screening and interventions tailored to females.
Collapse
Affiliation(s)
- Rose Crossin
- Department of Population Health, University of Otago Christchurch campus, 34 Gloucester Street, Christchurch, 8140, New Zealand; Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, Melbourne, Victoria, 3052, Australia.
| | - Shalini Arunogiri
- Central Clinical School, Monash Alfred Psychiatry Research Centre, Level 4, 607 St Kilda Road, Melbourne, Victoria, 3004, Australia; Turning Point, Eastern Health, 110 Church Street, Richmond, Melbourne, Victoria, 3121, Australia
| |
Collapse
|
48
|
Influence of prenatal stress on metabolic abnormalities induced by postnatal intake of a high-fat diet in BALB/c mice. J Dev Orig Health Dis 2020; 12:721-730. [PMID: 33118903 DOI: 10.1017/s2040174420000987] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Prenatal insults during fetal development result in increased likelihood of developing chronic disease. Obesity, the biggest risk factor for the development of metabolic disease, is affected by several genetic and environmental factors. High-fat diet (HFD) consumption is usually linked with the development of obesity. The main goal of this study was to analyze the impact of the exposure to a HFD in prenatally stressed animals. For this purpose, we subjected pregnant BALB/c mice to restraint stress for 2 h a day between gestational day (GD) 14 and GD 21. Prenatally stressed and control offspring of both sexes were postnatally exposed to a HFD for 24 weeks. We found that prenatal stress (PS) per se produced disturbances in males such as increased total blood cholesterol and triglycerides, with a decrease in mRNA expression of sirtuin-1. When these animals were fed a HFD, we observed a rise in glucose and insulin levels and an increase in visceral adipose tissue gene expression of leptin, resistin, and interleukin-1 beta. Although females proved to be more resilient to PS consequences, when they were fed a HFD, they showed significant metabolic impairment. In addition to the changes observed in males, females also presented an increase in body weight and adiposity and a rise in cholesterol levels.
Collapse
|
49
|
Arisaka O, Ichikawa G, Koyama S, Sairenchi T. Childhood obesity: rapid weight gain in early childhood and subsequent cardiometabolic risk. Clin Pediatr Endocrinol 2020; 29:135-142. [PMID: 33088012 PMCID: PMC7534524 DOI: 10.1297/cpe.29.135] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/06/2020] [Indexed: 12/19/2022] Open
Abstract
Dynamic changes in body weight have long been recognized as important indicators of risk
for human health. Many population-based observational studies have shown that rapid weight
gain during infancy, including a catch-up growth phenomenon or adiposity rebound in early
childhood, predisposes a person to the development of obesity, type 2 diabetes, and
cardiovascular diseases later in life. However, a consensus has not been established
regarding which period of weight gain contributes to future risks. This review evaluates
recent evidence on the relationship between early rapid growth and future obesity and
cardiometabolic risk, with a focus on the differential significance of rapid weight gain
in infancy and early childhood. Although there is a need for attention to childhood growth
during early infancy before 1 yr of age as it may be related to future obesity, emerging
evidence strongly suggests that toddlers showing an increase in body mass index (BMI)
before 3 yr of age, a period normally characterized by decreased BMI, are prone to
developing later cardiometabolic risk.
Collapse
Affiliation(s)
- Osamu Arisaka
- Department of Pediatrics, Nasu Red Cross Hospital, Tochigi, Japan.,Department of Pediatrics, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Go Ichikawa
- Department of Pediatrics, Nasu Red Cross Hospital, Tochigi, Japan.,Department of Pediatrics, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Satomi Koyama
- Department of Pediatrics, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Toshimi Sairenchi
- Department of Public Health, Dokkyo Medical University School of Medicine, Tochigi, Japan
| |
Collapse
|
50
|
Ong YY, Sadananthan SA, Aris IM, Tint MT, Yuan WL, Huang JY, Chan YH, Ng S, Loy SL, Velan SS, Fortier MV, Godfrey KM, Shek L, Tan KH, Gluckman PD, Yap F, Choo JTL, Ling LH, Tan K, Chen L, Karnani N, Chong YS, Eriksson JG, Wlodek ME, Chan SY, Lee YS, Michael N. Mismatch between poor fetal growth and rapid postnatal weight gain in the first 2 years of life is associated with higher blood pressure and insulin resistance without increased adiposity in childhood: the GUSTO cohort study. Int J Epidemiol 2020; 49:1591-1603. [PMID: 32851407 PMCID: PMC7116531 DOI: 10.1093/ije/dyaa143] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Using longitudinal ultrasounds as an improved fetal growth marker, we aimed to investigate if fetal growth deceleration followed by rapid postnatal weight gain is associated with childhood cardiometabolic risk biomarkers in a contemporary well-nourished population. METHODS We defined fetal growth deceleration (FGD) as ultrasound-measured 2nd-3rd-trimester abdominal circumference decrease by ≥0.67 standard deviation score (SDS) and rapid postnatal weight gain (RPWG) as 0-2-year-old weight increase by ≥0.67 SDS. In the GUSTO mother-offspring cohort, we grouped 797 children into four groups of FGD-only (14.2%), RPWG-only (23.3%), both (mismatch, 10.7%) or neither (reference, 51.8%). Adjusting for confounders and comparing with the reference group, we tested associations of these growth groups with childhood cardiometabolic biomarkers: magnetic resonance imaging (MRI)-measured abdominal fat (n = 262), liver fat (n = 216), intramyocellular lipids (n = 227), quantitative magnetic resonance-measured overall body fat % (BF%) (n = 310), homeostasis model assessment of insulin resistance (HOMA-IR) (n = 323), arterial wall thickness (n = 422) and stiffness (n = 443), and blood pressure trajectories (ages 3-6 years). RESULTS Mean±SD birthweights were: FGD-only (3.11 ± 0.38 kg), RPWG-only (3.03 ± 0.37 kg), mismatch (2.87 ± 0.31 kg), reference (3.30 ± 0.36 kg). FGD-only children had elevated blood pressure trajectories without correspondingly increased BF%. RPWG-only children had altered body fat partitioning, higher BF% [BF = 4.26%, 95% confidence interval (CI) (2.34, 6.19)], HOMA-IR 0.28 units (0.11, 0.45)] and elevated blood pressure trajectories. Mismatch children did not have increased adiposity, but had elevated ectopic fat, elevated HOMA-IR [0.29 units (0.04,0.55)] and the highest blood pressure trajectories. Associations remained even after excluding small-for-gestational-age infants from analyses. CONCLUSIONS Fetal growth deceleration coupled with rapid postnatal weight gain was associated with elevated childhood cardiometabolic risk biomarkers without correspondingly increased BF%.
Collapse
Affiliation(s)
- Yi Ying Ong
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suresh Anand Sadananthan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Izzuddin M Aris
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Mya Thway Tint
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wen Lun Yuan
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jonathan Y Huang
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Yiong Huak Chan
- Biostatistics Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sharon Ng
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - See Ling Loy
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Sendhil S Velan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
- Singapore Bioimaging Consortium, Agency for Science Technology and Research, Singapore, Singapore
| | - Marielle V Fortier
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Diagnostic and Interventional Imaging, KK Women's and Children's Hospital, Singapore, Singapore
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Lynette Shek
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Pediatrics, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
| | - Kok Hian Tan
- Duke-NUS Medical School, Singapore, Singapore
- Department of Maternal Fetal Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Peter D Gluckman
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Fabian Yap
- Duke-NUS Medical School, Singapore, Singapore
- Department of Pediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | | | - Lieng Hsi Ling
- Department of Cardiology, National University Heart Centre, Singapore, Singapore
| | - Karen Tan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
- Molecular Diagnosis Centre, Department of Laboratory Medicine, National University Health System, Singapore, Singapore
| | - Li Chen
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Neerja Karnani
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Yap-Seng Chong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Johan G Eriksson
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mary E Wlodek
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yung Seng Lee
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Pediatrics, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
| | - Navin Michael
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore
| |
Collapse
|