1
|
Phillips EA, Alharithi YJ, Kadam L, Coussens LM, Kumar S, Maloyan A. Metabolic abnormalities in the bone marrow cells of young offspring born to mothers with obesity. Int J Obes (Lond) 2024; 48:1542-1551. [PMID: 38937647 DOI: 10.1038/s41366-024-01563-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND/OBJECTIVES Intrauterine metabolic reprogramming occurs in mothers with obesity during gestation, putting the offspring at high risk of developing obesity and associated metabolic disorders even before birth. We have generated a mouse model of maternal high-fat diet-induced obesity that recapitulates the metabolic changes seen in humans born to women with obesity. METHODS Here, we profiled and compared the metabolic characteristics of bone marrow cells of newly weaned 3-week-old offspring of dams fed either a high-fat (Off-HFD) or a regular diet (Off-RD). We utilized a state-of-the-art flow cytometry, and targeted metabolomics approach coupled with a Seahorse metabolic analyzer. RESULTS We revealed significant metabolic perturbation in the offspring of HFD-fed vs. RD-fed dams, including utilization of glucose primarily via oxidative phosphorylation. We also show a reduction in levels of amino acids, a phenomenon previously linked to bone marrow aging. Using flow cytometry, we found changes in the immune complexity of bone marrow cells and identified a unique B cell population expressing CD19 and CD11b in the bone marrow of three-week-old offspring of high-fat diet-fed mothers. Our data also revealed increased expression of Cyclooxygenase-2 (COX-2) on myeloid CD11b, and on CD11bhi B cells. CONCLUSIONS Altogether, we demonstrate that the offspring of mothers with obesity show metabolic and immune changes in the bone marrow at a very young age and prior to any symptomatic metabolic disease.
Collapse
Affiliation(s)
- Elysse A Phillips
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA
- The University of California San Francisco, San Francisco, CA, USA
| | - Yem J Alharithi
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Leena Kadam
- Department of OB/GYN, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Lisa M Coussens
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Sushil Kumar
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
| | - Alina Maloyan
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
- The University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Hirsch A, Peled T, Schlesinger S, Sela HY, Grisaru-Granovsky S, Rottenstreich M. Impact of gestational diabetes mellitus on neonatal outcomes in small for gestational age infants: a multicenter retrospective study. Arch Gynecol Obstet 2024; 310:685-693. [PMID: 38874779 PMCID: PMC11258160 DOI: 10.1007/s00404-024-07587-y] [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: 03/27/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVE To evaluate obstetric and perinatal outcomes among small for gestational age (SGA) infants born to patients diagnosed with Gestational diabetes mellitus (GDM). MATERIALS AND METHODS A multicenter retrospective cohort study between 2005 and 2021. The perinatal outcomes of SGA infants born to patients with singleton pregnancy and GDM were compared to SGA infants born to patients without GDM. The primary outcome was a composite adverse neonatal outcome. Infants with known structural/genetic abnormalities or infections were excluded. A univariate analysis was conducted followed by a multivariate analysis (adjusted odds ratio [95% confidence interval]). RESULTS During the study period, 11,662 patients with SGA infants met the inclusion and exclusion criteria. Of these, 417 (3.6%) SGA infants were born to patients with GDM, while 11,245 (96.4%) were born to patients without GDM. Overall, the composite adverse neonatal outcome was worse in the GDM group (53.7% vs 17.4%, p < 0.01). Specifically, adverse neonatal outcomes such as a 5 min Apgar score < 7, meconium aspiration, seizures, and hypoglycemia were independently associated with GDM among SGA infants. In addition, patients with GDM and SGA infants had higher rates of overall and spontaneous preterm birth, unplanned cesarean, and postpartum hemorrhage. In a multivariate logistic regression assessing the association between GDM and neonatal outcomes, GDM was found to be independently associated with the composite adverse neonatal outcome (aOR 4.26 [3.43-5.3]), 5 min Apgar score < 7 (aOR 2 [1.16-3.47]), meconium aspiration (aOR 4.62 [1.76-12.13]), seizures (aOR 2.85 [1.51-5.37]) and hypoglycemia (aOR 16.16 [12.79-20.41]). CONCLUSIONS Our study demonstrates that GDM is an independent risk factor for adverse neonatal outcomes among SGA infants. This finding underscores the imperative for tailored monitoring and management strategies in those pregnancies.
Collapse
Affiliation(s)
- Ayala Hirsch
- Department of Obstetrics and Gynecology, Shaare Zedek Medical Center, Affiliated with the Hebrew University School of Medicine, 12 Bayit Street, 91031, Jerusalem, Israel
| | - Tzuria Peled
- Department of Obstetrics and Gynecology, Shaare Zedek Medical Center, Affiliated with the Hebrew University School of Medicine, 12 Bayit Street, 91031, Jerusalem, Israel.
| | - Shaked Schlesinger
- Department of Military Medicine and "Tzameret", Faculty of Medicine, Israel Defense Forces, Hebrew University of Jerusalem, and Medical Corps, Jerusalem, Israel
| | - Hen Y Sela
- Department of Obstetrics and Gynecology, Shaare Zedek Medical Center, Affiliated with the Hebrew University School of Medicine, 12 Bayit Street, 91031, Jerusalem, Israel
| | - Sorina Grisaru-Granovsky
- Department of Obstetrics and Gynecology, Shaare Zedek Medical Center, Affiliated with the Hebrew University School of Medicine, 12 Bayit Street, 91031, Jerusalem, Israel
| | - Misgav Rottenstreich
- Department of Obstetrics and Gynecology, Shaare Zedek Medical Center, Affiliated with the Hebrew University School of Medicine, 12 Bayit Street, 91031, Jerusalem, Israel
- Department of Nursing, Jerusalem College of Technology, Jerusalem, Israel
| |
Collapse
|
3
|
Chen B, Chen Y, Wang Y, Xin Q, Ma D. The association between rapid growth and lipid profile: a systematic review and meta-analysis. Front Endocrinol (Lausanne) 2024; 15:1353334. [PMID: 38577566 PMCID: PMC10991823 DOI: 10.3389/fendo.2024.1353334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 02/27/2024] [Indexed: 04/06/2024] Open
Abstract
Background & aims Metabolic disease prevalence has increased in many regions, and is closely associated with dyslipidemia. Rapid growth refers to a significant increase in growth velocity above the normal range, particularly in infants and children, and is highly prevalent in congenital deficiency infants. But the association between dyslipidemia and rapid growth remains controversial. We performed this meta-analysis to investigate the lipid profile in subjects with and without postnatal rapid growth, and to determine what are the confounding factors. Methods Medline, EMBASE, China National Knowledge Infrastructure Chinese citation database and WANFANG database were searched (last search in May 2021). Publication bias was examined by constructing funnel plots, Egger's linear regression test and Begg's rank correlation test. Results The fixed effects model would be adopted if I2 is less than 25%, otherwise random effects model would be used. There were 11 articles involved with a total of 1148 participants (539 boys and 609 girls, mean age=7.4 years). Pooled analysis found that rapid growth was negatively associated with high-density lipoprotein cholesterol (HDL-C) (weighted mean difference=-0.068, 95%CI [-0.117, -0.020]), but not associated with triglycerides (TG), total cholesterol (TC), or low-density lipoprotein cholesterol (LDL-C). Stratified analysis suggested that increased TG were found in rapid growth subjects from developing countries. Higher TC was observed for rapid growth participants of follow-up age ≤8 years old, rapid growth duration ≤2 years, preterm, low birth weight, and from developing countries. But decreased TC was observed in small for gestational age (SGA) rapid growth subjects. Decreased LDL-C had been documented in rapid growth subjects of follow-up age >8 years old, from developed countries, and SGA. At last, rapid growth groups had lower HDL-C in infants of rapid growth duration >2 years and from developed countries. Conclusion Rapid growth is associated with lipid profiles, particularly during early childhood, and this relationship is influenced by factors such as the duration of growth, the level of national development, and birth weight. These findings are significant for the development of strategies to prevent metabolic diseases.This review was registered in PROSPERO International Prospective Register of Systematic Reviews (www.crd.york.ac.uk/prospero/) with the registration number CRD42020154240.
Collapse
Affiliation(s)
- Botian Chen
- School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yunli Chen
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuyang Wang
- School of Public Health, Peking University Health Science Center, Beijing, China
| | - Qinghua Xin
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Defu Ma
- School of Public Health, Peking University Health Science Center, Beijing, China
| |
Collapse
|
4
|
Alina M, Phillips E, Alharithi Y, Kadam L, Coussens L, Kumar S. Metabolic abnormalities in the bone marrow cells of young offspring born to obese mothers. RESEARCH SQUARE 2024:rs.3.rs-3830161. [PMID: 38313293 PMCID: PMC10836107 DOI: 10.21203/rs.3.rs-3830161/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Intrauterine metabolic reprogramming occurs in obese mothers during gestation, putting the offspring at high risk of developing obesity and associated metabolic disorders even before birth. We have generated a mouse model of maternal high-fat diet-induced obesity that recapitulates the metabolic changes seen in humans born to obese women. Here, we profiled and compared the metabolic characteristics of bone marrow cells of newly weaned 3-week-old offspring of dams fed either a high-fat (Off-HFD) or a regular diet (Off-RD). We utilized a state-of-the-art targeted metabolomics approach coupled with a Seahorse metabolic analyzer. We revealed significant metabolic perturbation in the offspring of HFD-fed vs. RD-fed dams, including utilization of glucose primarily via oxidative phosphorylation. We also found a reduction in levels of amino acids, a phenomenon previously linked to bone marrow aging. Using flow cytometry, we identified a unique B cell population expressing CD19 and CD11b in the bone marrow of three-week-old offspring of high-fat diet-fed mothers, and found increased expression of Cyclooxygenase-2 (COX-2) on myeloid CD11b, and on CD11bhi B cells. Altogether, we demonstrate that the offspring of obese mothers show metabolic and immune changes in the bone marrow at a very young age and prior to any symptomatic metabolic disease.
Collapse
|
5
|
Phillips E, Alharithi Y, Kadam L, Coussens LM, Kumar S, Maloyan A. Metabolic abnormalities in the bone marrow cells of young offspring born to obese mothers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569274. [PMID: 38077037 PMCID: PMC10705475 DOI: 10.1101/2023.11.29.569274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
Intrauterine metabolic reprogramming occurs in obese mothers during gestation, putting the offspring at high risk of developing obesity and associated metabolic disorders even before birth. We have generated a mouse model of maternal high-fat diet-induced obesity that recapitulates the metabolic changes seen in humans. Here, we profiled and compared the metabolic characteristics of bone marrow cells of newly weaned 3-week-old offspring of dams fed either a high-fat (Off-HFD) or a regular diet (Off-RD). We utilized a state-of-the-art targeted metabolomics approach coupled with a Seahorse metabolic analyzer. We revealed significant metabolic perturbation in the offspring of HFD-fed vs. RD-fed dams, including utilization of glucose primarily via oxidative phosphorylation, and reduction in levels of amino acids, a phenomenon previously linked to aging. Furthermore, in the bone marrow of three-week-old offspring of high-fat diet-fed mothers, we identified a unique B cell population expressing CD19 and CD11b, and found increased expression of Cyclooxygenase-2 (COX-2) on myeloid CD11b, and on CD11b hi B cells, with all the populations being significantly more abundant in offspring of dams fed HFD but not a regular diet. Altogether, we demonstrate that the offspring of obese mothers show metabolic and immune changes in the bone marrow at a very young age and prior to any symptomatic metabolic disease.
Collapse
|
6
|
Fasoulakis Z, Koutras A, Antsaklis P, Theodora M, Valsamaki A, Daskalakis G, Kontomanolis EN. Intrauterine Growth Restriction Due to Gestational Diabetes: From Pathophysiology to Diagnosis and Management. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1139. [PMID: 37374343 DOI: 10.3390/medicina59061139] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/16/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023]
Abstract
Intrauterine growth restriction (IUGR) represents a condition where the fetal weight is less than the 10th percentile for gestational age, or the estimated fetal weight is lower than expected based on gestational age. IUGR can be caused by various factors such as maternal, placental or fetal factors and can lead to various complications for both the fetus and the mother, including fetal distress, stillbirth, preterm delivery, and maternal hypertension. Women with gestational diabetes are at an increased risk of developing IUGR. This article reviews the different aspects of gestational diabetes in addition to IUGR, the diagnostic methods available for IUGR detection, including ultrasound and Doppler studies, discusses the management strategies for women with IUGR and gestational diabetes and analyzes the importance of early detection and timely intervention to improve pregnancy outcomes.
Collapse
Affiliation(s)
- Zacharias Fasoulakis
- Department of Obstetrics and Gynecology, General Hospital of Athens 'Alexandra', National and Kapodistrian University of Athens, Lourou and Vasilissis Sofias Ave, 11528 Athens, Greece
| | - Antonios Koutras
- Department of Obstetrics and Gynecology, General Hospital of Athens 'Alexandra', National and Kapodistrian University of Athens, Lourou and Vasilissis Sofias Ave, 11528 Athens, Greece
| | - Panos Antsaklis
- Department of Obstetrics and Gynecology, General Hospital of Athens 'Alexandra', National and Kapodistrian University of Athens, Lourou and Vasilissis Sofias Ave, 11528 Athens, Greece
| | - Marianna Theodora
- Department of Obstetrics and Gynecology, General Hospital of Athens 'Alexandra', National and Kapodistrian University of Athens, Lourou and Vasilissis Sofias Ave, 11528 Athens, Greece
| | - Asimina Valsamaki
- Department of Internal Medicine, General Hospital of Larisa, Tsakalof 1, 41221 Larisa, Greece
| | - George Daskalakis
- Department of Obstetrics and Gynecology, General Hospital of Athens 'Alexandra', National and Kapodistrian University of Athens, Lourou and Vasilissis Sofias Ave, 11528 Athens, Greece
| | - Emmanuel N Kontomanolis
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 6th km Alexandroupolis-Makris, 68100 Alexandroupolis, Greece
| |
Collapse
|
7
|
Nordman H, Jääskeläinen J, Voutilainen R. Birth Size as a Determinant of Cardiometabolic Risk Factors in Children. Horm Res Paediatr 2021; 93:144-153. [PMID: 32846418 DOI: 10.1159/000509932] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 07/06/2020] [Indexed: 11/19/2022] Open
Abstract
The association between birth size and cardiometabolic disease risk may be U-shaped. Being born small for gestational age (SGA) has a definitive association with later cardiovascular risk, but the impact of being born large for gestational age (LGA) on cardiometabolic health is more controversial. In addition to birth size, early postnatal growth pattern and later weight gain affect cardiometabolic risk in adulthood. Most SGA-born children have catch-up and LGA-born children have catch-down growth during the first years of life. The extent of this early compensatory growth may contribute to the adverse health outcomes. Both SGA- and LGA-born children are at an increased risk for overweight and obesity. This may have a long-term impact on cardiometabolic health as overweight tends to track to adulthood. Other cardiometabolic risk factors, including alterations in glucose metabolism, dyslipidemia, hypertension, and low-grade inflammation are associated with birth weight. Many of these risk factors are related to overweight or adverse fat distribution. Since later cardiometabolic risk is often mediated by early growth pattern and later overweight in SGA and LGA children, it is important to focus on staying normal weight throughout life. Hence, effective interventions to reduce cardiometabolic risk in LGA and SGA children should be developed.
Collapse
Affiliation(s)
- Henrikki Nordman
- Department of Pediatrics, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland,
| | - Jarmo Jääskeläinen
- Department of Pediatrics, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Raimo Voutilainen
- Department of Pediatrics, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
8
|
Cerón NA, Gutiérrez OO, Cerón OM, Ortiz RA. Complicaciones cardiovasculares en relación con la programación fetal. REPERTORIO DE MEDICINA Y CIRUGÍA 2021. [DOI: 10.31260/repertmedcir.01217273.943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Introducción: la programación fetal ofrece nuevas perspectivas sobre el origen de las enfermedades cardiovasculares, relacionando su aparición con factores perinatales. Objetivo: exponer evidencia que vincule las alteraciones gestacionales con las enfermedades cardiovasculares en la vida adulta del feto. Metodología: búsqueda en las bases de datos EBSCO, COCHRANE, MEDLINE, PROQUEST y SciELO de los artículos de revisión e investigaciones originales en inglés publicados en los últimos diez años. Se utilizaron términos MeSH para búsqueda controlada y se evaluaron los estudios con STROBE y PRISMA según correspondía. Resultados: los hallazgos sugieren que nacer con menos de 2600 k guarda relación con diabetes mellitus (OR de 1.607 IC 95% 1.324-1.951), hipertensión arterial (OR de 1.15 IC 95% 1.043-1.288) y menor función endotelial (1.94+0.37 vs 2.68+0.41, p: 0.0001) en la adultez. La prematuridad se asocia con mayores presiones arteriales sistólicas (4.2 mmHg IC 95%; 2.8 - 5.7 p 0.001) y diastólicas (2.6 mmHg IC 95%; 1.2-4.0; p 0.001). Las alteraciones nutricionales maternas y la diabetes gestacional aumentan el riesgo de síndrome metabólico (OR 1.2 IC 95% 0.9-1.7) y sobrepeso en la edad escolar (OR 1.81 IC 95% 1.18 - 2.86). Conclusión: los resultados adversos en la gestación están relacionados con el desarrollo de enfermedades cardiovasculares en la vida adulta del feto expuesto.
Collapse
|
9
|
Barbieri MR, Fontes AM, Barbieri MA, Saraiva MCP, Simões VMF, Silva AAMD, Abraham KJ, Bettiol H. Effects of FTO and PPARγ variants on intrauterine growth restriction in a Brazilian birth cohort. ACTA ACUST UNITED AC 2021; 54:e10465. [PMID: 33729310 PMCID: PMC7945878 DOI: 10.1590/1414-431x202010465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/02/2021] [Indexed: 01/18/2023]
Abstract
Intrauterine growth restriction (IUGR) is related to a higher risk of neonatal mortality, minor cognitive deficit, metabolic syndrome, and cardiovascular disease in adulthood. In previous studies, genetic variants in the FTO (fat mass and obesity-associated) and PPARγ (peroxisome proliferator-activated receptor-gamma) genes have been associated with metabolic disease, body mass index, and obesity among other outcomes. We studied the association of selected FTO (rs1421085, rs55682395, rs17817449, rs8043757, rs9926289, and rs9939609) and PPARγ (rs10865710, rs17036263, rs35206526, rs1801282, rs28763894, rs41516544, rs62243567, rs3856806, and rs1805151) single-nucleotide polymorphisms (SNPs) with IUGR, through a case-control study in a cohort of live births that occurred from June 1978 to May 1979 in a Brazilian city. We selected 280 IUGR cases and 256 controls for analysis. Logistic regression was used to jointly analyze the SNPs as well as factors such as maternal smoking, age, and schooling. We found that the PPARγ rs41516544 increased the risk of IUGR for male offspring (OR 27.83, 95%CI 3.65-212.32) as well as for female offspring (OR=8.94, 95%CI: 1.96-40.88). The FTO rs9939609 TA genotype resulted in a reduced susceptibility to IUGR for male offspring only (OR=0.47, 95%CI: 0.26-0.86). In conclusion, we demonstrated that PPARγ SNP had a positive effect and FTO SNP had a negative effect on IUGR occurrence, and these effects were gender-specific.
Collapse
Affiliation(s)
- M R Barbieri
- Departamento de Puericultura e Pediatria, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - A M Fontes
- Departamento de Genética, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - M A Barbieri
- Departamento de Puericultura e Pediatria, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - M C P Saraiva
- Departamento de Clínica Infantil, Faculdade de Odontologia de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - V M F Simões
- Departamento de Saúde Pública, Centro de Ciências da Saúde, Universidade Federal do Maranhão, São Luís, MA, Brasil
| | - A A M da Silva
- Departamento de Saúde Pública, Centro de Ciências da Saúde, Universidade Federal do Maranhão, São Luís, MA, Brasil
| | - K J Abraham
- Departamento de Economia, Faculdade de Economia, Administração e Contabilidade de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - H Bettiol
- Departamento de Puericultura e Pediatria, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| |
Collapse
|
10
|
Vassilakou T. Childhood Malnutrition: Time for Action. CHILDREN-BASEL 2021; 8:children8020103. [PMID: 33546298 PMCID: PMC7913494 DOI: 10.3390/children8020103] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 01/22/2023]
Affiliation(s)
- Tonia Vassilakou
- Department of Public Health Policy, School of Public Health, University of West Attica, Athens University Campus, 196 Alexandras Ave, GR-11521 Athens, Greece
| |
Collapse
|
11
|
Chatterjee S, Ouidir M, Tekola-Ayele F. Pleiotropic genetic influence on birth weight and childhood obesity. Sci Rep 2021; 11:48. [PMID: 33420178 PMCID: PMC7794220 DOI: 10.1038/s41598-020-80084-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 12/09/2020] [Indexed: 01/09/2023] Open
Abstract
Childhood obesity is a global public health problem. Understanding the molecular mechanisms that underlie early origins of childhood obesity can facilitate interventions. Consistent phenotypic and genetic correlations have been found between childhood obesity traits and birth weight (a proxy for in-utero growth), suggesting shared genetic influences (pleiotropy). We aimed to (1) investigate whether there is significant shared genetic influence between birth weight and childhood obesity traits, and (2) to identify genetic loci with shared effects. Using a statistical approach that integrates summary statistics and functional annotations for paired traits, we found strong evidence of pleiotropy (P < 3.53 × 10–127) and enrichment of functional annotations (P < 1.62 × 10–39) between birth weight and childhood body mass index (BMI)/obesity. The pleiotropic loci were enriched for regulatory features in skeletal muscle, adipose and brain tissues and in cell lines derived from blood lymphocytes. At 5% false discovery rate, 6 loci were associated with birth weight and childhood BMI and 13 loci were associated with birth weight and childhood obesity. Out of these 19 loci, one locus (EBF1) was novel to childhood obesity and one locus (LMBR1L) was novel to both birth weight and childhood BMI/obesity. These findings give evidence of substantial shared genetic effects in the regulation of both fetal growth and childhood obesity.
Collapse
Affiliation(s)
- Suvo Chatterjee
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, Room 3204, Bethesda, 20892-7004, USA
| | - Marion Ouidir
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, Room 3204, Bethesda, 20892-7004, USA
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, Room 3204, Bethesda, 20892-7004, USA.
| |
Collapse
|
12
|
An J, Wang J, Guo L, Xiao Y, Lu W, Li L, Chen L, Wang X, Dong Z. The Impact of Gut Microbiome on Metabolic Disorders During Catch-Up Growth in Small-for-Gestational-Age. Front Endocrinol (Lausanne) 2021; 12:630526. [PMID: 33746902 PMCID: PMC7970190 DOI: 10.3389/fendo.2021.630526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Catch-up growth (CUG) in small for gestational age (SGA) leads to increased risk of metabolic syndrome and cardiovascular diseases in adults. It remains unclear if microbiota could play an important role in CUG-SGA independent of genetic or nutritional factors. The present study explored the role of gut microbiota in, and its association with, metabolic disorders during CUG-SGA. METHODS An SGA rat model was established by restricting food intake during pregnancy, and the rats were divided into catch-up growth (CUG-SGA) and non-catch-up growth (NCUG-SGA) groups based on body weight and length at the fourth postnatal week. High-throughput sequencing of 16S rRNA was conducted to detect the diversity and composition of the gut microbiota. Fecal short-chain fatty acids (SCFAs) were detected by gas chromatography-mass spectrometry. Transcriptome sequencing of liver tissue was performed and verified using real-time PCR. Concentrations of insulin and total cholesterol were determined using enzyme-linked immunosorbent assay. RESULTS The composition of gut microbiota in CUG-SGA rats differed from that of NCUG-SGA rats, with reduced abundance of Lactobacillus in the CUG-SGA group. The decrease in Lactobacillus was significantly associated with increased body weight and upregulated insulin and total cholesterol levels. Five SCFAs and two branched chain fatty acids were significantly higher in the CUG-SGA group than in the NCUG-SGA group. Additionally, SCFAs were positively associated with clinical indices such as weight, body mass index, insulin, and total cholesterol. Transcriptomic data revealed that insulin-like growth factor-2 expression was significantly decreased in CUG-SGA rats and was associated with a decrease in Lactobacillus bacteria. CONCLUSION Lactobacillus and SCFAs were associated with the metabolic disorders during CUG in SGA. Gut microbiome may play a certain role on metabolic disorders during catch-up growth in small-for-gestational-age.
Collapse
Affiliation(s)
- Jingjing An
- Department of Endocrine and Metabolic Disease, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junqi Wang
- Department of Endocrine and Metabolic Disease, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Guo
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, United States
| | - Yuan Xiao
- Department of Endocrine and Metabolic Disease, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenli Lu
- Department of Endocrine and Metabolic Disease, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lin Li
- Department of Endocrine and Metabolic Disease, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lifen Chen
- Department of Endocrine and Metabolic Disease, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinqiong Wang
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Zhiya Dong, ; Xinqiong Wang,
| | - Zhiya Dong
- Department of Endocrine and Metabolic Disease, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Pediatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Zhiya Dong, ; Xinqiong Wang,
| |
Collapse
|
13
|
Walczak M, Szalecki M, Horneff G, Lebl J, Kalina-Faska B, Giemza T, Moldovanu F, Nanu M, Zouater H. Long-term follow up of carbohydrate metabolism and adverse events after termination of Omnitrope® treatment in children born small for gestational age. Ther Adv Endocrinol Metab 2021; 12:20420188211013121. [PMID: 34104396 PMCID: PMC8111548 DOI: 10.1177/20420188211013121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/08/2021] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Recombinant human growth hormone (rhGH) therapy can affect carbohydrate metabolism and lead to impaired glucose tolerance during treatment. In addition, short children born small for gestational age (SGA) are predisposed to metabolic abnormalities. This study assessed the long-term safety of rhGH (Omnitrope®) use in short children born SGA. METHODS This was a follow-up observational study of patients from a phase IV study. The baseline visit was the final visit of the phase IV study. Further visits were planned after 6 months (F1), 1 year (F2), 5 years (F3), and 10 years (F4). The primary objective was to evaluate the long-term effect of rhGH treatment on the development of diabetes mellitus; secondary objectives included incidence/severity of adverse events (AEs). RESULTS In total, 130 subjects were enrolled in the follow-up study; 99 completed F1, 88 completed F2, and 13 completed F3 (no subject reached F4). The full analysis set for evaluation comprised 118 patients (64 female). Mean (standard deviation) duration of follow up was 39.6 (24.4) months. No subject was newly diagnosed with diabetes. The results for carbohydrate metabolism parameters were consistent with this finding. A total of 144 AEs were reported in 54 subjects; these were mostly of mild-to-moderate intensity (96.5%) and not suspected to be related to previous rhGH treatment (94.4%). Serious AEs (n = 18) were reported in eight patients; three (in one patient) were suspected as possibly related to previous rhGH treatment (anemia, menorrhagia, oligomenorrhoea). One fatal event occurred (sepsis), which was judged as not related to previous rhGH treatment. CONCLUSIONS None of the participating subjects, who had all been previously treated with Omnitrope® in a phase IV study, developed diabetes during this follow-up study. In addition, no other unexpected or concerning safety signals were observed.
Collapse
Affiliation(s)
- Mieczyslaw Walczak
- Department of Pediatrics, Endocrinology, Diabetology, Metabolic Diseases and Cardiology of the Developmental Age, Pomeranian Medical University, Szczecin, Zachodniopomorskie, Poland
| | - Mieczyslaw Szalecki
- Collegium Medicum UJK, Kielce, Children’s Memorial Health Institute, Warsaw, Poland
| | - Gerd Horneff
- Department of Pediatrics, Center for Pediatric Rheumatology, Asklepios Clinic Sankt Augustin, Sankt Augustin, Germany
- Department of Pediatric and Adolescents Medicine, University Hospital of Cologne, Cologne, Germany
| | - Jan Lebl
- Department of Pediatrics, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Barbara Kalina-Faska
- Department of Pediatrics and Pediatric Endocrinology, Medical University of Silesia, Faculty of Medical Science, Katowice, Slaskie, Poland
| | | | - Florentina Moldovanu
- National Institute for Mother and Child Health, ‘Alessandrescu Rusescu’, Bucharest, Romania
| | - Michaela Nanu
- National Institute for Mother and Child Health, ‘Alessandrescu Rusescu’, Bucharest, Romania
| | | |
Collapse
|
14
|
Marcos-Pasero H, Aguilar-Aguilar E, Ikonomopoulou MP, Loria-Kohen V. BDNF Gene as a Precision Skill of Obesity Management. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1331:233-248. [PMID: 34453302 DOI: 10.1007/978-3-030-74046-7_15] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The scarcity of the results obtained for the treatment of obesity leads us to consider new strategies, contemplating all the factors involved in the development of the disease. One of the key molecules for controlling body weight and energy homeostasis is the brain-derived neurotrophic factor (BDNF). This work summarizes the mechanisms in which BDNF gene regulates this multifactorial disease. In addition, we discuss the role of other BDNF polymorphisms as genetic determinants of obesity. In this context, a total of 14 SNPs near or inside BDNF/BDNF-AS related to BMI were identified in various GWASs. Finally, we assess gene-diet interaction as a novel tool to prevent obesity and formulate solid and personalized nutritional management. Our research group has performed the first study on the association of BDNF-AS rs925946 polymorphism and calcium intake as potential modulators of the nutritional status. Although these results should be confirmed in future studies, they open the path for new prevention opportunities.
Collapse
Affiliation(s)
- Helena Marcos-Pasero
- Nutrition and Clinical Trials Unit, GENYAL Platform, IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - Elena Aguilar-Aguilar
- Nutrition and Clinical Trials Unit, GENYAL Platform, IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - Maria P Ikonomopoulou
- Translational Venomics Group, IMDEA-Food, CEI UAM+CSIC, Madrid, Spain.,Institute for Molecular Bioscience, The University of Queensland, St Lucia, Australia
| | - Viviana Loria-Kohen
- Nutrition and Clinical Trials Unit, GENYAL Platform, IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain. .,Department of Nutrition and Food Science, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain.
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW A growing body of epidemiological and experimental data indicate that nutritional or environmental stressors during early development can induce long-term adaptations that increase risk of obesity, diabetes, cardiovascular disease, and other chronic conditions-a phenomenon termed "developmental programming." A common phenotype in humans and animal models is altered body composition, with reduced muscle and bone mass, and increased fat mass. In this review, we summarize the recent literature linking prenatal factors to future body composition and explore contributing mechanisms. RECENT FINDINGS Many prenatal exposures, including intrauterine growth restriction, extremes of birth weight, maternal obesity, and maternal diabetes, are associated with increased fat mass, reduced muscle mass, and decreased bone density, with effects reported throughout infancy and childhood, and persisting into middle age. Mechanisms and mediators include maternal diet, breastmilk composition, metabolites, appetite regulation, genetic and epigenetic influences, stem cell commitment and function, and mitochondrial metabolism. Differences in body composition are a common phenotype following disruptions to the prenatal environment, and may contribute to developmental programming of obesity and diabetes risk.
Collapse
Affiliation(s)
- Elvira Isganaitis
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
- Research Division, Joslin Diabetes Center, 1 Joslin Place, Room 655A, Boston, 02215, MA, USA.
| |
Collapse
|
16
|
Gupta MK, Sarojamma V, Vadde R. Diabetes and Pancreatic Cancer: A Bidirectional Relationship Perspective. EXPLORING PANCREATIC METABOLISM AND MALIGNANCY 2019:35-51. [DOI: 10.1007/978-981-32-9393-9_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
|
17
|
Lang L, Chemmalakuzhy R, Shay C, Teng Y. PFKP Signaling at a Glance: An Emerging Mediator of Cancer Cell Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1134:243-258. [PMID: 30919341 DOI: 10.1007/978-3-030-12668-1_13] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Phosphofructokinase-1 (PFK-1), a rate-determining enzyme of glycolysis, is an allosteric enzyme that regulates the oxidation of glucose in cellular respiration. Glycolysis phosphofructokinase platelet (PFKP) is the platelet isoform and works as an important mediator of cell metabolism. Considering that PFKP is a crucial player in many steps of cancer initiation and metastasis, we reviewed the specificities and complexities of PFKP and its biological roles in human diseases, especially malignant tumors. The possible use of PFKP as a diagnostic marker or a drug target in the prevention or treatment of cancer is also discussed.
Collapse
Affiliation(s)
- Liwei Lang
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Ron Chemmalakuzhy
- Department of Biology, College of Science and Mathematics, Augusta University, Augusta, GA, USA
| | - Chloe Shay
- The Robinson College of Business, Georgia State University, Atlanta, GA, USA
- Division of Endocrinology and Diabetes, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Yong Teng
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, USA.
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA.
- Department of Medical Laboratory, Imaging and Radiologic Sciences, College of Allied Health, Augusta University, Augusta, GA, USA.
| |
Collapse
|
18
|
Sureshchandra S, Wilson RM, Rais M, Marshall NE, Purnell JQ, Thornburg KL, Messaoudi I. Maternal Pregravid Obesity Remodels the DNA Methylation Landscape of Cord Blood Monocytes Disrupting Their Inflammatory Program. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:2729-2744. [PMID: 28887432 PMCID: PMC7384891 DOI: 10.4049/jimmunol.1700434] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 08/07/2017] [Indexed: 01/03/2023]
Abstract
Prepregnancy maternal obesity is associated with adverse outcomes for the offspring, including increased incidence of neonatal bacterial sepsis and necrotizing enterocolitis. We recently reported that umbilical cord blood (UCB) monocytes from babies born to obese mothers generate a reduced IL-6/TNF-α response to TLR 1/2 and 4 ligands compared to those collected from lean mothers. These observations suggest altered development of the offspring's immune system, which in turn results in dysregulated function. We therefore investigated transcriptional and epigenetic differences within UCB monocytes stratified by prepregnancy maternal body mass index. We show that UCB monocytes from babies born to obese mothers generate a dampened response to LPS stimulation compared with those born to lean mothers, at the level of secreted immune mediators and transcription. Because gene expression profiles of resting UCB monocytes from both groups were comparable, we next investigated the role of epigenetic differences. Indeed, we detected stark differences in methylation levels within promoters and regulatory regions of genes involved in TLR signaling in resting UCB monocytes. Interestingly, the DNA methylation status of resting cells was highly predictive of transcriptional changes post-LPS stimulation, suggesting that cytosine methylation is one of the dominant mechanisms driving functional inadequacy in UCB monocytes obtained from babies born to obese mothers. These data highlight a potentially critical role of maternal pregravid obesity-associated epigenetic changes in influencing the function of an offspring's monocytes at birth. These findings further our understanding of mechanisms that explain the increased risk of infection in neonates born to mothers with high prepregnancy body mass index.
Collapse
Affiliation(s)
- Suhas Sureshchandra
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA 92697
| | - Randall M Wilson
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA 92521
| | - Maham Rais
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA 92521
| | - Nicole E Marshall
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland, OR 97239; and
| | - Jonathan Q Purnell
- Department of Medicine, The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239
| | - Kent L Thornburg
- Department of Medicine, The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA 92697;
| |
Collapse
|
19
|
The importance of platelet phosphofructokinase (PFKP) rs6602024 polymorphism in pathogenesis of obesity. ACTA ACUST UNITED AC 2017. [DOI: 10.18794/aams/64163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
WstępCelem pracy było wykazanie potencjalnego związku między występowaniem nadwagi i otyłości a polimorfizmem rs6602024 genu fosfofruktokinazy płytkowej (PFKP) u kolejnych pacjentów zgłaszających się do lekarza pierwszego kontaktu w POZ.Materiał i metodyBadaniem objęto łącznie kolejnych 438 pacjentów z rejonu Polski południowej, którzy zgłaszali się do poradni ogólnej POZ. Genotypowanie polimorfizmu genu PFKP rs6602024 prowadzono z wykorzystaniem znakowanych fluorescencyjnie sond. Do obliczeń poszczególnych parametrów w ujęciu statystycznym zastosowano program Statistica 8,0.WynikiU osób z nadwagą oraz otyłością, a także w całej badanej grupie osób z nadwagą lub otyłością wykazano występowanie określonych zaburzeń metabolicznych. Wyniki porównano z rezultatami osób bez nadwagi i otyłości. U badanych osób nie stwierdzono istotnych statystycznie różnic w rozkładzie genotypów i alleli polimorfizmu rs6602024 PFKP w porównaniu z kontrolą. W grupie kontrolnej wykazano znamienną statystycznie różnicę pomiędzy kobietami i mężczyznami w rozkładzie alleli. Wśród kobiet stwierdzono znamienną statystycznie różnicę w rozkładzie alleli G/A między osobami z grupy kontrolnej a osobami z nadwagą oraz nadwagą i otyłością razem wziętych.Wnioski1. Nadwaga i otyłość bardzo często występują u osób zgłaszających się do lekarza POZ.
2. Występowaniu allelu A polimorfizmu rs6602024 genu PFKP u kobiet towarzyszy zwiększone ryzyko nadwagi i otyłości.
Collapse
|
20
|
Levy E, Saenger A, Steffes M, Delvin E. Pediatric Obesity and Cardiometabolic Disorders: Risk Factors and Biomarkers. EJIFCC 2017; 28:6-24. [PMID: 28439216 PMCID: PMC5387697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Obesity remains the most prevailing disorder in childhood males and females worldwide. Its high prevalence markedly predisposes children to insulin resistance, hypertension, hyperlipidemia and liver disorders while enhancing the risk of type 2 diabetes and cardiovascular diseases. In this review, the relationship of obesity with genetic and environmental factors will be described and the underlined causes will briefly be reported. As obesity in children constitutes an increasingly health concern, important potential biomarkers have been discussed for the diagnosis, treatment and follow-up of the wide range of overweight-related complications. Awareness about the applicability and limitations of these preventive and predictive biomarkers will intensify the research and medical efforts for new developments in order to efficiently struggle against childhood obesity.
Collapse
Affiliation(s)
- E. Levy
- Department of Nutrition, University of Montreal, Montreal, Quebec, Canada, CHU Sainte-Justine Research Center, Nutrition, Gastroenterology and Hepatology Division, University of Montreal, Montreal, Quebec, Canada
| | - A.K. Saenger
- Department of Laboratory Medicine and Pathology, University of Minnesota Health, Minneapolis, MN, USA
| | - M.W. Steffes
- Department of Laboratory Medicine and Pathology, University of Minnesota Health, Minneapolis, MN, USA
| | - E. Delvin
- CHU Sainte-Justine Research Center, Nutrition, Gastroenterology and Hepatology Division, University of Montreal, Montreal, Quebec, Canada, Department of Biochemistry, University of Montreal, Montreal, Quebec, Canada,Ph.D. CHU Sainte-Justine Research Center 3175 Côte Ste-Catherine Montréal, Québec, H3T 1C5 Canada (514) 345-4931 ext. 6268
| |
Collapse
|
21
|
Bassareo PP, Marras AR, Cugusi L, Zedda AM, Mercuro G. The reasons why cardiologists should consider prematurity at birth and intrauterine growth retardation among risk factors. J Cardiovasc Med (Hagerstown) 2017; 17:323-9. [PMID: 26627499 DOI: 10.2459/jcm.0000000000000338] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The survival percentage of infants born preterm has risen steadily worldwide thanks to the giant steps forward made in the field of perinatal (the period immediately after birth) and neonatal (the first 4 weeks of birth following delivery) medicine. However, prematurity at birth and consequent low birth weight still represent the major causes of neonatal morbidity and mortality. Infants born preterm are at high risk of developing neurological, ophthalmological, and gastrointestinal complications as well. Furthermore, extensive more recent epidemiological findings have demonstrated an increase in risk factors and a higher mortality rate due to cardiovascular causes in patients born preterm and/or with intrauterine growth restriction. The aim of this review is to provide scientific evidence about how the cardiovascular system may be negatively influenced by prematurity and by a low birth weight that should by rights be viewed as new cardiovascular risk factors. This condition is referred to as 'cardiovascular perinatal programming'. In the light of the above, an early, constant, and prolonged cardiovascular follow-up should be implemented in former preterm individuals.
Collapse
Affiliation(s)
- Pier P Bassareo
- Department of Medical Sciences 'M. Aresu', University of Cagliari, Cagliari, Italy
| | | | | | | | | |
Collapse
|
22
|
Giapros V, Vavva E, Siomou E, Kolios G, Tsabouri S, Cholevas V, Bairaktari E, Tzoufi M, Challa A. Low-birth-weight, but not catch-up growth, correlates with insulin resistance and resistin level in SGA infants at 12 months. J Matern Fetal Neonatal Med 2016; 30:1771-1776. [PMID: 27609490 DOI: 10.1080/14767058.2016.1224838] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To investigate the insulin resistance status in SGA infants at 12 months and its relationship with auxological and metabolic parameters. METHODS One group of 45 SGA and one of 50 appropriate for gestational age infants were followed from birth to the end of the first year of life. At 12 months, skinfold thickness, waist circumference, and blood levels of glucose, insulin, adiponectin, leptin, resistin, visfatin, retinol-binding protein 4, IGFs, lipids profile were determined, and the HOMA-IR index was calculated. RESULTS The SGAs had increased insulin (5.2 ± 2.7 versus 2.9 ± 2.4 μIU/ml, p = 0.012) and HOMA-IR (1.09 ± 0.9 versus 0.59 ± 0.55, p = 0.016). In multiple regression, insulin resistance indices were independently correlated with low-birth-weight (β = -2.92, p = 0.015 for insulin, β = -2.98, p = 0.011 for HOMA-IR) but not with catch-up growth in either height or weight or any other metabolic parameter. Resistin was higher in the SGAs (5.1 ± 2.1 versus 3.9 ± 2.1 ng/ml, p = 0.03) and independently correlated with low-birth-weight but not insulin resistance. Resistin was negatively correlated with total cholesterol (R = -0.33, p = 0.007) and positively with lipoprotein(a) (R = 0.49, p = 0.001). CONCLUSION Low-birth-weight, but not catch-up growth or adiposity tissue hormones, was correlated with insulin resistance at 12 months in non-obese SGA infants. The higher resistin in SGA infants and its correlation with total cholesterol and lipoprotein(a) need further clarification.
Collapse
Affiliation(s)
| | | | | | | | | | - Vasileios Cholevas
- c Pediatric Research Laboratory , University of Ioannina , Ioannina , Greece
| | | | - Meropi Tzoufi
- a Neonatal Intensive Care Unit, Child Health Department
| | - Anna Challa
- c Pediatric Research Laboratory , University of Ioannina , Ioannina , Greece
| |
Collapse
|
23
|
Gallo P, Cioffi L, Limauro R, Farris E, Bianco V, Sassi R, De Giovanni M, Gallo V, D’Onofrio A, Di Maio S. SGA Children in Pediatric Primary Care: What Is the Best Choice, Large or Small? A 10-Year Prospective Longitudinal Study. Glob Pediatr Health 2016; 3:2333794X16659993. [PMID: 27583297 PMCID: PMC4995669 DOI: 10.1177/2333794x16659993] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 05/31/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Epidemiologic evidences suggest a strong association between low birth weight and some diseases in adult life ( hypertension, diabetes, cardiovascular diseases).Aim of this study was to evaluate the obesity/overweight prevalence in a population of children born small for gestation age, SGA children 400, 208 males and 192 females compared to a population of children born appropriate for gestational age 6818 AGA children, 3502 males and 3316 females, during childhood. Our intention was also to build the natural history of weight gain during prepubertal age in children born SGA and AGA. DESIGN AND METHODS Observational prospective longitudinal study. We followed our patients from January2001 up to December 2010; weight, height and body mass index (BMI) were evaluated in all the SGA and AGA children. BMI z-score range for defining overweight and obesity was, respectively, 1.13 to 1.7 and >1.7 according to CDC growth charts. RESULTS In transversal evaluation, we prove that 10-year-old SGA females are twice obese and more overweight compared to equal age AGA females. In longitudinal evaluation, we highlight different observations: SGA children obese at 2 years are still obese at 10 years; the number of obese SGA children increases gradually until the age of 10; AGA children, appear to be less obese than SGA children at 10 years. CONCLUSION SGA males and females are more obese at 5 and 10 years compared to the AGA population. Primary care pediatricians, through early detection of the children at risk, can carry out an effective obesity prevention project in SGA children.
Collapse
Affiliation(s)
- Patrizia Gallo
- FIMP (Italian Federation of Pediatricians), Naples Section, Pediatric Primary Care Local Health Authority, Naples, Italy
| | - Luigi Cioffi
- FIMP (Italian Federation of Pediatricians), Naples Section, Pediatric Primary Care Local Health Authority, Naples, Italy
| | - Raffaele Limauro
- FIMP (Italian Federation of Pediatricians), Naples Section, Pediatric Primary Care Local Health Authority, Naples, Italy
| | - Evelina Farris
- FIMP (Italian Federation of Pediatricians), Naples Section, Pediatric Primary Care Local Health Authority, Naples, Italy
| | - Vincenzo Bianco
- FIMP (Italian Federation of Pediatricians), Naples Section, Pediatric Primary Care Local Health Authority, Naples, Italy
| | - Roberto Sassi
- FIMP (Italian Federation of Pediatricians), Naples Section, Pediatric Primary Care Local Health Authority, Naples, Italy
| | - Maria De Giovanni
- FIMP (Italian Federation of Pediatricians), Naples Section, Pediatric Primary Care Local Health Authority, Naples, Italy
| | | | - Antonietta D’Onofrio
- FIMP (Italian Federation of Pediatricians), Naples Section, Pediatric Primary Care Local Health Authority, Naples, Italy
| | | |
Collapse
|
24
|
Abstract
This review discusses available literature on the diagnosis and management of intrauterine growth restriction (IUGR) in women with type 1 diabetes. IUGR is diagnosed when ultrasound-estimated fetal weight is below the 10th percentile for gestational age. IUGR diagnosis implies a pathologic process behind low fetal weight. IUGR in pregnancy complicated by type 1 diabetes is usually caused by placental dysfunction related to maternal vasculopathy. Prevention of IUGR should ideally start before pregnancy. Strict glycemic control and intensive treatment of nephropathy and hypertension are essential. Low-dose aspirin initiated before 16 gestational weeks can also reduce IUGR risk in women with vasculopathy. Umbilical and uterine artery Doppler studies can guide diagnosis and surveillance of fetuses with IUGR. Decisions regarding the timing of delivery should be based on assessment of umbilical artery Doppler. The risk of prematurity and impaired fetal lung maturation should always be considered, especially in fetuses younger than 32 weeks.
Collapse
Affiliation(s)
- Paweł Gutaj
- Department of Obstetrics and Women’s Diseases, Poznan University of Medical Sciences, Polna 33, 60-535 Poznan, Poland
| | - Ewa Wender-Ozegowska
- Department of Obstetrics and Women’s Diseases, Poznan University of Medical Sciences, Polna 33, 60-535 Poznan, Poland
| |
Collapse
|
25
|
Campbell MK. Biological, environmental, and social influences on childhood obesity. Pediatr Res 2016; 79:205-11. [PMID: 26484623 DOI: 10.1038/pr.2015.208] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 09/30/2015] [Indexed: 01/04/2023]
Abstract
The prevalence of childhood obesity has increased globally over the past three decades, with evidence of recent leveling off in developed countries. Reduction in the, currently high, prevalence of obesity will require a full understanding of the biological and social pathways to obesity in order to develop appropriately targeted prevention strategies in early life. Determinants of childhood obesity include individual level factors, including biological, social, and behavioral risks, acting within the influence of the child's family environment, which is, in turn, imbedded in the context of the community environment. These influences act across childhood, with suggestions of early critical periods of biological and behavioral plasticity. There is evidence of sex and gender differences in the responses of boys and girls to their environments. The evidence that determinants of childhood obesity act at many levels and at different stages of childhood is of policy relevance to those planning early health promotion and primary prevention programs as it suggests the need to address the individual, the family, the physical environment, the social environment, and social policy. The purpose of this narrative review is to summarize current, and emerging, literature in a multilevel, life course framework.
Collapse
Affiliation(s)
- M Karen Campbell
- Department of Epidemiology and Biostatistics, The University of Western Ontario, London, Ontario, Canada.,Department of Pediatrics, The University of Western Ontario, London, Ontario, Canada.,Department of Obstetrics & Gynecology, The University of Western Ontario, London, Ontario, Canada.,Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
26
|
Wilson RM, Messaoudi I. The impact of maternal obesity during pregnancy on offspring immunity. Mol Cell Endocrinol 2015; 418 Pt 2:134-42. [PMID: 26232506 PMCID: PMC4674375 DOI: 10.1016/j.mce.2015.07.028] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 06/28/2015] [Accepted: 07/27/2015] [Indexed: 12/19/2022]
Abstract
In the United States, approximately 64% of women of childbearing age are either overweight or obese. Maternal obesity during pregnancy is associated with a greater risk for adverse maternal-fetal outcomes. Adverse health outcomes for the offspring can persist into adulthood, increasing the incidence of several chronic conditions including cardiovascular disease, diabetes, and asthma. Since these diseases have a significant inflammatory component, these observations are indicative of perturbation of the normal development and maturation of the immune system of the offspring in utero. This hypothesis is strongly supported by data from several rodent studies. Although the mechanisms of these perturbations are not fully understood, it is thought that increased placental inflammation due to obesity may directly affect neonatal development through alterations in nutrient transport. In this review we examine the impact of maternal obesity on the neonatal immune system, and potential mechanisms for the changes observed.
Collapse
Affiliation(s)
- Randall M Wilson
- Graduate Program in Cell, Molecular, and Developmental Biology, University of California, Riverside, USA
| | - Ilhem Messaoudi
- Graduate Program in Cell, Molecular, and Developmental Biology, University of California, Riverside, USA; Division of Biomedical Sciences, School of Medicine, University of California, Riverside, USA.
| |
Collapse
|
27
|
Su RN, Zhu WW, Wei YM, Wang C, Feng H, Lin L, Yang HX. Maternal and neonatal outcomes in multiple pregnancy: A multicentre study in the Beijing population. Chronic Dis Transl Med 2015; 1:197-202. [PMID: 29063007 PMCID: PMC5643594 DOI: 10.1016/j.cdtm.2015.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Indexed: 12/04/2022] Open
Abstract
Objective To compare the adverse maternal and neonatal outcomes of multiple pregnancy and singleton pregnancy from multiple medical centers in Beijing. Methods Data concerning maternal and neonatal adverse outcomes in multiple and singleton pregnancies were collected from 15 hospitals in Beijing by a systemic cluster sampling survey conducted from 20 June to 30 November 2013. The SPSS software (version 20.0) was used for data analysis. The χ2 test was used for statistical analyses. Results The rate of caesarean deliveries was much higher in women with multiple pregnancies (85.8%) than that in women with singleton pregnancies (42.6%, χ2 = 190.8, P < 0.001). The incidences of anemia (χ2 = 40.023, P < 0.001), preterm labor (χ2 = 1021.172, P < 0.001), gestational diabetes mellitus (χ2 = 9.311, P < 0.01), hypertensive disorders (χ2 = 122.708, P < 0.001) and post-partum hemorrhage (χ2 = 48.550, P < 0.001) was significantly increased with multiple pregnancy. In addition, multiple pregnancy was associated with a significantly higher rate of small-for-gestational-age infants (χ2 = 92.602, P < 0.001), low birth weight (χ2 = 1141.713, P < 0.001), and neonatal intensive care unit (NICU) admission (χ2 = 340.129, P < 0.001). Conclusions Multiple pregnancy is a significant risk factor for adverse maternal and neonatal outcomes in Beijing. Improving obstetric care for multiple pregnancy, particularly in reducing preterm labor, is required to reduce the risk to mothers and infants.
Collapse
Affiliation(s)
- Ri-Na Su
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing 100034, China
| | - Wei-Wei Zhu
- National Institute of Hospital Administration, Beijing 100191, China
| | - Yu-Mei Wei
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing 100034, China
| | - Chen Wang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing 100034, China
| | - Hui Feng
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing 100034, China
| | - Li Lin
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing 100034, China
| | - Hui-Xia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
28
|
Andraweera PH, Gatford KL, Dekker GA, Leemaqz S, Russell D, Thompson SD, McCowan L, Roberts CT. Insulin family polymorphisms in pregnancies complicated by small for gestational age infants. Mol Hum Reprod 2015; 21:745-52. [DOI: 10.1093/molehr/gav031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/01/2015] [Indexed: 11/12/2022] Open
|
29
|
Li J, Wang Z, Li R, Wu R. BAYESIAN GROUP LASSO FOR NONPARAMETRIC VARYING-COEFFICIENT MODELS WITH APPLICATION TO FUNCTIONAL GENOME-WIDE ASSOCIATION STUDIES. Ann Appl Stat 2015; 9:640-664. [PMID: 26478762 DOI: 10.1214/15-aoas808] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Although genome-wide association studies (GWAS) have proven powerful for comprehending the genetic architecture of complex traits, they are challenged by a high dimension of single-nucleotide polymorphisms (SNPs) as predictors, the presence of complex environmental factors, and longitudinal or functional natures of many complex traits or diseases. To address these challenges, we propose a high-dimensional varying-coefficient model for incorporating functional aspects of phenotypic traits into GWAS to formulate a so-called functional GWAS or fGWAS. Bayesian group lasso and the associated MCMC algorithms are developed to identify significant SNPs and estimate how they affect longitudinal traits through time-varying genetic actions. The model is generalized to analyze the genetic control of complex traits using subject-specific sparse longitudinal data. The statistical properties of the new model are investigated through simulation studies. We use the new model to analyze a real GWAS data set from the Framingham Heart Study, leading to the identification of several significant SNPs associated with age-specific changes of body mass index. The fGWAS model, equipped with Bayesian group lassso, will provide a useful tool for genetic and developmental analysis of complex traits or diseases.
Collapse
Affiliation(s)
- Jiahan Li
- Department of Applied and Computational, Mathematics and Statistics, The University of Notre Dame, Notre Dame, IN 46556.
| | - Zhong Wang
- Center for Computational Biology, Beijing Forestry University, Beijing, China 100083.
| | - Runze Li
- Department of Statistics, The Methodology Center, The Pennsylvania State University, University Park, PA 16802.
| | - Rongling Wu
- Center for Statistical Genetics, The Pennsylvania State University, Hershey, PA 17033.
| |
Collapse
|
30
|
Descamps OS, Tarantino E, Guilmot PF. Does FTO have a paradoxical effect in fetal life? BMC Genet 2014; 15:145. [PMID: 25539997 PMCID: PMC4332444 DOI: 10.1186/s12863-014-0145-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 12/05/2014] [Indexed: 01/21/2023] Open
Abstract
Background Low weight at birth is associated with obesity in later life. One hypothesis to explain such an association is that genetic variants that increase the risk of obesity also reduce fetal weight. Recently, obesity in adults was found to be associated with common variants of the fat mass and obesity-associated (FTO) gene. We examined the association between FTO polymorphisms and birth weight in a singleton, full-term birth cohort of 494 newborn-mother pairs without any complications. Results The risk alleles for obesity (“A” allele for the rs9939609 FTO variant and “G” allele for the rs9930506 FTO variant) were associated with low weight at birth. The mean differences per risk allele were −79 g (95% CI: −129 to −30; p = 0.002) for rs9939609 and −84 g (95% CI: −131 to −36; P < 0.001) for rs9930506. The level of association remained statistically significant after adjustment for the maternal risk allele and for variables usually associated with birth weight (−50 g, 95% CI: −99 to 0; p = 0.05 for rs9939609 and −48 g, 95% CI: −100 to 0; p = 0.05 for rs9930506). In the follow-up, the allelic difference in weight was attenuated over time. Conclusions The FTO variants that confer a predisposition to obesity later in life appear to be associated with low weight at birth. This finding favors the hypothesis of a common genetic denominator that predisposes to a low weight at birth and obesity in adults.
Collapse
Affiliation(s)
- Olivier S Descamps
- Center for Medical Research at Jolimont, 159 Rue Ferrer, B-7100, Haine Saint-Paul, Belgium. .,Department of Internal Medicine, Centre Hospitalier Jolimont-Lobbes, 159 Rue Ferrer, B-7100, Haine Saint-Paul, Belgium.
| | - Eric Tarantino
- Center for Medical Research at Jolimont, 159 Rue Ferrer, B-7100, Haine Saint-Paul, Belgium.
| | - Pierre-Francois Guilmot
- Department of Obstetrics and Gynecology, Centre Hospitalier Jolimont-Lobbes, 159 Rue Ferrer, B-7100, Haine Saint-Paul, Belgium.
| |
Collapse
|
31
|
Whiteman VE, August EM, Mogos M, Naik E, Garba M, Sanchez E, Weldeselasse HE, Salihu HM. Preterm birth in the first pregnancy and risk of neonatal death in the second pregnancy: a propensity score-weighted matching approach. J OBSTET GYNAECOL 2014; 35:30-6. [PMID: 25058689 DOI: 10.3109/01443615.2014.937328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The study purpose was to assess the relationship between various grades of preterm birth (moderate preterm: 33-36 weeks; severe preterm: 27-32 weeks; extreme preterm: ≤ 26 weeks) in the first pregnancy and neonatal mortality (death within 28 days of birth; early: 0-7 days; late: 8-28 days) in the second pregnancy. Using the Missouri maternally-linked dataset (1989-2005), a population-based, retrospective cohort analysis with propensity score-weighted matching was conducted on mothers with two consecutive singleton live births (n = 310,653 women). Women with a prior preterm birth were more likely to subsequently experience neonatal death. The odds increased in a dose-dependent pattern with ascending severity of the preterm event in the first pregnancy (moderate preterm: AOR = 1.32; 95% CI: 1.10-1.60; severe preterm: AOR = 2.62; 95% CI: 2.01-3.41; extreme preterm: AOR = 5.84; 95% CI: 4.28-7.97; p value for trend < 0.001). However, the pathway for the relationship between prior preterm birth and subsequent neonatal mortality may be the recurrence of preterm birth.
Collapse
Affiliation(s)
- V E Whiteman
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, College of Medicine
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Iglesias-Platas I, Martin-Trujillo A, Petazzi P, Guillaumet-Adkins A, Esteller M, Monk D. Altered expression of the imprinted transcription factor PLAGL1 deregulates a network of genes in the human IUGR placenta. Hum Mol Genet 2014; 23:6275-85. [PMID: 24993786 DOI: 10.1093/hmg/ddu347] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Genomic imprinting is the epigenetic process that results in monoallelic expression of genes depending on parental origin. These genes are known to be critical for placental development and fetal growth in mammals. Aberrant epigenetic profiles at imprinted loci, such as DNA methylation defects, are surprisingly rare in pregnancies with compromised fetal growth, while variations in transcriptional output from the expressed alleles of imprinted genes are more commonly reported in pregnancies complicated with intrauterine growth restriction (IUGR). To determine if PLAGL1 and HYMAI, two imprinted transcripts deregulated in Transient Neonatal Diabetes Mellitus, are involved in non-syndromic IUGR we compared the expression and DNA methylation levels in a large cohort of placental biopsies from IUGR and uneventful pregnancies. This revealed that despite appropriate maternal methylation at the shared PLAGL1/HYMAI promoter, there was a loss of correlation between PLAGL1 and HYMAI expression in IUGR. This incongruity was due to higher HYMAI expression in IUGR gestations, coupled with PLAGL1 down-regulation in placentas from IUGR girls, but not boys. The PLAGL1 protein is a zinc-finger transcription factor that has been shown to be a master coordinator of a genetic growth network in mice. We observe PLAGL1 binding to the H19/IGF2 shared enhancers in placentae, with significant correlations between PLAGL1 levels with H19 and IGF2 expression levels. In addition, PLAGL1 binding and expression also correlate with expression levels of metabolic regulator genes SLC2A4, TCF4 and PPARγ1. Our results strongly suggest that fetal growth can be influenced by altered expression of the PLAGL1 gene network in human placenta.
Collapse
Affiliation(s)
- Isabel Iglesias-Platas
- Servicio de Neonatología, Hospital Sant Joan de Déu, Fundació Sant Joan de Déu, Barcelona 08950, Spain,
| | | | - Paolo Petazzi
- Cancer Epigenetics Group, Cancer Epigenetic and Biology Program, Institut D'Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona 08907, Spain
| | - Amy Guillaumet-Adkins
- Servicio de Neonatología, Hospital Sant Joan de Déu, Fundació Sant Joan de Déu, Barcelona 08950, Spain, Imprinting and Cancer Group
| | - Manel Esteller
- Cancer Epigenetics Group, Cancer Epigenetic and Biology Program, Institut D'Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona 08907, Spain, Department of Physiological Sciences II, School of Medicine, University of Barcelona, Barcelona 08097, Spain and Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia 08010, Spain
| | | |
Collapse
|
33
|
Murphy R, Thompson JM, Tost J, Mitchell EA. No evidence for copy number and methylation variation in H19 and KCNQ10T1 imprinting control regions in children born small for gestational age. BMC MEDICAL GENETICS 2014; 15:67. [PMID: 24934635 PMCID: PMC4089969 DOI: 10.1186/1471-2350-15-67] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 06/11/2014] [Indexed: 11/24/2022]
Abstract
Background There is a substantial genetic component for birthweight variation, and although there are known associations between fetal genotype and birthweight, the role of common epigenetic variation in influencing the risk for small for gestational age (SGA) is unknown. The two imprinting control regions (ICRs) located on chromosome 11p15.5, involved in the overgrowth disorder Beckwith-Wiedemann syndrome (BWS) and the growth restriction disorder Silver-Russell syndrome (SRS), are prime epigenetic candidates for regulating fetal growth. We investigated whether common variation in copy number in the BWS/SRS 11p15 region or altered methylation levels at IGF2/H19 ICR or KCNQ10T1 ICR was associated with SGA. Methods We used a methylation-specific multiplex-ligation-dependent probe amplification assay to analyse copy number variation in the 11p15 region and methylation of IGF2/H19 and KCNQ10T1 ICRs in blood samples from 153 children (including 80 SGA), as well as bisulfite pyrosequencing to measure methylation at IGF2 differentially methylated region (DMR)0 and H19 DMR. Results No copy number variants were detected in the analyzed cohort. Children born SGA had 2.7% lower methylation at the IGF2 DMR0. No methylation differences were detected at the H19 or KCNQ10T1 DMRs. Conclusions We confirm that a small hypomethylation of the IGF2 DMR0 is detected in peripheral blood leucocytes of children born SGA at term. Copy number variation within the 11p15 BWS/SRS region is not an important cause of non-syndromic SGA at term.
Collapse
Affiliation(s)
- Rinki Murphy
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | | | | | | | | |
Collapse
|
34
|
Chakraborty A, Rakesh PS, Kumaran V, Prasad J, Alexander AM, George K. Risk of developing adulthood obesity among females born with low birth weight: Results from a non-concurrent study from rural Southern India. Indian J Endocrinol Metab 2014; 18:414-8. [PMID: 24944941 PMCID: PMC4056145 DOI: 10.4103/2230-8210.131214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE To determine the relationship between birth weight and the evolution of obesity in adult life in women from a rural developmental block in southern India. DESIGN Non-concurrent cohort. SETTING General community- a rural developmental block in southern India. PARTICIPANTS Two hundred and seventy one young healthy females were recruited from a birth cohort. The study subjects were 98 women in the age group of 19-23 years who had been born with low birth weight (LBW) and 173 women in the same age group who had been born with normal birth weight (NBW). MATERIALS AND METHODS Data collection involved interview using a structured questionnaire and anthropometric measurements. ANALYSIS Chi-square test to assess significance of association, independent sample t test to assess the difference between means, odds ratios for measuring magnitude of association, stratified analysis to identify various interactions and confounders, and multiple logistic regression models to identify the relationship between birth weight and young adult obesity (BMI > 25). RESULTS A crude odds ratio of 0.564 (95% CI 0.262 - 1.214) was obtained for the association between LBW and development of obesity later in life. In the final logistic regression model, it was found that a young adult female with low birth weight who belonged to a higher socio-economic group had a higher risk of developing obesity (Adjusted odds for the interaction term between LBW and high SES 6.251; 95% CI 1.236 - 31.611). CONCLUSION The study could not find any significant association between LBW and development of obesity later in life, but it found a higher probability of developing obesity later in life among low birth weight female children born in high socio-economic status families.
Collapse
Affiliation(s)
- Arup Chakraborty
- Department of Community Health, Christian Medical College, Vellore, Tamil Nadu, India
| | - P. S. Rakesh
- Department of Community Health, Christian Medical College, Vellore, Tamil Nadu, India
| | - V. Kumaran
- Department of Community Health, Christian Medical College, Vellore, Tamil Nadu, India
| | - Jasmin Prasad
- Department of Community Health, Christian Medical College, Vellore, Tamil Nadu, India
| | - Anu M. Alexander
- Department of Community Health, Christian Medical College, Vellore, Tamil Nadu, India
| | - Kuryan George
- Department of Community Health, Christian Medical College, Vellore, Tamil Nadu, India
| |
Collapse
|
35
|
Poppitt SD, Silvestre MP, Liu A. Etiology of Obesity Over the Life Span: Ecologic and Genetic Highlights from New Zealand Cohorts. Curr Obes Rep 2014; 3:38-45. [PMID: 26626466 DOI: 10.1007/s13679-013-0079-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The origins of the New Zealand population are highly diverse. New Zealand Māori are the indigenous peoples with a population of approximately half a million (~12 %), with the remainder comprising predominantly European/Caucasian (~50 %), Pacific Island Polynesian (~28 %) and Asian (~10 %) peoples. With a prevalence of overweight and obesity of 65 % for adults >15 years of age, of which 28 % have a BMI > 30 kg/m(2), New Zealand has been ranked third highest in a global OECD obesity review, behind only the US and Mexico. Levels of childhood obesity are also significant, with 31 % of New Zealand's children either overweight or obese. Few gender differences exist, but there are significant differences between ethnicities (Asian > European Caucasian > Māori > Pacific) with disproportionate representation by those poorer and with less formal education. A high 62 % of Pacifika are obese and virtually the entire adult population has a BMI >25 kg/m(2). Public health measures to limit progressive increases in weight are unsuccessful, and clearly should be priority for government focused on disease prevention.
Collapse
Affiliation(s)
- S D Poppitt
- Human Nutrition Unit, University of Auckland, 18 Carrick Place, Mt Eden, Auckland, 1024, New Zealand.
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.
- Department of Medicine, University of Auckland, Auckland, New Zealand.
- Riddet Institute, Palmerston North, New Zealand.
| | - M P Silvestre
- Human Nutrition Unit, University of Auckland, 18 Carrick Place, Mt Eden, Auckland, 1024, New Zealand
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - A Liu
- Human Nutrition Unit, University of Auckland, 18 Carrick Place, Mt Eden, Auckland, 1024, New Zealand
- Department of Medicine, University of Auckland, Auckland, New Zealand
| |
Collapse
|
36
|
Passarella G, Trifirò G, Gasparetto M, Moreolo GS, Milanesi O. Disorders in glucidic metabolism and congenital heart diseases: detection and prevention. Pediatr Cardiol 2013; 34:931-7. [PMID: 23229289 DOI: 10.1007/s00246-012-0577-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 10/12/2012] [Indexed: 11/24/2022]
Abstract
The identification of gestational diabetes (GDM) through appropriate screening and its subsequent treatment have not been demonstrated to limit neonatal malformations to date. This study aimed to detect congenital heart diseases in newborns of mothers with GDM by evaluating the existence of a correlation with maternal glycemic control. This observational prospective study investigated newborns of mothers with GDM enrolled during a period of 9 months. Four subgroups were considered according to the type of maternal glucidic alteration during pregnancy and the home treatment: impaired glucose tolerance, insulin-dependent gestational diabetes mellitus (IDDM), non-insulin-dependent gestational diabetes mellitus (NIDDM), and gestational diabetes not controlled (NC: untreated diabetes). Student's t test was used to compare the subgroups. The study enrolled 65 newborns (30 boys) born to 82 of mothers with impaired glucidic metabolism. Patent ductus arteriosus was observed in 11 patients (16.9 %), pulmonary stenosis of mild grade in 4 patients ( 6.2 %), and hypertrophy of the ventricular septum in 22 patients (33.8 %). A total of 14 patients had increased thickness in the left ventricle posterior wall, and 17 patients had an abnormal electrocardiogram. Hyperglycemia can influence the development of the fetal heart, affecting both its structure and its function. A treatment with insulin for women with GDM is supported by the study data.
Collapse
Affiliation(s)
- G Passarella
- Struttura Operativa Complessa di Pediatria, Ospedale Santa Maria della Misericordia, Viale Tre Martiri n° 140, CAP 45100, Rovigo, Italy.
| | | | | | | | | |
Collapse
|
37
|
Mumm H, Kamper-Jørgensen M, Nybo Andersen AM, Glintborg D, Andersen M. Birth weight and polycystic ovary syndrome in adult life: a register-based study on 523,757 Danish women born 1973–1991. Fertil Steril 2013. [DOI: 10.1016/j.fertnstert.2012.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
38
|
Maternal, perinatal, and postneonatal outcomes in women with chronic heart disease in Washington State. Obstet Gynecol 2013; 120:1283-90. [PMID: 23168751 DOI: 10.1097/aog.0b013e3182733d56] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To explore the association between the presence of maternal heart disease and maternal, perinatal, and infant outcomes. METHODS We conducted a population-based retrospective cohort study using Washington State birth certificates linked with hospital discharge records of mothers noted to have maternal congenital heart disease, ischemic heart disease, heart failure, or pulmonary hypertension. Women who gave birth between 1987 and 2009 (n=2,171) were compared with a sample of mothers without these conditions (n=21,710). We described characteristics of pregnant women with heart disease over time. Logistic regression estimated the association between chronic maternal heart disease and small-for-gestational-age (SGA) neonates as well as perinatal, postneonatal, and maternal death. RESULTS The proportion of births to women with reported heart disease increased 224% between the 1987 and 1994 and 2002 and 2009 calendar periods. Chronic maternal heart disease was associated with increased risk of SGA (62 additional SGA newborns per 1,000 births, 95% confidence interval [CI] 46-78; P<.001), perinatal death (14 additional deaths per 1,000 births, 95% CI 8-20; P<.001), postneonatal death (5 additional deaths per 1,000 births, 95% CI 2-9; P<.001), and maternal death (5 additional deaths per 1,000 births, 95% CI 2-9; P<.001). CONCLUSION The presence of chronic maternal heart disease is associated with elevated risk for poor maternal, perinatal, and postneonatal outcomes.
Collapse
|
39
|
Han DY, Murphy R, Morgan AR, Lam WJ, Thompson JMD, Wall CR, Waldie KE, Mitchell EA, Ferguson LR. Reduced genetic influence on childhood obesity in small for gestational age children. BMC MEDICAL GENETICS 2013; 14:10. [PMID: 23339409 PMCID: PMC3556300 DOI: 10.1186/1471-2350-14-10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 01/18/2013] [Indexed: 11/15/2022]
Abstract
Background Children born small-for-gestational-age (SGA) are at increased risk of developing obesity and metabolic diseases later in life, a risk which is magnified if followed by accelerated postnatal growth. We investigated whether common gene variants associated with adult obesity were associated with increased postnatal growth, as measured by BMI z-score, in children born SGA and appropriate for gestational age (AGA) in the Auckland Birthweight Collaborative. Methods A total of 37 candidate SNPs were genotyped on 547 European children (228 SGA and 319 AGA). Repeated measures of BMI (z-score) were used for assessing obesity status, and results were corrected for multiple testing using the false discovery rate. Results SGA children had a lower BMI z-score than non-SGA children at assessment age 3.5, 7 and 11 years. We confirmed 27 variants within 14 obesity risk genes to be individually associated with increasing early childhood BMI, predominantly in those born AGA. Conclusions Genetic risk variants are less important in influencing early childhood BMI in those born SGA than in those born AGA, suggesting that non-genetic or environmental factors may be more important in influencing childhood BMI in those born SGA.
Collapse
Affiliation(s)
- Dug Yeo Han
- Discipline of Nutrition, FM&HS, The University of Auckland, Auckland, New Zealand.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Stawerska R, Szałapska M, Borowiec M, Młynarski W, Antosik K, Hilczer M, Lewiński A. Frequency of the E23K polymorphism of the KCNJ11 gene in children born small for gestational age and its influence on auxological and metabolic parameters in the prepubertal period. J Pediatr Endocrinol Metab 2013; 26:457-62. [PMID: 23412854 DOI: 10.1515/jpem-2012-0196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 12/22/2012] [Indexed: 11/15/2022]
Abstract
BACKGROUND The E23K variant of the KCNJ11 gene is possibly responsible for changes in insulin secretion during the fetal life. We tried to assess the influence of the E23K variant on birth weight and metabolic profile in prepubertal children born small for gestational age (SGA). SUBJECTS One hundred and twenty-three SGA and 132 born appropriate for gestational age (AGA) children were genotyped for the E23K variant. Lipids, glucose, and insulin concentrations during oral glucose tolerance test were assessed in 112 SGA prepubertal children. RESULTS There were no significant differences between the frequency of the E23K variant in SGA and AGA children. In SGA children with E23K, the mean birth weight was significantly higher than in the E23E group. Body mass index, glucose, insulin, and lipids were not different between the E23K, E23E, and K23K groups. CONCLUSIONS The higher birth weight in SGA children with the E23K variant may be related to higher insulin concentrations in the fetal period. The E23K variant did not affect metabolic disorders in prepubertal SGA children.
Collapse
Affiliation(s)
- Renata Stawerska
- Department of Endocrinology and Metabolic Diseases, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | | | | | | | | | | | | |
Collapse
|
41
|
Veena SR, Krishnaveni GV, Fall CH. Newborn size and body composition as predictors of insulin resistance and diabetes in the parents: Parthenon Birth Cohort Study, Mysore, India. Diabetes Care 2012; 35:1884-90. [PMID: 22751963 PMCID: PMC3425002 DOI: 10.2337/dc12-0177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE We aimed to examine detailed neonatal measurements as predictors of later diabetes in both parents. RESEARCH DESIGN AND METHODS Babies (n = 617) born to nondiabetic parents in Holdsworth Memorial Hospital, Mysore, India, were measured at birth for weight; crown-to-heel length (CHL), crown-to-rump length (CRL), and leg length; skinfolds (triceps and subscapular); and circumferences (head, abdomen, and mid-upper-arm circumference [MUAC]). Nine and a half years later, glucose tolerance and fasting insulin were measured in their parents (469 mothers and 398 fathers). RESULTS Sixty-two (15.6%) fathers and 22 (4.7%) mothers had developed diabetes. There were linear inverse associations of the children's birth weight, CHL, CRL, MUAC, and skinfolds with paternal diabetes and insulin resistance (P < 0.05 for all). Offspring birth weight and adiposity (MUAC, abdominal circumference, and skinfolds) showed U-shaped associations with maternal diabetes (P for quadratic association <0.05 for all). These associations persisted after adjusting for the parents' current adiposity and maternal glucose concentrations and adiposity during pregnancy. Newborn adiposity was positively related to maternal insulin resistance; this association was nonsignificant after adjusting for maternal current adiposity. CONCLUSIONS Newborn size is a window into the future health of the parents. Small newborn size (especially soft-tissue body components) predicts an increased risk of later diabetes in both parents, suggesting a genetic or epigenetic link between parents' diabetes risk and reduced fetal growth in their children. The association of higher birth weight and newborn adiposity with later maternal diabetes suggests effects on fetal adiposity of the intrauterine environment in prediabetic mothers.
Collapse
|
42
|
Small size for gestational age and the risk for infant mortality in the subsequent pregnancy. Ann Epidemiol 2012; 22:764-71. [PMID: 22858049 DOI: 10.1016/j.annepidem.2012.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 07/05/2012] [Accepted: 07/06/2012] [Indexed: 11/23/2022]
Abstract
PURPOSE To examine the association between small for gestational age (SGA) in the first pregnancy and risk for infant mortality in the second pregnancy. METHODS This is a population-based, retrospective cohort study in which we used the Missouri maternally linked cohort dataset for 1978-2005. Analyses were restricted to women who had two singleton pregnancies during the study period. The exposure was SGA in the first pregnancy, whereas the primary outcome was infant mortality in the second pregnancy. Kaplan-Meier Estimate and Cox proportional hazard regression were conducted. RESULTS Infant mortality was significantly greater among mothers with previous SGA (P < .01). A persistent association of previous SGA with subsequent infant mortality was observed (adjusted hazard ratio [AHR] 1.35, 95% confidence interval [95% CI] 1.24-1.48). Race-specific data illustrated that black women with a previous SGA birth were 40% more likely to experience infant mortality (AHR 1.40, 95% CI 1.21-1.63) than their counterparts without a history of SGA, but white women with a previous SGA had an increased risk of 31% (AHR 1.31, 95% CI 1.17-1.46). CONCLUSIONS Women with previous SGA bear increased risks for subsequent infant mortality, which was greater among black mothers. Hence, SGA plays an important role in the black-white disparity in infant mortality. Women's previous childbearing experiences could serve as important criterion in determining appropriate interconception strategies to improve infant health and survival.
Collapse
|
43
|
Abstract
Onset of obesity has been anticipated at earlier ages, and prevalence has dramatically increased worldwide over the past decades. Epidemic obesity is mainly attributable to modern lifestyle, but family studies prove the significant role of genes in the individual's predisposition to obesity. Advances in genotyping technologies have raised great hope and expectations that genetic testing will pave the way to personalized medicine and that complex traits such as obesity will be prevented even before birth. In the presence of the pressing offer of direct-to-consumer genetic testing services from private companies to estimate the individual's risk for complex phenotypes including obesity, the present review offers pediatricians an update of the state of the art on genomics obesity in childhood. Discrepancies with respect to genomics of adult obesity are discussed. After an appraisal of findings from genome-wide association studies in pediatric populations, the rare variant-common disease hypothesis, the theoretical soil for next-generation sequencing techniques, is discussed as opposite to the common disease-common variant hypothesis. Next-generation sequencing techniques are expected to fill the gap of "missing heritability" of obesity, identifying rare variants associated with the trait and clarifying the role of epigenetics in its heritability. Pediatric obesity emerges as a complex phenotype, modulated by unique gene-environment interactions that occur in periods of life and are "permissive" for the programming of adult obesity. With the advent of next-generation sequencing techniques and advances in the field of exposomics, sensitive and specific tools to predict the obesity risk as early as possible are the challenge for the next decade.
Collapse
Affiliation(s)
- Melania Manco
- FACN, Scientific Directorate, Bambino Gesù Pediatric Hospital, Istituto Di Ricovero e Cura a Carattere Scientifico, Rome, Italy.
| | | |
Collapse
|
44
|
Saenger P, Reiter E. Genetic factors associated with small for gestational age birth and the use of human growth hormone in treating the disorder. INTERNATIONAL JOURNAL OF PEDIATRIC ENDOCRINOLOGY 2012; 2012:12. [PMID: 22587301 PMCID: PMC3511163 DOI: 10.1186/1687-9856-2012-12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 03/19/2012] [Indexed: 12/31/2022]
Abstract
The term small for gestational age (SGA) refers to infants whose birth weights and/or lengths are at least two standard deviation (SD) units less than the mean for gestational age. This condition affects approximately 3%–10% of newborns. Causes for SGA birth include environmental factors, placental factors such as abnormal uteroplacental blood flow, and inherited genetic mutations. In the past two decades, an enhanced understanding of genetics has identified several potential causes for SGA. These include mutations that affect the growth hormone (GH)/insulin-like growth factor (IGF)-1 axis, including mutations in the IGF-1 gene and acid-labile subunit (ALS) deficiency. In addition, select polymorphisms observed in patients with SGA include those involved in genes associated with obesity, type 2 diabetes, hypertension, ischemic heart disease and deletion of exon 3 growth hormone receptor (d3-GHR) polymorphism. Uniparental disomy (UPD) and imprinting effects may also underlie some of the phenotypes observed in SGA individuals. The variety of genetic mutations associated with SGA births helps explain the diversity of phenotype characteristics, such as impaired motor or mental development, present in individuals with this disorder. Predicting the effectiveness of recombinant human GH (hGH) therapy for each type of mutation remains challenging. Factors affecting response to hGH therapy include the dose and method of hGH administration as well as the age of initiation of hGH therapy. This article reviews the results of these studies and summarizes the success of hGH therapy in treating this difficult and genetically heterogenous disorder.
Collapse
Affiliation(s)
- Paul Saenger
- Albert Einstein College of Medicine, Winthrop University Hospital, 120 Mineola Boulevard, Mineola, NY, 13501, USA.
| | | |
Collapse
|
45
|
Abstract
Intrauterine growth restriction (IUGR) is prevalent worldwide and affects children and adults in multiple ways. These include predisposition to type 2 diabetes mellitus, the metabolic syndrome, cardiovascular disease, persistent reduction in stature, and possibly changes in the pattern of puberty. A review of recent literature confirms that the metabolic effects of being born small for gestational age are evident in the very young, persist with age, and are amplified by adiposity. Furthermore, the pattern of growth in the first few years of life has a significant bearing on a person's later health, with those that show increasing weight gain being at the greatest risk for future metabolic dysfunction. Treatment with exogenous human GH is used to improve height in children who remain short after being small for gestational age at birth, but the response of individuals remains variable and difficult to predict. The mechanisms involved in the metabolic programming of IUGR children are just beginning to be explored. It appears that IUGR leads to widespread changes in DNA methylation and that specific "epigenetic signatures" for IUGR are likely to be found in various fetal tissues. The challenge is to link such alterations with modifications in gene expression and ultimately the metabolic abnormalities of adulthood, and it represents one of the frontiers for research in the field.
Collapse
Affiliation(s)
- Steven D Chernausek
- Department of Pediatrics, University of Oklahoma Health Sciences Center, 1200 North Phillips Avenue, Suite 4500, Oklahoma City, Oklahoma 73104-4600, USA.
| |
Collapse
|
46
|
Mercuro G, Bassareo PP, Flore G, Fanos V, Dentamaro I, Scicchitano P, Laforgia N, Ciccone MM. Prematurity and low weight at birth as new conditions predisposing to an increased cardiovascular risk. Eur J Prev Cardiol 2012; 20:357-67. [PMID: 22345683 DOI: 10.1177/2047487312437058] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Although the survival rate for preterm subjects has improved considerably, due to the progress in the field of perinatal medicine, preterm birth is frequently the cause underlying a series of notorious complications: morphological, neurological, ophthalmological, and renal alterations. In addition, it has recently been demonstrated how low gestational age and reduced foetal growth contribute towards an increased cardiovascular risk in preterm neonates. In fact, cardiovascular mortality is higher among former preterm adults than those born at term. This condition is referred to as cardiovascular perinatal programming. In the light of the above, an early, constant, and prolonged cardiological followup programme should be implemented in former preterm individuals. The aim of this paper was to perform a comprehensive literature review about two new emerging conditions predisposing to an increased cardiovascular risk: prematurity and low weight at birth.
Collapse
|
47
|
|
48
|
Andraweera PH, Dekker GA, Thompson SD, North RA, McCowan LME, Roberts CT. A functional variant in the thrombospondin-1 gene and the risk of small for gestational age infants. J Thromb Haemost 2011; 9:2221-8. [PMID: 21883885 DOI: 10.1111/j.1538-7836.2011.04494.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Thrombospondin-1 (TSP-1) is a prothrombotic and anti-angiogenic glycoprotein expressed in the placenta. A functional single nucleotide polymorphism in the TSP-1 gene (TSP-1 A2210G) is a risk factor for familial premature myocardial infarction. Small for gestational age (SGA) infants are at increased risk of coronary artery disease in adult life and common genetic factors may underlie both conditions. We investigated the association of TSP-1 A2210G in SGA infants and their parents. METHOD The 3234 nulliparous pregnant women, their partners and babies were recruited in Adelaide and Auckland to a prospective multicenter cohort study. Amongst 2123 Caucasian women, 216 (10.2%) delivered an SGA infant, defined as birth weight < 10th customized centile adjusted for maternal height, weight, parity and ethnicity, as well as gestational age at delivery and infant sex. Uncomplicated pregnancies served as controls (n = 1185). DNA extracted from peripheral/cord blood or buccal swabs was genotyped using Sequenom MassARRAY. Multivariable logistic regression was used to compare the odds of SGA between the genotype groups adjusting for potential confounders. RESULTS Paternal (adjOR, 1.4; 95% CI 1.0-2.0) and neonatal (adjOR, 1.8; 95% CI, 1.1-2.7) TSP-1 A2210G associates with SGA. The maternal polymorphism approaches significance for an association with SGA (adjOR, 1.3; 95% CI, 0.9-1.9). Maternal TSP-1 A2210G associates with a reduced maternal birth weight adjusted for gestational age at delivery (P = 0.03). CONCLUSION The TSP-1 A2210G polymorphism, which is a risk factor for myocardial infarction, is associated with SGA pregnancies, suggesting that this polymorphism may associate with the risk of vascular disorders across the life course.
Collapse
Affiliation(s)
- P H Andraweera
- Discipline of Obstetrics and Gynaecology, Research Centre for Reproductive Health, University of Adelaide, Adelaide, Australia
| | | | | | | | | | | |
Collapse
|