1
|
Costantine MM, Tita ATN, Mele L, Casey BM, Peaceman AM, Varner MW, Reddy UM, Wapner RJ, Thorp JM, Saade GR, Rouse DJ, Sibai B, Mercer BM, Caritis SN. The Association between Infant Birth Weight, Head Circumference, and Neurodevelopmental Outcomes. Am J Perinatol 2024; 41:e1313-e1323. [PMID: 36791785 PMCID: PMC10425571 DOI: 10.1055/s-0043-1761920] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
OBJECTIVE The aim of this study was to evaluate whether being small for gestational age (SGA) or large for gestational age (LGA) or having a small or large head circumference (HC) at birth is associated with adverse neurodevelopmental outcomes. STUDY DESIGN This is a secondary analysis of a multicenter negative randomized trial of thyroxine therapy for subclinical hypothyroid disorders in pregnancy. The primary outcome was child intelligence quotient (IQ) at 5 years of age. Secondary outcomes included several neurodevelopmental measures. Associations between the outcomes in children with SGA (<10th percentile) or LGA (>90th percentile) birth weights, using ethnicity- and sex-specific population nomogram as well as nomograms from the National Fetal Growth (NFG) study, were compared with the referent of those with appropriate for gestational age (AGA) birth weight. Similar analyses were performed for HC. RESULTS Using the population nomogram, 90 (8.2%) were SGA and 112 (10.2%) were LGA. SGA neonates were more likely to be born preterm to mothers who were younger, smoked, and were less likely to have less than a high school education, whereas LGA neonates were more likely to be born to mothers who were older and have higher body mass index, compared with AGA neonates. SGA at birth was associated with a decrease in the child IQ at 5 years of age by 3.34 (95% confidence interval [CI], 0.54-6.14) points, and an increase in odds of child with an IQ < 85 (adjusted odds ratio [aOR], 1.9; 95% CI, 1.1-3.2). There was no association between SGA and other secondary outcomes, or between LGA and the primary or secondary outcomes. Using the NFG standards, SGA at birth remained associated with a decrease in the child IQ at 5 years of age by 3.14 (95% CI, 0.22-6.05) points and higher odds of an IQ < 85 (aOR, 2.3; 95% CI, 1.3-4.1), but none of the other secondary outcomes. HC was not associated with the primary outcome, and there were no consistent associations of these standards with the secondary outcomes. CONCLUSION In this cohort of pregnant individuals with hypothyroid disorders, SGA birth weight was associated with a decrease in child IQ and greater odds of child IQ < 85 at 5 years of age. Using a fetal growth standard did not appear to improve the detection of newborns at risk of adverse neurodevelopment.
Collapse
Affiliation(s)
- Maged M Costantine
- Department of Obstetrics and Gynecology, The Ohio State University, Columbus, Ohio
| | - Alan T N Tita
- University of Alabama at Birmingham, Birmingham, Alabama
| | - Lisa Mele
- George Washington University Biostatistics Center, Washington, District of Columbia
| | | | | | | | - Uma M Reddy
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland
| | | | - John M Thorp
- University of North Carolina, Chapel Hill, North Carolina
| | | | | | - Baha Sibai
- University of Texas - Houston, Houston, Texas
| | | | | |
Collapse
|
2
|
Shi J, Guo Q, Fang H, Cheng X, Ju L, Wei X, Zhao L, Cao Q, Yuan X, He L. The Relationship between Birth Weight and the Risk of Overweight and Obesity among Chinese Children and Adolescents Aged 7-17 Years. Nutrients 2024; 16:715. [PMID: 38474841 DOI: 10.3390/nu16050715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/25/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Obesity is a major public health issue in children and adolescents. Our study aimed to examine the impacts of birth weight on overweight and obesity among Chinese children and adolescents. Using data from the China National Nutrition and Health Surveillance of Children and Lactating Mothers in 2016-2017, we included 10,041 participants aged 7-17 years. According to birth weight, participants were categorized into six groups, and the birth weight category of 3000 to 3499 g was chosen as the reference group, containing the largest number of children. Logistic regression analyses were used to investigate the association of birth weight with the risk of being obese at 7 to 17 years of age in multivariable-adjusted models. A restricted cubic spline was utilized to show the odds ratios (ORs) of obesity at different birth weight levels. The adjusted ORs for overweight were 0.98 (95%CI 0.63, 1.53), 1.02 (95%CI 0.84, 1.25), 1.34 (95%CI 1.16, 1.55), 1.72 (95%CI 1.35, 2.18), and 1.17 (95%CI 0.71, 1.96) in several birth weight groups, compared with group C (3000-3499 g). The adjusted ORs for obesity were 0.82 (95%CI 0.48, 1.40), 0.77 (95%CI 0.60, 0.98), 1.33 (95%CI 1.13, 1.57), 1.97 (95%CI 1.53, 2.53), and 2.01 (95%CI 1.27, 3.19). Furthermore, children in the post-pubertal stage had a slightly higher risk of overweight and obesity than those in the pre-pubertal and pubertal stage. Moreover, these associations were stronger among boys. The lower part of normal birth weight range is associated with a lower risk of overweight and obesity in children and adolescents. However, higher levels of birth weight increase risk.
Collapse
Affiliation(s)
- Jiaqi Shi
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Qiya Guo
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Hongyun Fang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Xue Cheng
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Lahong Ju
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Xiaoqi Wei
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Liyun Zhao
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Qiuye Cao
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Xiaolin Yuan
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Li He
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| |
Collapse
|
3
|
Blomberg J, Luna Ramirez RI, Goyal D, Limesand SW, Goyal R. Sexual dimorphic gene expression profile of perirenal adipose tissue in ovine fetuses with growth restriction. Front Physiol 2023; 14:1179288. [PMID: 37601643 PMCID: PMC10437077 DOI: 10.3389/fphys.2023.1179288] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/06/2023] [Indexed: 08/22/2023] Open
Abstract
Worldwide, fetal growth restriction (FGR) affects 7%-10% of pregnancies, or roughly 20.5 million infants, each year. FGR increases not only neonatal mortality and morbidity but also the risk of obesity in later life. Currently, the molecular mechanisms by which FGR "programs" an obese phenotype are not well understood. Studies demonstrate that FGR females are more prone to obesity compared to males; however, the molecular mechanisms that lead to the sexually dimorphic programming of FGR are not known. Thus, we hypothesized that FGR leads to the sexually dimorphic programming of preadipocytes and reduces their ability to differentiate into mature adipocytes. To test the hypothesis, we utilized a maternal hyperthermia-induced placental insufficiency to restrict fetal growth in sheep. We collected perirenal adipose tissue from near-term (∼140 days gestation) male and female FGR and normal-weight fetal lambs (N = 4 to 5 in each group), examined the preadipocytes' differentiation potential, and identified differential mRNA transcript expression in perirenal adipose tissue. Male FGR fetuses have a lower cellular density (nuclei number/unit area) compared to control male fetuses. However, no difference was observed in female FGR fetuses compared to control female fetuses. In addition, the ability of preadipocytes to differentiate into mature adipocytes with fat accumulation was impaired in male FGR fetuses, but this was not observed in female FGR fetuses. Finally, we examined the genes and pathways involved in the sexually dimorphic programming of obesity by FGR. On enrichment of differentially expressed genes in males compared to females, the Thermogenesis KEGG Pathway was downregulated, and the Metabolic and Steroid Biosynthesis KEGG pathways were upregulated. On enrichment of differentially expressed genes in male FGR compared to male control, the Steroid Biosynthesis KEGG Pathway was downregulated, and the PPAR Signaling KEGG pathway was upregulated. No pathways were altered in females in response to growth restriction in perirenal adipose tissue. Thus, the present study demonstrates a sexually dimorphic program in response to growth restriction in sheep fetal perirenal adipose tissue.
Collapse
Affiliation(s)
| | | | | | | | - Ravi Goyal
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
4
|
Oliveira WR, Rigo CP, Ferreira ARO, Ribeiro MVG, Perres MNC, Palma-Rigo K. Precocious evaluation of cardiovascular risk and its correlation with perinatal condition. AN ACAD BRAS CIENC 2023; 95:e20201702. [PMID: 37377255 DOI: 10.1590/0001-3765202320201702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/28/2021] [Indexed: 06/29/2023] Open
Abstract
The cardiovascular disease is the main cause of worldwide death. This profile is potentialized by the increased severity of infections in people with obesity, type 2 diabetes and hypertension. Children and adolescents are target groups for the prevention of non-communicable diseases. The Developmental Origins of Health and Disease concept points that perinatal conditions are an important risk factor to development of non-communicable disease in adulthood. In this context, the present review identifies perinatal factor that induces precocious cardiovascular risk factors, related with cardiometabolic syndrome. The low or high birth weight and caesarean delivery are risk factors that induce increased occurrence of cardiovascular risk biomarkers in children and adolescents, while the breast feeding or feeding with breast milk from the birth until two years-old is a protector strategy. Evaluation of perinatal conditions associated with precocious identification of cardiovascular risk factors in children and adolescents is an efficient strategy to prevent and control cardiovascular mortality; through interventions, as lifestyle changes during vulnerable windows of development, able to set up the risk to cardiometabolic disease.
Collapse
Affiliation(s)
- Wanderson R Oliveira
- Faculdade Adventista Paranaense, PR-317, Km 119, Gleba, Rua Paiçandu, Lote 80, Zona Rural, 87130-000 Ivatuba, PR, Brazil
| | - Cleusa P Rigo
- Centro Universitário Filadélfia, Rua Alagoas, 2050, Centro, 86010-520 Londrina, PR, Brazil
| | - Anna R O Ferreira
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Av. Colombo, 5790, Zona 7, 87020-900 Maringá, PR, Brazil
| | - Maiara V G Ribeiro
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Av. Colombo, 5790, Zona 7, 87020-900 Maringá, PR, Brazil
| | - Maria N C Perres
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Av. Colombo, 5790, Zona 7, 87020-900 Maringá, PR, Brazil
| | - Kesia Palma-Rigo
- Universidade Estadual de Maringá, Departamento de Análises Clínicas e Biomedicina, Av. Colombo, 5790, Zona 7, 87020-900 Maringá, PR, Brazil
- Faculdade Adventista Paranaense, PR-317, Km 119, Gleba, Rua Paiçandu, Lote 80, Zona Rural, 87130-000 Ivatuba, PR, Brazil
| |
Collapse
|
5
|
Wathes DC. Developmental Programming of Fertility in Cattle-Is It a Cause for Concern? Animals (Basel) 2022; 12:2654. [PMID: 36230395 PMCID: PMC9558991 DOI: 10.3390/ani12192654] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
Cattle fertility remains sub-optimal despite recent improvements in genetic selection. The extent to which an individual heifer fulfils her genetic potential can be influenced by fetal programming during pregnancy. This paper reviews the evidence that a dam's age, milk yield, health, nutrition and environment during pregnancy may programme permanent structural and physiological modifications in the fetus. These can alter the morphology and body composition of the calf, postnatal growth rates, organ structure, metabolic function, endocrine function and immunity. Potentially important organs which can be affected include the ovaries, liver, pancreas, lungs, spleen and thymus. Insulin/glucose homeostasis, the somatotropic axis and the hypothalamo-pituitary-adrenal axis can all be permanently reprogrammed by the pre-natal environment. These changes may act directly at the level of the ovary to influence fertility, but most actions are indirect. For example, calf health, the timing of puberty, the age and body structure at first calving, and the ability to balance milk production with metabolic health and fertility after calving can all have an impact on reproductive potential. Definitive experiments to quantify the extent to which any of these effects do alter fertility are particularly challenging in cattle, as individual animals and their management are both very variable and lifetime fertility takes many years to assess. Nevertheless, the evidence is compelling that the fertility of some animals is compromised by events happening before they are born. Calf phenotype at birth and their conception data as a nulliparous heifer should therefore both be assessed to avoid such animals being used as herd replacements.
Collapse
Affiliation(s)
- D Claire Wathes
- Department for Pathobiology and Population Sciences, Royal Veterinary College, Hatfield AL9 7TA, UK
| |
Collapse
|
6
|
Jeje SO, Adenawoola M, Abosede C. Gestational Nutrition as a Predisposing Factor to Obesity Onset in Offspring: Role for Involvement of Epigenetic Mechanism. Niger J Physiol Sci 2022; 37:1-7. [PMID: 35947841 DOI: 10.54548/njps.v37i1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 04/12/2022] [Indexed: 06/15/2023]
Abstract
Maternal lifestyle has been implicated as a predisposing factor in the development of metabolic disorders in adulthood. This lifestyle includes the immediate environment, physical activity and nutrition. Maternal nutrition has direct influence on the developmental programming through biochemical alterations and can lead to modifications in the fetal genome through epigenetic mechanisms. Imbalance in basic micro or macro nutrients due to famine or food deficiency during delicate gestational periods can lead to onset of metabolic syndrome including obesity. A major example is the Dutch famine which led to a serious metabolic disorder in adulthood of affected infants. Notably due to gene variants, individualized responses to nutritional deficiencies are unconventional, therefore intensifying the need to study nutritional genomics during fetal programming. Epigenetic mechanisms can cause hereditary changes without changing the DNA sequence; the major mechanisms include small non-coding RNAs, histone modifications and most stable of all is DNA methylation. The significance association between obesity and DNA methylation is through regulation of genes implicated in lipid and glucose metabolism either directly or indirectly by hypomethylation or hypermethylation. Examples include CPT1A, APOA2, ADRB3 and POMC. Any maternal exposure to malnutrition or overnutrition that can affect genes regulating major metabolic pathways in the fetus, will eventually cause underlying changes that can predispose or cause the onset of metabolic disorder in adulthood. In this review, we examined the interaction between nutrition during gestation and epigenetic programming of metabolic syndrome.
Collapse
|
7
|
Zapaterini JR, Fonseca ARB, Bidinotto LT, Colombelli KT, Rossi ALD, Kass L, Justulin LA, Barbisan LF. Maternal Low-Protein Diet Deregulates DNA Repair and DNA Replication Pathways in Female Offspring Mammary Gland Leading to Increased Chemically Induced Rat Carcinogenesis in Adulthood. Front Cell Dev Biol 2022; 9:756616. [PMID: 35178394 PMCID: PMC8844450 DOI: 10.3389/fcell.2021.756616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Studies have shown that maternal malnutrition, especially a low-protein diet (LPD), plays a key role in the developmental mechanisms underlying mammary cancer programming in female offspring. However, the molecular pathways associated with this higher susceptibility are still poorly understood. Thus, this study investigated the adverse effects of gestational and lactational low protein intake on gene expression of key pathways involved in mammary tumor initiation after a single dose of N-methyl-N-nitrosourea (MNU) in female offspring rats. Pregnant Sprague-Dawley rats were fed a normal-protein diet (NPD) (17% protein) or LPD (6% protein) from gestational day 1 to postnatal day (PND) 21. After weaning (PND 21), female offspring (n = 5, each diet) were euthanized for histological analysis or received NPD (n = 56 each diet). At PND 28 or 35, female offspring received a single dose of MNU (25 mg/kg body weight) (n = 28 each diet/timepoint). After 24 h, some females (n = 10 each diet/timepoint) were euthanized for histological, immunohistochemical, and molecular analyses at PDN 29 or 36. The remaining animals (n = 18 each diet/timepoint) were euthanized when tumors reached ≥2 cm or at PND 250. Besides the mammary gland development delay observed in LPD 21 and 28 groups, the gene expression profile demonstrated that maternal LPD deregulated 21 genes related to DNA repair and DNA replication pathways in the mammary gland of LPD 35 group after MNU. We further confirmed an increased γ-H2AX (DNA damage biomarker) and in ER-α immunoreactivity in mammary epithelial cells in the LPD group at PND 36. Furthermore, these early postnatal events were followed by significantly higher mammary carcinogenesis susceptibility in offspring at adulthood. Thus, the results indicate that maternal LPD influenced the programming of chemically induced mammary carcinogenesis in female offspring through increase in DNA damage and deregulation of DNA repair and DNA replication pathways. Also, Cidea upregulation gene in the LPD 35 group may suggest that maternal LPD could deregulate genes possibly leading to increased risk of mammary cancer development and/or poor prognosis. These findings increase the body of evidence of early-transcriptional mammary gland changes influenced by maternal LPD, resulting in differential response to breast tumor initiation and susceptibility and may raise discussions about lifelong prevention of breast cancer risk.
Collapse
Affiliation(s)
- Joyce R Zapaterini
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | - Antonio R B Fonseca
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | - Lucas T Bidinotto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Botucatu, Brazil.,Barretos School of Health Sciences, Dr. Paulo Prata-FACISB, Barretos, Brazil
| | - Ketlin T Colombelli
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | | | - Laura Kass
- Instituto de Salud y Ambiente del Litoral (UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Luis A Justulin
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | - Luis F Barbisan
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| |
Collapse
|
8
|
Omere C, Goncharov DA, Pedroza C, Rosa MLA, Munn M, Chiossi G, Longo M, Saad AF. Randomized Trial Of Fundal Height Versus Point-Of-Care Ultrasound During Routine Antenatal Visits. Am J Obstet Gynecol MFM 2022; 4:100563. [PMID: 35032696 DOI: 10.1016/j.ajogmf.2022.100563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/28/2021] [Accepted: 01/06/2022] [Indexed: 10/19/2022]
Affiliation(s)
- Chasey Omere
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX USA.
| | - Daphne Arena Goncharov
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX USA
| | - Claudia Pedroza
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX USA
| | - Mauricio LA Rosa
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX USA
| | - Mary Munn
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX USA
| | - Giuseppe Chiossi
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX USA
| | - Monica Longo
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX USA
| | - Antonio F Saad
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX USA
| |
Collapse
|
9
|
Fanous M, Caputo MP, Lee YJ, Rund LA, Best-Popescu C, Kandel ME, Johnson RW, Das T, Kuchan MJ, Popescu G. Quantifying myelin content in brain tissue using color Spatial Light Interference Microscopy (cSLIM). PLoS One 2020; 15:e0241084. [PMID: 33211727 PMCID: PMC7676665 DOI: 10.1371/journal.pone.0241084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 10/08/2020] [Indexed: 12/18/2022] Open
Abstract
Deficient myelination of the brain is associated with neurodevelopmental delays, particularly in high-risk infants, such as those born small in relation to their gestational age (SGA). New methods are needed to further study this condition. Here, we employ Color Spatial Light Interference Microscopy (cSLIM), which uses a brightfield objective and RGB camera to generate pathlength-maps with nanoscale sensitivity in conjunction with a regular brightfield image. Using tissue sections stained with Luxol Fast Blue, the myelin structures were segmented from a brightfield image. Using a binary mask, those portions were quantitatively analyzed in the corresponding phase maps. We first used the CLARITY method to remove tissue lipids and validate the sensitivity of cSLIM to lipid content. We then applied cSLIM to brain histology slices. These specimens are from a previous MRI study, which demonstrated that appropriate for gestational age (AGA) piglets have increased internal capsule myelination (ICM) compared to small for gestational age (SGA) piglets and that a hydrolyzed fat diet improved ICM in both. The identity of samples was blinded until after statistical analyses.
Collapse
Affiliation(s)
- Michael Fanous
- Quantitative Light Imaging Laboratory, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Megan P. Caputo
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Young Jae Lee
- Quantitative Light Imaging Laboratory, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Laurie A. Rund
- Laboratory of Integrative Immunology & Behavior, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Catherine Best-Popescu
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Mikhail E. Kandel
- Quantitative Light Imaging Laboratory, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Rodney W. Johnson
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Laboratory of Integrative Immunology & Behavior, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Tapas Das
- Abbott Nutrition, Discovery Research, Columbus, OH, United States of America
| | - Matthew J. Kuchan
- Abbott Nutrition, Strategic Research, Columbus, OH, United States of America
| | - Gabriel Popescu
- Quantitative Light Imaging Laboratory, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
- * E-mail:
| |
Collapse
|
10
|
L. Aubuchon-Endsley N, E. Swann-Thomsen H, Douthit N. Maternal Prenatal Cortisol and Breastfeeding Predict Infant Growth. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17218233. [PMID: 33171766 PMCID: PMC7664389 DOI: 10.3390/ijerph17218233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/17/2020] [Accepted: 11/04/2020] [Indexed: 11/16/2022]
Abstract
Fetal/infant growth affects adult obesity and morbidities/mortality and has been associated with prenatal exposure to cortisol. Bidirectional relations between maternal stress and breastfeeding suggest that they interact to influence offspring growth. No models have tested this hypothesis, particularly regarding longer-term offspring outcomes. We used a subset of the IDAHO Mom Study (n = 19-95) to examine associations among maternal prenatal cortisol (cortisol awakening response (CAR) and area under the curve), and standardized weight-for-length (WLZ) and length-for-age (LAZ) z-scores from birth-18 months, and main and interactive effects of prenatal cortisol and breastfeeding on infant growth from birth-6 months. CAR was negatively associated with LAZ at birth (r = -0.247, p = 0.039) but positively associated at 13-14 months (r = 0.378, p = 0.033), suggesting infant catch-up growth with lower birth weights, likely related to elevated cortisol exposure, continues beyond early infancy. A negative correlation between breastfeeding and 10-month WLZ (r = -0.344, p = 0.037) and LAZ (r = -0.468, p = 0.005) suggests that breastfeeding assists in managing infant growth. WLZ and LAZ increased from birth to 6 months (ps < 0.01), though this was unrelated to interactions between prenatal cortisol and breastfeeding (i.e., no significant moderation), suggesting that other factors played a role, which should be further investigated. Findings add to our understanding of the predictors of infant growth.
Collapse
Affiliation(s)
- Nicki L. Aubuchon-Endsley
- Department of Psychology, Idaho State University, Pocatello, ID 83209, USA
- Correspondence: ; Tel.: +1-208-282-2574
| | | | | |
Collapse
|
11
|
Alejandro EU, Jo S, Akhaphong B, Llacer PR, Gianchandani M, Gregg B, Parlee SD, MacDougald OA, Bernal-Mizrachi E. Maternal low-protein diet on the last week of pregnancy contributes to insulin resistance and β-cell dysfunction in the mouse offspring. Am J Physiol Regul Integr Comp Physiol 2020; 319:R485-R496. [PMID: 32877242 PMCID: PMC7717124 DOI: 10.1152/ajpregu.00284.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022]
Abstract
Maternal low-protein diet (LP) throughout gestation affects pancreatic β-cell fraction of the offspring at birth, thus increasing their susceptibility to metabolic dysfunction and type 2 diabetes in adulthood. The present study sought to strictly examine the effects of LP during the last week of gestation (LP12.5) alone as a developmental window for β-cell programming and metabolic dysfunction in adulthood. Islet morphology analysis revealed normal β-cell fraction in LP12.5 newborns. Normal glucose tolerance was observed in 6- to 8-wk-old male and female LP12.5 offspring. However, male LP12.5 offspring displayed glucose intolerance and reduced insulin sensitivity associated with β-cell dysfunction with aging. High-fat diet exposure of metabolically normal 12-wk-old male LP12.5 induced glucose intolerance due to increased body weight, insulin resistance, and insufficient β-cell mass adaptation despite higher insulin secretion. Assessment of epigenetic mechanisms through microRNAs (miRs) by a real-time PCR-based microarray in islets revealed elevation in miRs that regulate insulin secretion (miRs 342, 143), insulin resistance (miR143), and obesity (miR219). In the islets, overexpression of miR143 reduced insulin secretion in response to glucose. In contrast to the model of LP exposure throughout pregnancy, islet protein levels of mTOR and pancreatic and duodenal homeobox 1 were normal in LP12.5 islets. Collectively, these data suggest that LP diet during the last week of pregnancy is critical and sufficient to induce specific and distinct developmental programming effects of tissues that control glucose homeostasis, thus causing permanent changes in specific set of microRNAs that may contribute to the overall vulnerability of the offspring to obesity, insulin resistance, and type 2 diabetes.
Collapse
Affiliation(s)
- Emilyn U Alejandro
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, Brehm Center for Diabetes Research, University of Michigan Medical School, Ann Arbor, Michigan
| | - Seokwon Jo
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota
| | - Brian Akhaphong
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota
| | - Pau Romaguera Llacer
- Division of Endocrinology, Metabolism and Diabetes, University of Miami, Miami, Florida
| | - Maya Gianchandani
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, Brehm Center for Diabetes Research, University of Michigan Medical School, Ann Arbor, Michigan
| | - Brigid Gregg
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Sebastian D Parlee
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Ormond A MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Ernesto Bernal-Mizrachi
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, Brehm Center for Diabetes Research, University of Michigan Medical School, Ann Arbor, Michigan
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
- Miami Veterans Affairs Healthcare System University of Miami, Miami, Florida
- Division of Endocrinology, Metabolism and Diabetes, University of Miami, Miami, Florida
| |
Collapse
|
12
|
Chassen SS, Ferchaud-Roucher V, Palmer C, Li C, Jansson T, Nathanielsz PW, Powell TL. Placental fatty acid transport across late gestation in a baboon model of intrauterine growth restriction. J Physiol 2020; 598:2469-2489. [PMID: 32338384 PMCID: PMC7384518 DOI: 10.1113/jp279398] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/14/2020] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS Intrauterine growth restriction (IUGR) is associated with perinatal morbidity and increased risk of lifelong disease, including neurodevelopmental impairment. Fatty acids (FA) are critical for normal brain development, although their transport across the placenta in IUGR pregnancies is poorly understood. The present study used a baboon model of IUGR (maternal nutrient restriction, MNR) to investigate placental expression of FA transport and binding proteins, and to determine gestational age-related changes in maternal and fetal plasma FA concentrations. We found MNR to be associated with increased placental expression of FA binding and transport proteins in late gestation, with fetal plasma FA concentrations that were similar to those of control animals. The present study is the first to report a profile of fetal and maternal plasma FA concentrations in a baboon model of growth restriction with data that suggest adaptation of placental transport to maintain delivery of critically needed FA. ABSTRACT Intrauterine growth restriction (IUGR) is associated with specific changes in placental transport of amino acids, folate and ions. However, little is known about placental fatty acid (FA) transport in IUGR. We hypothesized that placental FA transport proteins (FATP) and FA binding proteins (FABP) are up-regulated and fetal plasma FA concentrations are decreased at term in a baboon model of IUGR. Pregnant baboons were fed control or maternal nutrient restricted (MNR) diet (70% of control calories) from gestation day (GD) 30 (term 184 days). Plasma and placental samples were collected at GD120 (control n = 8, MNR n = 9), GD140 (control n = 6, MNR n = 7) and GD170 (control n = 6, MNR n = 6). Placentas were homogenized, and syncytiotrophoblast microvillous plasma membrane (MVM) and basal plasma membranes (BM) were isolated. Protein expression of FABP1, 3, 4 and 5 (homogenate) and FATP2, 4, and 6 (MVM, BM) was determined by Western blotting. FA content in maternal and umbilical vein plasma was measured by gas chromatography-mass spectrometry. Placental FABP1 and FABP5 expression was increased in MNR compared to controls at GD170, as was MVM FATP2 and FATP6 expression at GD140 and FATP2 expression at GD170. BM FATP4 and FATP6 expression was increased in MNR at GD140. Fetal plasma FA concentrations were similar in controls and MNR. These data suggest the adaptation of placental transport when aiming to maintain delivery of critically needed FAs for fetal growth and brain development.
Collapse
Affiliation(s)
- Stephanie S Chassen
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Veronique Ferchaud-Roucher
- University of Nantes, CHU Nantes, INRA, UMR 1280 Physiology of Nutritional Adaptations, Nantes, France
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Claire Palmer
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Cun Li
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
- Southwest National Primate Research Center, San Antonio, TX, USA
| | - Thomas Jansson
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Peter W Nathanielsz
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
- Southwest National Primate Research Center, San Antonio, TX, USA
| | - Theresa L Powell
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
13
|
Metagenome-wide association of gut microbiome features for schizophrenia. Nat Commun 2020; 11:1612. [PMID: 32235826 PMCID: PMC7109134 DOI: 10.1038/s41467-020-15457-9] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 03/12/2020] [Indexed: 02/06/2023] Open
Abstract
Evidence is mounting that the gut-brain axis plays an important role in mental diseases fueling mechanistic investigations to provide a basis for future targeted interventions. However, shotgun metagenomic data from treatment-naïve patients are scarce hampering comprehensive analyses of the complex interaction between the gut microbiota and the brain. Here we explore the fecal microbiome based on 90 medication-free schizophrenia patients and 81 controls and identify a microbial species classifier distinguishing patients from controls with an area under the receiver operating characteristic curve (AUC) of 0.896, and replicate the microbiome-based disease classifier in 45 patients and 45 controls (AUC = 0.765). Functional potentials associated with schizophrenia include differences in short-chain fatty acids synthesis, tryptophan metabolism, and synthesis/degradation of neurotransmitters. Transplantation of a schizophrenia-enriched bacterium, Streptococcus vestibularis, appear to induces deficits in social behaviors, and alters neurotransmitter levels in peripheral tissues in recipient mice. Our findings provide new leads for further investigations in cohort studies and animal models. Gut microbiome has been linked to neurogenerative diseases. Here, the authors present a metagenome-wide association study of schizophrenia (SZ) in human cohorts and identify SZ-associated specific gut-brain functional modules and pathways including SCFAs and neurotransmitters.
Collapse
|
14
|
Kabiri D, Romero R, Gudicha DW, Hernandez-Andrade E, Pacora P, Benshalom-Tirosh N, Tirosh D, Yeo L, Erez O, Hassan SS, Tarca AL. Prediction of adverse perinatal outcome by fetal biometry: comparison of customized and population-based standards. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2020; 55:177-188. [PMID: 31006913 PMCID: PMC6810752 DOI: 10.1002/uog.20299] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/09/2019] [Accepted: 04/12/2019] [Indexed: 05/15/2023]
Abstract
OBJECTIVE To compare the predictive performance of estimated fetal weight (EFW) percentiles, according to eight growth standards, to detect fetuses at risk for adverse perinatal outcome. METHODS This was a retrospective cohort study of 3437 African-American women. Population-based (Hadlock, INTERGROWTH-21st , World Health Organization (WHO), Fetal Medicine Foundation (FMF)), ethnicity-specific (Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)), customized (Gestation-Related Optimal Weight (GROW)) and African-American customized (Perinatology Research Branch (PRB)/NICHD) growth standards were used to calculate EFW percentiles from the last available scan prior to delivery. Prediction performance indices and relative risk (RR) were calculated for EFW < 10th and > 90th percentiles, according to each standard, for individual and composite adverse perinatal outcomes. Sensitivity at a fixed (10%) false-positive rate (FPR) and partial (FPR < 10%) and full areas under the receiver-operating-characteristics curves (AUC) were compared between the standards. RESULTS Ten percent (341/3437) of neonates were classified as small-for-gestational age (SGA) at birth, and of these 16.4% (56/341) had at least one adverse perinatal outcome. SGA neonates had a 1.5-fold increased risk of any adverse perinatal outcome (P < 0.05). The screen-positive rate of EFW < 10th percentile varied from 6.8% (NICHD) to 24.4% (FMF). EFW < 10th percentile, according to all standards, was associated with an increased risk for each of the adverse perinatal outcomes considered (P < 0.05 for all). The highest RRs associated with EFW < 10th percentile for each adverse outcome were 5.1 (95% CI, 2.1-12.3) for perinatal mortality (WHO); 5.0 (95% CI, 3.2-7.8) for perinatal hypoglycemia (NICHD); 3.4 (95% CI, 2.4-4.7) for mechanical ventilation (NICHD); 2.9 (95% CI, 1.8-4.6) for 5-min Apgar score < 7 (GROW); 2.7 (95% CI, 2.0-3.6) for neonatal intensive care unit (NICU) admission (NICHD); and 2.5 (95% CI, 1.9-3.1) for composite adverse perinatal outcome (NICHD). Although the RR CIs overlapped among all standards for each individual outcome, the RR of composite adverse perinatal outcome in pregnancies with EFW < 10th percentile was higher according to the NICHD (2.46; 95% CI, 1.9-3.1) than the FMF (1.47; 95% CI, 1.2-1.8) standard. The sensitivity for composite adverse perinatal outcome varied substantially between standards, ranging from 15% for NICHD to 32% for FMF, due mostly to differences in FPR; this variation subsided when the FPR was set to the same value (10%). Analysis of AUC revealed significantly better performance for the prediction of perinatal mortality by the PRB/NICHD standard (AUC = 0.70) compared with the Hadlock (AUC = 0.66) and FMF (AUC = 0.64) standards. Evaluation of partial AUC (FPR < 10%) demonstrated that the INTERGROWTH-21st standard performed better than the Hadlock standard for the prediction of NICU admission and mechanical ventilation (P < 0.05 for both). Although fetuses with EFW > 90th percentile were also at risk for any adverse perinatal outcome according to the INTERGROWTH-21st (RR = 1.4; 95% CI, 1.0-1.9) and Hadlock (RR = 1.7; 95% CI, 1.1-2.6) standards, many times fewer cases (2-5-fold lower sensitivity) were detected by using EFW > 90th percentile, rather than EFW < 10th percentile, in screening by these standards. CONCLUSIONS Fetuses with EFW < 10th percentile or EFW > 90th percentile were at increased risk of adverse perinatal outcomes according to all or some of the eight growth standards, respectively. The RR of a composite adverse perinatal outcome in pregnancies with EFW < 10th percentile was higher for the most-stringent (NICHD) compared with the least-stringent (FMF) standard. The results of the complementary analysis of AUC suggest slightly improved detection of adverse perinatal outcome by more recent population-based (INTERGROWTH-21st ) and customized (PRB/NICHD) standards compared with the Hadlock and FMF standards. Published 2019. This article is a U.S. Government work and is in the public domain in the USA.
Collapse
Affiliation(s)
- Doron Kabiri
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS)
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI
- Corresponding authors: Roberto Romero, MD, D.Med.Sci., Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women’s Hospital, 3990 John R Street, 4 Brush, Detroit, Michigan 48201; telephone: (313) 993-2700; fax: (313) 577-6294; . Adi L. Tarca, PhD, Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women’s Hospital, 3990 John R Street, 4 Brush, Detroit, Michigan 48201; telephone: (313) 577-5305; fax: (313) 577-6294;
| | - Dereje W. Gudicha
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS)
| | - Edgar Hernandez-Andrade
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Percy Pacora
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Neta Benshalom-Tirosh
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Dan Tirosh
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Lami Yeo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Maternity Department “D”, Division of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, faculty of Health Sciences, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Sonia S. Hassan
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI
| |
Collapse
|
15
|
Aguiar-Oliveira MH, Bartke A. Growth Hormone Deficiency: Health and Longevity. Endocr Rev 2019; 40:575-601. [PMID: 30576428 PMCID: PMC6416709 DOI: 10.1210/er.2018-00216] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/07/2018] [Indexed: 12/13/2022]
Abstract
The important role of GH in the control of mammalian longevity was first deduced from extended longevity of mice with genetic GH deficiency (GHD) or GH resistance. Mice with isolated GHD (IGHD) due to GHRH or GHRH receptor mutations, combined deficiency of GH, prolactin, and TSH, or global deletion of GH receptors live longer than do their normal siblings. They also exhibit multiple features of delayed and/or slower aging, accompanied by extension of healthspan. The unexpected, remarkable longevity benefit of severe endocrine defects in these animals presumably represents evolutionarily conserved trade-offs among aging, growth, maturation, fecundity, and the underlying anabolic processes. Importantly, the negative association of GH signaling with longevity extends to other mammalian species, apparently including humans. Data obtained in humans with IGHD type 1B, owing to a mutation of the GHRH receptor gene, in the Itabaianinha County, Brazil, provide a unique opportunity to study the impact of severe reduction in GH signaling on age-related characteristics, health, and functionality. Individuals with IGHD are characterized by proportional short stature, doll facies, high-pitched voices, and central obesity. They have delayed puberty but are fertile and generally healthy. Moreover, these IGHD individuals are partially protected from cancer and some of the common effects of aging and can attain extreme longevity, 103 years of age in one case. We think that low, but detectable, residual GH secretion combined with life-long reduction of circulating IGF-1 and with some tissue levels of IGF-1 and/or IGF-2 preserved may account for the normal longevity and apparent extension of healthspan in these individuals.
Collapse
Affiliation(s)
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, Illinois
| |
Collapse
|
16
|
Wixey JA, Lee KM, Miller SM, Goasdoue K, Colditz PB, Tracey Bjorkman S, Chand KK. Neuropathology in intrauterine growth restricted newborn piglets is associated with glial activation and proinflammatory status in the brain. J Neuroinflammation 2019; 16:5. [PMID: 30621715 PMCID: PMC6323795 DOI: 10.1186/s12974-018-1392-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/17/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The fetal brain is particularly vulnerable to intrauterine growth restriction (IUGR) conditions evidenced by neuronal and white matter abnormalities and altered neurodevelopment in the IUGR infant. To further our understanding of neurodevelopment in the newborn IUGR brain, clinically relevant models of IUGR are required. This information is critical for the design and implementation of successful therapeutic interventions to reduce aberrant brain development in the IUGR newborn. We utilise the piglet as a model of IUGR as growth restriction occurs spontaneously in the pig as a result of placental insufficiency, making it a highly relevant model of human IUGR. The purpose of this study was to characterise neuropathology and neuroinflammation in the neonatal IUGR piglet brain. METHODS Newborn IUGR (< 5th centile) and normally grown (NG) piglets were euthanased on postnatal day 1 (P1; < 18 h) or P4. Immunohistochemistry was utilised to examine neuronal, white matter and inflammatory responses, and PCR for cytokine analysis in parietal cortex of IUGR and NG piglets. RESULTS The IUGR piglet brain displayed less NeuN-positive cells and reduced myelination at both P1 and P4 in the parietal cortex, indicating neuronal and white matter disruption. A concurrent decrease in Ki67-positive proliferative cells and increase in cell death (caspase-3) in the IUGR piglet brain was also apparent on P4. We observed significant increases in the number of both Iba-1-positive microglia and GFAP-positive astrocytes in the white matter in IUGR piglet brain on both P1 and P4 compared with NG piglets. These increases were associated with a change in activation state, as noted by altered glial morphology. This inflammatory state was further evident with increased expression levels of proinflammatory cytokines (interleukin-1β, tumour necrosis factor-α) and decreased levels of anti-inflammatory cytokines (interleukin-4 and -10) observed in the IUGR piglet brains. CONCLUSIONS These findings suggest that the piglet model of IUGR displays the characteristic neuropathological outcomes of neuronal and white matter impairment similar to those reported in the IUGR human brain. The activated glial morphology and elevated proinflammatory cytokines is indicative of an inflammatory response that may be associated with neuronal damage and white matter disruption. These findings support the use of the piglet as a pre-clinical model for studying mechanisms of altered neurodevelopment in the IUGR newborn.
Collapse
Affiliation(s)
- Julie A Wixey
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, 4029, Australia.
| | - Kah Meng Lee
- Institute of Health Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, Australia
| | - Stephanie M Miller
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, 4029, Australia
| | - Kate Goasdoue
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, 4029, Australia
| | - Paul B Colditz
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, 4029, Australia.,Perinatal Research Centre, Royal Brisbane and Women's Hospital, Herston, QLD, 4029, Australia
| | - S Tracey Bjorkman
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, 4029, Australia
| | - Kirat K Chand
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, 4029, Australia
| |
Collapse
|
17
|
Spada E, Chiossi G, Coscia A, Monari F, Facchinetti F. Effect of maternal age, height, BMI and ethnicity on birth weight: an Italian multicenter study. J Perinat Med 2018; 46:1016-1021. [PMID: 29257759 DOI: 10.1515/jpm-2017-0102] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 11/07/2017] [Indexed: 11/15/2022]
Abstract
AIM To assess the effect of maternal age, height, early pregnancy body mass index (BMI) and ethnicity on birth weight. SUBJECTS AND METHODS A cross-sectional study was conducted on more than 42,000 newborns. Ethnicity was defined by maternal country of birth or, when missing (<0.6% of records), by citizenship. The effect of maternal characteristics on birth weight was evaluated with general linear models. RESULTS Maternal height and BMI, although not age, significantly affected birth weight. Among Italian babies, 4.7% of newborns were classified as appropriate-for-gestational age (AGA) (birth weight between the 10th and the 90th centile) according to the country-specific Italian Neonatal Study (INeS) charts and were re-classified as either large-(LGA) (birth weight >90th centile) or small-(SGA) (birth weight <10th centile) for gestational age (GA) after adjustment for maternal characteristics. On the contrary, 1.6% of Italian newborns were classified as SGA or LGA according to the INeS charts and re-classified as AGA after adjustment. Maternal ethnicity had a significant impact on birth weight. Specifically, babies born to Senegalese mothers were the lightest, whilst babies born to Chinese mothers were the heaviest. CONCLUSIONS Maternal height and early pregnancy BMI, should be considered in the evaluation of birth weight. The effect of ethnicity suggests the appropriateness of ethnic-specific charts. Further studies are necessary to determine if changes in birth weight classification, may translate into improved detection of subjects at risk of adverse outcomes.
Collapse
Affiliation(s)
- Elena Spada
- University of Turin, Department of Public Health and Pediatrics, Neonatology Unit, Torino, Italy
| | - Giuseppe Chiossi
- University of Texas Medical Branch, Department of Obstetrics and Gynecology, Galveston, TX, USA
| | - Alessandra Coscia
- University of Turin, Department of Public Health and Pediatrics, Neonatology Unit, Torino, Italy
| | - Francesca Monari
- University of Modena and Reggio Emilia, Obstetric Unit, Mother-Infant Department, Modena, Italy
| | - Fabio Facchinetti
- University of Modena and Reggio Emilia, Obstetric Unit, Mother-Infant Department, Modena, Italy
| |
Collapse
|
18
|
Guo JM, Lin HC, Ou P. [Prevalence of simple obesity and its high-risk factors in preschool children in Fuzhou, China]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20. [PMID: 30477626 PMCID: PMC7389030 DOI: 10.7499/j.issn.1008-8830.2018.11.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE To investigate the prevalence of simple obesity and its risk factors in preschool children in Fuzhou, China. METHODS The physical examination data of 14 kindergartens in Fuzhou, China were collected by stratified cluster random sampling. The detection rate of simple obesity was calculated. The children with normal body weight were selected as the control group by 1:1 case-control method. The risk factors for childhood simple obesity were investigated by self-made questionnaire and multivariate logistic regression analysis. RESULTS A total of 5 767 children aged 3-6 years were enrolled in this study. A total of 289 (5.01%) children with simple obesity were screened out, including 153 with mild obesity and 136 with moderate to severe obesity. The prevalence rate of simple obesity gradually increased with the age of children. The multivariate logistic regression analysis showed that the following factors were independent risk factors for simple obesity: preference for fried food (OR=4.789, P<0.05), caregivers' over-concern about diet (OR=4.620, P<0.05), eating before sleep (OR=4.006, P<0.05), eating fast (OR=3.221, P<0.05), preference for sweets (OR=2.282, P<0.05), high birth weight (OR=2.202, P<0.05), overweight or obesity in father (OR=2.074, P<0.05), overweight or obesity in mother (OR=2.047, P<0.05), more than 1.2 times the food intake at the same age (OR=2.013, P<0.05), watching TV (OR=1.665, P<0.05), and lack of exercise (OR=1.463, P<0.05). CONCLUSIONS The prevalence rate of simple obesity is 5.01% in preschool children in Fuzhou, China. The development of simple obesity is multifactorial. It is suggested that doctors, parents, and teachers should strengthen health education for preschool children, help them develop good living and eating habits, and encourage them to take more exercise, in order to reduce the development of simple obesity.
Collapse
Affiliation(s)
- Jing-Min Guo
- Department of Child Health Care, Fujian Provincial Maternity and Children's Hospital/Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China.
| | | | | |
Collapse
|
19
|
Cox LA, Olivier M, Spradling-Reeves K, Karere GM, Comuzzie AG, VandeBerg JL. Nonhuman Primates and Translational Research-Cardiovascular Disease. ILAR J 2018; 58:235-250. [PMID: 28985395 DOI: 10.1093/ilar/ilx025] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in the United States. Human epidemiological studies provide challenges for understanding mechanisms that regulate initiation and progression of CVD due to variation in lifestyle, diet, and other environmental factors. Studies describing metabolic and physiologic aspects of CVD, and those investigating genetic and epigenetic mechanisms influencing CVD initiation and progression, have been conducted in multiple Old World nonhuman primate (NHP) species. Major advantages of NHPs as models for understanding CVD are their genetic, metabolic, and physiologic similarities with humans, and the ability to control diet, environment, and breeding. These NHP species are also genetically and phenotypically heterogeneous, providing opportunities to study gene by environment interactions that are not feasible in inbred animal models. Each Old World NHP species included in this review brings unique strengths as models to better understand human CVD. All develop CVD without genetic manipulation providing multiple models to discover genetic variants that influence CVD risk. In addition, as each of these NHP species age, their age-related comorbidities such as dyslipidemia and diabetes are accelerated proportionally 3 to 4 times faster than in humans.In this review, we discuss current CVD-related research in NHPs focusing on selected aspects of CVD for which nonprimate model organism studies have left gaps in our understanding of human disease. We include studies on current knowledge of genetics, epigenetics, calorie restriction, maternal calorie restriction and offspring health, maternal obesity and offspring health, nonalcoholic steatohepatitis and steatosis, Chagas disease, microbiome, stem cells, and prevention of CVD.
Collapse
Affiliation(s)
- Laura A Cox
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas.,Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | - Michael Olivier
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas.,Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | | | - Genesio M Karere
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas
| | - Anthony G Comuzzie
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas
| | - John L VandeBerg
- South Texas Diabetes and Obesity Center, School of Medicine, University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, Texas
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Type 2 diabetes is a growing concern worldwide with increasing incidence in youth. Development of preventive strategies in earlier stages of life is crucial. We aimed to examine epidemiological evidence of early-life exposures and their associations with childhood and later risk of obesity and diabetes, and to discuss potential mechanisms. RECENT FINDINGS Parental obesity and diabetes in the preconception period may influence offspring's obesity risk via epigenetic mechanisms influencing gametogenesis and early development that could have significant transgenerational effects. A more comprehensive understanding of these effects is needed to identify possible avenues for interventions in both fathers and mothers to be. In addition, current evidence suggests that growth and body weight trajectories in infancy and childhood are useful indicators of later obesity and type 2 diabetes. Moreover, the composition and variations in the microbiome in early life are associated with long-term health and could mediate associations between several early-life exposures and later risk of diseases. Altogether, the epidemiological evidence supports the need for preconception and early-life interventions to reduce the obesity and diabetes burden in later life.
Collapse
Affiliation(s)
- Véronique Gingras
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Landmark Center, 401 Park Drive Suite 401 East, Boston, MA, 02215, USA.
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Landmark Center, 401 Park Drive Suite 401 East, Boston, MA, 02215, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Landmark Center, 401 Park Drive Suite 401 East, Boston, MA, 02215, USA
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
21
|
Jarman M, Mathe N, Ramazani F, Pakseresht M, Robson PJ, Johnson ST, Bell RC. Dietary Patterns Prior to Pregnancy and Associations with Pregnancy Complications. Nutrients 2018; 10:nu10070914. [PMID: 30018227 PMCID: PMC6073508 DOI: 10.3390/nu10070914] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 01/02/2023] Open
Abstract
Few studies have explored pre-pregnancy diet and its relationship with pregnancy outcomes. The objectives of this study were to: (1) derive pre-pregnancy dietary patterns for women enrolled in a prospective cohort in the province of Alberta, Canada; (2) describe associations between dietary patterns and socio-demographic characteristics; and (3) describe associations between dietary patterns and pregnancy complications. Upon enrolment into the Alberta Pregnancy Outcomes and Nutrition (APrON) study (median age of gestation, 17 weeks), women (n = 1545) completed a validated 142-item food frequency questionnaire recording food and beverages consumed “in the 12 months prior to pregnancy”. Other assessments included pre-pregnancy body mass index (BMI), gestational weight gain, gestational hypertension, gestational diabetes, and socio-demographic characteristics. Dietary patterns were derived using principal components analysis. Scores were calculated to represent adherence with each dietary pattern retained. Four dietary patterns were retained, accounting for 22.9% of the variation in the overall diet. Dietary patterns were named the “healthy”, “meat and refined carbohydrate”, “beans, cheese and salad” or “tea and coffee” patterns. Higher “healthy” pattern scores prior to pregnancy were associated with lower odds of developing gestational hypertension during pregnancy (adjusted Odds Ratio (OR): 0.6, 95% Confidence Intervals (CI): 0.4, 0.9). Diet prior to pregnancy is an important target for interventions and may reduce the likelihood of developing complications such as gestational hypertension during pregnancy.
Collapse
Affiliation(s)
- Megan Jarman
- Li Ka Shing Centre for Health Research Innovation, Department of Agricultural, Food and Nutritional Sciences, Division of Human Nutrition, University of Alberta, Edmonton, AB T6G 2E1, Canada; (M.J.); (M.P.)
| | - Nonsikelelo Mathe
- Alliance for Health Outcomes Research in Diabetes (ACHORD), University of Alberta, Edmonton, AB T6G 2E1, Canada; (N.M.); (F.R.); (S.T.J.)
| | - Fatemeh Ramazani
- Alliance for Health Outcomes Research in Diabetes (ACHORD), University of Alberta, Edmonton, AB T6G 2E1, Canada; (N.M.); (F.R.); (S.T.J.)
| | - Mohammadreza Pakseresht
- Li Ka Shing Centre for Health Research Innovation, Department of Agricultural, Food and Nutritional Sciences, Division of Human Nutrition, University of Alberta, Edmonton, AB T6G 2E1, Canada; (M.J.); (M.P.)
- Cancer Research, CancerControl Alberta, Alberta Health Services, Edmonton, AB T5J 3H1, Canada;
| | - Paula J. Robson
- Cancer Research, CancerControl Alberta, Alberta Health Services, Edmonton, AB T5J 3H1, Canada;
| | - Steven T. Johnson
- Alliance for Health Outcomes Research in Diabetes (ACHORD), University of Alberta, Edmonton, AB T6G 2E1, Canada; (N.M.); (F.R.); (S.T.J.)
- Faculty of Health Disciplines, Athabasca University, Athabasca, AB T9S 3A3, Canada
| | - Rhonda C. Bell
- Li Ka Shing Centre for Health Research Innovation, Department of Agricultural, Food and Nutritional Sciences, Division of Human Nutrition, University of Alberta, Edmonton, AB T6G 2E1, Canada; (M.J.); (M.P.)
- Correspondence:
| | - the APrON and ENRICH study teams
- Li Ka Shing Centre for Health Research Innovation, Department of Agricultural, Food and Nutritional Sciences, Division of Human Nutrition, University of Alberta, Edmonton, AB T6G 2E1, Canada; (M.J.); (M.P.)
| |
Collapse
|
22
|
How does the social environment during life course embody in and influence the development of cancer? Int J Public Health 2018; 63:811-821. [DOI: 10.1007/s00038-018-1131-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 05/19/2018] [Accepted: 06/01/2018] [Indexed: 12/21/2022] Open
|
23
|
Wixey JA, Chand KK, Pham L, Colditz PB, Bjorkman ST. Therapeutic potential to reduce brain injury in growth restricted newborns. J Physiol 2018; 596:5675-5686. [PMID: 29700828 DOI: 10.1113/jp275428] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/28/2018] [Indexed: 12/27/2022] Open
Abstract
Brain injury in intrauterine growth restricted (IUGR) infants is a major contributing factor to morbidity and mortality worldwide. Adverse outcomes range from mild learning difficulties, to attention difficulties, neurobehavioral issues, cerebral palsy, epilepsy, and other cognitive and psychiatric disorders. While the use of medication to ameliorate neurological deficits in IUGR neonates has been identified as warranting urgent research for several years, few trials have been reported. This review summarises clinical trials focusing on brain protection in the IUGR newborn as well as therapeutic interventions trialled in animal models of IUGR. Therapeutically targeting mechanisms of brain injury in the IUGR neonate is fundamental to improving long-term neurodevelopmental outcomes. Inflammation is a key mechanism in neonatal brain injury; and therefore an appealing target. Ibuprofen, an anti-inflammatory drug currently used in the preterm neonate, may be a potential therapeutic candidate to treat brain injury in the IUGR neonate. To better understand the potential of ibuprofen and other therapeutic agents to be neuroprotective in the IUGR neonate, long-term follow-up information of neurodevelopmental outcomes must be studied. Where agents such as ibuprofen are shown to be effective, have a good safety profile and are relatively inexpensive, they can be widely adopted and lead to improved outcomes.
Collapse
Affiliation(s)
- Julie A Wixey
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| | - Kirat K Chand
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| | - Lily Pham
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| | - Paul B Colditz
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| | - S Tracey Bjorkman
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, 4029, Australia
| |
Collapse
|
24
|
Aranda F, Udry S, Perés Wingeyer S, Amshoff LC, Bogdanova N, Wieacker P, Latino JO, Markoff A, de Larrañaga G. Maternal carriers of the ANXA5 M2 haplotype are exposed to a greater risk for placenta-mediated pregnancy complications. J Assist Reprod Genet 2018; 35:921-928. [PMID: 29497952 DOI: 10.1007/s10815-018-1142-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/15/2018] [Indexed: 01/28/2023] Open
Abstract
PURPOSE Annexin A5 (ANXA5) is a protein abundantly expressed in normal placenta where it contributes to the healthy outcome of a pregnancy. Lower ANXA5 levels have been observed in M2/ANXA5 haplotype carrying chorion. Consequently, this study aimed to assess the potential association of M2 maternal carrier status with the risk of recurrent pregnancy loss (RPL), the timing of miscarriages, and other obstetric complications, for the first time in a population from Latin America. METHODS This study was designed as a prospective recruitment of RPL patients with post hoc analysis. The distribution of the M2/ANXA5 haplotype was compared between a group of 229 Argentine women with RPL and 100 parous controls, and was further analyzed in subgroups of patients stratified according to the timing of miscarriages and in relation to other obstetric complications. RESULTS No significant differences were found in the distribution of M2 haplotype among either RPL patients or the subgroups with embryonic, early fetal, or late fetal losses compared to parous controls. Notwithstanding, maternal M2/ANXA5 was found to be independently associated with a higher risk of suffering intrauterine growth restriction (IUGR) and/or preeclampsia (PE). Simultaneously, the presence of inherited and/or acquired thrombophilia also proved to be an independent risk factor for these. CONCLUSIONS The association found between the maternal carriage of the M2/ANXA5 haplotype and an elevated risk of IUGR and/or PE supports the hypothesis that carrier status of this haplotype and the consequently reduced placental ANXA5 expression might be responsible, at least partially, for the onset of these gestational vascular complications.
Collapse
Affiliation(s)
- Federico Aranda
- Hemostasis and Thrombosis Laboratory, Hospital of Infectious Diseases "Dr Francisco J. Muñiz", Uspallata 2272, C1282AEN, Buenos Aires, Argentina.
| | - Sebastián Udry
- Section of Autoimmune Diseases, Thrombophilia and Pregnancy, Acute Care Hospital "Dr Carlos G. Durand", Av. Díaz Vélez 5044, C1405DCS, Buenos Aires, Argentina
| | - Silvia Perés Wingeyer
- Hemostasis and Thrombosis Laboratory, Hospital of Infectious Diseases "Dr Francisco J. Muñiz", Uspallata 2272, C1282AEN, Buenos Aires, Argentina
| | - Lea Christina Amshoff
- Institute of Human Genetics, University Clinic Muenster, Vesaliusweg 12, 48149, Muenster, Germany
| | - Nadja Bogdanova
- Institute of Human Genetics, University Clinic Muenster, Vesaliusweg 12, 48149, Muenster, Germany
| | - Peter Wieacker
- Institute of Human Genetics, University Clinic Muenster, Vesaliusweg 12, 48149, Muenster, Germany
| | - José Omar Latino
- Section of Autoimmune Diseases, Thrombophilia and Pregnancy, Acute Care Hospital "Dr Carlos G. Durand", Av. Díaz Vélez 5044, C1405DCS, Buenos Aires, Argentina
| | - Arseni Markoff
- Institute of Human Genetics, University Clinic Muenster, Vesaliusweg 12, 48149, Muenster, Germany
| | - Gabriela de Larrañaga
- Hemostasis and Thrombosis Laboratory, Hospital of Infectious Diseases "Dr Francisco J. Muñiz", Uspallata 2272, C1282AEN, Buenos Aires, Argentina
| |
Collapse
|
25
|
Lawson SP, Helmreich SL, Rehan SM. Effects of nutritional deprivation on development and behavior in the subsocial bee Ceratina calcarata (Hymenoptera: Xylocopinae). ACTA ACUST UNITED AC 2017; 220:4456-4462. [PMID: 28970348 DOI: 10.1242/jeb.160531] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 09/26/2017] [Indexed: 01/11/2023]
Abstract
By manipulating resources or dispersal opportunities, mothers can force offspring to remain at the nest to help raise siblings, creating a division of labor. In the subsocial bee Ceratina calcarata, mothers manipulate the quantity and quality of pollen provided to the first female offspring, producing a dwarf eldest daughter that is physically smaller and behaviorally subordinate. This daughter forages for her siblings and forgoes her own reproduction. To understand how the mother's manipulation of pollen affects the physiology and behavior of her offspring, we manipulated the amount of pollen provided to offspring and measured the effects of pollen quantity on offspring development, adult body size and behavior. We found that by experimentally manipulating pollen quantities we could recreate the dwarf eldest daughter phenotype, demonstrating how nutrient deficiency alone can lead to the development of a worker-like daughter. Specifically, by reducing the pollen and nutrition to offspring, we significantly reduced adult body size and lipid stores, creating significantly less aggressive, subordinate individuals. Worker behavior in an otherwise solitary bee begins to explain how maternal manipulation of resources could lead to the development of social organization and reproductive hierarchies, a major step in the transition to highly social behaviors.
Collapse
Affiliation(s)
- Sarah P Lawson
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| | | | - Sandra M Rehan
- Department of Biological Sciences, University of New Hampshire, Durham, NH 03824, USA
| |
Collapse
|
26
|
Somatic growth, aging, and longevity. NPJ Aging Mech Dis 2017; 3:14. [PMID: 28970944 PMCID: PMC5622030 DOI: 10.1038/s41514-017-0014-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 09/06/2017] [Accepted: 09/13/2017] [Indexed: 02/01/2023] Open
Abstract
Although larger species of animals typically live longer than smaller species, the relationship of body size to longevity within a species is generally opposite. The longevity advantage of smaller individuals can be considerable and is best documented in laboratory mice and in domestic dogs. Importantly, it appears to apply broadly, including humans. It is not known whether theses associations represent causal links between various developmental and physiological mechanisms affecting growth and/or aging. However, variations in growth hormone (GH) signaling are likely involved because GH is a key stimulator of somatic growth, and apparently also exerts various “pro-aging” effects. Mechanisms linking GH, somatic growth, adult body size, aging, and lifespan likely involve target of rapamycin (TOR), particularly one of its signaling complexes, mTORC1, as well as various adjustments in mitochondrial function, energy metabolism, thermogenesis, inflammation, and insulin signaling. Somatic growth, aging, and longevity are also influenced by a variety of hormonal and nutritional signals, and much work will be needed to answer the question of why smaller individuals may be likely to live longer.
Collapse
|
27
|
Chiossi G, Pedroza C, Costantine MM, Truong VTT, Gargano G, Saade GR. Customized vs population-based growth charts to identify neonates at risk of adverse outcome: systematic review and Bayesian meta-analysis of observational studies. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2017; 50:156-166. [PMID: 27935148 DOI: 10.1002/uog.17381] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 11/26/2016] [Accepted: 11/30/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVE To compare the effectiveness of customized vs population-based growth charts for the prediction of adverse pregnancy outcomes. METHODS MEDLINE, ClinicalTrials.gov and The Cochrane Library were searched up to 31 May 2016 to identify interventional and observational studies comparing adverse outcomes among large- (LGA) and small- (SGA) for-gestational-age neonates, when classified according to customized vs population-based growth charts. Perinatal mortality and admission to the neonatal intensive care unit (NICU) of both SGA and LGA neonates, intrauterine fetal demise (IUFD) and neonatal mortality of SGA neonates, and neonatal shoulder dystocia and hypoglycemia as well as maternal third- and fourth-degree perineal lacerations in LGA pregnancies were evaluated. RESULTS The electronic search identified 237 records that were examined based on title and abstract, of which 27 full-text articles were examined for eligibility. After excluding seven articles, 20 observational studies were included in a Bayesian meta-analysis. Neonates classified as SGA according to customized growth charts had higher risks of IUFD (odds ratio (OR), 7.8 (95% CI, 4.2-12.3)), neonatal death (OR, 3.5 (95% CI, 1.1-8.0)), perinatal death (OR, 5.8 (95% CI, 3.8-7.8)) and NICU admission (OR, 3.6 (95% CI, 2.0-5.5)) than did non-SGA cases. Neonates classified as SGA according to population-based growth charts also had increased risk for adverse outcomes, albeit the point estimates of the pooled ORs were smaller: IUFD (OR, 3.3 (95% CI, 1.9-5.0)), neonatal death (OR, 2.9 (95% CI, 1.2-4.5)), perinatal death (OR, 4.0 (95% CI, 2.8-5.1)) and NICU admission (OR, 2.4 (95% CI, 1.7-3.2)). For LGA vs non-LGA, there were no differences in pooled ORs for perinatal death, NICU admission, hypoglycemia and maternal third- and fourth-degree perineal lacerations when classified according to either the customized or the population-based approach. In contrast, both approaches indicated that LGA neonates are at increased risk for shoulder dystocia than are non-LGA ones (OR, 7.4 (95% CI, 4.9-9.8) using customized charts; OR, 8.0 (95% CI, 5.3-10.1) using population-based charts). CONCLUSIONS Both customized and population-based growth charts can identify SGA neonates at risk for adverse outcomes. Although the point estimates of the pooled ORs may differ for some outcomes, the overlapping CIs and lack of direct comparisons prevent conclusions from being drawn on the superiority of one method. Future clinical trials should compare directly the two approaches in the management of fetuses of abnormal size. Copyright © 2016 ISUOG. Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- G Chiossi
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - C Pedroza
- Center for Clinical Research and Evidence-Based Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - M M Costantine
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - V T T Truong
- Center for Clinical Research and Evidence-Based Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - G Gargano
- Department of Neonatology, Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - G R Saade
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
28
|
Li C, Jenkins S, Mattern V, Comuzzie AG, Cox LA, Huber HF, Nathanielsz PW. Effect of moderate, 30 percent global maternal nutrient reduction on fetal and postnatal baboon phenotype. J Med Primatol 2017; 46:293-303. [PMID: 28744866 DOI: 10.1111/jmp.12290] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2017] [Indexed: 01/24/2023]
Abstract
BACKGROUND Most developmental programming studies on maternal nutrient reduction (MNR) are in altricial rodents whose maternal nutritional burden and offspring developmental trajectory differ from precocial non-human primates and humans. METHODS Control (CTR) baboon mothers ate ad libitum; MNR mothers ate 70% global control diet in pregnancy and lactation. RESULTS We present offspring morphometry, blood cortisol, and adrenocorticotropin (ACTH) during second half of gestation (G) and first three postnatal years. Moderate MNR produced intrauterine growth restriction (IUGR). IUGR males (n=43) and females (n=28) were smaller than CTR males (n=50) and females (n=47) in many measurements at many ages. In CTR, fetal ACTH increased 228% and cortisol 48% between 0.65G and 0.9G. IUGR ACTH was elevated at 0.65G and cortisol at 0.9G. 0.9G maternal gestational weight gain, fetal weight, and placenta weight were correlated. CONCLUSIONS Moderate IUGR decreased body weight and morphometric measurements at key time points and altered hypothalamo-pituitary-adrenal function.
Collapse
Affiliation(s)
- Cun Li
- Texas Pregnancy and Life-course Health Center, Department of Animal Sciences, University of Wyoming, Laramie, WY, USA.,Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Susan Jenkins
- Texas Pregnancy and Life-course Health Center, Department of Animal Sciences, University of Wyoming, Laramie, WY, USA
| | - Vicki Mattern
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Laura A Cox
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Hillary F Huber
- Texas Pregnancy and Life-course Health Center, Department of Animal Sciences, University of Wyoming, Laramie, WY, USA
| | - Peter W Nathanielsz
- Texas Pregnancy and Life-course Health Center, Department of Animal Sciences, University of Wyoming, Laramie, WY, USA.,Texas Biomedical Research Institute, San Antonio, TX, USA
| |
Collapse
|
29
|
Bartke A, Sun L, Fang Y, Hill C. Growth hormone actions during development influence adult phenotype and longevity. Exp Gerontol 2016; 86:22-27. [PMID: 26752217 PMCID: PMC4930735 DOI: 10.1016/j.exger.2015.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 11/22/2022]
Abstract
There is considerable evidence that exposure to undernutrition, overnutrition, stress or endocrine disruptors during fetal development can increase the probability of obesity, hypertension, cardiovascular disease and other problems in adult life. In contrast to these findings, reducing early postnatal growth by altering maternal diet or number of pups in a litter can increase longevity. In hypopituitary Ames dwarf mice, which are remarkably long lived, a brief period of growth hormone therapy starting at 1 or 2weeks of age reduces longevity and normalizes ("rescues") multiple aging-related traits. Collectively, these findings indicate that nutritional and hormonal signals during development can have profound impact on the trajectory of aging. We suspect that altered "programming" of aging during development may represent one of the mechanisms of the Developmental Origins of Health and Disease (DOHaD) and the detrimental effects of "catch-up" growth.
Collapse
Affiliation(s)
- A Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States.
| | - L Sun
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Y Fang
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - C Hill
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States
| |
Collapse
|
30
|
Wixey JA, Chand KK, Colditz PB, Bjorkman ST. Review: Neuroinflammation in intrauterine growth restriction. Placenta 2016; 54:117-124. [PMID: 27916232 DOI: 10.1016/j.placenta.2016.11.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 12/21/2022]
Abstract
Disruption to the maternal environment during pregnancy from events such as hypoxia, stress, toxins, inflammation, and reduced placental blood flow can affect fetal development. Intrauterine growth restriction (IUGR) is commonly caused by chronic placental insufficiency, interrupting supply of oxygen and nutrients to the fetus resulting in abnormal fetal growth. IUGR is a major cause of perinatal morbidity and mortality, occurring in approximately 5-10% of pregnancies. The fetal brain is particularly vulnerable in IUGR and there is an increased risk of long-term neurological disorders including cerebral palsy, epilepsy, learning difficulties, behavioural difficulties and psychiatric diagnoses. Few studies have focused on how growth restriction interferes with normal brain development in the IUGR neonate but recent studies in growth restricted animal models demonstrate increased neuroinflammation. This review describes the role of neuroinflammation in the progression of brain injury in growth restricted neonates. Identifying the mediators responsible for alterations in brain development in the IUGR infant is key to prevention and treatment of brain injury in these infants.
Collapse
Affiliation(s)
- Julie A Wixey
- The University of Queensland, Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland 4029, Australia.
| | - Kirat K Chand
- The University of Queensland, Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland 4029, Australia
| | - Paul B Colditz
- The University of Queensland, Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland 4029, Australia
| | - S Tracey Bjorkman
- The University of Queensland, Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland 4029, Australia
| |
Collapse
|
31
|
Groom KM, McCowan LM, Stone PR, Chamley LC, McLintock C. Enoxaparin for the prevention of preeclampsia and intrauterine growth restriction in women with a prior history - an open-label randomised trial (the EPPI trial): study protocol. BMC Pregnancy Childbirth 2016; 16:367. [PMID: 27876004 PMCID: PMC5120461 DOI: 10.1186/s12884-016-1162-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 11/15/2016] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Preeclampsia and intrauterine fetal growth restriction (IUGR) are two of the most common causes of maternal and perinatal morbidity and mortality. Current methods of predicting those at most risk of these conditions remain relatively poor, and in clinical practice past obstetric history remains the most commonly used tool. Aspirin and, in women at risk of preeclampsia only, calcium have been demonstrated to have a modest effect on risk reduction. Several observational studies and randomised trials suggest that low molecular weight heparin (LMWH) therapy may confer some benefit. METHODS/DESIGN This is a multicentre open label randomised controlled trial to determine the effect of the LMWH, enoxaparin, on the prevention of recurrence of preeclampsia and/or IUGR in women at high risk due to their past obstetric history in addition to standard high risk care for all participants. INCLUSION CRITERIA A singleton pregnancy >6+0 and <16+0 weeks gestation with most recent prior pregnancy with duration >12 weeks having; (1) preeclampsia delivered <36+0 weeks, (2) Small for gestational age (SGA) infant <10th customised birthweight centile delivered <36+0 weeks or, (3) SGA infant ≤3rd customised birthweight centile delivered at any gestation. Randomisation is stratified for maternal thrombophilia status and women are randomly assigned to 'standard high risk care' or 'standard high risk care' plus enoxaparin 40 mg from recruitment until 36+0 weeks or delivery, whichever occurs sooner. Standard high risk care includes the use of aspirin 100 mg daily and calcium 1000-1500 mg daily (unless only had previous SGA with no preeclampsia). The primary outcome is preeclampsia and/or SGA <5th customised birthweight centile. Analysis will be by intention to treat. DISCUSSION The EPPI trial has more focussed and clinically relevant inclusion criteria than other randomised trials with a more restricted composite primary outcome. The inclusion of standard use of aspirin (and calcium) for all participants will help to ensure that any differences observed in outcome are likely to be related to enoxaparin use. These data will make a significant contribution to future meta-analyses and systematic reviews on the use of LMWH for the prevention of placental mediated conditions. TRIAL REGISTRATION ACTRN12609000699268 Australian New Zealand Clinical Trials Registry. Date registered 13/Aug/2009 (prospective registration).
Collapse
Affiliation(s)
- K. M. Groom
- Department of Obstetrics and Gynaecology, Faculty of Medical Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
- National Women’s Health, Auckland City Hospital, Private Bag 92024, Auckland, New Zealand
| | - L. M. McCowan
- Department of Obstetrics and Gynaecology, Faculty of Medical Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
- National Women’s Health, Auckland City Hospital, Private Bag 92024, Auckland, New Zealand
| | - P. R. Stone
- Department of Obstetrics and Gynaecology, Faculty of Medical Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - L. C. Chamley
- Department of Obstetrics and Gynaecology, Faculty of Medical Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - C. McLintock
- National Women’s Health, Auckland City Hospital, Private Bag 92024, Auckland, New Zealand
| | - the EPPI trial Study Group
- Department of Obstetrics and Gynaecology, Faculty of Medical Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
- National Women’s Health, Auckland City Hospital, Private Bag 92024, Auckland, New Zealand
| |
Collapse
|
32
|
Ziauddeen H, Garcia-Rizo C, Bernardo M, Kirkpatrick B, Ozanne SE, Jones PB, Fernandez-Egea E. Association of birth weight and the development of antipsychotic induced adiposity in individuals with treatment resistant schizophrenia. Eur Neuropsychopharmacol 2016; 26:972-8. [PMID: 27107738 DOI: 10.1016/j.euroneuro.2016.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 02/16/2016] [Accepted: 03/18/2016] [Indexed: 11/19/2022]
Abstract
Though weight gain is a common side effect of antipsychotic treatment, there are no useful predictors of which patients are likely to be affected and to what degree. It has been shown that exposure to adverse conditions during intra-uterine life confers a vulnerability to the development of later life metabolic complications and low birth weight for gestational age has been shown to be a robust marker of such prenatal adversity. We hypothesised that patients with schizophrenia with a lower birth weight will have increased vulnerability to the weight inducing effects of antipsychotic treatment. The relationship between birth weight and total and central adiposity, measured as body mass index (BMI) and waist-to-hip ratio (WHR) respectively, was examined in three groups: drug naïve first episode of psychosis (FEP) patients (n=41), treatment resistant schizophrenia (TRS) patients (n=42) and matched healthy volunteers (n=72). All analyses were controlled for age, gender and duration of treatment exposure. We found that a lower birth weight was associated with higher BMI and WHR only in TRS patients but not in FEP or controls, suggesting that prenatal adversity, as indicated by the surrogate marker of a lower birth weight, confers an increased vulnerability to clozapine induced weight gain.
Collapse
Affiliation(s)
- Hisham Ziauddeen
- Department of Psychiatry, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0SZ, UK; Cambridgeshire and Peterborough Foundation Trust, Fulbourn Hospital, Cambridge CB21 5EF, UK; University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, CB2 0QQ UK
| | - Clemente Garcia-Rizo
- Barcelona Clinic Schizophrenia Unit, Neuroscience Institute, Hospital Clinic of Barcelona and Department of Psychiatry and Clinical Psychobiology, University of Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain; Institut d׳Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Spain
| | - Miquel Bernardo
- Barcelona Clinic Schizophrenia Unit, Neuroscience Institute, Hospital Clinic of Barcelona and Department of Psychiatry and Clinical Psychobiology, University of Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain; Institut d׳Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Spain
| | - Brian Kirkpatrick
- Department of Psychiatry & Behavioral Sciences, University of Nevada School of Medicine, Reno, NV, United States
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, CB2 0QQ UK
| | - Peter B Jones
- Department of Psychiatry, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0SZ, UK; Cambridgeshire and Peterborough Foundation Trust, Fulbourn Hospital, Cambridge CB21 5EF, UK
| | - Emilio Fernandez-Egea
- Department of Psychiatry, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0SZ, UK; Cambridgeshire and Peterborough Foundation Trust, Fulbourn Hospital, Cambridge CB21 5EF, UK; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain.
| |
Collapse
|
33
|
Lopes GAD, Ribeiro VLB, Barbisan LF, Marchesan Rodrigues MA. Fetal developmental programing: insights from human studies and experimental models. J Matern Fetal Neonatal Med 2016; 30:722-728. [DOI: 10.1080/14767058.2016.1183635] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
| | | | - Luís Fernando Barbisan
- Department of Morphology, Institute of Biosciences, UNESP – Univ. Estadual Paulista, Botucatu, SP, Brazil
| | | |
Collapse
|
34
|
Pollock AJ, Allen DB, Wiebe D, Eickhoff J, MacDonald M, Baker M. Development of filter paper hemoglobin A1c assay applicable to newborn screening. Clin Chim Acta 2016; 457:24-6. [PMID: 27016455 DOI: 10.1016/j.cca.2016.03.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/18/2016] [Accepted: 03/18/2016] [Indexed: 01/12/2023]
Abstract
PURPOSE Gestational diabetes influences risk for future metabolic disease including type 2 diabetes. Hemoglobin A1c (HbA1c) measurement assesses hemoglobin A glycosylation, and could theoretically be used as a test to estimate gestational glucose exposure. HbA1c assay on dried blood spots (DBS) is needed before potential application to statewide newborn screening (NBS) population studies. The study aimed to establish a reliable method to measure HbA1c on NBS DBS specimens. De-identified blood was used to generate trials to evaluate stability of HbA1c in DBS, optimal elution time, and stability of eluted blood. RESULTS Analysis of DBS stability HbA1c measurements from 3 to 6days after collection overestimated HbA1c values by a bias factor between 0.83 and 0.87. Sixty minutes of elution time produced maximal reproducibility and minimal bias of results. Within assay standard deviation: 0.058; average bias: -0.02%. Stability of eluted blood did not vary significantly between days 0-2 after DBS elution. CONCLUSIONS Measurement of HbA1c levels on DBS from human blood is feasible. Results suggest new method using DBS to measure HbA1c level with the following characteristics: optimal time for sample analysis 3-6days after collection, elution time of 60min and eluted blood analysis within 2days of elution. Measurement of neonatal HbA1c could provide insight regarding the infant's in utero exposure to glucose.
Collapse
Affiliation(s)
- Allison J Pollock
- University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792, USA.
| | - David B Allen
- University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792, USA
| | - Donald Wiebe
- University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792, USA
| | - Jens Eickhoff
- University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792, USA
| | - Michael MacDonald
- University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792, USA
| | - Mei Baker
- University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792, USA; Wisconsin State Laboratory of Hygiene, 2601 Agriculture Dr, Madison, WI 53718, USA
| |
Collapse
|
35
|
Keswani SG, Balaji S, Katz AB, King A, Omar K, Habli M, Klanke C, Crombleholme TM. Intraplacental gene therapy with Ad-IGF-1 corrects naturally occurring rabbit model of intrauterine growth restriction. Hum Gene Ther 2015; 26:172-82. [PMID: 25738403 DOI: 10.1089/hum.2014.065] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Intrauterine growth restriction (IUGR) due to placental insufficiency is a leading cause of perinatal complications for which there is no effective prenatal therapy. We have previously demonstrated that intraplacental injection of adenovirus-mediated insulin-like growth factor-1 (Ad-IGF-1) corrects fetal weight in a murine IUGR model induced by mesenteric uterine artery branch ligation. This study investigated the effect of intraplacental Ad-IGF-1 gene therapy in a rabbit model of naturally occurring IUGR (runt) due to placental insufficiency, which is similar to the human IUGR condition with onset in the early third trimester, brain sparing, and a reduction in liver weight. Laparotomy was performed on New Zealand White rabbits on day 21 of 30 days of gestation and litters were divided into five groups: Control (first position)+phosphate-buffered saline (PBS), control+Ad-IGF-1, runt (third position)+PBS, runt+Ad-IGF-1, and runt+Ad-LacZ. The effect of IGF-1 gene therapy on fetal, placental, liver, heart, lung, and musculoskeletal weights of the growth-restricted pups was examined. Protein expression after gene transfer was seen along the maternal-fetal placenta interface (n=12) 48 hr after gene therapy. There was minimal gene transfer detected in the pups or maternal organs. At term, compared with the normally grown first-position control, the runted third-position pups demonstrated significantly lower fetal, placental, liver, lung, and musculoskeletal weights. The fetal, liver, and musculoskeletal weights were restored to normal by intraplacental Ad-IGF-1 gene therapy (p<0.01), with no change in the placental weight. Intraplacental gene therapy is a novel strategy for the treatment of IUGR caused by placental insufficiency that takes advantage of an organ that will be discarded at birth. Development of nonviral IGF-1 gene delivery using placenta-specific promoters can potentially minimize toxicity to the mother and fetus and facilitate clinical translation of this novel therapy.
Collapse
Affiliation(s)
- Sundeep G Keswani
- 1 Center for Molecular Fetal Therapy, Division of Pediatric, General, Thoracic, and Fetal Surgery, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine , Cincinnati, OH 45229-3039
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Kirkpatrick B, Miller B, García-Rizo C, Fernandez-Egea E. Schizophrenia: a systemic disorder. ACTA ACUST UNITED AC 2015; 8:73-9. [PMID: 23518782 DOI: 10.3371/csrp.kimi.031513] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The concept of schizophrenia that is most widely taught is that it is a disorder in which psychotic symptoms are the main problem, and a dysregulation of dopamine signaling is the main feature of pathophysiology. However, this concept limits clinical assessment, the treatments offered to patients, research, and the development of therapeutics. A more appropriate conceptual model is that: 1) schizophrenia is not a psychotic disorder, but a disorder of essentially every brain function in which psychosis is present; 2) it is not a brain disease, but a disorder with impairments throughout the body; 3) for many patients, neuropsychiatric problems other than psychosis contribute more to impairment in function and quality of life than does psychosis; and, 4) some conditions that are considered to be comorbid are integral parts of the illness. In conclusion, students, patients, and family members should be taught this model, along with its implications for assessment, research, and therapeutics.
Collapse
Affiliation(s)
- Brian Kirkpatrick
- Department of Psychiatry and Behavioral Sciences, University of Nevada, Reno, NV
| | - Brian Miller
- Department of Psychiatry and Health Behavior, Georgia Health Sciences University, Augusta, GA
| | - Clemente García-Rizo
- Schizophrenia Program, Department of Psychiatry, Neuroscience Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Emilio Fernandez-Egea
- Department of Psychiatry, Behavioural and Clinical Neuroscience Institute (BCNI), University of Cambridge, Good Outcome Schizophrenia Clinic, Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK, Huntingdon, UK
| |
Collapse
|
37
|
Olson G, Weiner SJ, Rouse DJ, Reddy UM, Mercer BM, Varner MW, Leveno KJ, Iams JD, Wapner RJ, Ramin SM, Malone FD, Carpenter MW, O’Sullivan MJ, Dinsmoor MJ, Hankins GDV, Caritis SN. Relation between birth weight and weight and height at the age of 2 in children born preterm. Am J Perinatol 2015; 32:591-8. [PMID: 25730133 PMCID: PMC4485423 DOI: 10.1055/s-0035-1544947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE The aim of the study was to evaluate associations between fetal growth and weight at 2 years in infants born preterm using a customized approach for birth weight. STUDY DESIGN This is a secondary analysis of a multicenter trial that included a 2-year follow-up of children born prematurely. Customized birth weight percentiles were calculated using the Gardosi model for a U.S. population, and the relation between customized percentile and weight and height at 2 years (adjusted for gender using z-score) was determined using regression analysis and by comparing z-scores for children with birth weight <10th versus ≥10th percentile. RESULTS Weight z-score at 2 years was significantly lower in the <10th than in the ≥10th percentile group (median [interquartile range, IQR]: -0.66 [-1.58, -0.01] vs. -0.23 [-1.05, 0.55]; p < 0.001), and remained after adjusting for maternal education (p < 0.001). A similar relationship was noted for height z-score between groups (median [IQR]: -0.56 [-1.29, 0.19] vs. -0.24 [-0.99, 0.37]; p < 0.001). Positive relationships between customized birth weight percentile and weight and height at 2 years were noted (p < 0.001 for both), but were not strong (R (2) = 0.04 and 0.02, respectively). CONCLUSION Customized birth weight percentile is a minor determinant of weight at 2 years among children born preterm.
Collapse
Affiliation(s)
- Gayle Olson
- Departments of Obstetrics and Gynecology of the University of Texas Medical Branch Galveston, Galveston, TX
| | - Steven J. Weiner
- George Washington University Biostatistics Center, Washington, DC
| | | | - Uma M. Reddy
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, M.D
| | - Brian M. Mercer
- Case Western Reserve University-MetroHealth Medical Center, Cleveland, OH and University of Tennessee, Memphis, TN
| | | | | | | | - Ronald J. Wapner
- Thomas Jefferson University and Drexel University, Philadelphia, PA
| | - Susan M. Ramin
- University of Texas Health Science Center at Houston, Houston, TX
| | | | | | | | | | | | | |
Collapse
|
38
|
Pham H, Duy AP, Pansiot J, Bollen B, Gallego J, Charriaut-Marlangue C, Baud O. Impact of inhaled nitric oxide on white matter damage in growth-restricted neonatal rats. Pediatr Res 2015; 77:563-9. [PMID: 25580736 DOI: 10.1038/pr.2015.4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 09/30/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND Fetal growth restriction is the second leading cause of perinatal morbidity and mortality, and neonates with intrauterine growth retardation (IUGR) have increased neurocognitive and neuropsychiatric morbidity. These neurocognitive impairments are mainly related to injury of the developing brain associated with IUGR. Growing evidence from preclinical models of brain injury in both adult and neonatal rodents supports the view that nitric oxide can promote neuroprotection. METHODS In a model of IUGR induced by protracted gestational hypoxia leading to diffuse white matter injury, we subjected neonatal rats to low dose (5 ppm) but long-lasting (7 d) exposure to inhaled NO (iNO). We used a combination of techniques, including immunohistochemistry, quantitative PCR, and cognitive assessment, to assess neuroprotection. RESULTS Antenatal hypoxia-induced IUGR was associated with severe neuroinflammation and delayed myelination. iNO exposure during the first postnatal week significantly attenuated cell death and microglial activation, enhanced oligodendroglial proliferation and finally improved myelination. Remarkably, iNO was associated with the specific upregulation of P27kip1, which initiates oligodendrocytic differentiation. Finally, iNO counteracted the deleterious effects of hypoxia on learning abilities. CONCLUSION This study provides new evidence that iNO could be effective in preventing brain damage and/or enhancing repair of the developing brain.
Collapse
Affiliation(s)
- Hoa Pham
- 1] INSERM UMR1141, Université Paris Diderot, Paris, France [2] PremUP Foundation, Paris, France
| | - An Phan Duy
- 1] INSERM UMR1141, Université Paris Diderot, Paris, France [2] PremUP Foundation, Paris, France
| | - Julien Pansiot
- 1] INSERM UMR1141, Université Paris Diderot, Paris, France [2] PremUP Foundation, Paris, France
| | - Bieke Bollen
- 1] INSERM UMR1141, Université Paris Diderot, Paris, France [2] PremUP Foundation, Paris, France
| | - Jorge Gallego
- 1] INSERM UMR1141, Université Paris Diderot, Paris, France [2] PremUP Foundation, Paris, France
| | | | - Olivier Baud
- 1] INSERM UMR1141, Université Paris Diderot, Paris, France [2] PremUP Foundation, Paris, France [3] Neonatal Intensive Care Unit, Robert Debré Children's Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| |
Collapse
|
39
|
Abuzgaia AM, Hardy DB, Arany E. Regulation of postnatal pancreatic Pdx1 and downstream target genes after gestational exposure to protein restriction in rats. Reproduction 2015; 149:293-303. [DOI: 10.1530/rep-14-0245] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The study carried out in our laboratory demonstrated that protein restriction (low protein, LP) during fetal and neonatal life alters pancreatic development and impairs glucose tolerance later in life. In this study, we examined the role of the transcription factorPdx1, a master regulator of β-cell differentiation and function along with its downstream target genes insulin,Glut2and glucokinase (GK). The role(s) of these genes and protein products on the pancreata of male offspring from mothers exposed to LP diets were assessed during gestation, weaning, and adult life. Pregnant rats were allocated to two dietary treatments: control (C) 20% protein diet or LP, 8% protein diet. At birth, offspring were divided into four groups: C received control diet all life, LP1 received LP diet all life, LP2 changed the LP diet to C at weaning, and LP3 switched to C after being exposed to LP during gestation only. Body weights (bw) were significantly (P<0.001) decreased in all LP groups at birth. At weaning, only the LP3 offspring had their body weight restored to control levels.Pdx1or any of thePdx1-target genes were similar in all diets at day 21. However, at d130Pdx1mRNA expression and protein abundance were significantly decreased (P<0.05) in all LP groups. In addition, insulin mRNA and protein were decreased in LP1 and LP3 groups compared with C,Glut2mRNA and GLUT2 protein levels were decreased in LP3 and GK did not change between groups. Intraperitoneal glucose tolerance test revealed impaired glucose tolerance in LP3 males, concomitant with decreased β-cell mass, islet area, and PDX1 nuclear protein localization. Collectively, this study suggests that restoring proteins in the diet after birth in LP offspring dramatically impairs glucose homeostasis in early adulthood, by alteringPdx1expression and downstream-target genes increasing the risk to develop type 2 diabetes.
Collapse
|
40
|
Body composition and behaviour in adult rats are influenced by maternal diet, maternal age and high-fat feeding. J Nutr Sci 2015; 4:e3. [PMID: 26090100 PMCID: PMC4463023 DOI: 10.1017/jns.2014.64] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 10/14/2014] [Accepted: 11/04/2014] [Indexed: 01/23/2023] Open
Abstract
Fetal exposure to maternal undernutrition has lifelong consequences for physiological and
metabolic function. Maternal low-protein diet is associated with an age-related phenotype
in rats, characterised by a period of resistance to development of obesity in early
adulthood, giving way to an obesity-prone, insulin-resistant state in later adulthood.
Offspring of rats fed a control (18 % casein) or low-protein (9 % casein; LP) diet in
pregnancy were challenged with a high-fat diet at 9 months of age. To assess whether other
maternal factors modulated the programming effects of nutrition, offspring were studied
from young (2–4 months old) and older (6–9 months old) mothers. Weight gain with a
high-fat diet was attenuated in male offspring of older mothers fed LP (interaction of
maternal age and diet; P = 0·011) and adipose tissue deposition was lower
with LP feeding in both males and females (P < 0·05). Although the
resistance to weight gain and adiposity was partially explained by lower energy intake in
offspring of LP mothers (P < 0·001 males only), it was apparent
that energy expenditure must be influenced by maternal diet and age. Assessment of
locomotor activity indicated that energy expenditure associated with physical activity was
unlikely to explain resistance to weight gain, but showed that offspring of older mothers
were more anxious than those of younger mothers, with more rearing observed in a novel
environment and on the elevated plus-maze. The data showed that in addition to maternal
undernutrition, greater maternal age may influence development and long-term body
composition in the rat.
Collapse
|
41
|
Sex-specific effects of low protein diet on in utero programming of renal G-protein coupled receptors. J Dev Orig Health Dis 2015; 5:36-44. [PMID: 24847689 DOI: 10.1017/s2040174413000524] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Intrauterine growth restriction (IUGR) is an important risk factor for development of hypertension, diabetes and the metabolic syndrome. Maternal low protein (LP) intake during rat pregnancy leads to IUGR in male and female offspring, although females may be resistant to the development of effect. Current evidence suggests that changes in the renin-angiotensin system (RAS) in utero contribute to this programmed hypertension, via sex-specific mechanisms. The previously orphaned G-protein coupled receptor (GPR91) was identified as a central player in the development of hypertension in adult mice, through a RAS-dependent pathway. However, whether the GPR91 pathway contributes to fetal programming is unknown. Furthermore, the nature of involvement of downstream modulators of the RAS including Gqα/11α and GαS has not been investigated in IUGR-LP rats. Therefore, we postulated that renal GPR91, in conjunction with RAS, is differentially impacted in a sex-specific manner from LP-induced IUGR rats. Pregnant Wistar rats were fed control (C, 20% protein) or LP (8% protein) diet until embryonic day 19 (E19) or postnatal d21. At E19, GPR91 protein and mRNA were increased in both male and female LP kidneys (P<0.05), whereas renin and angiotensin converting enzyme (ACE) were only increased in males (P=0.06 and P<0.05, respectively). On d21, AT1R and Gqα/11α were increased in LP males, while in LP females, AT2R protein was elevated and renin expression was decreased (P<0.05). This study demonstrates that in IUGR-LP rats, up regulation of GPR91 in fetal kidney is mirrored by increased ACE and renin in males. These in utero alterations, when combined with postnatal increases in AT1R-Gqα/11α specifically in male offspring, may predispose to the development of hypertension.
Collapse
|
42
|
Ngo HT, Hetland RB, Steffensen IL. The intrauterine and nursing period is a window of susceptibility for development of obesity and intestinal tumorigenesis by a high fat diet in Min/+ mice as adults. J Obes 2015; 2015:624023. [PMID: 25874125 PMCID: PMC4383426 DOI: 10.1155/2015/624023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 12/14/2014] [Accepted: 12/16/2014] [Indexed: 11/18/2022] Open
Abstract
We studied how obesogenic conditions during various life periods affected obesity and intestinal tumorigenesis in adult C57BL/6J-Min (multiple intestinal neoplasia)/+ mice. The mice were given a 10% fat diet throughout life (negative control) or a 45% fat diet in utero, during nursing, during both in utero and nursing, during adult life, or during their whole life-span, and terminated at 11 weeks for tumorigenesis (Min/+) or 23 weeks for obesogenic effect (wild-type). Body weight at 11 weeks was increased after a 45% fat diet during nursing, during both in utero and nursing, and throughout life, but had normalized at 23 weeks. In the glucose tolerance test, the early exposure to a 45% fat diet in utero, during nursing, or during both in utero and nursing, did not affect blood glucose, whereas a 45% fat diet given to adults or throughout life did. However, a 45% fat diet during nursing or during in utero and nursing increased the number of small intestinal tumors. So did exposures to a 45% fat diet in adult life or throughout life, but without increasing the tumor numbers further. The intrauterine and nursing period is a window of susceptibility for dietary fat-induced obesity and intestinal tumor development.
Collapse
Affiliation(s)
- Ha Thi Ngo
- Department of Food, Water and Cosmetics, Division of Environmental Medicine, Norwegian Institute of Public Health, P.O. Box 4404 Nydalen, 0403 Oslo, Norway
| | - Ragna Bogen Hetland
- Department of Food, Water and Cosmetics, Division of Environmental Medicine, Norwegian Institute of Public Health, P.O. Box 4404 Nydalen, 0403 Oslo, Norway
| | - Inger-Lise Steffensen
- Department of Food, Water and Cosmetics, Division of Environmental Medicine, Norwegian Institute of Public Health, P.O. Box 4404 Nydalen, 0403 Oslo, Norway
- *Inger-Lise Steffensen:
| |
Collapse
|
43
|
Santangeli L, Sattar N, Huda SS. Impact of maternal obesity on perinatal and childhood outcomes. Best Pract Res Clin Obstet Gynaecol 2014; 29:438-48. [PMID: 25497183 DOI: 10.1016/j.bpobgyn.2014.10.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 10/24/2014] [Indexed: 12/15/2022]
Abstract
Maternal obesity is of major consequence, affecting every aspect of maternity care including both short- and long-term effects on the health of the offspring. Obese mothers are at a higher risk of developing gestational diabetes and pre-eclampsia, potentially exposing the foetus to an adverse intrauterine environment. Maternal obesity is linked to foetal macrosomia, resulting in increased neonatal and maternal morbidity. Foetal macrosomia is a result of a change in body composition in the neonate with an increase in both percentage fat and fat mass. Maternal obesity and gestational weight gain are associated with childhood obesity, and this effect extends into adulthood. Childhood obesity in turn increases chances of later life obesity, thus type 2 diabetes, and cardiovascular disease in the offspring. Further clinical trials of lifestyle and, potentially, pharmacological interventions in obese pregnant women are required to determine whether short- and long-term adverse effects for the mother and child can be reduced.
Collapse
Affiliation(s)
- Louise Santangeli
- ST7 Obstetrics and Gynaecology Speciality Doctor, Wishaw General Hospital, Lanarkshire, Glasgow, UK.
| | - Naveed Sattar
- Professor of Metabolic Medicine, Institute of Cardiovascular and Medical Sciences, RC214 Level C2, Institute of C&MS, BHF GCRC, Glasgow G12 8TA, UK.
| | - Shahzya S Huda
- Consultant Obstetrician and Gynaecologist and Honorary Senior Clinical Lecturer, Women and Children, Forth Valley Royal Hospital, Larbert, UK.
| |
Collapse
|
44
|
Amirian M, Sajadi E, Rostami P, Chaloosi M. Effect of prenatal stress (immobilization) on blood glucose levels and body weight. Int J Diabetes Dev Ctries 2014. [DOI: 10.1007/s13410-014-0214-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
45
|
Lopes GAD, Fan WYC, Ciol H, Bidinotto LT, Rodrigues MAM, Barbisan LF. Maternal western style diet increases susceptibility to chemically-induced mammary carcinogenesis in female rats offspring. Nutr Cancer 2014; 66:1293-303. [PMID: 25333700 DOI: 10.1080/01635581.2014.956256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The present study investigated whether maternal exposure to western style diet (WD) increases susceptibility to mammary carcinogenesis induced by N-methyl-N-nitrosourea (MNU) in female offspring. Pregnant female Sprague-Dawley rats received WD diet or control diet from gestational day 12 until postnatal day (PND) 21. At PND 21, female offspring received a single dose of MNU (50 mg/kg body weight) and were fed chow diet until PND 110. Mammary gland structures were assessed on whole-mount preparations in the offspring at PND 21, and tumor morphology was examined at PND 110. Immunohistochemical analysis for cell proliferation (PCNA), apoptosis (cleaved caspase-3) and estrogen receptor alpha (ER-α) was performed in mammary terminal end buds (TEBs) at PND 21, and PCNA, ER-α, and p63 analysis in mammary tumors at PND 110. Maternal WD intake induced a significant increase in the number of TEBs (P = 0.024) and in PCNA labeling index (P < 0.020) in the mammary glands at PND 21. Tumor multiplicity, tumor weight, and PCNA labeling indexes were significantly higher in the WD offspring than that of the control offspring (P < 0.05). These findings indicate that maternal western style diet potentially enhanced the development of mammary tumors induced by MNU in female offspring, possibly by affecting the mammary gland differentiation.
Collapse
Affiliation(s)
- Gisele A D Lopes
- a Department of Pathology, Botucatu Medical School , São Paulo State University , São Paulo , Brazil
| | | | | | | | | | | |
Collapse
|
46
|
Small for gestational age and adulthood risk of disability pension: The contribution of childhood and adulthood conditions. Soc Sci Med 2014; 119:249-57. [DOI: 10.1016/j.socscimed.2013.11.052] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 08/22/2013] [Accepted: 11/20/2013] [Indexed: 11/22/2022]
|
47
|
Sex-specific impact of maternal-fetal risk factors on depression and cardiovascular risk 40 years later. J Dev Orig Health Dis 2014; 2:353-64. [PMID: 23378891 DOI: 10.1017/s2040174411000651] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Major depressive disorder (MDD) and cardiovascular disease (CVD) represent leading causes of morbidity and mortality worldwide. We tested the hypothesis that growth restriction and preeclampsia (referred to as fetal risk) are significant predictors of these conditions, with women at higher risk in adulthood. Adult offspring exposed to fetal risk factors and their discordant siblings were from two prenatal cohorts, whose mothers were followed through pregnancy and whom we recruited as adults 40 years later (n = 538; 250 males and 288 females). Subjects were psychiatrically diagnosed and underwent a stress challenge during which parasympathetic regulation was assessed by electrocardiogram, operationalized as high-frequency R-R interval variability (HF-RRV). Linear mixed models and generalized estimating equations were used to examine the relationship of fetal risk on HF-RRV, MDD and comorbidity of low HF-RRV (lowest 25th percentile) and MDD, including interactions with sex and socioeconomic status (SES). Fetal risk was significantly associated with low HF-RRV response (F = 3.64, P = 0.05), particularly among low SES (interaction: F = 4.31, P < 0.04). When stratified by MDD, the fetal risk impact was three times greater among MDD compared with non-MDD subjects (effect size: 0.21 v. 0.06). Females had a significantly higher risk for the comorbidity of MDD and low HF-RRV than males (relative risk (RR) = 1.36, 95% CI: 1.07-1.73), an association only seen among those exposed to fetal risk (RR = 1.38, 95% CI: 1.04-1.83). Findings suggest that these are shared fetal antecedents to the comorbidity of MDD and CVD risk 40 years later, an association stronger in females than in males.
Collapse
|
48
|
Serum βhCG and Lipid Profile in Early Second Trimester as Predictors of Pregnancy-Induced Hypertension. J Obstet Gynaecol India 2013; 64:169-74. [PMID: 24966499 DOI: 10.1007/s13224-013-0490-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 11/04/2013] [Indexed: 10/25/2022] Open
Abstract
OBJECTIVES A variety of biological, biochemical, and biophysical markers implicated in the pathophysiology of pre-eclampsia during the last two decades have instigated the growing interest in this study to include both βhCG and lipid profile studies in the early second trimester as early predictors of pregnancy-induced hypertension. Early identification of at-risk women may help in taking timely preventive and curative management to prevent or delay complications associated with pregnancy-induced hypertension. METHOD A prospective study was performed on 120 patients attending the outpatient department of the Obstetrics and Gynaecology of the Maharaja Agrasen Hospital. All the patients were screened for serum βhCG and serum lipid profile in their early second trimester (14-20 weeks) and followed up till their delivery. Comparative studies of serum βhCG and serum lipid profile were performed between those who remain normotensive (group I) and those who developed pregnancy-induced hypertension (group II). RESULTS TG, total cholesterol, VLDL, and LDL values for those women who developed PIH (group II) were significantly higher than those who remain normotensive (group I), with p value of <0.05 which is statistically significant. HDL and βhCG values for group II were not higher than those in group I with p value >0.05 which is statistically insignificant. CONCLUSION Maternal lipid profile in second trimester is very good noninvasive test which can be used for prediction of pregnancy-induced hypertension before its clinical onset. However, there is no correlation between maternal serum βhCG and pregnancy-induced hypertension.
Collapse
|
49
|
Karbalaei N, Ghasemi A, Hedayati M, Godini A, Zahediasl S. The possible mechanisms by which maternal hypothyroidism impairs insulin secretion in adult male offspring in rats. Exp Physiol 2013; 99:701-14. [DOI: 10.1113/expphysiol.2013.073825] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Narges Karbalaei
- Department of Physiology; Faculty of Medicine; Shiraz University of Medical Sciences; Shiraz Iran
| | | | - Mehdi Hedayati
- Cellular & Molecular Endocrine Research Center; Research Institute for Endocrine Sciences; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Aliashraf Godini
- Department of Physiology; Faculty of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | | |
Collapse
|
50
|
Phan Duy A, El Khabbaz F, Renolleau C, Aberchich J, Heneau A, Pham H, Baud O. Retard de croissance intra-utérin et cerveau en développement. Arch Pediatr 2013; 20:1034-8. [DOI: 10.1016/j.arcped.2013.06.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 06/17/2013] [Indexed: 10/26/2022]
|