1
|
Dumolt JH, Rosario FJ, Barentsen K, Urschitz J, Powell TL, Jansson T. Trophoblast-specific overexpression of adiponectin receptor 2 causes fetal growth restriction in pregnant mice. FASEB J 2024; 38:e70100. [PMID: 39387608 PMCID: PMC11508969 DOI: 10.1096/fj.202302143r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 08/07/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024]
Abstract
Maternal obesity in pregnancy is strongly associated with complications such as fetal overgrowth and infants of obese mothers have an increased risk to develop obesity, diabetes, and cardiovascular disease later in life. However, the underlying mechanisms are not well established. Circulating levels of adiponectin are low in obese pregnant women and maternal circulating adiponectin is negatively associated with birth weight. We have reported that normalizing maternal adiponectin in obese pregnant mice prevents placental dysfunction, fetal overgrowth, and programming of offspring cardio-metabolic disease. However, the mechanistic link between maternal adiponectin, placental function, and fetal growth remains to be established. We hypothesized that trophoblast-specific overexpression of the adiponectin receptor 2 (Adipor2) in healthy pregnant mice inhibits placental mTORC1 signaling and nutrient transport, resulting in fetal growth restriction. Using lentiviral transduction of blastocysts with a mammalian gene expression lentiviral vector for up-regulation of Adipor2 (Adipor2-OX), we achieved a ~ 3-fold increase in placenta Adipor2 mRNA levels and a 2-fold increase of the ADIPOR2 protein in the trophoblast plasma membrane. Placenta-specific Adipor2-OX increased placental peroxisome proliferator-activated receptor-α phosphorylation, ceramide synthase expression and ceramide concentrations. Furthermore, Adipor2-OX inhibited placental mTORC1 signaling and reduced in vivo placental transport of glucose and amino acids. Lastly, Adipor2-OX reduced fetal weight by 11%. These data provide mechanistic evidence that placental Adipor2 signaling directly affects fetal growth. We propose that low circulating adiponectin in maternal obesity causes fetal overgrowth and programs the offspring for cardio-metabolic disease mediated by a direct effect on placental function.
Collapse
Affiliation(s)
- Jerad H Dumolt
- Division of Reproductive Sciences, Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Fredrick J Rosario
- Division of Reproductive Sciences, Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kenneth Barentsen
- Division of Reproductive Sciences, Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Johann Urschitz
- Division of Reproductive Sciences, Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Institute for Biogenesis Research, University of Hawaii, Honolulu, HI, USA
| | - Theresa L Powell
- Division of Reproductive Sciences, Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
2
|
Thornburg KL, Valent AM. Maternal Malnutrition and Elevated Disease Risk in Offspring. Nutrients 2024; 16:2614. [PMID: 39203750 PMCID: PMC11357549 DOI: 10.3390/nu16162614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/31/2024] [Accepted: 08/03/2024] [Indexed: 09/03/2024] Open
Abstract
US populations have seen dramatic increases in the prevalence of chronic disease over the past three generations. Rapid increases in type 2 diabetes and obesity have occurred in all the states but have been particularly striking in the Deep South. These increases have contributed to decreases in life expectancy and to painful elevations in health care costs. The causes of worsening population health are complex and incompletely understood. However, there is strong evidence that vulnerability to chronic conditions is determined in early life. Most chronic diseases are developmentally driven. There are specific stressors experienced in early life that influence epigenetic and structural changes during development. These include malnutrition, severe levels of social stress, toxic chemicals, and low oxygen levels. Most US populations have experienced a decrease in the quality of the food they consume as industrial foods have replaced garden-grown foods. Thus, the consumption of too few nutrients before and during pregnancy and during lactation influences the growth of the placenta and fetal organs and their level of resilience when faced with stresses in postnatal life and particularly as adults. Animal studies have shown that the effects of poor nutrition can be passed on to future generations. The most powerful way that the current epidemics of obesity and insulin resistance can be reversed is by providing key nutrients to prospective mothers and those already pregnant.
Collapse
Affiliation(s)
- Kent L. Thornburg
- OHSU Bob and Charlee Moore Institute for Nutrition and Wellness, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA;
- Center for Developmental Health, Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Medicine, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amy M. Valent
- OHSU Bob and Charlee Moore Institute for Nutrition and Wellness, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA;
- Department of Obstetrics & Gynecology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
3
|
Sullivan EL, Molloy KR, Dunn GA, Balanzar AL, Young AS, Loftis JM, Ablow JC, Nigg JT, Gustafsson HC. Adipokines measured during pregnancy and at birth are associated with infant negative affect. Brain Behav Immun 2024; 120:34-43. [PMID: 38772428 PMCID: PMC11401062 DOI: 10.1016/j.bbi.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 03/18/2024] [Accepted: 05/18/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Increased adiposity during pregnancy may be related to offspring risk for mental health disorders, although the biological mechanisms are poorly understood. One promising hypothesis is that factors secreted from adipocytes such as leptin and adiponectin may explain this association. The current study examined whether pregnancy or umbilical cord blood concentrations of leptin and/or adiponectin a) predict elevated infant negative affect at 6 months (an early life marker of risk for psychopathology); and b) help explain the association between pregnancy adiposity and increased infant negative affect. METHODS Data came from a prospective cohort (N = 305) of pregnant individuals and their offspring. Second trimester adiposity was assessed using air displacement plethysmography. Concentrations of leptin and adiponectin were measured in second trimester plasma and umbilical cord plasma. Infant negative affect was assessed by standardized observation at 6 months. Second trimester inflammation was assessed using a comprehensive panel of cytokines. RESULTS Lower second trimester adiponectin was associated with elevated infant negative affect, and mediated the effect of pregnancy adiposity on infant negative affect. This association was independent of the effect of second trimester inflammation. Umbilical cord leptin also predicted higher infant negative affect and mediated the association between pregnancy adiposity and infant negative affect. CONCLUSIONS This is the first study to link pregnancy adiponectin or cord blood leptin to infant markers of risk for psychopathology, and the first to demonstrate that these adipokines mediate the association between pregnancy adiposity and offspring behavioral outcomes, suggesting novel markers of risk and potential mechanisms of effect.
Collapse
Affiliation(s)
- Elinor L Sullivan
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States; Division of Neuroscience, Oregon National Primary Research Center, United States.
| | - Kelly R Molloy
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Geoffrey A Dunn
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Adriana L Balanzar
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Anna S Young
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Jennifer M Loftis
- Department of Psychiatry, Oregon Health & Science University, United States; VA Portland Health Care System, United States
| | | | - Joel T Nigg
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| | - Hanna C Gustafsson
- Department of Psychiatry, Oregon Health & Science University, United States; Center for Mental Health Innovation, Oregon Health & Science University, United States
| |
Collapse
|
4
|
De Toro V, Alberti G, Dominguez A, Carrasco-Negüe K, Ferrer P, Valenzuela R, Garmendia ML, Casanello P. Growth patterns in infants born to women with pregestational overweight/obesity supplemented with docosahexaenoic acid during pregnancy. J Pediatr Gastroenterol Nutr 2024; 79:371-381. [PMID: 38922906 DOI: 10.1002/jpn3.12294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 05/12/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Previous studies of maternal docosahexaenoic acid (DHA) supplementation during pregnancy have controversial and contrasting results on the short and long-term effects on early child growth. The impact of this nutritional intervention on the postnatal growth patterns in the offspring of women with pregestational overweight/obesity (PGO) also remains controversial. OBJECTIVE To analyze the postnatal growth patterns during the first 4 months of life in the offspring of women with PGO randomly supplemented with 800 mg/day (PGO-800) compared with normative doses of 200 mg/day (PGO-200) of DHA during pregnancy (<15 weeks of gestation until delivery). METHODS This study evaluated the growth patterns during the first 4 months of life of 169 infants of the women that participated in the MIGHT study (NCT02574767). We included the infants of women from the PGO-200 (n = 81) and PGO-800 group (n = 88). The growth patterns (weight, length, and head circumference) and change in z-score (World health Organization charts) were evaluated. RESULTS Throughout the first 4 months of life, the infants of the PGO-800 group had lower weight-for-length z-score (coef. -0.65, 95% confidence interval [CI] -1.07, -0.22, p = 0.003) and lower body mass index-for-age z-score (coef. -0.56, 95% CI -0.99, -0.12, p = 0.012) compared with the PGO-200 group adjusted by maternal body mass index, gestational weight gain, gestational age, insulin in cord blood and infant feeding (exclusive breastfed, not breastfed, and partially breastfed). CONCLUSIONS Maternal supplementation with DHA during pregnancy could beneficially limit the offspring's postnatal weight gain during the first 4 months of life.
Collapse
Affiliation(s)
- Valeria De Toro
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gigliola Alberti
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angelica Dominguez
- Department of Public Health, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karina Carrasco-Negüe
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pedro Ferrer
- Department of Nutrition, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Maria Luisa Garmendia
- Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Santiago, Chile
| | - Paola Casanello
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Neonatology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
5
|
Golden TN, Mani S, Linn RL, Leite R, Trigg NA, Wilson A, Anton L, Mainigi M, Conine CC, Kaufman BA, Strauss JF, Parry S, Simmons RA. Extracellular vesicles alter trophoblast function in pregnancies complicated by COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.17.580824. [PMID: 38464046 PMCID: PMC10925147 DOI: 10.1101/2024.02.17.580824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and resulting coronavirus disease (COVID-19) causes placental dysfunction, which increases the risk of adverse pregnancy outcomes. While abnormal placental pathology resulting from COVID-19 is common, direct infection of the placenta is rare. This suggests that pathophysiology associated with maternal COVID-19, rather than direct placental infection, is responsible for placental dysfunction and alteration of the placental transcriptome. We hypothesized that maternal circulating extracellular vesicles (EVs), altered by COVID-19 during pregnancy, contribute to placental dysfunction. To examine this hypothesis, we characterized maternal circulating EVs from pregnancies complicated by COVID-19 and tested their effects on trophoblast cell physiology in vitro . We found that the gestational timing of COVID-19 is a major determinant of circulating EV function and cargo. In vitro trophoblast exposure to EVs isolated from patients with an active infection at the time of delivery, but not EVs isolated from Controls, altered key trophoblast functions including hormone production and invasion. Thus, circulating EVs from participants with an active infection, both symptomatic and asymptomatic cases, can disrupt vital trophoblast functions. EV cargo differed between participants with COVID-19 and Controls, which may contribute to the disruption of the placental transcriptome and morphology. Our findings show that COVID-19 can have effects throughout pregnancy on circulating EVs and circulating EVs are likely to participate in placental dysfunction induced by COVID-19.
Collapse
|
6
|
Dawid M, Pich K, Mlyczyńska E, Respekta-Długosz N, Wachowska D, Greggio A, Szkraba O, Kurowska P, Rak A. Adipokines in pregnancy. Adv Clin Chem 2024; 121:172-269. [PMID: 38797542 DOI: 10.1016/bs.acc.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Reproductive success consists of a sequential events chronology, starting with the ovum fertilization, implantation of the embryo, placentation, and cellular processes like proliferation, apoptosis, angiogenesis, endocrinology, or metabolic changes, which taken together finally conduct the birth of healthy offspring. Currently, many factors are known that affect the regulation and proper maintenance of pregnancy in humans, domestic animals, or rodents. Among the determinants of reproductive success should be distinguished: the maternal microenvironment, genes, and proteins as well as numerous pregnancy hormones that regulate the most important processes and ensure organism homeostasis. It is well known that white adipose tissue, as the largest endocrine gland in our body, participates in the synthesis and secretion of numerous hormones belonging to the adipokine family, which also may regulate the course of pregnancy. Unfortunately, overweight and obesity lead to the expansion of adipose tissue in the body, and its excess in both women and animals contributes to changes in the synthesis and release of adipokines, which in turn translates into dramatic changes during pregnancy, including those taking place in the organ that is crucial for the proper progress of pregnancy, i.e. the placenta. In this chapter, we are summarizing the current knowledge about levels of adipokines and their role in the placenta, taking into account the physiological and pathological conditions of pregnancy, e.g. gestational diabetes mellitus, preeclampsia, or intrauterine growth restriction in humans, domestic animals, and rodents.
Collapse
Affiliation(s)
- Monika Dawid
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Karolina Pich
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Ewa Mlyczyńska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Natalia Respekta-Długosz
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Dominka Wachowska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Aleksandra Greggio
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Oliwia Szkraba
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Patrycja Kurowska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Agnieszka Rak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland.
| |
Collapse
|
7
|
Lizárraga D, Gómez-Gil B, García-Gasca T, Ávalos-Soriano A, Casarini L, Salazar-Oroz A, García-Gasca A. Gestational diabetes mellitus: genetic factors, epigenetic alterations, and microbial composition. Acta Diabetol 2024; 61:1-17. [PMID: 37660305 DOI: 10.1007/s00592-023-02176-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/18/2023] [Indexed: 09/05/2023]
Abstract
Gestational diabetes mellitus (GDM) is a common metabolic disorder, usually diagnosed during the third trimester of pregnancy that usually disappears after delivery. In GDM, the excess of glucose, fatty acids, and amino acids results in foetuses large for gestational age. Hyperglycaemia and insulin resistance accelerate the metabolism, raising the oxygen demand, and creating chronic hypoxia and inflammation. Women who experienced GDM and their offspring are at risk of developing type-2 diabetes, obesity, and other metabolic or cardiovascular conditions later in life. Genetic factors may predispose the development of GDM; however, they do not account for all GDM cases; lifestyle and diet also play important roles in GDM development by modulating epigenetic signatures and the body's microbial composition; therefore, this is a condition with a complex, multifactorial aetiology. In this context, we revised published reports describing GDM-associated single-nucleotide polymorphisms (SNPs), DNA methylation and microRNA expression in different tissues (such as placenta, umbilical cord, adipose tissue, and peripheral blood), and microbial composition in the gut, oral cavity, and vagina from pregnant women with GDM, as well as the bacterial composition of the offspring. Altogether, these reports indicate that a number of SNPs are associated to GDM phenotypes and may predispose the development of the disease. However, extrinsic factors (lifestyle, nutrition) modulate, through epigenetic mechanisms, the risk of developing the disease, and some association exists between the microbial composition with GDM in an organ-specific manner. Genes, epigenetic signatures, and microbiota could be transferred to the offspring, increasing the possibility of developing chronic degenerative conditions through postnatal life.
Collapse
Affiliation(s)
- Dennise Lizárraga
- Laboratory of Molecular and Cell Biology, Centro de Investigación en Alimentación y Desarrollo, Avenida Sábalo Cerritos s/n, 82112, Mazatlán, Sinaloa, Mexico
| | - Bruno Gómez-Gil
- Laboratory of Microbial Genomics, Centro de Investigación en Alimentación y Desarrollo, Avenida Sábalo Cerritos s/n, 82112, Mazatlán, Sinaloa, Mexico
| | - Teresa García-Gasca
- Laboratory of Molecular and Cellular Biology, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Avenida de las Ciencias s/n, 76230, Juriquilla, Querétaro, Mexico
| | - Anaguiven Ávalos-Soriano
- Laboratory of Molecular and Cell Biology, Centro de Investigación en Alimentación y Desarrollo, Avenida Sábalo Cerritos s/n, 82112, Mazatlán, Sinaloa, Mexico
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, via G. Campi 287, 41125, Modena, Italy
| | - Azucena Salazar-Oroz
- Maternal-Fetal Department, Instituto Vidalia, Hospital Sharp Mazatlán, Avenida Rafael Buelna y Dr. Jesús Kumate s/n, 82126, Mazatlán, Sinaloa, Mexico
| | - Alejandra García-Gasca
- Laboratory of Molecular and Cell Biology, Centro de Investigación en Alimentación y Desarrollo, Avenida Sábalo Cerritos s/n, 82112, Mazatlán, Sinaloa, Mexico.
| |
Collapse
|
8
|
Zhu M, Lv Y, Peng Y, Wu Y, Feng Y, Jia T, Xu S, Li S, Wang W, Tian J, Sun L. GCKR and ADIPOQ gene polymorphisms in women with gestational diabetes mellitus. Acta Diabetol 2023; 60:1709-1718. [PMID: 37524927 PMCID: PMC10587232 DOI: 10.1007/s00592-023-02165-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/18/2023] [Indexed: 08/02/2023]
Abstract
AIMS To investigate the associations of GCKR and ADIPOQ variants with the risk of gestational diabetes mellitus (GDM) in Chinese women. METHODS GCKR rs1260326, ADIPOQ rs266729, and rs1501299 were selected and genotyped in 519 GDM patients and 498 controls. Candidate SNPs were genotyped using multiplex polymerase chain reaction (PCR) combined with next-generation sequencing methods, and the association of these SNPs with GDM was analyzed. RESULTS We found that GCKR rs1260326 was significantly associated with an increased risk of GDM in the allele model, the codominant model (CC vs. TT), the dominant model, the recessive model, and the genotypic model distributions (p = 0.0029, p = 0.0022, p = 0.0402, p = 0.0038, and p = 0.0028, respectively). The rs1260326 polymorphism was shown to be associated with 1 h-OGTT level and gravidity in GDM patients (CC vs. TT: p = 0.0475 and p = 0.0220, respectively). Diastolic blood pressure (DBP) was significantly higher in the GDM patients with the rs266729 GG genotype compared to those with the CC or CG genotype (p = 0.0444 and p = 0.0339, respectively). The DBP of the GDM patients with the rs1501299 GT genotype was lower than that of those with the GG genotype (p = 0.0197). There was a weak linkage disequilibrium value between the GCKR and ADIPOQ SNPs. CONCLUSIONS The genes GCKR and ADIPOQ may be involved in the pathophysiology of GDM.
Collapse
Affiliation(s)
- Manning Zhu
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yaer Lv
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yanqing Peng
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yingnan Wu
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yanan Feng
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Tianshuang Jia
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Songcheng Xu
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Songxue Li
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wei Wang
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiawei Tian
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
Starodubtseva NL, Tokareva AO, Volochaeva MV, Kononikhin AS, Brzhozovskiy AG, Bugrova AE, Timofeeva AV, Kukaev EN, Tyutyunnik VL, Kan NE, Frankevich VE, Nikolaev EN, Sukhikh GT. Quantitative Proteomics of Maternal Blood Plasma in Isolated Intrauterine Growth Restriction. Int J Mol Sci 2023; 24:16832. [PMID: 38069155 PMCID: PMC10706154 DOI: 10.3390/ijms242316832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/17/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Intrauterine growth restriction (IUGR) remains a significant concern in modern obstetrics, linked to high neonatal health problems and even death, as well as childhood disability, affecting adult quality of life. The role of maternal and fetus adaptation during adverse pregnancy is still not completely understood. This study aimed to investigate the disturbance in biological processes associated with isolated IUGR via blood plasma proteomics. The levels of 125 maternal plasma proteins were quantified by liquid chromatography-multiple reaction monitoring mass spectrometry (LC-MRM MS) with corresponding stable isotope-labeled peptide standards (SIS). Thirteen potential markers of IUGR (Gelsolin, Alpha-2-macroglobulin, Apolipoprotein A-IV, Apolipoprotein B-100, Apolipoprotein(a), Adiponectin, Complement C5, Apolipoprotein D, Alpha-1B-glycoprotein, Serum albumin, Fibronectin, Glutathione peroxidase 3, Lipopolysaccharide-binding protein) were found to be inter-connected in a protein-protein network. These proteins are involved in plasma lipoprotein assembly, remodeling, and clearance; lipid metabolism, especially cholesterol and phospholipids; hemostasis, including platelet degranulation; and immune system regulation. Additionally, 18 proteins were specific to a particular type of IUGR (early or late). Distinct patterns in the coagulation and fibrinolysis systems were observed between isolated early- and late-onset IUGR. Our findings highlight the complex interplay of immune and coagulation factors in IUGR and the differences between early- and late-onset IUGR and other placenta-related conditions like PE. Understanding these mechanisms is crucial for developing targeted interventions and improving outcomes for pregnancies affected by IUGR.
Collapse
Affiliation(s)
- Natalia L. Starodubtseva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.O.T.); (M.V.V.); (A.S.K.); (A.G.B.); (A.E.B.); (A.V.T.); (E.N.K.); (V.L.T.); (N.E.K.); (V.E.F.); (G.T.S.)
- Moscow Institute of Physics and Technology, 141700 Moscow, Russia
| | - Alisa O. Tokareva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.O.T.); (M.V.V.); (A.S.K.); (A.G.B.); (A.E.B.); (A.V.T.); (E.N.K.); (V.L.T.); (N.E.K.); (V.E.F.); (G.T.S.)
| | - Maria V. Volochaeva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.O.T.); (M.V.V.); (A.S.K.); (A.G.B.); (A.E.B.); (A.V.T.); (E.N.K.); (V.L.T.); (N.E.K.); (V.E.F.); (G.T.S.)
| | - Alexey S. Kononikhin
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.O.T.); (M.V.V.); (A.S.K.); (A.G.B.); (A.E.B.); (A.V.T.); (E.N.K.); (V.L.T.); (N.E.K.); (V.E.F.); (G.T.S.)
| | - Alexander G. Brzhozovskiy
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.O.T.); (M.V.V.); (A.S.K.); (A.G.B.); (A.E.B.); (A.V.T.); (E.N.K.); (V.L.T.); (N.E.K.); (V.E.F.); (G.T.S.)
| | - Anna E. Bugrova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.O.T.); (M.V.V.); (A.S.K.); (A.G.B.); (A.E.B.); (A.V.T.); (E.N.K.); (V.L.T.); (N.E.K.); (V.E.F.); (G.T.S.)
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Angelika V. Timofeeva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.O.T.); (M.V.V.); (A.S.K.); (A.G.B.); (A.E.B.); (A.V.T.); (E.N.K.); (V.L.T.); (N.E.K.); (V.E.F.); (G.T.S.)
| | - Evgenii N. Kukaev
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.O.T.); (M.V.V.); (A.S.K.); (A.G.B.); (A.E.B.); (A.V.T.); (E.N.K.); (V.L.T.); (N.E.K.); (V.E.F.); (G.T.S.)
- V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Victor L. Tyutyunnik
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.O.T.); (M.V.V.); (A.S.K.); (A.G.B.); (A.E.B.); (A.V.T.); (E.N.K.); (V.L.T.); (N.E.K.); (V.E.F.); (G.T.S.)
| | - Natalia E. Kan
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.O.T.); (M.V.V.); (A.S.K.); (A.G.B.); (A.E.B.); (A.V.T.); (E.N.K.); (V.L.T.); (N.E.K.); (V.E.F.); (G.T.S.)
| | - Vladimir E. Frankevich
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.O.T.); (M.V.V.); (A.S.K.); (A.G.B.); (A.E.B.); (A.V.T.); (E.N.K.); (V.L.T.); (N.E.K.); (V.E.F.); (G.T.S.)
- Laboratory of Translational Medicine, Siberian State Medical University, 634050 Tomsk, Russia
| | - Evgeny N. Nikolaev
- V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Gennady T. Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.O.T.); (M.V.V.); (A.S.K.); (A.G.B.); (A.E.B.); (A.V.T.); (E.N.K.); (V.L.T.); (N.E.K.); (V.E.F.); (G.T.S.)
| |
Collapse
|
10
|
Miturski A, Gęca T, Stupak A, Kwaśniewski W, Semczuk-Sikora A. Influence of Pre-Pregnancy Obesity on Carbohydrate and Lipid Metabolism with Selected Adipokines in the Maternal and Fetal Compartment. Nutrients 2023; 15:2130. [PMID: 37432262 DOI: 10.3390/nu15092130] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 07/12/2023] Open
Abstract
A higher body mass index (BMI) before pregnancy is associated with an increased risk of maternal and perinatal complications. This study aimed to analyze selected parameters of carbohydrate and lipid metabolism, including adipokines, in obese pre-pregnant women, and their influence on the birth weight of newborns. MATERIALS AND METHODS The study group (O) consisted of 34 pregnant women with higher BMI (obese) before pregnancy. The control group (C) was 27 pregnant women with target BMI and physiological pregnancy. The BMI index: body weight [kg]/(height [m]2 was assessed on the first obstetrical visit. The research material was the serum of pregnant women collected in the third trimester of pregnancy and umbilical cord blood collected immediately after delivery. Selected parameters of carbohydrate and lipid metabolism and adipokines were determined. RESULTS There were no statistically significant differences between the study group and the control group concerning the concentrations of insulin, glucose, VLDL, adiponectin, TNF-α, HOMA-IR, as well as LDH and cholesterol in maternal blood serum and umbilical cord blood serum. Total cholesterol and HDL in both maternal blood serum and umbilical cord blood were statistically significantly lower than those in the control group. The concentration of triglycerides (TG) and resistin in the blood serum of obese mothers were higher than those in the control group (p < 0.05). However, no statistically significant differences were found between the two groups regarding the concentrations of TG and resistin in the umbilical cord blood. The concentration of LDL cholesterol in the umbilical blood serum in the obese group was statistically significantly lower than that in the control group. The concentration of leptin in maternal blood serum and umbilical cord blood serum in the study group was statistically significantly higher than that in the control group. CONCLUSIONS Pregestational obesity does not substantially affect the basic parameters of carbohydrate metabolism in pregnant women, but it disturbs the lipid profile, which is manifested by a significant increase in triglycerides and a decrease in the level of HDL cholesterol in the serum. Preexisting obesity increases the concentration of leptin and resistin in the serum of pregnant women, which may be caused by the increased volume of adipose tissue. The concentrations of leptin and resistin in the blood of pregnant women correlate positively, and the concentrations of adiponectin and TNF-α negatively correlate with pre-pregnancy BMI values. There is a positive correlation between the concentration of leptin in the serum of umbilical cord blood and the birth weight of the newborn, which suggests that this parameter contributes to the pathomechanism of macrosomia.
Collapse
Affiliation(s)
- Andrzej Miturski
- Department of Gynaecology, 1st Clinical Military Hospital in Lublin, Al. Racławickie 23, 20-049 Lublin, Poland
| | - Tomasz Gęca
- Chair and Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, Staszica 16 Street, 20-081 Lublin, Poland
| | - Aleksandra Stupak
- Chair and Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, Staszica 16 Street, 20-081 Lublin, Poland
| | - Wojciech Kwaśniewski
- Department of Gynecologic Oncology and Gynecology, Medical University of Lublin, Staszica 16 Street, 20-081 Lublin, Poland
| | - Anna Semczuk-Sikora
- Chair and Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, Staszica 16 Street, 20-081 Lublin, Poland
| |
Collapse
|
11
|
Donato J. Programming of metabolism by adipokines during development. Nat Rev Endocrinol 2023:10.1038/s41574-023-00828-1. [PMID: 37055548 DOI: 10.1038/s41574-023-00828-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 04/15/2023]
Abstract
The intrauterine and early postnatal periods represent key developmental stages in which an organism is highly susceptible to being permanently influenced by maternal factors and nutritional status. Strong evidence indicates that either undernutrition or overnutrition during development can predispose individuals to disease later in life, especially type 2 diabetes mellitus and obesity, a concept known as metabolic programming. Adipose tissue produces important signalling molecules that control energy and glucose homeostasis, including leptin and adiponectin. In addition to their well-characterized metabolic effects in adults, adipokines have been associated with metabolic programming by affecting different aspects of development. Therefore, alterations in the secretion or signalling of adipokines, caused by nutritional insults in early life, might lead to metabolic diseases in adulthood. This Review summarizes and discusses the potential role of several adipokines in inducing metabolic programming through their effects during development. The identification of the endocrine factors that act in early life to permanently influence metabolism represents a key step in understanding the mechanisms behind metabolic programming. Thus, future strategies aiming to prevent and treat these metabolic diseases can be designed, taking into consideration the relationship between adipokines and the developmental origins of health and disease.
Collapse
Affiliation(s)
- Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
12
|
Hojeij B, Rousian M, Sinclair KD, Dinnyes A, Steegers-Theunissen RPM, Schoenmakers S. Periconceptional biomarkers for maternal obesity: a systematic review. Rev Endocr Metab Disord 2023; 24:139-175. [PMID: 36520252 PMCID: PMC10023635 DOI: 10.1007/s11154-022-09762-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2022] [Indexed: 12/23/2022]
Abstract
Periconceptional maternal obesity is linked to adverse maternal and neonatal outcomes. Identifying periconceptional biomarkers of pathways affected by maternal obesity can unravel pathophysiologic mechanisms and identify individuals at risk of adverse clinical outcomes. The literature was systematically reviewed to identify periconceptional biomarkers of the endocrine, inflammatory and one-carbon metabolic pathways influenced by maternal obesity. A search was conducted in Embase, Ovid Medline All, Web of Science Core Collection and Cochrane Central Register of Controlled Trials databases, complemented by manual search in PubMed until December 31st, 2020. Eligible studies were those that measured biomarker(s) in relation to maternal obesity, overweight/obesity or body mass index (BMI) during the periconceptional period (14 weeks preconception until 14 weeks post conception). The ErasmusAGE score was used to assess the quality of included studies. Fifty-one articles were included that evaluated over 40 biomarkers. Endocrine biomarkers associated with maternal obesity included leptin, insulin, thyroid stimulating hormone, adiponectin, progesterone, free T4 and human chorionic gonadotropin. C-reactive protein was associated with obesity as part of the inflammatory pathway, while the associated one-carbon metabolism biomarkers were folate and vitamin B12. BMI was positively associated with leptin, C-reactive protein and insulin resistance, and negatively associated with Free T4, progesterone and human chorionic gonadotropin. Concerning the remaining studied biomarkers, strong conclusions could not be established due to limited or contradictory data. Future research should focus on determining the predictive value of the optimal set of biomarkers for their use in clinical settings. The most promising biomarkers include leptin, adiponectin, human chorionic gonadotropin, insulin, progesterone and CRP.
Collapse
Affiliation(s)
- Batoul Hojeij
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Melek Rousian
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Kevin D Sinclair
- School of Biosciences, Sutton Bonnington Campus, University of Nottingham, Leicestershire, LE12 6HD, UK
| | - Andras Dinnyes
- BioTalentum Ltd., Godollo, 2100, Hungary
- Department of Cell Biology and Molecular Medicine, University of Szeged, Szeged, 6720, Hungary
- Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Godollo, 2100, Hungary
| | | | - Sam Schoenmakers
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, 3015GD, The Netherlands.
| |
Collapse
|
13
|
Moyce Gruber BL, Dolinsky VW. The Role of Adiponectin during Pregnancy and Gestational Diabetes. Life (Basel) 2023; 13:301. [PMID: 36836658 PMCID: PMC9958871 DOI: 10.3390/life13020301] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Pregnancy involves a range of metabolic adaptations to supply adequate energy for fetal growth and development. Gestational diabetes (GDM) is defined as hyperglycemia with first onset during pregnancy. GDM is a recognized risk factor for both pregnancy complications and long-term maternal and offspring risk of cardiometabolic disease development. While pregnancy changes maternal metabolism, GDM can be viewed as a maladaptation by maternal systems to pregnancy, which may include mechanisms such as insufficient insulin secretion, dysregulated hepatic glucose output, mitochondrial dysfunction and lipotoxicity. Adiponectin is an adipose-tissue-derived adipokine that circulates in the body and regulates a diverse range of physiologic mechanisms including energy metabolism and insulin sensitivity. In pregnant women, circulating adiponectin levels decrease correspondingly with insulin sensitivity, and adiponectin levels are low in GDM. In this review, we summarize the current state of knowledge about metabolic adaptations to pregnancy and the role of adiponectin in these processes, with a focus on GDM. Recent studies from rodent model systems have clarified that adiponectin deficiency during pregnancy contributes to GDM development. The upregulation of adiponectin alleviates hyperglycemia in pregnant mice, although much remains to be understood for adiponectin to be utilized clinically for GDM.
Collapse
Affiliation(s)
- Brittany L. Moyce Gruber
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM), Research Theme of the Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| | - Vernon W. Dolinsky
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM), Research Theme of the Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| |
Collapse
|
14
|
Yung HW, Zhao X, Glover L, Burrin C, Pang PC, Jones CJ, Gill C, Duhig K, Olovsson M, Chappell LC, Haslam SM, Dell A, Burton GJ, Charnock-Jones DS. Perturbation of placental protein glycosylation by endoplasmic reticulum stress promotes maladaptation of maternal hepatic glucose metabolism. iScience 2023; 26:105911. [PMID: 36660474 PMCID: PMC9843443 DOI: 10.1016/j.isci.2022.105911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/30/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Placental hormones orchestrate maternal metabolic adaptations to support pregnancy. We hypothesized that placental ER stress, which characterizes early-onset pre-eclampsia (ePE), compromises glycosylation, reducing hormone bioactivity and these maladaptations predispose the mother to metabolic disease in later life. We demonstrate ER stress reduces the complexity and sialylation of trophoblast protein N-glycosylation, while aberrant glycosylation of vascular endothelial growth factor reduced its bioactivity. ER stress alters the expression of 66 of the 146 genes annotated with "protein glycosylation" and reduces the expression of sialyltransferases. Using mouse placental explants, we show ER stress promotes the secretion of mis-glycosylated glycoproteins. Pregnant mice carrying placentas with junctional zone-specific ER stress have reduced blood glucose, anomalous hepatic glucose metabolism, increased cellular stress and elevated DNA methyltransferase 3A. Using pregnancy-specific glycoproteins as a readout, we also demonstrate aberrant glycosylation of placental proteins in women with ePE, thus providing a mechanistic link between ePE and subsequent maternal metabolic disorders.
Collapse
Affiliation(s)
- Hong Wa Yung
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Xiaohui Zhao
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Luke Glover
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Charlotte Burrin
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Poh-Choo Pang
- Department of Life Sciences, Imperial College London, London, UK
| | - Carolyn J.P. Jones
- Maternal and Fetal Health Centre, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Carolyn Gill
- Department of Women and Children’s Health, King’s College London, London, UK
| | - Kate Duhig
- Maternal and Fetal Health Centre, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
- Department of Women and Children’s Health, King’s College London, London, UK
| | - Matts Olovsson
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden
| | - Lucy C. Chappell
- Department of Women and Children’s Health, King’s College London, London, UK
| | - Stuart M. Haslam
- Department of Life Sciences, Imperial College London, London, UK
| | - Anne Dell
- Department of Life Sciences, Imperial College London, London, UK
| | - Graham J. Burton
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - D. Stephen Charnock-Jones
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge CB2 0SW, UK
| |
Collapse
|
15
|
Yalinbas EE, Akcilar R. Serum adiponectin levels and adiponectin +276 G/T gene polymorphism in newborns with large and small birth weights. J Matern Fetal Neonatal Med 2022; 35:10638-10646. [PMID: 36415042 DOI: 10.1080/14767058.2022.2150073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
PURPOSE Adiponectin may be an important indicator in the regulation of fetal and neonatal growth due to its metabolism, energy balance, and insulin-sensitizing action. The current study's goal was to determine if there is a link between adiponectin +276 G/T gene polymorphism and serum adiponectin level in newborns classified as appropriate for gestational age (AGA), small for gestational age (SGA), or large for gestational age (LGA). METHODS The study included newborns classified as AGA (n = 65), SGA (n = 65), or LGA (n = 65) according to their gestational age or birth weight. To determine the presence of adiponectin +276 G/T gene polymorphism, genotyping was done using polymerase chain reaction-restriction fragment length polymorphism. Enzyme-linked immunosorbent assay was used to determine the level of adiponectin in the blood. RESULTS The SGA newborns had significantly lower levels of serum adiponectin than the AGA and LGA newborns. There were statistically significant differences between the genotype frequencies (GG, GT, TT) of the SGA newborns (29.9%, 45.1%, 13.9%), the AGA newborns (41.6%, 20.7%, 44.4%), and the LGA newborns (28.6%, 34.1%, 41.7%) (chi-square = 15.8; degree of freedom = 4; p = .003). The newborns carrying the GT genotype had an increased risk of being SGA compared to those carrying the GG and TT genotypes (odds ratio [OR] = 3.07; confidence interval [CI] = 95% (1.38-6.64); p = .005 and OR = 6.96; CI = 95% (2.19-22.1); p < .001, respectively). The newborns carrying the GG and TT genotypes had better protection against being SGA than those carrying the GT genotype (OR = 0.33; CI = 95% (0.15-0.72); p = .005 and OR = 0.14; CI = 95% (0.05-0.46); p < .001, respectively). The newborns carrying the GT genotype had lower birth weights, head circumferences, and ponderal indices than those carrying the TT genotype (p < .001). The serum adiponectin levels between adiponectin +276 G/T genotypes did not differ significantly (p = .429). In addition, serum adiponectin level showed a significant positive correlation with birth weight, birth length, head circumference, and ponderal index in all newborns. CONCLUSION The results of the current study suggest that the adiponectin +276 G/T gene polymorphism was associated with an increased chance of being born SGA or LGA. The effect of this polymorphism on newborn birth size was independently associated with serum adiponectin levels. Adiponectin may play a role in fetal growth.
Collapse
Affiliation(s)
| | - Raziye Akcilar
- Department of Physiology, Faculty of Medicine, Kütahya Health Sciences University, Kütahya, Turkey
| |
Collapse
|
16
|
Cechinel LR, Batabyal RA, Freishtat RJ, Zohn IE. Parental obesity-induced changes in developmental programming. Front Cell Dev Biol 2022; 10:918080. [PMID: 36274855 PMCID: PMC9585252 DOI: 10.3389/fcell.2022.918080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Many studies support the link between parental obesity and the predisposition to develop adult-onset metabolic syndromes that include obesity, high blood pressure, dyslipidemia, insulin resistance, and diabetes in the offspring. As the prevalence of obesity increases in persons of childbearing age, so does metabolic syndrome in their descendants. Understanding how parental obesity alters metabolic programs in the progeny, predisposing them to adult-onset metabolic syndrome, is key to breaking this cycle. This review explores the basis for altered metabolism of offspring exposed to overnutrition by focusing on critical developmental processes influenced by parental obesity. We draw from human and animal model studies, highlighting the adaptations in metabolism that occur during normal pregnancy that become maladaptive with obesity. We describe essential phases of development impacted by parental obesity that contribute to long-term alterations in metabolism in the offspring. These encompass gamete formation, placentation, adipogenesis, pancreas development, and development of brain appetite control circuits. Parental obesity alters the developmental programming of these organs in part by inducing epigenetic changes with long-term consequences on metabolism. While exposure to parental obesity during any of these phases is sufficient to alter long-term metabolism, offspring often experience multiple exposures throughout their development. These insults accumulate to increase further the susceptibility of the offspring to the obesogenic environments of modern society.
Collapse
|
17
|
Adiponectin Deficiency Alters Placenta Function but Does Not Affect Fetal Growth in Mice. Int J Mol Sci 2022; 23:ijms23094939. [PMID: 35563332 PMCID: PMC9101632 DOI: 10.3390/ijms23094939] [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: 04/08/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 01/25/2023] Open
Abstract
Adiponectin administration to pregnant mice decreases nutrient transport and fetal growth. An adiponectin deficiency, on the other hand, as seen in obese women during pregnancy, alters fetal growth; however, the mechanism is unclear. To determine the role of adiponectin on placenta function and fetal growth, we used adiponectin knockout, adiponectin heterozygote that displays reduced adiponectin levels, and wild-type mice on a control diet or high fat/high sucrose (HF/HS) diet. Triglycerides (TGs) in the serum, liver, and placenta were measured using colorimetric assays. Gene expression was measured using quantitative RT-PCR. Adiponectin levels did not affect fetal weight, but it reduced adiponectin levels, increased fetal serum and placenta TG content. Wildtype dams on a HF/HS diet protected the fetuses from fatty acid overload as judged by increased liver TGs in dams and normal serum and liver TG levels in fetuses, while low adiponectin was associated with increased fetal liver TGs. Low maternal adiponectin increased the expression of genes involved in fatty acid transport; Lpl and Cd36 in the placenta. Adiponectin deficiency does not affect fetal growth but induces placental dysfunction and increases fetal TG load, which is enhanced with obesity. This could lead to imprinting effects on the fetus and the development of metabolic dysfunction in the offspring.
Collapse
|
18
|
Pippen J, Stetson B, Doherty L, Varner MW, Casey BM, Reddy UM, Wapner RJ, Rouse DJ, Tita ATN, Thorp JM, Chien EK, Saade GR, Blackwell SC. Neonatal Birthweight, Infant Feeding, and Childhood Metabolic Markers. Am J Perinatol 2022; 39:584-591. [PMID: 34918330 PMCID: PMC9106839 DOI: 10.1055/s-0041-1740056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Antenatal and early neonatal nutritional environment may influence later metabolic health. Infants of mothers with gestational diabetes mellitus (GDM) have higher risk for childhood obesity and metabolic syndrome (MetS). Leptin and adiponectin are known biomarkers for MetS and may guide interventions to reduce later obesity. We sought to examine the relationship between birthweight, early infancy feeding practices, and biomarkers for MetS in offspring of women with mild GDM. STUDY DESIGN Secondary analysis of a prospective observational follow-up study on the offspring of women who participated in a multicenter randomized treatment trial on mild GDM. Children were evaluated by research coordinators and biospecimens collected at the age of 5 to 10. Plasma concentrations of leptin and adiponectin were compared between large for gestational age (LGA) and average birthweight (AGA) infants, and according to whether solid foods were introduced early (<6 months of age) or at the recommended age (≥6 months of age). Multivariable analysis adjusted for fetal sex, race/ethnicity, and maternal body mass index. RESULTS Leptin and adiponectin were measured in 336 plasma samples. In bivariate analysis, compared with AGA children, LGA children had lower leptin (5.0 ng/mL [3.6-6.0] vs. 5.8 ng/mL [4.5 = 6.6], p = 0.01) and similar adiponectin (6.3 µg/mL [5.1-7.9] vs. 6.4 µg/mL [5.3-8.6], p = 0.49) concentrations. Maternal/child characteristics were similar between the early/delayed solid feeding groups. Leptin and adiponectin concentrations were similar in the early fed and delayed feeding groups (5.8 ng/mL [4.6-6.7] vs. 5.6 ng/mL [4.2-6.6], p = 0.50 and 6.4 µg/mL [5.4-8.1] vs. 6.4 µg/mL [5.1-8.8], p = 0.85, respectively). After controlling for covariates, children who were LGA and AGA at birth had similar leptin concentrations. CONCLUSION Birthweight and early infancy feeding practice are not associated with alterations in leptin and adiponectin in children of women with mild GDM. KEY POINTS · Adipocytokines are markers of metabolic status.. · Children of women with mild GDM may be at risk for MetS.. · Biomarkers similar in LGA and AGA groups.. · Biomarkers similar in early and delayed solid-fed groups.. · Nonhuman milk does not modify effect of feeding practice..
Collapse
Affiliation(s)
- Jessica Pippen
- Department of Obstetrics and Gynecology of The Ohio State University, Columbus, Ohio
| | - Bethany Stetson
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, Illinois
| | - Lindsay Doherty
- Department of Obstetrics and Gynecology, The George Washington University Biostatistics Center, Washington, Dist. of Columbia
| | - Michael W. Varner
- Department of Obstetrics and Gynecology, The University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Brian M. Casey
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Uma M. Reddy
- Department of Obstetrics and Gynecology, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Rockville, Maryland
| | - Ronald J. Wapner
- Department of Obstetrics and Gynecology, Columbia University, New York, New York
| | - Dwight J. Rouse
- Department of Obstetrics and Gynecology, Brown University, Providence, Rhode Island
| | | | - John M. Thorp
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Edward K. Chien
- MetroHealth Medical Center-Case Western Reserve University, Cleveland, Ohio
| | | | - Sean C. Blackwell
- University of Texas Health Science Center at Houston-Children’s Memorial Hermann Hospital, Houston, Texas
| | | |
Collapse
|
19
|
Tan K, Tint MT, Michael N, Yap F, Chong YS, Tan KH, Godfrey KM, Larbi A, Lee YS, Chan SY, Fortier MV, Eriksson JG, Karnani N. Determinants of cord blood adipokines and association with neonatal abdominal adipose tissue distribution. Int J Obes (Lond) 2022; 46:637-645. [PMID: 34864815 PMCID: PMC8873009 DOI: 10.1038/s41366-021-00975-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 09/09/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cord blood leptin and adiponectin are adipokines known to be associated with birth weight and overall infant adiposity. However, few studies have investigated their associations with abdominal adiposity in neonates. We examined maternal factors associated with cord blood leptin and adiponectin, and the association of these adipokines with neonatal adiposity and abdominal fat distribution measured by magnetic resonance imaging (MRI) in an Asian mother-offspring cohort. METHODS Growing Up in Singapore Towards healthy Outcomes (GUSTO), is a prospective mother-offspring birth cohort study in Singapore. Cord blood plasma leptin and adiponectin concentrations were measured using Luminex and Enzyme-Linked Immunosorbent Assay respectively in 816 infants. A total of 271 neonates underwent MRI within the first 2-weeks after delivery. Abdominal superficial (sSAT), deep subcutaneous (dSAT), and intra-abdominal (IAT) adipose tissue compartment volumes were quantified from MRI images. Multivariable regression analyses were performed. RESULTS Indian or Malay ethnicity, female sex, and gestational age were positively associated with cord blood leptin and adiponectin concentrations. Maternal gestational diabetes (GDM) positively associated with cord blood leptin concentrations but inversely associated with cord blood adiponectin concentrations. Maternal pre-pregnancy body mass index (BMI) showed a positive relationship with cord blood leptin but not with adiponectin concentrations. Each SD increase in cord blood leptin was associated with higher neonatal sSAT, dSAT and IAT; differences in SD (95% CI): 0.258 (0.142, 0.374), 0.386 (0.254, 0.517) and 0.250 (0.118, 0.383), respectively. Similarly, each SD increase in cord blood adiponectin was associated with higher neonatal sSAT and dSAT; differences in SD (95% CI): 0.185 (0.096, 0.274) and 0.173 (0.067, 0.278), respectively. The association between cord blood adiponectin and neonatal adiposity was observed in neonates of obese mothers only. CONCLUSIONS Cord blood leptin and adiponectin concentrations were associated with ethnicity, maternal BMI and GDM, sex and gestational age. Both adipokines showed positive association with neonatal abdominal adiposity.
Collapse
Affiliation(s)
- Karen Tan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR) Singapore, Singapore, Singapore
| | - Mya Thway Tint
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR) Singapore, Singapore, Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Navin Michael
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR) Singapore, Singapore, Singapore
| | - Fabian Yap
- Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
- Department of Pediatric Endocrinology, KK Women's and Children's Hospital, Singapore, Singapore
- National Technological University (NTU)-Lee Kong Chian School of Medicine, Singapore, Singapore
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR) Singapore, Singapore, Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kok Hian Tan
- Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
- National Technological University (NTU)-Lee Kong Chian School of Medicine, Singapore, Singapore
- Department of Maternal Fetal Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital, Southampton, United Kingdom
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yung Seng Lee
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR) Singapore, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR) Singapore, Singapore, Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Obstetrics and Gynaecology, National University Hospital, Singapore, Singapore
| | - Marielle V Fortier
- Department of Diagnostic and Interventional Imaging, KK Women's and Children's Hospital, Singapore, Singapore
| | - Johan G Eriksson
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR) Singapore, Singapore, Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Folkhälsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| | - Neerja Karnani
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR) Singapore, Singapore, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
20
|
Wang Y, Liu H, Wang J, Hu X, Wang A, Nie Z, Xu H, Li J, Xin H, Zhang J, Zhang H, Wang Y, Lyu Z. Development and validation of a new predictive model for macrosomia at late-term pregnancy: A prospective study. Front Endocrinol (Lausanne) 2022; 13:1019234. [PMID: 36465629 PMCID: PMC9713232 DOI: 10.3389/fendo.2022.1019234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Fetal macrosomia is defined as a birth weight more than 4,000 g and is associated with maternal and fetal complications. This early metabolic disease may influence the entire life of the infant. Currently, macrosomia is predicted by using the estimated fetal weight (EFW). However, the EFW is inaccurate when the gestational week is gradually increasing. To assess precisely the risk of macrosomia, we developed a new predictive model to estimate the risk of macrosomia. METHODS We continuously collected data on 655 subjects who attended regular antenatal visits and delivered at the Second Hospital of Hebei Medical University (Shijiazhuang, China) from November 2020 to September 2021. A total of 17 maternal features and 2 fetal ultrasonographic features were included at late-term pregnancy. The 655 subjects were divided into a model training set and an internal validation set. Then, 450 pregnant women were recruited from Handan Central Hospital (Handan, China) from November 2021 to March 2022 as the external validation set. The least absolute shrinkage and selection operator method was used to select the most appropriate predictive features and optimize them via 10-fold cross-validation. The multivariate logistical regressions were used to build the predictive model. Receiver operating characteristic (ROC) curves, C-indices, and calibration plots were obtained to assess model discrimination and accuracy. The model's clinical utility was evaluated via decision curve analysis (DCA). RESULTS Four predictors were finally included to develop this new model: prepregnancy obesity (prepregnancy body mass index ≥ 30 kg/m2), hypertriglyceridemia, gestational diabetes mellitus, and fetal abdominal circumference. This model afforded moderate predictive power [area under the ROC curve 0.788 (95% confidence interval [CI] 0.736, 0.840) for the training set, 0.819 (95% CI 0.744,0.894) for the internal validation set, and 0.773 (95% CI 0.713,0.833) for the external validation set]. On DCA, the model evidenced a good fit with, and positive net benefits for, both the internal and external validation sets. CONCLUSIONS We developed a predictive model for macrosomia and performed external validation in other regions to further prove the discrimination and accuracy of this predictive model. This novel model will aid clinicians in easily identifying those at high risk of macrosomia and assist obstetricians to plan accordingly.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Endocrinology, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Hongzhou Liu
- Department of Endocrinology, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Department of Endocrinology, First Hospital of Handan City, Handan, Hebei, China
| | - Jincheng Wang
- Department of Epidemiology, The George Washington University, Washington, DC, United States
| | - Xiaodong Hu
- Department of Endocrinology, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Anning Wang
- Department of Endocrinology, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhimei Nie
- Department of Endocrinology, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Huaijin Xu
- Department of Endocrinology, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Jiefei Li
- Department of Endocrinology, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Hong Xin
- Department of Obstetrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiamei Zhang
- Department of Ultrasound Diagnosis, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Han Zhang
- Department of Ultrasound Diagnosis, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yueheng Wang
- Department of Ultrasound Diagnosis, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhaohui Lyu
- Department of Endocrinology, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Zhaohui Lyu,
| |
Collapse
|
21
|
Espinoza C, Fuenzalida B, Leiva A. Increased Fetal Cardiovascular Disease Risk: Potential Synergy Between Gestational Diabetes Mellitus and Maternal Hypercholesterolemia. Curr Vasc Pharmacol 2021; 19:601-623. [PMID: 33902412 DOI: 10.2174/1570161119666210423085407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 03/16/2021] [Indexed: 01/25/2023]
Abstract
Cardiovascular diseases (CVD) remain a major cause of death worldwide. Evidence suggests that the risk for CVD can increase at the fetal stages due to maternal metabolic diseases, such as gestational diabetes mellitus (GDM) and maternal supraphysiological hypercholesterolemia (MSPH). GDM is a hyperglycemic, inflammatory, and insulin-resistant state that increases plasma levels of free fatty acids and triglycerides, impairs endothelial vascular tone regulation, and due to the increased nutrient transport, exposes the fetus to the altered metabolic conditions of the mother. MSPH involves increased levels of cholesterol (mainly as low-density lipoprotein cholesterol) which also causes endothelial dysfunction and alters nutrient transport to the fetus. Despite that an association has already been established between MSPH and increased CVD risk, however, little is known about the cellular processes underlying this relationship. Our knowledge is further obscured when the simultaneous presentation of MSPH and GDM takes place. In this context, GDM and MSPH may substantially increase fetal CVD risk due to synergistic impairment of placental nutrient transport and endothelial dysfunction. More studies on the separate and/or cumulative role of both processes are warranted to suggest specific treatment options.
Collapse
Affiliation(s)
- Cristian Espinoza
- Faculty of Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Barbara Fuenzalida
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| | - Andrea Leiva
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Providencia 7510157, Chile
| |
Collapse
|
22
|
Mallardo M, Ferraro S, Daniele A, Nigro E. GDM-complicated pregnancies: focus on adipokines. Mol Biol Rep 2021; 48:8171-8180. [PMID: 34652617 PMCID: PMC8604848 DOI: 10.1007/s11033-021-06785-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/15/2021] [Indexed: 12/27/2022]
Abstract
Gestational diabetes mellitus (GDM) is a serious complication of pregnancy and is defined as a state of glucose intolerance that is first diagnosed and arises during gestation. Although the pathophysiology of GDM has not yet been thoroughly clarified, insulin resistance and pancreatic β-cell dysfunction are considered critical components of its etiopathogenesis. To sustain fetus growth and guarantee mother health, many significant changes in maternal metabolism are required in normal and high-risk pregnancy accompanied by potential complications. Adipokines, adipose tissue-derived hormones, are proteins with pleiotropic functions including a strong metabolic influence in physiological conditions and during pregnancy too. A growing number of studies suggest that various adipokines including adiponectin, leptin, visfatin, resistin and tumor necrosis factor α (TNF-α) are dysregulated in GDM and might have pathological significance and a prognostic value in this pregnancy disorder. In this review, we will focus on the current knowledge on the role that the aforementioned adipokines play in the development and progression of GDM.
Collapse
Affiliation(s)
- Marta Mallardo
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, Università degli Studi della Campania "Luigi Vanvitelli", Via G. Vivaldi 42, 81100, Caserta, Italy
- CEINGE-Biotecnologie Avanzate S.c.a r.l., Via G. Salvatore 486, 80145, Naples, Italy
| | - Sara Ferraro
- CEINGE-Biotecnologie Avanzate S.c.a r.l., Via G. Salvatore 486, 80145, Naples, Italy
- Dipartimento di Sanità Pubblica, Università degli Studi di Napoli "Federico II", Via Pansini 5, 80145, Naples, Italy
| | - Aurora Daniele
- CEINGE-Biotecnologie Avanzate S.c.a r.l., Via G. Salvatore 486, 80145, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Naples "Federico II", Naples, Italy
| | - Ersilia Nigro
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, Università degli Studi della Campania "Luigi Vanvitelli", Via G. Vivaldi 42, 81100, Caserta, Italy.
- CEINGE-Biotecnologie Avanzate S.c.a r.l., Via G. Salvatore 486, 80145, Naples, Italy.
- CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145, Naples, Italy.
| |
Collapse
|
23
|
Wu Y, Chanclón B, Micallef P, Stener-Victorin E, Wernstedt Asterholm I, Benrick A. Maternal adiponectin prevents visceral adiposity and adipocyte hypertrophy in prenatal androgenized female mice. FASEB J 2021; 35:e21299. [PMID: 33715227 DOI: 10.1096/fj.202002212r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/26/2020] [Accepted: 12/07/2020] [Indexed: 01/17/2023]
Abstract
Hyperandrogenism is the main characteristic of polycystic ovary syndrome, which affects placental function and fetal growth, and leads to reproductive and metabolic dysfunction in female offspring. Adiponectin acts on the placenta and may exert endocrine effects on the developing fetus. This study aims to investigate if maternal and/or fetal adiponectin can prevent metabolic and reproductive dysfunction in prenatal androgenized (PNA) female offspring. Adiponectin transgenic (APNtg) and wild-type dams received dihydrotestosterone/vehicle injections between gestational days 16.5-18.5 to induce PNA offspring, which were followed for 4 months. Offspring from APNtg dams were smaller than offspring from wild-type dams, independent of genotype. Insulin sensitivity was higher in wild-type mice from APNtg dams compared to wild-types from wild-type dams, and insulin sensitivity correlated with fat mass and adipocyte size. PNA increased visceral fat% and adipocyte size in wild-type offspring from wild-type dams, while wild-type and APNtg offspring from APNtg dams were protected against this effect. APNtg mice had smaller adipocytes than wild-types and this morphology was associated with an increased expression of genes regulating adipogenesis (Ppard, Pparg, Cebpa, and Cebpb) and metabolism (Chrebp and Lpl). Anogenital distance was increased in all PNA-exposed wild-type offspring, but there was no increase in PNA APNtg offspring, suggesting that adiponectin overexpression protects against this effect. In conclusion, elevated adiponectin levels in utero improve insulin sensitivity, reduce body weight and fat mass gain in the adult offspring and protect against PNA-induced visceral adiposity. In conclusion, these data suggest that PNA offspring benefit from prenatal adiponectin supplementation.
Collapse
Affiliation(s)
- Yanling Wu
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Belén Chanclón
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter Micallef
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Ingrid Wernstedt Asterholm
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Benrick
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,School of Health Sciences, University of Skövde, Skövde, Sweden
| |
Collapse
|
24
|
Abstract
Almost 2 billion adults in the world are overweight, and more than half of them are classified as obese, while nearly one-third of children globally experience poor growth and development. Given the vast amount of knowledge that has been gleaned from decades of research on growth and development, a number of questions remain as to why the world is now in the midst of a global epidemic of obesity accompanied by the "double burden of malnutrition," where overweight coexists with underweight and micronutrient deficiencies. This challenge to the human condition can be attributed to nutritional and environmental exposures during pregnancy that may program a fetus to have a higher risk of chronic diseases in adulthood. To explore this concept, frequently called the developmental origins of health and disease (DOHaD), this review considers a host of factors and physiological mechanisms that drive a fetus or child toward a higher risk of obesity, fatty liver disease, hypertension, and/or type 2 diabetes (T2D). To that end, this review explores the epidemiology of DOHaD with discussions focused on adaptations to human energetics, placental development, dysmetabolism, and key environmental exposures that act to promote chronic diseases in adulthood. These areas are complementary and additive in understanding how providing the best conditions for optimal growth can create the best possible conditions for lifelong health. Moreover, understanding both physiological as well as epigenetic and molecular mechanisms for DOHaD is vital to most fully address the global issues of obesity and other chronic diseases.
Collapse
Affiliation(s)
- Daniel J Hoffman
- Department of Nutritional Sciences, Program in International Nutrition, and Center for Childhood Nutrition Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Theresa L Powell
- Department of Pediatrics and Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Emily S Barrett
- Department of Biostatistics and Epidemiology, School of Public Health and Division of Exposure Science and Epidemiology, Rutgers Environmental and Occupational Health Sciences Institute, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Daniel B Hardy
- Department of Biostatistics and Epidemiology, School of Public Health and Division of Exposure Science and Epidemiology, Rutgers Environmental and Occupational Health Sciences Institute, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
25
|
Kristiansen O, Zucknick M, Reine TM, Kolset SO, Jansson T, Powell TL, Haugen G, Henriksen T, Michelsen TM. Mediators Linking Maternal Weight to Birthweight and Neonatal Fat Mass in Healthy Pregnancies. J Clin Endocrinol Metab 2021; 106:1977-1993. [PMID: 33713406 PMCID: PMC8692236 DOI: 10.1210/clinem/dgab166] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Indexed: 12/16/2022]
Abstract
CONTEXT Lifestyle interventions have not efficaciously reduced complications caused by maternal weight on fetal growth, requiring insight into explanatory mediators. OBJECTIVE We hypothesized that maternal mediators, including adiponectin, leptin, insulin, and glucose, mediate effects of pregestational BMI (pBMI) and gestational weight gain (GWG) on birthweight and neonatal fat mass percentage (FM%) through placental weight and fetal mediators, including insulin levels (Ifv) and venous-arterial glucose difference (ΔGfva). Hypothesized confounders were maternal age, gestational age, and parity. METHODS A cross-sectional study of healthy mother-offspring-pairs (n = 165) applying the 4-vessel in vivo sampling method at Oslo University Hospital, Norway. We obtained pBMI, GWG, birthweight, and placental weight. FM% was available and calculated for a subcohort (n = 84). We measured circulating levels of adiponectin, leptin, glucose, and insulin and performed path analysis and traditional mediation analyses based on linear regression models. RESULTS The total effect of pBMI and GWG on newborn size was estimated to be 30 g (range, 16-45 g) birthweight and 0.17 FM% (range, 0.04-0.29 FM%) per kg∙m-2 pBMI and 31 g (range, 18-44 g) and 0.24 FM% (range, 0.10-0.37 FM%) per kg GWG. The placental weight was the main mediator, mediating 25-g birthweight and 0.11 FM% per kg∙m-2 pBMI and 25-g birthweight and 0.13 FM% per kg GWG. The maternal mediators mediated a smaller part of the effect of pBMI (3.8-g birthweight and 0.023 FM% per kg∙m-2 pBMI) but not GWG. CONCLUSION Placental weight was the main mediator linking pBMI and GWG to birthweight and FM%. The effect of pBMI, but not GWG, on birthweight and FM%, was also mediated via the maternal and fetal mediators.
Collapse
Affiliation(s)
- Oddrun Kristiansen
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital, Oslo, 0424Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0372Norway
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0372Norway
- Correspondence: Oddrun Kristiansen, PhD, Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital, Postboks 4956 Nydalen, Oslo, 0424 Norway. ;
| | - Manuela Zucknick
- Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0372Norway
| | - Trine M Reine
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0372Norway
- Institute of Cancer Genetics and Informatics, Oslo University Hospital, Oslo, 0379Norway
| | - Svein O Kolset
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, 0372Norway
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Colorado 80045, USA
| | - Theresa L Powell
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Colorado 80045, USA
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Colorado 80045, USA
| | - Guttorm Haugen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0372Norway
- Department of Fetal Medicine, Division of Obstetrics and Gynecology, Oslo University Hospital, Oslo, 0424Norway
| | - Tore Henriksen
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital, Oslo, 0424Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0372Norway
| | - Trond M Michelsen
- Department of Obstetrics, Division of Obstetrics and Gynecology, Oslo University Hospital, Oslo, 0424Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0372Norway
| |
Collapse
|
26
|
The effect of adiponectin and its receptors in placental development of diabetic rats. Biologia (Bratisl) 2021. [DOI: 10.1007/s11756-021-00742-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
27
|
Pádua ECR, Daher S, Sampaio IDPC, Araujo Júnior E, Guazzelli CF. Evaluation of the Blood Level of Adiponectin in Pregnant Adolescents. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2021; 43:429-435. [PMID: 34077989 PMCID: PMC10411129 DOI: 10.1055/s-0041-1730288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 02/18/2021] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVE To evaluate serum levels of adiponectin in pregnant adolescents between 30 and 36 weeks of gestation. METHOD A prospective cross-sectional study enrolled 67 normal pregnant women between 30 and 36 weeks of gestation and eutrophic (body mass index [BMI]: 18.5-25 kg/m2), of which 36 were adolescents (< 20 years old) and 31 adults (≥ 20 years old). Serum adiponectin levels were determined by enzyme-linked immunosorbent assay (ELISA). The t-student or Mann-Whitney tests were used for intergroup comparison. RESULTS Pregnant adolescents showed significantly higher serum adiponectin concentrations compared with pregnant adults (p = 0.04). No differences were observed in adiponectin levels in younger pregnant adolescents (< 16 years old) compared with older pregnant adolescents (≥ 16 years old). Adiponectin values were divided into 3 subgroups: < 3,000 ng/mL, between 3,000 and 5,000 ng/mL, and > 5,000 ng/mL. Birthweight was significantly higher in women > 5,000 ng/mL when compared with < 3,000 ng/mL in the adolescent group. No association between pregestational adiponectin levels and BMI, gestational weight gain, and gestational age was observed; however, there was a positive relation with birthweight (p = 0.0239). CONCLUSION Serum adiponectin values in pregnant adolescents between 30 and 36 weeks of gestation were higher compared with pregnant adults; however, no differences between younger and older pregnant adolescents were observed.
Collapse
Affiliation(s)
- Elaine Cristina Rocha Pádua
- Department of Obstetrics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo-SP, Brazil
| | - Silvia Daher
- Department of Obstetrics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo-SP, Brazil
| | - Isa de Pádua Cintra Sampaio
- Department of Pediatrics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo-SP, Brazil
| | - Edward Araujo Júnior
- Department of Obstetrics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo-SP, Brazil
| | - Cristina Falbo Guazzelli
- Department of Obstetrics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo-SP, Brazil
| |
Collapse
|
28
|
Buck CO, Li N, Eaton CB, Kelsey KT, Cecil KM, Kalkwarf HJ, Yolton K, Lanphear BP, Chen A, Braun JM. Neonatal and Adolescent Adipocytokines as Predictors of Adiposity and Cardiometabolic Risk in Adolescence. Obesity (Silver Spring) 2021; 29:1036-1045. [PMID: 34029449 PMCID: PMC8567116 DOI: 10.1002/oby.23160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/30/2021] [Accepted: 02/25/2021] [Indexed: 11/05/2022]
Abstract
OBJECTIVE This study aimed to examine associations of changes in leptin and adiponectin concentrations from birth to age 12 years with adolescent adiposity and cardiometabolic risk in the Health Outcomes and Measures of Environment (HOME) Study, a prospective birth cohort (Cincinnati, Ohio; N = 166). METHODS Adiposity and cardiometabolic risk factors were assessed at age 12 years using anthropometry, dual-energy x-ray absorptiometry, and fasting serum biomarkers. Cardiometabolic risk scores were calculated by summing age- and sex- standardized z scores for individual cardiometabolic risk factors. RESULTS Most serum adipocytokine concentrations at birth were not associated with adiposity or cardiometabolic risk outcomes. Leptin and adiponectin concentrations at age 12 years were associated with all outcomes in the expected direction. Adolescents with increasing (β: 4.2; 95% CI: 3.2 to 5.2) and stable (β: 2.2; 95% CI: 1.2 to 3.2) leptin concentrations from birth to age 12 years had higher cardiometabolic risk scores than adolescents with decreasing concentrations (reference group). Adolescents with increasing (e.g., fat mass index = β: -1.04; 95% CI: -1.27 to -0.80) and stable (β: 0.66; 95% CI: -0.92 to -0.40) adiponectin/leptin ratios had more favorable adiposity outcomes than adolescents with decreasing ratios. CONCLUSIONS In this cohort, changes in leptin concentrations and adiponectin/leptin ratios over childhood were associated with adiposity and cardiometabolic risk scores, indicating that adipocytokine concentrations are potential biomarkers for predicting excess adiposity and cardiometabolic risk in adolescence.
Collapse
Affiliation(s)
| | - Nan Li
- Department of Epidemiology, School of Public Health, Brown University, Providence, RI
| | - Charles B. Eaton
- Department of Epidemiology, School of Public Health, Brown University, Providence, RI
- Department of Family Medicine, Alpert Medical School of Brown University, Providence, RI
- Kent Memorial Hospital, Warwick, RI
| | - Karl T. Kelsey
- Department of Epidemiology, School of Public Health, Brown University, Providence, RI
- Department of Pathology and Laboratory Medicine, Brown University, Providence
| | - Kim M. Cecil
- Imaging Research Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Departments of Pediatrics and Radiology, University of Cincinnati College of Medicine, Cincinnati
| | - Heidi J. Kalkwarf
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati
| | - Kimberly Yolton
- Department of Pediatrics, Division of General and Community Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati
| | - Bruce P. Lanphear
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Aimin Chen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Joseph M. Braun
- Department of Epidemiology, School of Public Health, Brown University, Providence, RI
| |
Collapse
|
29
|
Infant body composition relationship to maternal adipokines and fat mass: the PONCH study. Pediatr Res 2021; 89:1756-1764. [PMID: 32927470 DOI: 10.1038/s41390-020-01115-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Infant adiposity is linked to both high maternal fat mass (FM) and excessive gestational FM gain, whereas the association with maternal adipokines is less clear. The aim was to determine how levels of maternal leptin, the soluble leptin receptor (sOB-R), adiponectin, and FM during pregnancy were linked to infant FM in normal-weight (NW) women and women with obesity (OB). METHODS Body composition and serum levels of leptin, adiponectin, and sOB-R were determined three times during pregnancy in 80 NW and 46 OB women. For infants, body composition was measured at 1 and 12 weeks of age. RESULTS Maternal leptin and sOB-R levels increased during pregnancy. For NW women, infant FM at 1 week was inversely associated with changes in maternal leptin and at 12 weeks inversely associated with absolute maternal sOB-R levels throughout pregnancy, as well as changes in sOB-R levels in early pregnancy. For OB women, infant FM at both 1 and 12 weeks were best explained by maternal FM. CONCLUSIONS Leptin and sOB-R, thought to regulate leptin bioavailability, are associated with fat accumulation in infants born to NW women. In OB women, maternal FM in early pregnancy is more important than leptin in determining infant fat accumulation. IMPACT In normal-weight women, the regulation of maternal leptin bioavailability during pregnancy has a role in infant fat mass accumulation. In women with obesity, however, pre-pregnancy maternal fat mass seems more important for infant fat mass. This is the first study of maternal adipokines and fat mass including longitudinal measurements in both mothers and their children. Understanding the relationship between maternal factors and infant fat mass is of great importance as obesity is programmed over the generations, and it is important to learn what regulates this programming.
Collapse
|
30
|
Słabuszewska-Jóźwiak A, Malinowska M, Kloska A, Jakóbkiewicz-Banecka J, Gujski M, Bojar I, Raczkiewicz D, Jakiel G. Global Changes of 5-mC/5h-mC Ratio and Methylation of Adiponectin and Leptin Gene in Placenta Depending on Mode of Delivery. Int J Mol Sci 2021; 22:3195. [PMID: 33801130 PMCID: PMC8004251 DOI: 10.3390/ijms22063195] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/08/2021] [Accepted: 03/18/2021] [Indexed: 01/03/2023] Open
Abstract
It was suggested that the epigenetic alterations of the placenta are associated with obesity, as well as the delivery mode. This study aimed to assess the effect of maternal outcome and delivery procedure on global placental DNA methylation status, as well as selected 5'-Cytosine-phosphate-Guanine-3' (CpG) sites in ADIPOQ and LEP genes. Global DNA methylation profile in the placenta was assessed using the 5-methylcytosine (5-mC) and 5-hydroxymethylcytosine (5-hmC) ratio evaluated with the ELISA, followed by target gene methylation patterns at selected gene regions which were determined using methylation-specific qPCR in 70 placentas from healthy, pregnant women with single pregnancy. We found no statistically significant differences in 5-mC/5-hmC ratio between intrapartum cesarean sections (CS) and vaginal deliveries (p = 0.214), as well as between elective cesarean sections and vaginal deliveries (p = 0.221). In intrapartum cesarean sections, the ADIPOQ demethylation index was significantly higher (the average: 1.75) compared to elective cesarean section (the average: 1.23, p = 0.010) and vaginal deliveries (the average: 1.23, p = 0.011). The LEP demethylation index did not significantly differ among elective CS, intrapartum CS, and vaginal delivery groups. The demethylation index of ADIPOQ correlated negatively with LEP in the placenta in the vaginal delivery group (r = -0.456, p = 0.017), but not with the global methylation. The methylation of a singular locus might be different depending on the mode of delivery and uterine contractions. Further studies should be conducted with locus-specific analysis of the whole genome to detect the methylation index of specific genes involved in metabolism.
Collapse
Affiliation(s)
- Aneta Słabuszewska-Jóźwiak
- First Department of Obstetrics and Gynaecology, Centre of Postgraduate Medical Education, Żelazna 90, 01-004 Warsaw, Poland;
| | - Marcelina Malinowska
- Department of Medical Biology and Genetics, Faculty of Biology University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (M.M.); (A.K.); (J.J.-B.)
| | - Anna Kloska
- Department of Medical Biology and Genetics, Faculty of Biology University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (M.M.); (A.K.); (J.J.-B.)
| | - Joanna Jakóbkiewicz-Banecka
- Department of Medical Biology and Genetics, Faculty of Biology University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (M.M.); (A.K.); (J.J.-B.)
| | - Mariusz Gujski
- Department of Public Health, Medical University of Warsaw, Nielubowicza 5, 02-097 Warsaw, Poland;
| | - Iwona Bojar
- Department of Women’s Health, Institute of Rural Health in Lublin, Jaczewskiego 2, 20-950 Lublin, Poland;
| | - Dorota Raczkiewicz
- Department of Medical Statistics, School of Public Health, Center of Postgraduate Medical Education, Kleczewska 61/63, 01-826 Warsaw, Poland;
| | - Grzegorz Jakiel
- First Department of Obstetrics and Gynaecology, Centre of Postgraduate Medical Education, Żelazna 90, 01-004 Warsaw, Poland;
| |
Collapse
|
31
|
Quercetin improved histological structure and upregulated adiponectin and adiponectin receptors in the placenta of rats with gestational diabetes mellitus. Placenta 2021; 106:49-57. [PMID: 33640737 DOI: 10.1016/j.placenta.2021.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 01/24/2021] [Accepted: 02/15/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Gestational diabetes mellitus (GDM) is a metabolic syndrome among pregnant mothers that increases the risk of developing growth disorders in the fetus and the placenta. Adiponectin is an adipokine, which plays a central role in the regulation of glucose and lipid metabolism, energy homeostasis, and insulin resistance in various tissues. Quercetin is a natural flavonoid with beneficial effects in the diabetic animal model, but data related to its effect on histological change and adiponectin system in the placenta of GDM are limited. In the current study, some histological changes and expression of adiponectin and its two receptors in the placenta of rats with GDM were investigated. METHODS This study was carried out on placentas from the rodent model. To induce GDM, female rats were treated with a single dose of STZ. Placenta tissue was harvested and stained by PAS method. Protein and mRNA levels of adiponectin and its two receptors were assessed by immunohistochemistry and Real time PCR analysis, respectively. RESULTS The results showed the increased number of glycogen cells and thickness of the labyrinth interhemal membrane (LIM) in the embryonic part of the placenta in diabetic rats, while the use of quercetin significantly prevented their increase in diabetic rats. Treatment of the diabetic group with quercetin caused significantly increased adiponectin expression and decreased its receptors.The immunohistochemical study revealed the expression of AdipoR2 in the cytoplasm of syncytiotrophoblast and cytotrophoblast cells. DISCUSSION The results indicated that quercetin in pregnant diabetic rats could attenuate the histological abnormalities and improved adiponectin system dysregulation in the placenta.
Collapse
|
32
|
Pheiffer C, Dias S, Jack B, Malaza N, Adam S. Adiponectin as a Potential Biomarker for Pregnancy Disorders. Int J Mol Sci 2021; 22:1326. [PMID: 33572712 PMCID: PMC7866110 DOI: 10.3390/ijms22031326] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/28/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022] Open
Abstract
Adiponectin is an adipocyte-derived hormone that plays a critical role in energy homeostasis, mainly attributed to its insulin-sensitizing properties. Accumulating studies have reported that adiponectin concentrations are decreased during metabolic diseases, such as obesity and type 2 diabetes, with an emerging body of evidence providing support for its use as a biomarker for pregnancy complications. The identification of maternal factors that could predict the outcome of compromised pregnancies could act as valuable tools that allow the early recognition of high-risk pregnancies, facilitating close follow-up and prevention of pregnancy complications in mother and child. In this review we consider the role of adiponectin as a potential biomarker of disorders associated with pregnancy. We discuss common disorders associated with pregnancy (gestational diabetes mellitus, preeclampsia, preterm birth and abnormal intrauterine growth) and highlight studies that have investigated the potential of adiponectin to serve as biomarkers for these disorders. We conclude the review by recommending strategies to consider for future research.
Collapse
Affiliation(s)
- Carmen Pheiffer
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, Cape Town 7505, South Africa; (S.D.); (B.J.); (N.M.)
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, P.O. Box 19063, Tygerberg, Cape Town 7505, South Africa
| | - Stephanie Dias
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, Cape Town 7505, South Africa; (S.D.); (B.J.); (N.M.)
| | - Babalwa Jack
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, Cape Town 7505, South Africa; (S.D.); (B.J.); (N.M.)
| | - Nompumelelo Malaza
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, Cape Town 7505, South Africa; (S.D.); (B.J.); (N.M.)
- Department of Reproductive Biology, University of Pretoria, Private Bag X169, Pretoria 0001, South Africa
| | - Sumaiya Adam
- Department of Obstetrics and Gynaecology, University of Pretoria, Private Bag X169, Pretoria 0001, South Africa;
| |
Collapse
|
33
|
Shen L, Zhu Y, Xiao J, Qian B, Jiang T, Deng J, Peng G, Yu S, Cao S, Zuo Z, Ma X, Zhong Z, Ren Z, Wang Y, Zhou Z, Liu H, Zong X, Hu Y. Relationships between placental adiponectin, leptin, visfatin and resistin and birthweight in cattle. Reprod Fertil Dev 2021; 32:402-408. [PMID: 31739842 DOI: 10.1071/rd18247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 07/01/2019] [Indexed: 01/05/2023] Open
Abstract
Adipokines can affect intrauterine development while calf birthweight (CBW) is a breeding standard of calves, which reflects the status of fetal intrauterine development. To explore the correlation between placental adipokines and CBW, 54 healthy Chinese Holstein cows were used in the present study. The cows were grouped according to the CBW of their calves. Placentas were collected immediately after delivery and enzyme-linked immunosorbent assay and reverse transcription-polymerase chain reaction were used to detect the placental expression levels of adiponectin, leptin, visfatin and resistin. Our results show that the mRNA transcription and blood placental content of adiponectin, leptin, visfatin and resistin increased with increasing CBW. The analysis showed that the mRNA transcription levels of placental adiponectin, leptin and resistin were positively correlated with CBW. The mRNA and protein expression levels of adiponectin, leptin and visfatin between the three groups were significantly correlated. Placental resistin mRNA levels correlated positively with adiponectin mRNA, but not leptin or visfatin. The protein expression levels of resistin were significantly positively correlated with those of adiponectin, leptin and visfatin. These results suggest that placental adipokines play important roles in regulating calf intrauterine growth.
Collapse
Affiliation(s)
- Liuhong Shen
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Yingkun Zhu
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Jinbang Xiao
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Bolin Qian
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Tao Jiang
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Junliang Deng
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Guangneng Peng
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Shumin Yu
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Suizhong Cao
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China; and Corresponding author.
| | - Zhicai Zuo
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Xiaoping Ma
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Zhijun Zhong
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Zhihua Ren
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Ya Wang
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Ziyao Zhou
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Haifeng Liu
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| | - Xiaolan Zong
- Sichuan Agricultural University, Chengdu Campus, Academic Affairs Office, Chengdu, Sichuan, 611130, China
| | - Yanchun Hu
- Sichuan Agricultural University, Chengdu Campus, The Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Chengdu, Sichuan, 611130, China
| |
Collapse
|
34
|
Role of Peroxisome Proliferator-Activated Receptors (PPARs) in Trophoblast Functions. Int J Mol Sci 2021; 22:ijms22010433. [PMID: 33406768 PMCID: PMC7795665 DOI: 10.3390/ijms22010433] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 12/23/2020] [Accepted: 12/29/2020] [Indexed: 12/11/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARα, PPARβ/δ, and PPARγ) belong to the transcription factor family, and they are highly expressed in all types of trophoblast during pregnancy. The present review discusses currently published papers that are related to the regulation of PPARs via lipid metabolism, glucose metabolism, and amino acid metabolism to affect trophoblast physiological conditions, including differentiation, maturation, secretion, fusion, proliferation, migration, and invasion. Recent pieces of evidence have proven that the dysfunctions of PPARs in trophoblast lead to several related pregnancy diseases such as recurrent miscarriage, preeclampsia, intrauterine growth restriction, and gestational diabetes mellitus. Moreover, the underlying mechanisms of PPARs in the control of these processes have been discussed as well. Finally, this review's purposes are to provide more knowledge about the role of PPARs in normal and disturbed pregnancy with trophoblast, so as to find PPAR ligands as a potential therapeutic target in the treatment and prevention of adverse pregnancy outcomes.
Collapse
|
35
|
Han L, Li B, Xu X, Liu S, Li Z, Li M, Wang D. Umbilical Cord Blood Adiponectin, Leptin, Insulin, and Ghrelin in Premature Infants and Their Association With Birth Outcomes. Front Endocrinol (Lausanne) 2021; 12:738964. [PMID: 34659122 PMCID: PMC8515017 DOI: 10.3389/fendo.2021.738964] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/26/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Premature/low-birth-weight infants are at significant risk of metabolic diseases in adulthood, which may be related to the levels of fetal adipokine. Here, we investigated the differences in the levels of umbilical cord blood adiponectin, leptin, insulin, and ghrelin in preterm and term infants and sought to elucidate the link between these hormones and fetal growth. We also evaluated the interrelationship among these metabolic hormones in both groups of newborns. METHODS A total of 149 mother-infant pairs (100 in the preterm group and 49 in the term group) were enrolled in the study. The preterm group was further subdivided according to birth weight (≤1,500, 1,501-2,000, 2,001-2,500, and >2,500 g), gestational age (<34 vs. ≥34 weeks), and appropriate for gestational age (AGA) vs. small for gestational age (SGA). The general condition of the mothers and the growth parameters of the newborns at birth were recorded. RESULTS The levels of adiponectin, leptin, and ghrelin were lower in the preterm group than those in the term group (p < 0.05). In the preterm group, the leptin levels of infants with gestational age ≥34 weeks were significantly higher than those of infants with gestational age <34 weeks (mean ln leptin = 0.63 vs. 0.36 ng/ml, p = 0.009). The levels of adiponectin were lower in the SGA group than those in the AGA group (mean ln adiponectin = 2.26 vs. 2.84 µg/ml, p = 0.001), whereas those of ghrelin displayed the opposite trend (mean ln ghrelin = 6.29 vs. 5.71 pg/ml, p < 0.001). Leptin was significantly correlated with insulin both in preterm infants with birth weight (BW) >2,000 g and in term infants. Umbilical cord blood leptin was positively correlated with the BW, birth length, and head circumference of newborns (r = 0.460, 0.311, and 0.310, respectively, all p < 0.05), whereas ghrelin was negatively correlated with the same parameters (r = -0.372, -0.415, and -0.373, respectively, all p > 0.05). CONCLUSIONS The lack of maturation of adipose tissue and the gastrointestinal tract by the fetus due to prematurity is associated with changes in the levels of cord blood adiponectin, leptin, and ghrelin. The dysregulation of these hormones in preterm infants may be a risk factor for fetal growth and future metabolic diseases.
Collapse
Affiliation(s)
- Luyan Han
- Department of Pediatrics, The First Hospital of Tsinghua University, Beijing, China
- Department of Pediatrics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Bo Li
- Department of Endocrinology, NHC Key Laboratory of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaojing Xu
- Department of Pediatrics, The First Hospital of Tsinghua University, Beijing, China
| | - Shufang Liu
- Department of Pediatrics, The First Hospital of Tsinghua University, Beijing, China
| | - Zhenghong Li
- Department of Pediatrics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Ming Li
- Department of Endocrinology, NHC Key Laboratory of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Ming Li, ; Danhua Wang,
| | - Danhua Wang
- Department of Pediatrics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Ming Li, ; Danhua Wang,
| |
Collapse
|
36
|
Solis Paredes JM, Perichart Perera O, Montoya Estrada A, Reyes Muñoz E, Espino y Sosa S, Ortega Castillo V, Medina Bastidas D, Tolentino Dolores M, Sanchez Martinez M, Nava Salazar S, Estrada Gutierrez G. Gestational Weight Gain Influences the Adipokine-Oxidative Stress Association during Pregnancy. Obes Facts 2021; 14:604-612. [PMID: 34535612 PMCID: PMC8740011 DOI: 10.1159/000518639] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/21/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION AND OBJECTIVE The weight gained during pregnancy could determine the immediate and future health of the mother-child dyad. Excessive gestational weight gain (EGWG) due to abnormal adipose tissue (AT) accumulation is strongly associated with adverse perinatal outcomes as gestational diabetes, macrosomia, obesity, and hypertension further in life. Dysregulation of adipokine, AT dysfunction, and an imbalance in the prooxidant-antioxidant systems are critical features in altered AT accumulation. This study was aimed to investigate the association between adipokines and oxidative stress markers in pregnant women and the influence of the GWG on this association. METHODS Maternal blood samples were obtained in the third trimester of pregnancy (n = 74) and serum adipokines (adiponectin, leptin, and resistin), oxidative damage markers: 8-oxo-2'-deoxyguanosine (8-oxodG), lipohydroperoxides (LOOH), malondialdehyde (MDA), and carbonylated proteins (CP), and glucose a metabolic marker were measured. RESULTS Women with EGWG had low adiponectin levels than women with adequate weight gain (AWG) or insufficient weight gain (IWG). Multiple linear regression models revealed a positive association between adiponectin and 8-oxodG in women with AWG (B = 1.09, 95% CI: 164-222, p = 0.027) and IWG (B = 0.860, 95% CI: 0.199-1.52, p = 0.013) but not in women with EGWG. In women with EGWG, leptin was positively associated with LOOH (p = 0.018), MDA (p = 0.005), and CP (p = 0.010) oxidative markers. CONCLUSION Our findings suggest that concurrent mechanisms regulate adipokine production and oxidative stress in pregnant women and that this regulation is influenced by GWG, probably due to an excessive AT accumulation.
Collapse
Affiliation(s)
- Juan Mario Solis Paredes
- Department of Human Genetics and Genomics, Instituto Nacional de Perinatologia, Mexico City, Mexico
| | - Otilia Perichart Perera
- Instituto Nacional de Perinatologia, Nutrition and Bioprogramming Coordination, Mexico City, Mexico
| | - Araceli Montoya Estrada
- Instituto Nacional de Perinatologia, Coordination of Gynecological and Perinatal Endocrinology, Mexico City, Mexico
| | - Enrique Reyes Muñoz
- Instituto Nacional de Perinatologia, Coordination of Gynecological and Perinatal Endocrinology, Mexico City, Mexico
| | | | | | - Diana Medina Bastidas
- Instituto Nacional de Perinatologia– Facultad de Quimica-UNAM, Unidad de Investigación en Reproducción Humana, Mexico City, Mexico
| | | | | | - Sonia Nava Salazar
- Department of Inmunobiochemistry, Instituto Nacional de Perinatologia, Mexico City, Mexico
| | | |
Collapse
|
37
|
Balachandiran M, Bobby Z, Dorairajan G, Gladwin V, Vinayagam V, Packirisamy RM. Decreased maternal serum adiponectin and increased insulin-like growth factor-1 levels along with increased placental glucose transporter-1 expression in gestational diabetes mellitus: Possible role in fetal overgrowth. Placenta 2020; 104:71-80. [PMID: 33285436 DOI: 10.1016/j.placenta.2020.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/02/2020] [Accepted: 11/24/2020] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The placental glucose transporter - 1 (GLUT-1) is involved in the transplacental glucose transport to the fetus. GLUT-1 expressions are increased in diabetic pregnancies and associated with altered fetal growth. However, the factors regulating the GLUT-1 expressions are largely unknown. We hypothesised that maternal adipokines and insulin-like growth factor-1 (IGF1) modulate the placental expressions of GLUT-1 through the activation of insulin/IGF-1 signalling which may contribute to a fetal overgrowth in GDM. METHODS Maternal blood, cord blood and placental samples were collected from GDM and control pregnant women (CPW). The biochemical parameters, IGF1, adipokines, and high sensitive C- reactive protein were measured. We analysed the placental expressions of GLUT-1 and proteins related to insulin/IGF-1 signalling - insulin receptor -β, insulin receptor substrate - 1, phosphatidylinositol-3-kinase p110α, phospho Akt-1, phospho extracellular signal-regulated kinase 1/2, and nuclear factor-κB p65 in GDM and CPW. RESULTS Increased maternal IGF-1 and decreased adiponectin levels were found in the GDM women. Maternal IGF-1 levels were positively correlated, whereas adiponectin levels were negatively correlated with the birth weight of GDM newborns. Increased phosphorylation of Akt and ERK 1/2 was found in the placenta of GDM women. Placental expressions of GLUT-1 were significantly higher in the GDM women and positively correlated to the maternal IGF-1 levels in the GDM group. DISCUSSION Decreased maternal adiponectin and increased IGF-1 levels might have caused increased GLUT-1 expression via the increased activation of insulin/IGF-1 signalling in the placenta of GDM women which might have influenced the fetal growth.
Collapse
Affiliation(s)
- Manoharan Balachandiran
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Zachariah Bobby
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India.
| | - Gowri Dorairajan
- Department of Obstetrics & Gynaecology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Victorraj Gladwin
- Department of Anatomy, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Vickneshwaran Vinayagam
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Rajaa Muthu Packirisamy
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| |
Collapse
|
38
|
The Influence of Maternal Levels of Vitamin D and Adiponectin on Anthropometrical Measures and Bone Health in Offspring. ACTA ACUST UNITED AC 2020; 40:91-98. [PMID: 32109213 DOI: 10.2478/prilozi-2020-0008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND From the conception onward, certain parameters associated with maternal health may affect foetal body composition, growth and bone mineral content. The objective of the study was to determine the association between maternal vitamin D and adiponectin status with the anthropometrical measures of newborns, and bone health status measured by Quantitative Ultrasound (QUS) at birth. METHODS Circulating 25OHD and adiponectin concentration were measured in 73 pregnant women. Correlations with the anthropometrical measures and bone health status in their infants were studied. Bone health was evaluated using QUS with the measurements of speed of sound (SOS, in m/s) and Z score on the right tibia. RESULTS There was no significant association between maternal 25OHD and newborn's anthropometrical measures at birth (weight p=0.35, length p=0.59 and head circumference p=0.47). There was a significant negative correlation between a maternal serum adiponectin and a) weight of infants at birth (R= -0.37, p=0.002); b) birth length (R= -0.31, p=0.008) and c) head circumference (R= -0.29, p=0.014). There was no significant correlation between maternal 25OHD blood levels during pregnancy and SOS in newborns (p=0.48). Additionally, a correlation between maternal adiponectin concentration during pregnancy and SOS in newborns was not significant (p=0.82). CONCLUSION Although a high prevalence of low 25OHD level among pregnant women was found, maternal vitamin D status did not influence growth and bone health of their offspring at birth. Maternal adiponectin levels in plasma showed an inverse relationship with anthropometrical measures of infants at birth, while no correlation with the newborn's bone health was found.
Collapse
|
39
|
Fetal Growth Trajectories and Their Association with Maternal, Cord Blood, and 5-year Child Adipokines. J Nutr Metab 2020; 2020:4861523. [PMID: 33029393 PMCID: PMC7530496 DOI: 10.1155/2020/4861523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 07/22/2020] [Accepted: 08/23/2020] [Indexed: 11/18/2022] Open
Abstract
Background The growth of the fetus is a complex process influenced by multiple factors. Studies have highlighted the important role of biochemical growth markers such as leptin and adiponectin on fetal growth. Objective To compare fetal growth trajectories with biochemical growth markers from maternal blood samples at 28 weeks' gestation, cord blood samples at birth, and in child blood samples at 5 years of age from mother-infant pairs who were part of the longitudinal ROLO study. Methods 781 mother-infant pairs from the ROLO and ROLO Kids study were included. Ultrasound measurements and birth weight were used to develop fetal growth trajectory groups for estimated abdominal circumference and estimated weight. Blood serum levels of leptin, adiponectin, insulin, TNF-alpha, and IL-6 from maternal, cord, and 5-year child samples were recorded. ANOVA and chi-square tests were applied to test the associations between fetal growth trajectory membership and maternal and child biochemical growth indicators. The influence of child sex was also investigated. Results Male sex was associated with a faster weight trajectory compared to females (p=0.001). At 28 weeks' gestation, maternal leptin levels were significantly higher in mothers with a fetus on a slower estimated abdominal circumference trajectory compared to fast (25616 [IQR: 11656.0 to 35341.0] vs. 14753.8 [IQR: 8565.4 to 24308.1], p < 0.001) and maternal adiponectin levels were lower in fetuses on a slower estimated abdominal circumference trajectory compared to a fast trajectory (22.4 [IQR: 13.6 to 35.9] vs. 27.6 [IQR: 17.6 to 46.3], p=0.027). No associations were noted with inflammatory markers. No associations were identified between fetal growth trajectories and growth markers at 5 years of age. Conclusions This study shows that male sex is associated with an accelerated estimated weight trajectory. Furthermore, high leptin and low adiponectin in maternal serum in late gestation are associated with a slower fetal growth trajectory. No associations were identified with blood growth markers after pregnancy.
Collapse
|
40
|
Kothari C, Diorio C, Durocher F. The Importance of Breast Adipose Tissue in Breast Cancer. Int J Mol Sci 2020; 21:ijms21165760. [PMID: 32796696 PMCID: PMC7460846 DOI: 10.3390/ijms21165760] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue is a complex endocrine organ, with a role in obesity and cancer. Adipose tissue is generally linked to excessive body fat, and it is well known that the female breast is rich in adipose tissue. Hence, one can wonder: what is the role of adipose tissue in the breast and why is it required? Adipose tissue as an organ consists of adipocytes, an extracellular matrix (ECM) and immune cells, with a significant role in the dynamics of breast changes throughout the life span of a female breast from puberty, pregnancy, lactation and involution. In this review, we will discuss the importance of breast adipose tissue in breast development and its involvement in breast changes happening during pregnancy, lactation and involution. We will focus on understanding the biology of breast adipose tissue, with an overview on its involvement in the various steps of breast cancer development and progression. The interaction between the breast adipose tissue surrounding cancer cells and vice-versa modifies the tumor microenvironment in favor of cancer. Understanding this mutual interaction and the role of breast adipose tissue in the tumor microenvironment could potentially raise the possibility of overcoming breast adipose tissue mediated resistance to therapies and finding novel candidates to target breast cancer.
Collapse
Affiliation(s)
- Charu Kothari
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1T 1C2, Canada;
- Cancer Research Centre, CHU de Quebec Research Centre, Quebec, QC G1V 4G2, Canada;
| | - Caroline Diorio
- Cancer Research Centre, CHU de Quebec Research Centre, Quebec, QC G1V 4G2, Canada;
- Department of Preventive and Social Medicine, Faculty of Medicine, Laval University, Quebec, QC G1T 1C2, Canada
| | - Francine Durocher
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1T 1C2, Canada;
- Cancer Research Centre, CHU de Quebec Research Centre, Quebec, QC G1V 4G2, Canada;
- Correspondence: ; Tel.: +1-(418)-525-4444 (ext. 48508)
| |
Collapse
|
41
|
Bahia W, Soltani I, Haddad A, Radhouani A, Mahdhi A, Ferchichi S, Almawi WY. Contribution of ADIPOQ Variants to the Genetic Susceptibility of Recurrent Pregnancy Loss. Reprod Sci 2020; 28:263-270. [PMID: 32748222 DOI: 10.1007/s43032-020-00274-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 07/23/2020] [Indexed: 02/02/2023]
Abstract
Adiponectin is a hormone implicated in regulating energy, lipid, and glucose metabolism and is encoded by the ADIPOQ gene. ADIPOQ variants can regulate the circulating levels of adiponectin. Irregular adiponectin concentrations have been associated with numerous reproductive diseases including recurrent pregnancy loss (RPL). The main objective of this study was to determine whether the 14 selected polymorphisms of the ADIPOQ gene are linked with RPL. The retrospective case-control study comprised a total of 332 women with RPL, adjusted as more than three consecutive abortions of unknown etiology, and 286 healthy controls. They were genotyped for the ADIPOQ variants using allele exclusion method on real-time PCR. Significantly higher rs1501299 minor allele frequencies (MAF) and lower rs2241767 and rs2241766 MAF were seen among RPL women, thereby assigning disease susceptibility and protective aspect to the mentioned variants, respectively. Different associations of ADIPOQ genotypes with RPL were noticed according to the genetic model exploited: rs1501299 and rs2241767 were significantly linked with RPL under the three models, while rs17366568 and rs2241766 were associated with RPL under codominant and dominant models, and rs7649121 was related to RPL under the dominant and recessive models. rs4632532 was linked according to the recessive model only. Based on LD pattern, 2-haplotype blocks were specified. Reduced frequency of AGG and GAGG and increased frequency of TAAG were noted in cases, compared with controls, hence indicating these haplotypes as RPL-protective and RPL-susceptible, respectively. These results support a significant role of ADIPOQ as an RPL candidate locus.
Collapse
Affiliation(s)
- Wael Bahia
- Research Unit of Clinical and Molecular Biology (UR17ES29), Department of Biochemistry, Faculty of Pharmacy, University of Monastir, 5000, Monastir, Tunisia.
| | - Ismael Soltani
- Molecular and Cellular Hematology Laboratory, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Anis Haddad
- Department of Obstetrics and Gynecology, Fattouma Bourguiba University Hospital, Monastir, Tunisia
| | - Assala Radhouani
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Abdelkarim Mahdhi
- Laboratory of Analysis, Treatment and Valorization of Pollutants of the Environment and Products, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Salima Ferchichi
- Research Unit of Clinical and Molecular Biology (UR17ES29), Department of Biochemistry, Faculty of Pharmacy, University of Monastir, 5000, Monastir, Tunisia
| | - Wassim Y Almawi
- College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
42
|
Associations between Maternal and Fetal Levels of Total Adiponectin, High Molecular Weight Adiponectin, Selected Somatomedins, and Birth Weight of Infants of Smoking and Non-Smoking Mothers. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17134781. [PMID: 32635165 PMCID: PMC7370151 DOI: 10.3390/ijerph17134781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/23/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022]
Abstract
The aim of the study was to determine the relationships between maternal smoking, total adiponectin, high molecular weight adiponectin (HMW adiponectin), selected somatomedins, and the birth weight of newborns. A total of 78 women with a healthy, singleton pregnancy, 41 active smokers and 37 non-smokers, and their offspring were studied. Total and HMW adiponectin, insulin-like growth factor I (IGF-I), and insulin-like growth factor binding protein-1 (IGFBP-1) and 2 (IGFBP-2) were determined in maternal and cord blood by enzyme-link immunosorbent assay. Serum levels of total and HMW adiponectin were lower in smokers compared to the tobacco abstinent in both the mothers (p = 0.013; p = 0.006) and the infants (p = 0.001; p = 0.047). In smoking women and their children, serum concentrations of IGF-I were significantly lower (p = 0.014; p = 0.042), IGFBP-1 significantly higher (p = 0.009; p = 0.039), and IGFBP-2 did not differ from that observed in the non-smoking group. In multivariate analysis performed on the whole group of mothers, the highest impact of serum cotinine and IGFBP-2 levels were indicated for adiponectin and cotinine and the number of cigarettes/day for HMW adiponectin concentration. In correlation analysis estimated separately for smokers and non-smokers, neonatal birth weight was positively associated with total and HMW adiponectin concentrations in umbilical cord blood. Birth weight was also inversely associated with IGFBP-1 and positively correlated with IGF-I levels in maternal serum as well as in cord blood (r = −0.317, p = 0.005; r = −0.294, p = 0.004; r = 0.245, p = 0.031; r = 0.271, p = 0.009, respectively). The present study showed the levels of total and HMW adiponectin in umbilical cord blood may have a significant effect on fetal development. Both IGF-I and IGFBP-1 concentrations also play an essential role in fetal growth, which is an important predictor of birth weight. Cigarette smoking during pregnancy negatively affected adiponectin and the insulin growth factor profile in the serum of women and the cord blood and may be the reason for the lower birth weight of the smokers newborns compared with the nonsmokers offspring.
Collapse
|
43
|
Kumar TR, Reusch JEB, Kohrt WM, Regensteiner JG. Sex Differences Across the Lifespan: A Focus on Cardiometabolism. J Womens Health (Larchmt) 2020; 29:899-909. [PMID: 32423340 DOI: 10.1089/jwh.2020.8408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Women's health and sex differences research remain understudied. In 2016, to address the topic of sex differences, the Center for Women' s Health Research (CWHR) at the University of Colorado (cwhr@ucdenver.edu) held its inaugural National Conference, "Sex Differences Across the Lifespan: A Focus on Metabolism" and published a report summarizing the presentations. Two years later, in 2018, CWHR organized the 2nd National Conference. The research presentations and discussions from the 2018 conference also addressed sex differences across the lifespan with a focus on cardiometabolism and expanded the focus by including circadian physiology and effects of sleep on cardiometabolic health. Over 100 participants, including basic scientists, clinicians, policymakers, advocacy group leaders, and federal agency leadership participated. The meeting proceedings reveal that although exciting advances in the area of sex differences have taken place, significant questions and gaps remain about women's health and sex differences in critical areas of health. Identifying these gaps and the subsequent research that will result may lead to important breakthroughs.
Collapse
Affiliation(s)
- T Rajendra Kumar
- Department of Obstetrics and Gynecology and University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jane E B Reusch
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Center for Women's Health Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Veterans Administration Eastern Colorado Health Care System, Denver, Colorado, USA
| | - Wendy M Kohrt
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Center for Women's Health Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Judith G Regensteiner
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Center for Women's Health Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
44
|
Abstract
Worldwide obesity is increasing at an alarming rate in children and adolescents, with the consequent emergence of co-morbidities. Moreover, the maternal environment during pregnancy plays an important role in obesity, contributing to transgenerational transmission of the same and metabolic dysfunction. White adipose tissue represents a prime target of metabolic programming induced by maternal milieu. In this article, we review adipose tissue physiology and development, as well as maternal influences during the perinatal period that may lead to obesity in early postnatal life and adulthood. First, we describe the adipose tissue cell composition, distribution and hormonal action, together with the evidence of hormonal factors participating in fetal/postnatal programming. Subsequently, we describe the critical periods of adipose tissue development and the relationship of gestational and early postnatal life with healthy fetal adipose tissue expansion. Furthermore, we discuss the evidence showing that adipose tissue is an important target for nutritional, hormonal and epigenetic signals to modulate fetal growth. Finally, we describe nutritional, hormonal, epigenetic and microbiome changes observed in maternal obesity, and whether their disruption alters fetal growth and adiposity. The presented evidence supports the developmental origins of health and disease concept, which proposes that the homeostatic system is affected during gestational and postnatal development, impeding the ability to regulate body weight after birth, thereby resulting in adult obesity. Consequently, we anticipate that promoting a healthy early-life programming of adipose tissue and increasing the knowledge of the mechanisms by which maternal factors affect the health of future generations may offer novel strategies for explaining and addressing worldwide health problems such as obesity.
Collapse
|
45
|
Chattrapiban T, Smit HA, Wijga AH, Brunekreef B, Vonk JM, Gehring U, van Rossem L. The joint effect of maternal smoking during pregnancy and maternal pre-pregnancy overweight on infants' term birth weight. BMC Pregnancy Childbirth 2020; 20:132. [PMID: 32106826 PMCID: PMC7047372 DOI: 10.1186/s12884-020-2816-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 02/17/2020] [Indexed: 11/20/2022] Open
Abstract
Background It is well known that maternal smoking during pregnancy and maternal pre-pregnancy overweight have opposite effects on the infants’ birth weight. We report on the association of the combination between both risk factors and the infants’ birth weight. Methods We studied 3241 infants born at term in the PIAMA birth cohort. Maternal smoking during pregnancy and pre-pregnancy height and weight were self-reported. Multivariable regression analysis was performed to assess the associations between infants of mothers who only smoked during pregnancy, who only had pre-pregnancy overweight and who had both risk factors simultaneously, on term birth weight and the risk of being SGA or LGA. Results Of 3241 infants, 421 infants (13%) were born to smoking, non-overweight mothers, 514 (15.8%) to non-smoking, overweight mothers, 129 (4%) to smoking and overweight mothers and 2177 (67%) to non-smoking, non-overweight mothers (reference group). Infants of mothers who smoked and also had pre-pregnancy overweight had similar term birth weight (− 26.6 g, 95%CI: − 113.0, 59.8), SGA risk (OR = 1.06, 95%CI: 0.56, 2.04), and LGA risk (OR = 1.09, 95%CI: 0.61, 1.96) as the reference group. Conclusions Our findings suggested that the effects of maternal smoking during pregnancy and maternal pre-pregnancy overweight on infants’ birth weight cancel each other out. Therefore, birth weight may not be a good indicator of an infant’s health status in perinatal practice because it may mask potential health risks due to these maternal risk factors when both present together.
Collapse
Affiliation(s)
- Thanin Chattrapiban
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, STR 6.118, Utrecht, 3508, GA, The Netherlands
| | - Henriette A Smit
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, STR 6.118, Utrecht, 3508, GA, The Netherlands
| | - Alet H Wijga
- Center for Prevention and Health Services Research, National Institute of Public Health and the Environment, P.O Box 1, 3720, BA, Bilthoven, The Netherlands
| | - Bert Brunekreef
- Institute for Risk Assessment Science (IRAS), Utrecht University, Yalelaan 2, 3584, CM, Utrecht, The Netherlands
| | - Judith M Vonk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, The Netherlands
| | - Ulrike Gehring
- Institute for Risk Assessment Science (IRAS), Utrecht University, Yalelaan 2, 3584, CM, Utrecht, The Netherlands
| | - Lenie van Rossem
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, STR 6.118, Utrecht, 3508, GA, The Netherlands. .,Department of Obstretrics and Gynecology, Erasmus MC University Medical Center, Dr. Molewaterplein 50, 3015, GD, Rotterdam, the Netherlands.
| |
Collapse
|
46
|
Insulin Resistance in Pregnancy: Implications for Mother and Offspring. CONTEMPORARY ENDOCRINOLOGY 2020. [DOI: 10.1007/978-3-030-25057-7_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
47
|
Arroyo-Jousse V, Jaramillo A, Castaño-Moreno E, Lépez M, Carrasco-Negüe K, Casanello P. Adipokines underlie the early origins of obesity and associated metabolic comorbidities in the offspring of women with pregestational obesity. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165558. [PMID: 31654701 DOI: 10.1016/j.bbadis.2019.165558] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 02/07/2023]
Abstract
Maternal pregestational obesity is a well-known risk factor for offspring obesity, metabolic syndrome, cardiovascular disease and type 2 diabetes. The mechanisms by which maternal obesity can induce alterations in fetal and later neonatal metabolism are not fully elucidated due to its complexity and multifactorial causes. Two adipokines, leptin and adiponectin, are involved in fetal and postnatal growth trajectories, and both are altered in women with pregestational obesity. The placenta synthesizes leptin, which goes mainly to the maternal circulation and in lesser amount to the developing fetus. Maternal pregestational obesity and hyperleptinemia are associated with placental dysfunction and changes in nutrient transporters which directly affect fetal growth and development. By the other side, the embryo can produce its own leptin from early in development, which is associated to fetal weight and adiposity. Adiponectin, an insulin-sensitizing adipokine, is downregulated in maternal obesity. High molecular weight (HMW) adiponectin is the most abundant form and with most biological actions. In maternal obesity lower total and HMW adiponectin levels have been described in the mother, paralleled with high levels in the umbilical cord. Several studies have found that cord blood adiponectin levels are related with postnatal growth trajectories, and it has been suggested that low adiponectin levels in women with pregestational obesity enhance placental insulin sensitivity and activation of placental amino acid transport systems, supporting fetal overgrowth. The possible mechanisms by which maternal pregestational obesity, focusing in the actions of leptin and adiponectin, affects the fetal development and postnatal growth trajectories in their offspring are discussed.
Collapse
Affiliation(s)
| | | | | | - M Lépez
- School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - K Carrasco-Negüe
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - P Casanello
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile; Department of Neonatology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
48
|
O’Brien CM, Louise J, Deussen A, Dodd JM. Maternal cardiometabolic markers are associated with fetal growth: a secondary exploratory analysis of the LIMIT randomised trial. BMC Endocr Disord 2019; 19:97. [PMID: 31601214 PMCID: PMC6785858 DOI: 10.1186/s12902-019-0416-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/07/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To determine the association between maternal cardiometabolic and inflammatory markers with measures of fetal biometry and adiposity. METHODS Women included in this exploratory analysis were randomised to the 'Standard Care' group (N = 911) from the LIMIT randomised trial involving a total of 2212 pregnant women who were overweight or obese (ACTRN12607000161426, Date of registration 9/03/2007, prospectively registered). Fetal biometry including abdominal circumference (AC), estimated fetal weight (EFW), and adiposity measurements (mid-thigh fat mass, subscapular fat mass, abdominal fat mass) were obtained from ultrasound assessments at 28 and 36 weeks' gestation. Maternal markers included C reactive protein (CRP), leptin and adiponectin concentrations, measured at 28 and 36 weeks' gestation and fasting triglycerides and glucose concentrations measured at 28 weeks' gestation. RESULTS There were negative associations identified between maternal serum adiponectin and fetal ultrasound markers of biometry and adiposity. After adjusting for confounders, a 1-unit increase in log Adiponectin was associated with a reduction in the mean AC z score [- 0.21 (- 0.35, - 0.07), P = 0.004] and EFW [- 0.23 (- 0.37, - 0.10), P < 0.001] at 28 weeks gestation. Similarly, a 1-unit increase in log Adiponectin was association with a reduction in the mean AC z score [- 0.30 (- 0.46, - 0.13), P < 0.001] and EFW [- 0.24 (- 0.38, - 0.10), P < 0.001] at 36 weeks gestation. There were no consistent associations between maternal cardiometabolic and inflammatory markers with measurements of fetal adiposity. CONCLUSION Adiponectin concentrations are associated with measures of fetal growth. Our findings contribute to further understanding of fetal growth in the setting of women who are overweight or obesity.
Collapse
Affiliation(s)
- Cecelia M. O’Brien
- School of Paediatrics and Reproductive Health, and Robinson Research Institute, University of Adelaide, Adelaide, Australia
- Women’s and Children’s Hospital, The University of Adelaide, 72 King William Road, North Adelaide, SA 5006 Australia
| | - Jennie Louise
- School of Paediatrics and Reproductive Health, and Robinson Research Institute, University of Adelaide, Adelaide, Australia
- School of Public Health, University of Adelaide, Adelaide, Australia
| | - Andrea Deussen
- School of Paediatrics and Reproductive Health, and Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Jodie M. Dodd
- School of Paediatrics and Reproductive Health, and Robinson Research Institute, University of Adelaide, Adelaide, Australia
- Department of Perinatal Medicine, Women’s and Babies Division, Women’s and Children’s Hospital, Adelaide, Australia
| |
Collapse
|
49
|
Fazeli Daryasari SR, Tehranian N, Kazemnejad A, Razavinia F, Tork Tatari F, Pahlavan F. Adiponectin levels in maternal serum and umbilical cord blood at birth by mode of delivery: relationship to anthropometric measurements and fetal sex. BMC Pregnancy Childbirth 2019; 19:344. [PMID: 31590631 PMCID: PMC6781401 DOI: 10.1186/s12884-019-2460-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 08/15/2019] [Indexed: 02/04/2023] Open
Abstract
Background The rate of cesarean section is increasing worldwide. Adiponectin is a hormone related to anti-inflammatory and anti-atherogenic effects; and it′s concentrations may change in response to inflammatory situations including surgical intervention. The aim of the current study was to investigate serum adiponectin levels in maternal and umbilical cord blood according to different modes of delivery and their relationship with anthropometric measurements and fetal sex. Methods The study population initially comprised 90 healthy pregnant women referred to the teaching hospital. Eventually, 40 participants in the vaginal delivery group and 35 subjects in the cesarean delivery group were recruited in to the study. Umbilical cord blood and maternal serum samples were analyzed according to the standard protocol from the manufacturer. The collected data were analyzed using SPSS-16 software. P-value < 0.05 was considered as the significance level for all tests. Results Our results indicated a significant association between maternal adiponectin and the mode of delivery, with adiponectin levels significantly higher in vaginal delivery compared to cesarean section (P < 0.001). However, no difference was found in umbilical cord blood adiponectin between the two groups (P = 0.51). A significant positive correlation was found between maternal serum adiponectin in the first day after birth and umbilical cord blood adiponectin in the vaginal delivery group (P = 0.007). Nevertheless, this correlation was not statistically significant in the cesarean delivery group (P = 0.62). There was also no significant correlation between fetal sex and anthropometric measurements with maternal adiponectin (P = 0.44) and umbilical cord blood adiponectin (P = 0.86). Conclusions The result of the current study revealed that maternal adiponectin concentration was significantly higher in vaginal delivery compared to cesarean section, which might be due to the increased levels of maternal adiponectin release during labor.
Collapse
Affiliation(s)
- Seyedeh Razieh Fazeli Daryasari
- Department of Midwifery & Reproductive Health, Faculty of Medical Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, P.O.Box: 14115-111, Tehran, Iran
| | - Najmeh Tehranian
- Department of Midwifery & Reproductive Health, Faculty of Medical Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, P.O.Box: 14115-111, Tehran, Iran.
| | - Anoshirvan Kazemnejad
- Department of Biostatistics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Razavinia
- Department of Midwifery & Reproductive Health, Faculty of Medical Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, P.O.Box: 14115-111, Tehran, Iran
| | - Fatemeh Tork Tatari
- Department of Midwifery & Reproductive Health, Faculty of Medical Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, P.O.Box: 14115-111, Tehran, Iran
| | - Fattaneh Pahlavan
- Department of Midwifery & Reproductive Health, Faculty of Medical Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, P.O.Box: 14115-111, Tehran, Iran
| |
Collapse
|
50
|
Longitudinal changes in adipokines and free leptin index during and after pregnancy in women with obesity. Int J Obes (Lond) 2019; 44:675-683. [PMID: 31551485 DOI: 10.1038/s41366-019-0452-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 08/30/2019] [Accepted: 09/06/2019] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Detailed data on adipokines and body composition during and after pregnancy in women of different BMI categories are lacking. Furthermore, adipokine regulation during pregnancy and the factors contributing to gestational insulin resistance are not completely understood. The objective was to longitudinally determine adipokine levels, body composition, and insulin sensitivity during and after pregnancy in women of healthy weight (HW) and with obesity (OB), and identify factors associated with insulin resistance. DESIGN Women (30 HW, 19 OB) underwent blood sampling and body composition examination, by air-displacement plethysmography, longitudinally during pregnancy (trimesters 1, 2, 3) and after pregnancy (6, 12, 18 months postpartum). Serum leptin, soluble leptin receptor (sOB-R), and adiponectin levels were measured and free leptin index (FLI) and homeostatic model assessment of insulin resistance (HOMA-IR) determined. RESULTS Fat mass and leptin increased during pregnancy in the HW (p < 0.01) but not in the OB group. sOB-R increased during pregnancy in both groups (p < 0.001). Thus, FLI was unchanged in HW throughout pregnancy but reduced in OB (p = 0.001), although consistently higher in OB. Adiponectin decreased in both groups during pregnancy (p < 0.001 for HW, p = 0.01 for OB). After pregnancy, adiponectin increased in both groups, but more markedly in OB where it reached trimester 1 levels. Multivariable regression identified FLI as the variable most strongly associated with HOMA-IR in all trimesters, but not after pregnancy. CONCLUSIONS Leptin, sOB-R, adiponectin, and FLI undergo marked changes during and after pregnancy with differences in women of different BMI. We suggest that leptin activity is regulated by its soluble receptor and that this is an important factor for optimizing fat mass and insulin sensitivity during pregnancy.
Collapse
|