1
|
Fasoulopoulos A, Varras M, Varra FN, Philippou A, Myoteri D, Varra VK, Kouroglou E, Gryparis A, Papadopetraki A, Vlachos I, Papadopoulos K, Koutsilieris M, Konstantinidou AE. Expression of the IGF‑1Ea isoform in human placentas from third trimester normal and idiopathic intrauterine growth restriction singleton pregnancies: Correlations with clinical and histopathological parameters. Mol Med Rep 2025; 31:69. [PMID: 39791214 PMCID: PMC11751665 DOI: 10.3892/mmr.2025.13434] [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: 05/21/2024] [Accepted: 10/17/2024] [Indexed: 01/12/2025] Open
Abstract
Intrauterine growth restriction (IUGR) is the second most common obstetric complication after preterm labor. Appropriate trophoblast differentiation and placental structure, growth and function are key for the maintenance of pregnancy and normal fetal growth, development and survival. Extravillous trophoblast cell proliferation, migration and invasion are regulated by molecules produced by the fetomaternal interface, including autocrine factors produced by the trophoblast, such as insulin‑like growth factor (IGF)‑1. The aim of the present study was to investigate expression patterns of IGF‑1Ea isoform in IUGR placenta compared with appropriate for gestational age (AGA) pregnancies. Placental frozen tissues were collected from 13 AGA and 15 IUGR third trimester pregnancies for detection of IGF‑1Ea mRNA expression using reverse transcription‑quantitative PCR. Formalin‑fixed paraffin‑embedded samples from 15 AGA and 47 IUGR pregnancies were analyzed immunohistochemically for the identification and localization of the IGF‑1Ea peptide and comparison of clinical and histopathological parameters. To the best of our knowledge, the present study is the first to show IGF‑1Ea expression in third trimester human placenta. The results indicated that similar IGF‑1Ea mRNA expression levels were present in placental specimens from both groups. Cytoplasmic IGF‑1Ea expression was localized in the perivillous syncytiotrophoblast, extravillous trophoblast and endothelium of the villous and decidual vessels in both groups. No significant difference in the scores and intensity of IGF‑1Ea expression in perivillous syncytiotrophoblasts were noted in the IUGR vs. AGA pregnancies. Most IUGR cases showed negative IGF‑1Ea expression in the extravillous trophoblast, whereas AGA pregnancies showed predominantly positive immunostaining. A sex‑specific expression pattern was noted in the extravillous trophoblast, with negative IGF‑1Ea expression in the placentas of female IUGR cases. Additionally, positive immunostaining for IGF‑1Ea peptide in fetal villous and maternal decidual vessels, was more frequently observed in the IUGR group compared with AGA. In conclusion, no difference in total IGF‑1Ea mRNA placental expression was observed between IUGR and AGA pregnancies, likely due to heterogeneity of histological structures expressing this isoform. Negative IGF‑1Ea immunohistological expression in the extravillous trophoblast from IUGR placentas, associated with histological changes of maternal malperfusion, may reflect the involvement of this isoform in defective placentation. The presence of IGF‑1Ea peptide in the endothelium of the villous vessels in IUGR placentas may indicate a reactive autocrine regulation to compensate for malperfused villi in IUGR pregnancy by regulating angiogenesis and vasodilation. The observed sex differences in IGF‑1Ea expression between IUGR and AGA placentas may indicate interactions between sex hormones and selective IGF‑1 binding proteins in regulating IGF‑1Ea synthesis; however, this requires further elucidation.
Collapse
Affiliation(s)
- Apostolos Fasoulopoulos
- Fourth Obstetrics and Gynecology Department, ‘Elena Venizelou’ General Hospital of Athens, 11521 Athens, Greece
| | - Michail Varras
- Fourth Obstetrics and Gynecology Department, ‘Elena Venizelou’ General Hospital of Athens, 11521 Athens, Greece
| | - Fani-Niki Varra
- Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Anastasios Philippou
- Department of Physiology, Medical School, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Despina Myoteri
- Department of Pathology, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | | | - Evgenia Kouroglou
- Fourth Obstetrics and Gynecology Department, ‘Elena Venizelou’ General Hospital of Athens, 11521 Athens, Greece
| | - Alexandros Gryparis
- Department of Speech and Language Therapy, University of Ioannina, 45500 Ioannina, Greece
| | - Argyro Papadopetraki
- Department of Physiology, Medical School, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Iakovos Vlachos
- Department of Pathology, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Konstantinos Papadopoulos
- Department of Pathology, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Anastasia Evangelia Konstantinidou
- Department of Pathology, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
- First Department of Pathology, Unit of Pediatric-Perinatal Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
2
|
Luo Y, An C, Zhong K, Zhou P, Li D, Liu H, Guo Q, Wei W, Pan H, Min Z, Li R, Yu Y, Fan Y. Exploring the impacts of senescence on implantation and early embryonic development using totipotent cell-derived blastoids. J Adv Res 2025; 68:115-129. [PMID: 38402947 DOI: 10.1016/j.jare.2024.02.011] [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: 09/18/2023] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 02/27/2024] Open
Abstract
INTRODUCTION Advanced maternal age is associated with reduced implantation and pregnancy rates, yet the underlying mechanisms remain poorly understood, and research models are limited. OBJECTIVES Here, we aim to elucidate the impacts of senescence on implantation ability by employing blastoids to construct a novel research model. METHODS We used a novel three-dimensional system with totipotent blastomere-like cells (TBLCs) to construct TBL-blastoids and established senescence-related embryo models derived from oxidative stress-induced TBLCs. RESULTS Morphological and transcriptomic analyses revealed that TBL-blastoids exhibited characteristic blastocyst morphology, cell lineages, and a higher consistency in developmental rate. TBL-blastoids demonstrated the ability to develop into postimplantation structures in vitro and successfully implanted into mouse uteri, inducing decidualization and forming embryonic tissues. Importantly, senescence impaired the implantation potential of TBL-blastoids, effectively mimicking the impaired implantation ability and reduced pregnancy rates associated with advanced age. Furthermore, analysis of differentially expressed genes (DEGs) in human homologous deciduae revealed enrichment in multiple fertility-related diseases and other complications of pregnancy. The genes implicated in these diseases and the common DEGs identified in the lineage-like cells of the two types of TBL-blastoids and deciduae may represent potential targets for addressing impaired implantation potential. CONCLUSION These results unveiled that TBL blastoids are an improved model for investigating implantation and early postimplantation, offering valuable insights into pregnancy-related disorders in women with advanced age and potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Yuxin Luo
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing 100191, China
| | - Chenrui An
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Ke Zhong
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Ping Zhou
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing 100191, China
| | - Dan Li
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Hui Liu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Qing Guo
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Wei Wei
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Hen Pan
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing 100191, China
| | - Zheying Min
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China.
| | - Rong Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing 100191, China.
| | - Yang Yu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China.
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China.
| |
Collapse
|
3
|
Schwartz NE, Schmill MP, Cadney MD, Castro AA, Hillis DA, McNamara MP, Rashid JO, Lampman W, DeLaCruz DF, Tran BD, Trutalli NL, Garland T. Maternal exercise opportunity before, during, and after pregnancy alters maternal care behavior and offspring development and survival, but has few effects on offspring physical activity or body composition. Physiol Behav 2024; 291:114752. [PMID: 39549866 DOI: 10.1016/j.physbeh.2024.114752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
Early-life experiences, especially during critical periods of development and growth, can have long-lasting effects on adult phenotypes. Parents are a crucial part of the offspring early-life environment, particularly in mammals (e.g., via pregnancy), and parental behaviors (e.g., maternal exercise) can modify the early-life environment experienced by offspring. Such changes might be beneficial or detrimental, depending on how they affect offspring development and growth or interact with other key parental behaviors (e.g., nursing). We used mice from a long-term artificial selection experiment for high voluntary wheel-running behavior to determine whether maternal exercise opportunity affected (1) maternal physical activity, (2) maternal care behavior, or (3) offspring physical activity and body composition. Eighty prospective dams (40 from 4 selectively bred High Runner [HR] lines and 40 from 4 non-selected Control [CON] lines) were housed with continuous wheel access starting two weeks prior to breeding and ending 10 days postpartum, after which dams were housed without wheels until offspring weaning (21 days postpartum). An additional 100 dams (50 HR, 50 CON) were housed without wheels. Prospective dams from HR lines ran more revolutions/day (mainly by running faster) than those from CON lines when individually housed and in the days leading up to, but not after, birth. During postpartum days 1-5, HR and CON dams with wheels tended to exhibit less maternal behavior than those without (PWheel = 0.0672). During post-partum days 6-10, HR dams with wheels continued to exhibit less maternal behavior than those without, whereas CON dams with wheels exhibited more than those without (PLinetype*Wheel = 0.0218). The proportion of dams giving birth did not differ among groups. However, CON dams with wheels were less likely to have litter death between birth and weaning than those without wheels, whereas the opposite was true for HR dams (PLinetype*Wheel = 0.0447). Both HR and CON dams with wheels had litters with a higher proportion of females at weaning than those without wheels (PWheel = 0.0129). Maternal wheel access had few statistically significant effects on offspring, but may have resulted in developmental delays (e.g., delayed eye opening and decreased lean mass at weaning and sexual maturity). Additionally, maternal wheel access and sex may have interacted to affect wheel-running distance (PSex*Wheel = 0.0683) and duration (PSex*Wheel = 0.0926); female offspring from dams with wheels ran fewer revolutions per day, by running fewer minutes per day, than from dams without wheels, whereas males ran more. However, maternal exercise had no statistically significant effects on offspring food consumption (mass-adjusted), home-cage activity, open-field behavior, the reproductive characteristics of offspring, their adult body composition, nor relative organ masses; nor did maternal wheel access have statistically significant effects on grand-offspring food consumption, body composition or voluntary exercise behavior. Overall, our results provide some support for maternal exercise opportunity altering maternal care behavior. Altered maternal care could explain the observed trends in offspring survival, development, and voluntary exercise behavior. However, these effects did not have apparent long-lasting impacts on offspring or grand-offspring body composition or reproductive characteristics.
Collapse
Affiliation(s)
- Nicole E Schwartz
- Department of Evolution, Ecology, and Organismal Biology, University of California - Riverside, Riverside, CA, USA.
| | - Margaret P Schmill
- Neuroscience Graduate Program, University of California - Riverside, Riverside, CA, USA
| | - Marcell D Cadney
- Department of Evolution, Ecology, and Organismal Biology, University of California - Riverside, Riverside, CA, USA
| | - Alberto A Castro
- Department of Evolution, Ecology, and Organismal Biology, University of California - Riverside, Riverside, CA, USA
| | - David A Hillis
- Genetics, Genomics, and Bioinformatics Graduate Program, University of California - Riverside, Riverside, CA, USA
| | - Monica P McNamara
- Department of Evolution, Ecology, and Organismal Biology, University of California - Riverside, Riverside, CA, USA
| | - Jaanam O Rashid
- Department of Evolution, Ecology, and Organismal Biology, University of California - Riverside, Riverside, CA, USA
| | - William Lampman
- Department of Evolution, Ecology, and Organismal Biology, University of California - Riverside, Riverside, CA, USA
| | - Dorothea F DeLaCruz
- Department of Evolution, Ecology, and Organismal Biology, University of California - Riverside, Riverside, CA, USA
| | - Bao D Tran
- Department of Evolution, Ecology, and Organismal Biology, University of California - Riverside, Riverside, CA, USA
| | - Natalie L Trutalli
- Department of Evolution, Ecology, and Organismal Biology, University of California - Riverside, Riverside, CA, USA
| | - Theodore Garland
- Department of Evolution, Ecology, and Organismal Biology, University of California - Riverside, Riverside, CA, USA
| |
Collapse
|
4
|
Al Zein M, Akomolafe AF, Mahmood FR, Khrayzat A, Sahebkar A, Pintus G, Kobeissy F, Eid AH. Leptin is a potential biomarker of childhood obesity and an indicator of the effectiveness of weight-loss interventions. Obes Rev 2024; 25:e13807. [PMID: 39044542 DOI: 10.1111/obr.13807] [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: 10/08/2023] [Revised: 05/12/2024] [Accepted: 07/02/2024] [Indexed: 07/25/2024]
Abstract
Childhood obesity represents a significant public health concern, imposing a substantial burden on the healthcare system. Furthermore, weight-loss programs often exhibit reduced effectiveness in adults who have a history of childhood obesity. Therefore, early intervention against childhood obesity is imperative. Presently, the primary method for diagnosing childhood obesity relies on body mass index (BMI), yet this approach has inherent limitations. Leptin, a satiety hormone produced by adipocytes, holds promise as a superior tool for predicting both childhood and subsequent adulthood obesity. In this review, we elucidate the tools employed for assessing obesity in children, delve into the biological functions of leptin, and examine the factors governing its expression. Additionally, we discuss maternal and infantile leptin levels as predictors of childhood obesity. By exploring the relationship between leptin levels and weight loss, we present leptin as a potential indicator of the effectiveness of obesity interventions.
Collapse
Affiliation(s)
- Mohammad Al Zein
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | | | - Fathima R Mahmood
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ali Khrayzat
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
5
|
Valencia-Ortega J, Castillo-Santos A, Molerés-Orduña M, Solis-Paredes JM, Saucedo R, Estrada-Gutierrez G, Camacho-Arroyo I. Influence of Maternal Adipokines on Anthropometry, Adiposity, and Neurodevelopmental Outcomes of the Offspring. Int J Mol Sci 2024; 25:11655. [PMID: 39519203 PMCID: PMC11547085 DOI: 10.3390/ijms252111655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/17/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Pregnancy is distinguished by a multitude of intricate interactions between the mother and the new individual, commencing at implantation and persisting until the maturation and integration of the fetal apparatus and systems. The physiological increase in fat mass during pregnancy and the association of maternal obesity with adverse neonatal outcomes have directed attention to the study of maternal adipokines as participants in fetal development. Interestingly, maternal concentrations of certain adipokines such as adiponectin, leptin, tumor necrosis factor-alpha, and interleukin-6 have been found to be associated with offspring anthropometry and adiposity at birth and at three months of age, even with neurodevelopmental alterations later in life. This is partly explained by the functions of these adipokines in the regulation of maternal metabolism and placental nutrient transport. This review compiles, organizes, and analyzes the most relevant studies on the association between maternal adipokines with anthropometry, adiposity, and neurodevelopmental outcomes of the offspring. Furthermore, it proposes the underlying mechanisms involved in this association.
Collapse
Affiliation(s)
- Jorge Valencia-Ortega
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, Mexico;
| | - Andrea Castillo-Santos
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (A.C.-S.); (M.M.-O.); (J.M.S.-P.)
| | - Miranda Molerés-Orduña
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (A.C.-S.); (M.M.-O.); (J.M.S.-P.)
| | - Juan Mario Solis-Paredes
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (A.C.-S.); (M.M.-O.); (J.M.S.-P.)
| | - Renata Saucedo
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Guadalupe Estrada-Gutierrez
- Department of Immunobiochemistry, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico;
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, Mexico;
| |
Collapse
|
6
|
Suwaydi MA, Lai CT, Warden AH, Perrella SL, McEachran JL, Wlodek ME, Geddes DT, Gridneva Z. Investigation of Relationships between Intakes of Human Milk Total Lipids and Metabolic Hormones and Infant Sex and Body Composition. Nutrients 2024; 16:2739. [PMID: 39203875 PMCID: PMC11357482 DOI: 10.3390/nu16162739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
Human milk (HM) composition, including metabolic hormones and lipids, is influenced by various factors, including lactation stage and, potentially, infant sex, which may affect infant body composition (BC) development. We aimed to: (a) characterize the longitudinal concentration and intake profiles of HM leptin, adiponectin, insulin, and total lipids; (b) determine if their concentrations and intakes differ by infant sex; and (c) explore the intakes relationships with the development of infant BC. Milk samples (n = 501) were collected from 82 mother-infant dyads during the first 6 months postpartum. Infant 24 h HM intake was measured, and the average cumulative HM component intakes were calculated. The statistical analysis used linear mixed modeling. Intakes of HM leptin, adiponectin, insulin, and total lipids increased to 1 month postpartum and then remained stable. HM intake and total lipids intake but not hormone intakes were positively associated with infant BC (fat-free mass, fat-free mass index, fat mass, fat mass index, percentage fat mass, and fat mass to fat-free mass ratio). HM component concentrations and intakes did not differ by sex. These findings advance our understanding of the temporal nature of HM components, emphasizing the role of infant 24 h HM and total lipids intake in development of infant lean and adipose tissue.
Collapse
Affiliation(s)
- Majed A. Suwaydi
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (M.A.S.); (C.T.L.); (A.H.W.); (S.L.P.); (J.L.M.); (M.E.W.); (D.T.G.)
- School of Applied Medical Sciences, Jazan University, Jazan 45142, Saudi Arabia
- ABREAST Network, Perth, WA 6000, Australia
- UWA Centre for Human Lactation Research and Translation, Crawley, WA 6009, Australia
| | - Ching Tat Lai
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (M.A.S.); (C.T.L.); (A.H.W.); (S.L.P.); (J.L.M.); (M.E.W.); (D.T.G.)
- ABREAST Network, Perth, WA 6000, Australia
- UWA Centre for Human Lactation Research and Translation, Crawley, WA 6009, Australia
| | - Ashleigh H. Warden
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (M.A.S.); (C.T.L.); (A.H.W.); (S.L.P.); (J.L.M.); (M.E.W.); (D.T.G.)
- ABREAST Network, Perth, WA 6000, Australia
- UWA Centre for Human Lactation Research and Translation, Crawley, WA 6009, Australia
| | - Sharon L. Perrella
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (M.A.S.); (C.T.L.); (A.H.W.); (S.L.P.); (J.L.M.); (M.E.W.); (D.T.G.)
- ABREAST Network, Perth, WA 6000, Australia
- UWA Centre for Human Lactation Research and Translation, Crawley, WA 6009, Australia
| | - Jacki L. McEachran
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (M.A.S.); (C.T.L.); (A.H.W.); (S.L.P.); (J.L.M.); (M.E.W.); (D.T.G.)
- ABREAST Network, Perth, WA 6000, Australia
- UWA Centre for Human Lactation Research and Translation, Crawley, WA 6009, Australia
| | - Mary E. Wlodek
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (M.A.S.); (C.T.L.); (A.H.W.); (S.L.P.); (J.L.M.); (M.E.W.); (D.T.G.)
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Donna T. Geddes
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (M.A.S.); (C.T.L.); (A.H.W.); (S.L.P.); (J.L.M.); (M.E.W.); (D.T.G.)
- ABREAST Network, Perth, WA 6000, Australia
- UWA Centre for Human Lactation Research and Translation, Crawley, WA 6009, Australia
| | - Zoya Gridneva
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (M.A.S.); (C.T.L.); (A.H.W.); (S.L.P.); (J.L.M.); (M.E.W.); (D.T.G.)
- ABREAST Network, Perth, WA 6000, Australia
- UWA Centre for Human Lactation Research and Translation, Crawley, WA 6009, Australia
| |
Collapse
|
7
|
Bhopatkar AA, Mandal S, Williams JM. Combination of reduced uterine perfusion pressure and leptin infusion as a novel model of preeclampsia. Am J Physiol Heart Circ Physiol 2024; 327:H507-H508. [PMID: 39058436 DOI: 10.1152/ajpheart.00476.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Affiliation(s)
- Anukool A Bhopatkar
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Sautan Mandal
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Jan M Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
8
|
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
|
9
|
Jiménez-Cortegana C, Gutiérrez-García C, Sánchez-Jiménez F, Vilariño-García T, Flores-Campos R, Pérez-Pérez A, Garnacho C, Sánchez-León ML, García-Domínguez DJ, Hontecillas-Prieto L, Palazón-Carrión N, De La Cruz-Merino L, Sánchez-Margalet V. Impact of obesity‑associated myeloid‑derived suppressor cells on cancer risk and progression (Review). Int J Oncol 2024; 65:79. [PMID: 38940351 PMCID: PMC11251741 DOI: 10.3892/ijo.2024.5667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/12/2024] [Indexed: 06/29/2024] Open
Abstract
Obesity is a chronic disease caused by the accumulation of excessive adipose tissue. This disorder is characterized by chronic low‑grade inflammation, which promotes the release of proinflammatory mediators, including cytokines, chemokines and leptin. Simultaneously, chronic inflammation can predispose to cancer development, progression and metastasis. Proinflammatory molecules are involved in the recruitment of specific cell populations in the tumor microenvironment. These cell populations include myeloid‑derived suppressor cells (MDSCs), a heterogeneous, immature myeloid population with immunosuppressive abilities. Obesity‑associated MDSCs have been linked with tumor dissemination, progression and poor clinical outcomes. A comprehensive literature review was conducted to assess the impact of obesity‑associated MDSCs on cancer in both preclinical models and oncological patients with obesity. A secondary objective was to examine the key role that leptin, the most important proinflammatory mediator released by adipocytes, plays in MDSC‑driven immunosuppression Finally, an overview is provided of the different therapeutic approaches available to target MDSCs in the context of obesity‑related cancer.
Collapse
Affiliation(s)
- Carlos Jiménez-Cortegana
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Cristian Gutiérrez-García
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Flora Sánchez-Jiménez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Teresa Vilariño-García
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Rocio Flores-Campos
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Antonio Pérez-Pérez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Carmen Garnacho
- Department of Normal and Pathological Histology and Cytology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Maria L. Sánchez-León
- Oncology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Daniel J. García-Domínguez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Natalia Palazón-Carrión
- Oncology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Luis De La Cruz-Merino
- Oncology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, CSIC, University of Seville, Seville 41013, Spain
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, CSIC, University of Seville, Seville 41013, Spain
| |
Collapse
|
10
|
Rusu CC, Kacso I, Moldovan D, Potra A, Tirinescu D, Ticala M, Orasan R, Budurea C, Anton F, Valea A, Bondor CI, Carsote M. Leptin Is Associated with Testosterone, Nutritional Markers, and Vascular Muscular Dysfunction in Chronic Kidney Disease. Int J Mol Sci 2024; 25:7646. [PMID: 39062887 PMCID: PMC11277084 DOI: 10.3390/ijms25147646] [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: 06/11/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Chronic kidney disease (CKD) causes specific hormonal disturbances, such as variations in leptin and testosterone levels and function. These disturbances can promote errors in signaling interaction and cellular information processing and can be implicated in the pathogenesis of atherosclerosis. This study investigates the factors that affect leptin in CKD patients and examines how leptin is related to markers of vascular disease. We conducted a cross-sectional study of 162 patients with CKD in pre-dialysis and dialysis stages. We recorded clinical and laboratory data, including leptin, testosterone, and subclinical atherosclerosis markers like brachial-ankle pulse wave velocity (ba PWV) in pre-dialysis CKD patients and flow-mediated vasodilation (FMD) and nitroglycerin-mediated vasodilation (NMD) in hemodialysis (HD) patients. Leptin was significantly correlated with testosterone in CKD pre-dialysis stages (p < 0.001) and also in HD (p = 0.026), with adipose tissue mass in pre-dialysis stages (p < 0.001), and also in HD (p < 0.001). In women HD patients, leptin correlated with NMD (p = 0.039; r = -0.379); in all HD patients, leptin correlated with C reactive protein (p = 0.007; r = 0.28) and parathormone (p = 0.039; r = -0.220). Our research emphasizes the connection between leptin, adipose tissue, and testosterone in all stages of CKD. Leptin was associated with NMD in HD women and correlated with inflammatory syndrome and parathyroid hormone in all HD patients.
Collapse
Affiliation(s)
- Crina Claudia Rusu
- Department of Nephrology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Ina Kacso
- Department of Nephrology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Diana Moldovan
- Department of Nephrology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Alina Potra
- Department of Nephrology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Dacian Tirinescu
- Department of Nephrology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Maria Ticala
- Department of Nephrology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
- Department of Nephrology, County Emergency Clinical Hospital Cluj, 3-5 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Remus Orasan
- Nefromed Dialysis Center, 40 Ana Aslan Street, 400528 Cluj-Napoca, Romania
| | - Cristian Budurea
- Nefromed Dialysis Center, 40 Ana Aslan Street, 400528 Cluj-Napoca, Romania
| | - Florin Anton
- Department of Cardiology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
| | - Ana Valea
- Department of Endocrinology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 8 Victor Babes, Street, 400012 Cluj-Napoca, Romania
| | - Cosmina Ioana Bondor
- Department of Medical Informatics and Biostatistics, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj, 6 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Mara Carsote
- Department of Endocrinology, "Carol Davila" University of Medicine and Pharmacy, Dionisie Lupu Street, Number 37, Sector 1, 020021 Bucharest, Romania
- Department of Clinical Endocrinology V, "C.I. Parhon" National Institute of Endocrinology, Aviatorilor Ave 34-36, Sector 1, 011863 Bucharest, Romania
| |
Collapse
|
11
|
Singh H, Almabhouh FA, Alshaikhli HSI, Hassan MJM, Daud S, Othman R, Md Salleh MFRR. Leptin in reproduction and hypertension in pregnancy. Reprod Fertil Dev 2024; 36:RD24060. [PMID: 39038160 DOI: 10.1071/rd24060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/27/2024] [Indexed: 07/24/2024] Open
Abstract
Leptin has important roles in numerous physiological functions, including those in the regulation of energy balance, and in immune and reproductive systems. However, in the recent years, evidence has implicated it in a number of obesity-related diseases, where its concentrations in serum are significantly elevated. Elevated serum leptin concentrations and increased placental leptin secretion have been reported in women with hypertensive disorders of pregnancy. Whether leptin is responsible for this disorder remains to be established. Leptin injections in healthy rats and mice during pregnancy result in endothelial activation, increased blood pressure and proteinuria. A potential role for leptin in the pathogenesis of pre-eclampsia is hypothesised, particularly in women who are overweight or obese where serum leptin concentrations are often elevated. This review summarises pertinent information in the literature on the role of leptin in puberty, pregnancy, and hypertensive disorders of pregnancy. In particular, the possible mechanism that may be involved in leptin-induced increase in blood pressure and proteinuria during pregnancy and the potential role of marinobufagenin in this disease entity. We hypothesise a significant role for oxidative stress in this, and propose a conceptual framework on the events that lead to endothelial activation, raised blood pressure and proteinuria following leptin administration.
Collapse
Affiliation(s)
- Harbindarjeet Singh
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Sg Buloh, Selangor, Malaysia
| | - Fayez A Almabhouh
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Sg Buloh, Selangor, Malaysia; and Department of Biology and Biotechnology, Faculty of Science Islamic University of Gaza, Gaza Strip, Palestine
| | | | | | - Suzanna Daud
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Sg Buloh, Selangor, Malaysia
| | - Rosfayati Othman
- Department of Physiology, Faculty of Medicine, MAHSA University, Bandar Saujana Putra, Kuala Langat, Selangor, Malaysia
| | - Muhd Fakh Rur Razi Md Salleh
- Department of Physiology, Faculty of Medicine, MAHSA University, Bandar Saujana Putra, Kuala Langat, Selangor, Malaysia
| |
Collapse
|
12
|
Dugershaw‐Kurzer B, Bossart J, Buljan M, Hannig Y, Zehnder S, Gupta G, Kissling VM, Nowak‐Sliwinska P, van Beijnum JR, Griffioen AW, Masjosthusmann S, Zühr E, Fritsche E, Hornung R, Rduch T, Buerki‐Thurnherr T. Nanoparticles Dysregulate the Human Placental Secretome with Consequences on Angiogenesis and Vascularization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401060. [PMID: 38767187 PMCID: PMC11267331 DOI: 10.1002/advs.202401060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/05/2024] [Indexed: 05/22/2024]
Abstract
Exposure to nanoparticles (NPs) in pregnancy is increasingly linked to adverse effects on embryo-fetal development and health later in life. However, the developmental toxicity mechanisms of NPs are largely unknown, in particular potential effects on the placental secretome, which orchestrates many developmental processes pivotal for pregnancy success. This study demonstrates extensive material- and pregnancy stage-specific deregulation of placental signaling from a single exposure of human placental explants to physiologically relevant concentrations of engineered (silica (SiO2) and titanium dioxide (TiO2) NPs) and environmental NPs (diesel exhaust particles, DEPs). This includes a multitude of secreted inflammatory, vascular, and endocrine placental factors as well as extracellular vesicle (EV)-associated proteins. Moreover, conditioned media (CM) from NP-exposed explants induce pronounced anti-angiogenic and anti-vasculogenic effects, while early neurodevelopmental processes are only marginally affected. These findings underscore the potential of metal oxide NPs and DEPs for widespread interference with the placental secretome and identify vascular morphogenesis as a sensitive outcome for the indirect developmental toxicity of different NPs. Overall, this work has profound implications for the future safety assessment of NPs for industrial, commercial, or medical applications in pregnancy, which should consider placenta-mediated toxicity by holistic secretomics approaches to ensure the development of safe nanotechnologies.
Collapse
Affiliation(s)
- Battuja Dugershaw‐Kurzer
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
- Department of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
| | - Jonas Bossart
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
- Department of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
- SIBSwiss Institute of BioinformaticsLausanne1015Switzerland
| | - Marija Buljan
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
- SIBSwiss Institute of BioinformaticsLausanne1015Switzerland
| | - Yvette Hannig
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
| | - Sarah Zehnder
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
| | - Govind Gupta
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
| | - Vera M. Kissling
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
| | - Patrycja Nowak‐Sliwinska
- Institute of Pharmaceutical Sciences of Western SwitzerlandGeneva1211Switzerland
- School of Pharmaceutical SciencesUniversity of GenevaGeneva1205Switzerland
| | - Judy R. van Beijnum
- Angiogenesis LaboratoryDepartment of Medical OncologyUMC loacation Vrije Universiteit AmsterdamAmsterdam1081The Netherlands
| | - Arjan W. Griffioen
- Angiogenesis LaboratoryDepartment of Medical OncologyUMC loacation Vrije Universiteit AmsterdamAmsterdam1081The Netherlands
| | | | - Etta Zühr
- IUF—Leibniz Research Institute for Environmental Medicine40225DuesseldorfGermany
| | - Ellen Fritsche
- IUF—Leibniz Research Institute for Environmental Medicine40225DuesseldorfGermany
- Medical FacultyHeinrich Heine University40225DuesseldorfGermany
- DNTOX GmbH40223DuesseldorfGermany
| | - René Hornung
- Department of Gynaecology and ObstetricsCantonal Hospital St.Gallen (KSSG)St. Gallen9007Switzerland
| | - Thomas Rduch
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
- Department of Gynaecology and ObstetricsCantonal Hospital St.Gallen (KSSG)St. Gallen9007Switzerland
| | - Tina Buerki‐Thurnherr
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
| |
Collapse
|
13
|
Cinzori ME, Pacyga DC, Rosas L, Whalen J, Smith S, Park JS, Geiger SD, Gardiner JC, Braun JM, Schantz SL, Strakovsky RS. Associations of per- and polyfluoroalkyl substances with maternal metabolic and inflammatory biomarkers in early-to-mid-pregnancy. ENVIRONMENTAL RESEARCH 2024; 250:118434. [PMID: 38346483 PMCID: PMC11102845 DOI: 10.1016/j.envres.2024.118434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) can disrupt metabolism. Early-to-mid pregnancy is characterized by amplified metabolic processes and inflammation to support maternal adaptations and fetal growth. Thus, we cross-sectionally evaluated whether PFAS are individually and jointly associated with these processes in early-to-mid pregnancy. METHODS Pregnant Illinois women (n = 452) provided fasted blood samples at median 17 weeks gestation. We quantified serum perfluorononanoic (PFNA), perfluorooctane sulfonic (PFOS), perfluorooctanoic (PFOA), methyl-perfluorooctane sulfonamide acetic acid (Me-PFOSA-AcOH), perfluorohexanesulfonic (PFHxS), perfluorodecanoic (PFDeA), and perfluoroundecanoic (PFUdA) acid. Key outcomes were plasma glucose, insulin, C-peptide, insulin-like growth factor 1 (IGF-1), adiponectin, leptin, triglycerides, free fatty acids, total cholesterol, high-density lipoprotein (HDL) cholesterol, C-reactive protein, tumor necrosis factor alpha (TNF-α), monocyte chemoattractant protein-1 (MCP-1), and interleukin 6. We calculated homeostatic model assessment for insulin resistance (HOMA-IR), low-density lipoprotein (LDL) cholesterol, and very low-density lipoprotein (VLDL). We evaluated associations of PFAS with each metabolic/inflammatory biomarker individually using covariate-adjusted linear regression and jointly using quantile-based g-computation. RESULTS In linear regression, all PFAS (except Me-PFOSA-AcOH) were negatively associated with insulin, HOMA-IR, and leptin, whereas all PFAS were positively associated with HDL cholesterol. We also observed negative associations of some PFAS with TNF-α and MCP-1; positive associations with adiponectin and total cholesterol also emerged. Additionally, PFOS was positively, whereas Me-PFOSA-AcOH was negatively, associated with triglycerides and VLDL. Each 25% increase in the PFAS mixture was associated with -31.3% lower insulin (95%CI: -45.8, -12.9), -31.9% lower HOMA-IR (95%CI: -46.4, -13.4), and -9.4% lower leptin (95%CI: -17.3, -0.8), but 7.4% higher HDL cholesterol (95%CI: 4.6, 10.3). For most outcomes, the major contributors to the PFAS mixture often differed compared to single-PFAS analyses. IMPLICATIONS Individual and joint PFAS exposures were associated with markers of maternal metabolism and inflammation in pregnancy. Further investigation is needed to elucidate possible mechanisms and consequences of these findings.
Collapse
Affiliation(s)
- Maria E Cinzori
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, 48824, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, 48824, USA
| | - Diana C Pacyga
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, 48824, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Libeth Rosas
- The Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, IL, 61801, USA
| | - Jason Whalen
- Michigan Diabetes Research Center Chemistry Laboratory, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sabrina Smith
- Environmental Chemistry Laboratory, Department of Toxic Substances Control, California Environmental Protection Agency, Berkeley, CA, 94710, USA
| | - June-Soo Park
- Environmental Chemistry Laboratory, Department of Toxic Substances Control, California Environmental Protection Agency, Berkeley, CA, 94710, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94158, USA
| | - Sarah D Geiger
- The Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, IL, 61801, USA; Department of Kinesiology and Community Health, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Joseph C Gardiner
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, 48824, USA
| | - Joseph M Braun
- Department of Epidemiology, Brown University, Providence, RI, 02912, USA
| | - Susan L Schantz
- The Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, IL, 61801, USA; Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, IL 61802, USA
| | - Rita S Strakovsky
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, 48824, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
14
|
Rumer KK, Sehgal S, Kramer A, Bogart KP, Winn VD. The effects of leptin on human cytotrophoblast invasion are gestational age and dose-dependent. Front Endocrinol (Lausanne) 2024; 15:1386309. [PMID: 38846494 PMCID: PMC11154010 DOI: 10.3389/fendo.2024.1386309] [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: 02/15/2024] [Accepted: 05/08/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Leptin and its receptors are expressed by the human placenta throughout gestation, yet the role of leptin in early human placental development is not well characterized. Leptin is overexpressed in the placentas from preeclamptic (PE) pregnancies. PE can result from the impaired invasion of fetal placental cells, cytotrophoblasts (CTBs), into the maternal decidua. We hypothesized that elevated leptin levels would impair human CTB invasion. Methods The effects of leptin on the invasion of human CTBs were evaluated in three cell models, HTR-8/SVneo cells, primary CTBs, and placental villous explants using invasion assays. Further, leptin receptor expression was characterized in all three cell models using RT-PCR. Further phosphokinase assays were performed in HTR-8/SVneo cells to determine signaling pathways involved in CTB invasion in response to differential leptin doses. Results We found that, prior to 8 weeks gestation, leptin promoted CTB invasion in the explant model. After 11 weeks gestation in explants, primary CTBs and in HTR-8/SVneo cells, leptin promoted invasion at moderate but not at high concentrations. Further, leptin receptor characterization revealed that leptin receptor expression did not vary over gestation, however, STAT, PI3K and MAPK pathways showed different signaling in response to varied leptin doses. Discussion These data suggest that the excess placental leptin observed in PE may cause impaired CTB invasion as a second-trimester defect. Leptin's differential effect on trophoblast invasion may explain the role of hyperleptinemia in preeclampsia pathogenesis.
Collapse
Affiliation(s)
- Kristen K. Rumer
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, United States
- Department of Molecular and Cellular Medicine, University of Colorado, Aurora, CO, United States
| | - Shilpi Sehgal
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Anita Kramer
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, United States
| | - Kevin P. Bogart
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, United States
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, United States
- Department of Molecular and Cellular Medicine, University of Colorado, Aurora, CO, United States
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
15
|
Jiang J, Wei S, Chen M, Tan Y, Yang Z, Yang G, Feng W, Han Z, Wei X, Luo X. Characterizing the Dynamic Expression of C1q/TNF-α-Related Protein 6 (CTRP6) during Pregnancy in Humans and Mice with Gestational Diabetes Mellitus. Biomedicines 2024; 12:1128. [PMID: 38791090 PMCID: PMC11117843 DOI: 10.3390/biomedicines12051128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
AIM C1q/TNF-related protein 6 (CTRP6) is a novel adipokine involved in insulin resistance. Thus, we aim to investigate the expression profile of CTRP6 in the plasma, adipose tissue and placenta of GDM patients and mice. METHODS Chinese Han pregnant women (GDM n = 9, control n = 10) with a scheduled caesarean section delivery were recruited. A number of high-fat diet (HFD) induced-pregnancy C57BL/6 mice were chosen as an animal model of GDM. Circulating levels of CTRP6 and adiponectin were examined by ELISA. CTRP6 expression in adipose tissue and placenta were detected by real-time qPCR and WB. RESULT Plasma CTRP6 levels were decreased during the first and second trimesters in mice, as well as the second and third trimesters in patients, while they were increased at delivery in GDM patients and mice. Plasma CTRP6 levels were significantly correlated with WBC, systolic pressure, diastolic pressure and fasting blood glucose. Moreover, CTRP6 mRNA expression in the subcutaneous (sWAT) and omental white adipose tissue (oWAT), as well as in the placenta, was significantly higher in GDM human patients at cesarean delivery. Furthermore, the mRNA expression of Ctrp6 was increased in the sWAT and visceral WAT (vWAT), whilst decreased in the interscapular brown adipose tissue (iBAT), of GDM mice at cesarean delivery. CONCLUSION Dynamically expressed CTRP6 may be served as a candidate target for treatment of GDM.
Collapse
Affiliation(s)
- Jianan Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (J.J.); (Y.T.); (W.F.)
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an Jiaotong University, Xi’an 710061, China
| | - Shuangyu Wei
- Clinical Laboratory, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China;
| | - Miao Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China; (M.C.); (Z.Y.); (G.Y.); (Z.H.)
| | - Yutian Tan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (J.J.); (Y.T.); (W.F.)
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an Jiaotong University, Xi’an 710061, China
| | - Zhao Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China; (M.C.); (Z.Y.); (G.Y.); (Z.H.)
| | - Guiying Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China; (M.C.); (Z.Y.); (G.Y.); (Z.H.)
| | - Weijie Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (J.J.); (Y.T.); (W.F.)
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an Jiaotong University, Xi’an 710061, China
| | - Zhen Han
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an 710061, China; (M.C.); (Z.Y.); (G.Y.); (Z.H.)
| | - Xiaojing Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (J.J.); (Y.T.); (W.F.)
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an Jiaotong University, Xi’an 710061, China
| | - Xiao Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (J.J.); (Y.T.); (W.F.)
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
16
|
Yau-Qiu ZX, Galmés S, Castillo P, Picó C, Palou A, Rodríguez AM. Maternal choline supplementation mitigates premature foetal weight gain induced by an obesogenic diet, potentially linked to increased amniotic fluid leptin levels in rats. Sci Rep 2024; 14:11366. [PMID: 38762543 PMCID: PMC11102553 DOI: 10.1038/s41598-024-62229-2] [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: 02/22/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024] Open
Abstract
Placental leptin may impact foetal development. Maternal overnutrition has been linked to increased plasma leptin levels and adverse effects on offspring, whereas choline, an essential nutrient for foetal development, has shown promise in mitigating some negative impacts of maternal obesity. Here, we investigate whether a maternal obesogenic diet alters foetal growth and leptin levels in the foetal stomach, amniotic fluid (AF), and placenta in late gestation and explore the potential modulating effects of maternal choline supplementation. Female rats were fed a control (CD) or a western diet (WD) four weeks before mating and during gestation, half of them supplemented with choline (pregnancy days 11-17). Leptin levels (in foetal stomach, AF, and placenta) and leptin gene expression (in placenta) were assessed on gestation days 20 and 21. At day 20, maternal WD feeding resulted in greater leptin levels in foetal stomach, placenta, and AF. The increased AF leptin levels were associated with a premature increase in foetal weight in both sexes. Maternal choline supplementation partially prevented these alterations, but effects differed in CD dams, causing increased AF leptin levels and greater weight in male foetuses at day 20. Maternal choline supplementation effectively mitigates premature foetal overgrowth induced by an obesogenic diet, potentially linked to increased AF leptin levels. Further research is needed to explore the sex-specific effects.
Collapse
Affiliation(s)
- Zhi Xin Yau-Qiu
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation-NuBE), University of the Balearic Islands (UIB), Cra. Valldemossa Km 7.5, 07122, Palma, Balearic Islands, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Sebastià Galmés
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation-NuBE), University of the Balearic Islands (UIB), Cra. Valldemossa Km 7.5, 07122, Palma, Balearic Islands, Spain.
- Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain.
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Pedro Castillo
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation-NuBE), University of the Balearic Islands (UIB), Cra. Valldemossa Km 7.5, 07122, Palma, Balearic Islands, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation-NuBE), University of the Balearic Islands (UIB), Cra. Valldemossa Km 7.5, 07122, Palma, Balearic Islands, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation-NuBE), University of the Balearic Islands (UIB), Cra. Valldemossa Km 7.5, 07122, Palma, Balearic Islands, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Ana María Rodríguez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation-NuBE), University of the Balearic Islands (UIB), Cra. Valldemossa Km 7.5, 07122, Palma, Balearic Islands, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| |
Collapse
|
17
|
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
|
18
|
Crespi BJ. Nausea, vomiting and conflict in pregnancy: The adaptive significance of Growth-Differentiation Factor 15. Evol Med Public Health 2024; 12:75-81. [PMID: 38711789 PMCID: PMC11071683 DOI: 10.1093/emph/eoae008] [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: 01/17/2024] [Revised: 03/05/2024] [Indexed: 05/08/2024] Open
Abstract
Nausea and vomiting in pregnancy (NVP) is heritable, common and aversive, and its extreme, hyperemesis gravidarum (HG), can be highly deleterious to the mother and fetus. Recent influential studies have demonstrated that HG is caused predominantly by high levels of Growth-Differentiation Factor 15 (GDF15), a hormone produced by the placenta in substantial amounts. This work has led to calls for therapeutic modulation of this hormone to reduce GDF15 levels and ameliorate HG risk. I describe three main lines of evidence relevant to the hypothesis that GDF15 production is typically adaptive for the fetus, in the context of enhanced placental invasion, reduced rates of miscarriage and preterm birth and higher birth weight. These considerations highlight the medical implications of maternal-fetal conflict, in the context of tradeoffs between aversive symptoms during gestation, rare disorders of pregnancy with major adverse effects and moderate fitness-enhancing benefits to fetuses.
Collapse
Affiliation(s)
- Bernard J Crespi
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
19
|
Abundis EM, Hernandez-Landero F, Escobar-Calderon G, Gomez-Crisostomo N, Contreras-Paredes A, de la Cruz-Hernandez E. Gene expression of cardiovascular risk markers in mononuclear cells of pregnant woman in relation to plasma leptin and homocysteine levels: A cross sectional study. Int J Gynaecol Obstet 2024; 165:350-360. [PMID: 38126620 DOI: 10.1002/ijgo.15302] [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: 05/30/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE To investigate the relationship between anthropometric, biochemical, and hematologic parameters and serum leptin and homocysteine (Hcy) levels. Also, to determine the effect of leptin and Hcy on expression of genes associated with cardiovascular disease susceptibility (APOA1, LRP1, COX-1, and COX-2) in mononuclear cells of healthy pregnant women. METHODS Between August 2018 and January 2020, a cross-sectional study was conducted on 161 healthy pregnant women in Tabasco, southeastern Mexico. The study population was classified by trimester, according to gestational pregnancy. Anthropometric, biochemical (leptin and homocysteine), and hematologic data were obtained under fasting conditions. APOA1, LRP1, COX-1, and COX-2 expression in mononuclear cells was evaluated using RT-qPCR. RESULTS Red cell indices (hemoglobin, hematocrit, and erythrocytes) were negatively and positively correlated with leptin and Hcy levels, respectively, in the first- and second-trimester groups. Increased leptin levels and low red cell indices were significantly associated with BMI <25.0 in the second-trimester group; however, no significant differences were observed in Hcy levels. Increased leptin and Hcy levels were significantly associated with high lipid indicators in the first- and third-trimester groups, respectively. High APOA1 and COX-2 expression was significantly associated with reduced leptin and increased Hcy levels in the second- and third-trimester groups. CONCLUSION Increased leptin and Hcy levels during pregnancy, mainly associated with modifications in erythrocytes and lipid indices, may lead to early modification of genes related to lipid metabolism (APOA1) and proinflammatory response (COX-2) and, thereby, increase cardiovascular disease risk.
Collapse
Affiliation(s)
- Eduardo Martínez Abundis
- Laboratorio de investigacion en Enfermedades Metabolicas e Infecciosas, Division Academica Multidisciplinaria de Comalcalco, Universidad Juarez Autonoma de Tabasco, Comalcalco, Tabasco, Mexico
| | - Fernanda Hernandez-Landero
- Laboratorio de investigacion en Enfermedades Metabolicas e Infecciosas, Division Academica Multidisciplinaria de Comalcalco, Universidad Juarez Autonoma de Tabasco, Comalcalco, Tabasco, Mexico
| | - Grecia Escobar-Calderon
- Laboratorio de investigacion en Enfermedades Metabolicas e Infecciosas, Division Academica Multidisciplinaria de Comalcalco, Universidad Juarez Autonoma de Tabasco, Comalcalco, Tabasco, Mexico
| | - Nancy Gomez-Crisostomo
- Laboratorio de investigacion en Enfermedades Metabolicas e Infecciosas, Division Academica Multidisciplinaria de Comalcalco, Universidad Juarez Autonoma de Tabasco, Comalcalco, Tabasco, Mexico
| | - Adriana Contreras-Paredes
- Unidad de Investigacion Biomedica en Cancer, Instituto Nacional de Cancerología-Universidad Nacional Autonoma de Mexico, Ciudad de Mexico, Mexico
| | - Erick de la Cruz-Hernandez
- Laboratorio de investigacion en Enfermedades Metabolicas e Infecciosas, Division Academica Multidisciplinaria de Comalcalco, Universidad Juarez Autonoma de Tabasco, Comalcalco, Tabasco, Mexico
| |
Collapse
|
20
|
Fang H, Li Q, Wang H, Ren Y, Zhang L, Yang L. Maternal nutrient metabolism in the liver during pregnancy. Front Endocrinol (Lausanne) 2024; 15:1295677. [PMID: 38572473 PMCID: PMC10987773 DOI: 10.3389/fendo.2024.1295677] [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: 09/17/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024] Open
Abstract
The liver plays pivotal roles in nutrient metabolism, and correct hepatic adaptations are required in maternal nutrient metabolism during pregnancy. In this review, hepatic nutrient metabolism, including glucose metabolism, lipid and cholesterol metabolism, and protein and amino acid metabolism, is first addressed. In addition, recent progress on maternal hepatic adaptations in nutrient metabolism during pregnancy is discussed. Finally, the factors that regulate hepatic nutrient metabolism during pregnancy are highlighted, and the factors include follicle-stimulating hormone, estrogen, progesterone, insulin-like growth factor 1, prostaglandins fibroblast growth factor 21, serotonin, growth hormone, adrenocorticotropic hormone, prolactin, thyroid stimulating hormone, melatonin, adrenal hormone, leptin, glucagon-like peptide-1, insulin glucagon and thyroid hormone. Our vision is that more attention should be paid to liver nutrient metabolism during pregnancy, which will be helpful for utilizing nutrient appropriately and efficiently, and avoiding liver diseases during pregnancy.
Collapse
Affiliation(s)
- Hongxu Fang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Qingyang Li
- College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Haichao Wang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Ying Ren
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Leying Zhang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Ling Yang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| |
Collapse
|
21
|
Lai YM, Tan GC, Shah SA, Abd Rahman R, Mohd Saleh MF, Mansor S, Khong TY, Wong YP. Non-hypertensive gestational diabetes mellitus: Placental histomorphology and its association with perinatal outcomes. Placenta 2024; 147:21-27. [PMID: 38278001 DOI: 10.1016/j.placenta.2024.01.012] [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: 10/09/2023] [Revised: 12/12/2023] [Accepted: 01/22/2024] [Indexed: 01/28/2024]
Abstract
INTRODUCTION Gestational diabetes mellitus (GDM) exerts a great impact on the placenta and reflects changes on placentas both morphological and functionally. The aims of this study are to evaluate the prevalence of placental histopathological lesions in pregnancies complicated by GDM compared to gestational age-matched controls, and their association with maternal and fetal complications. METHODS Fifty-four singleton GDM-complicated pregnancies were recruited and compared to 33 consecutive normal pregnancies. Two pathologists, blinded to all clinical data, reviewed and evaluated all histological samples of the placentas in accordance with Amsterdam criteria. Relevant demographic, clinical data and primary birth outcomes were recorded. RESULTS A myriad of histomorphological abnormalities, including chronic inflammation (n = 9/54, p = 0.031), histological chorioamnionitis (n = 23/54, p < 0.001), umbilical/chorionic vasculitis (n = 9/54, p = 0.031), changes related to maternal vascular malperfusion (n = 22/54, p = 0.003), chorangiosis (n = 10/54, p = 0.046) and villous dysmaturity (n = 9/54, p = 0.012) were observed more frequently in the GDM placentas compared to the controls. Additionally, GDM significantly increased the risk of fetal complications, including macrosomia/fetal growth restriction (n = 13/54, p = 0.004). DISCUSSION Histoarchitectural abnormalities were observed more frequently in placentas of GDM pregnancies compared to the controls. Our findings support the hypothesis that diabetic-induced damage in the placental function may be associated with the increased in fetal growth disorders in GDM-complicated pregnancies.
Collapse
Affiliation(s)
- Yip Meng Lai
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Geok Chin Tan
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Shamsul Azhar Shah
- Department of Community Health, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Rahana Abd Rahman
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Muhamad Fakhri Mohd Saleh
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Suhada Mansor
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Teck Yee Khong
- Department of Pathology, Women's and Children's Hospital, Adelaide, SA 5006, Australia
| | - Yin Ping Wong
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
22
|
Kallak TK, Serapio S, Visser N, Lager S, Skalkidou A, Ahlsson F. Differential gene expression in two consecutive pregnancies between same sex siblings and implications on maternal constraint. Sci Rep 2024; 14:4210. [PMID: 38378837 PMCID: PMC10879170 DOI: 10.1038/s41598-024-54724-3] [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: 10/13/2023] [Accepted: 02/15/2024] [Indexed: 02/22/2024] Open
Abstract
The objective of this study was to investigate how placental gene expression differs in two consecutive pregnancies in same sex siblings, and its possible association with the "maternal constraint" hypothesis. Material was gathered from the BASIC study (Biological, Affect, Stress, Imaging, and Cognition in Pregnancy and the Puerperium), a population based prospective study that was started in 2009 in Uppsala. Over 900 specimens of placenta biopsies were collected and out of these 10 women gave birth twice, to the same sex child, and were included in this study. The total RNA was isolated and prepared from frozen villous tissue from the placenta and further analyzed by use of Ion AmpliSeq Human Transcriptome Gene Expression kit. A total of 234 genes differed significantly between the first and second pregnancy placentas, when adjusting for delivery mode, maternal BMI and gestational age. Of special interest was the down-regulated group of genes in the second pregnancy. Exemplified by Pentraxin 3, SRY-Box Transcription Factor 9, and Serum Amyloid A1, which all were associated with biological processes involved in the immune system and inflammation. Further, protein-protein interaction analysis visualized them as hub genes interacting with several of the other differentially expressed genes. How these altered gene expressions affect maternal constraint during pregnancy needs further validation in lager study cohorts and also future validation in functional assays.
Collapse
Affiliation(s)
| | - Solveig Serapio
- Department of Women's and Children's Health, Uppsala University, 751 85, Uppsala, Sweden
| | - Nadja Visser
- Department of Women's and Children's Health, Uppsala University, 751 85, Uppsala, Sweden
| | - Susanne Lager
- Department of Women's and Children's Health, Uppsala University, 751 85, Uppsala, Sweden
| | - Alkistis Skalkidou
- Department of Women's and Children's Health, Uppsala University, 751 85, Uppsala, Sweden
| | - Fredrik Ahlsson
- Department of Women's and Children's Health, Uppsala University, 751 85, Uppsala, Sweden
| |
Collapse
|
23
|
Vilariño-García T, Polonio-González ML, Pérez-Pérez A, Ribalta J, Arrieta F, Aguilar M, Obaya JC, Gimeno-Orna JA, Iglesias P, Navarro J, Durán S, Pedro-Botet J, Sánchez-Margalet V. Role of Leptin in Obesity, Cardiovascular Disease, and Type 2 Diabetes. Int J Mol Sci 2024; 25:2338. [PMID: 38397015 PMCID: PMC10888594 DOI: 10.3390/ijms25042338] [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: 12/29/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Diabetes mellitus (DM) is a highly prevalent disease worldwide, estimated to affect 1 in every 11 adults; among them, 90-95% of cases are type 2 diabetes mellitus. This is partly attributed to the surge in the prevalence of obesity, which has reached epidemic proportions since 2008. In these patients, cardiovascular (CV) risk stands as the primary cause of morbidity and mortality, placing a substantial burden on healthcare systems due to the potential for macrovascular and microvascular complications. In this context, leptin, an adipocyte-derived hormone, plays a fundamental role. This hormone is essential for regulating the cellular metabolism and energy balance, controlling inflammatory responses, and maintaining CV system homeostasis. Thus, leptin resistance not only contributes to weight gain but may also lead to increased cardiac inflammation, greater fibrosis, hypertension, and impairment of the cardiac metabolism. Understanding the relationship between leptin resistance and CV risk in obese individuals with type 2 DM (T2DM) could improve the management and prevention of this complication. Therefore, in this narrative review, we will discuss the evidence linking leptin with the presence, severity, and/or prognosis of obesity and T2DM regarding CV disease, aiming to shed light on the potential implications for better management and preventive strategies.
Collapse
Affiliation(s)
- Teresa Vilariño-García
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, Virgen del Rocio University Hospital, University of Seville, Seville 41013, Spain;
| | - María L. Polonio-González
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009, Spain; (M.L.P.-G.); (A.P.-P.)
| | - Antonio Pérez-Pérez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009, Spain; (M.L.P.-G.); (A.P.-P.)
| | - Josep Ribalta
- Departament de Medicina i Cirurgia, University Rovira i Vigili, IISPV, CIBERDEM, 43007 Tarragona, Spain;
| | - Francisco Arrieta
- Endocrinology and Nutrition Service, Ramón y Cajal University Hospital, 28034 Madrid, Spain;
| | - Manuel Aguilar
- Endocrinology and Nutrition Service, Puerta del Mar University Hospital, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Cádiz University (UCA), 11001 Cádiz, Spain;
| | - Juan C. Obaya
- Chopera Helath Center, Alcobendas Primary Care,Alcobendas 28100 Madrid, Spain;
| | - José A. Gimeno-Orna
- Endocrinology and Nutrition Department, Hospital Clinico Universitario Lozano Blesa, 15 50009 Zaragoza, Spain;
| | - Pedro Iglesias
- Endocrinology and Nutrition Service, Puerta de Hierro University Hospital, Majadahonda, 28220 Madrid, Spain;
| | - Jorge Navarro
- Hospital Clínico Universitario de Valencia,46011 Valencia, Spain;
| | - Santiago Durán
- Endodiabesidad Clínica Durán & Asociados,41018 Seville, Spain;
| | - Juan Pedro-Botet
- Lipids and Cardiovascular Risk Unit, Hospital del Mar, Autonomous University of Barcelona, 08003 Barcelona, Spain;
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009, Spain; (M.L.P.-G.); (A.P.-P.)
- Institute of Biomedicine of Seville (IBIS), Hospital Universitario Virgen del Rocío/Virgen Macarena, CSIC, Universidad de Sevilla, 41013 Seville, Spain
| |
Collapse
|
24
|
Williams RP, Lesseur C, Cheng H, Li Q, Deyssenroth M, Molteno CD, Meintjes EM, Jacobson SW, Jacobson JL, Wainwright H, Hao K, Chen J, Carter RC. RNA-seq analysis reveals prenatal alcohol exposure is associated with placental inflammatory cells and gene expression. Gene 2024; 894:147951. [PMID: 37918548 DOI: 10.1016/j.gene.2023.147951] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/21/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Fetal alcohol spectrum disorders (FASD) are the most common preventable cause of birth defects and neurodevelopmental disorders worldwide. The placenta is the crucial interface between mother and fetus. Prenatal alcohol exposure (PAE) has been shown to alter placental structure and expression of genes in bulk placental tissue samples, but prior studies have not examined effects on placental cell-type composition or taken cell-type into consideration in transcriptome analyses. METHODS We leveraged an existent placenta single-cell RNA-seq dataset to perform cell-type deconvolution of bulk placental RNA-seq data from 35 heavy drinking pregnant women and 33 controls in a prospective birth cohort in Cape Town, South Africa. We used bivariate analyses and multivariable adjusted linear regression models to assess the relation of PAE on inferred placental cell-type proportions. We also examined differential expression of inflammatory response genes and PAE, using multivariable adjusted linear models. RESULTS Deconvolution analyses showed heterogeneous placenta cell-type composition in which stromal (27 %), endothelial (26 %) and cytotrophoblasts (18 %) were the predominant cell-types. PAE around conception was associated with a higher proportion of Hofbauer cells (B = 0.51, p = 0.035) in linear models adjusted for maternal age, infant sex, and gestational age. Among the 652 inflammatory genes examined, 35 were differential expressed in alcohol exposed placentas (FDR p < 0.05). CONCLUSIONS Our findings suggest that heavy alcohol exposure during pregnancy can influence the proportion of fetal placental villi macrophages (Hofbauer cells) and increased expression of inflammatory genes. Future studies are needed to further characterize these effects and to assess the potential functional roles of placental inflammation in FASD.
Collapse
Affiliation(s)
- Randy P Williams
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Corina Lesseur
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Haoxiang Cheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qian Li
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maya Deyssenroth
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Christopher D Molteno
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ernesta M Meintjes
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | - Sandra W Jacobson
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa; Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Joseph L Jacobson
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Helen Wainwright
- Department of Pathology, National Health Laboratory Service, Cape Town, South Africa
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - R Colin Carter
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa; Departments of Emergency Medicine and Pediatrics, Institute of Human Nutrition, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
25
|
Savino F, Sardo A, Gambarino S, Dini M, Clemente A, Pau A, Galliano I, Bergallo M. Leptin and Leptin Receptor Polymorphisms in Infants and Their Parents: Correlation with Preterm Birth. Genes (Basel) 2024; 15:139. [PMID: 38275620 PMCID: PMC10815013 DOI: 10.3390/genes15010139] [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: 12/22/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
It has been proven that single-nucleotide polymorphisms (SNPs) in LEP and LEPR genes could predispose individuals to an increased risk of pregnancy adverse outcomes (PAOs) such as recurrent pregnancy loss (RPL) and pre-eclampsia. Preterm birth (PTB) is the leading cause of infant mortality. We decided to investigate the correlation between PTB and LEP and LEPR SNPs. The study cohort included families who underwent spontaneous PTB and control samples of families who had at-term-born (≥37 weeks of gestational age) children. Swabs were performed by rubbing the sticky end for about 30 s on the gum and on the inside of the cheek, allowing us to collect the flaking cells of the oral mucosa. Genotyping of the three SNPs-LEPRA668G, LEPG2548A and A19G-was carried out via an ARMS-MAMA real-time PCR procedure, as previously described. Regarding LEPG2548A, we found that the most expressed genotype in infants both in the preterm and the at-term group was AG; however, we did not discover any statistically significant difference (p = 0.97). Considering LEPA19G, none among the infants and parents were found to carry the AA genotype. No statistically significant differences were found between children, mothers and fathers belonging to preterm and at-term groups. We did not find a statistically significant association in newborns and their mother, but our results show a statistical correlation with the LEPRA668G genotype GG of the father. This fact can contribute to defining genetic risk factors for PTB. Further studies are certainly needed to better clarify the role of genetics in influencing preterm delivery.
Collapse
Affiliation(s)
- Francesco Savino
- Early Infancy Special Care Unit, Regina Margherita Children Hospital, A.O.U. Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (F.S.); (A.S.)
| | - Allegra Sardo
- Early Infancy Special Care Unit, Regina Margherita Children Hospital, A.O.U. Città della Salute e della Scienza di Torino, 10126 Torino, Italy; (F.S.); (A.S.)
| | - Stefano Gambarino
- Department of Public Health and Pediatric Sciences, Immunopathology Laboratory, Medical School, University of Turin, 10126 Turin, Italy; (S.G.); (M.D.); (A.C.); (A.P.); (I.G.)
| | - Maddalena Dini
- Department of Public Health and Pediatric Sciences, Immunopathology Laboratory, Medical School, University of Turin, 10126 Turin, Italy; (S.G.); (M.D.); (A.C.); (A.P.); (I.G.)
| | - Anna Clemente
- Department of Public Health and Pediatric Sciences, Immunopathology Laboratory, Medical School, University of Turin, 10126 Turin, Italy; (S.G.); (M.D.); (A.C.); (A.P.); (I.G.)
| | - Anna Pau
- Department of Public Health and Pediatric Sciences, Immunopathology Laboratory, Medical School, University of Turin, 10126 Turin, Italy; (S.G.); (M.D.); (A.C.); (A.P.); (I.G.)
| | - Ilaria Galliano
- Department of Public Health and Pediatric Sciences, Immunopathology Laboratory, Medical School, University of Turin, 10126 Turin, Italy; (S.G.); (M.D.); (A.C.); (A.P.); (I.G.)
| | - Massimiliano Bergallo
- Department of Public Health and Pediatric Sciences, Immunopathology Laboratory, Medical School, University of Turin, 10126 Turin, Italy; (S.G.); (M.D.); (A.C.); (A.P.); (I.G.)
| |
Collapse
|
26
|
Wan R, Yao P, Wang Y, Zhang L, Guo W, Du M, Wang Y, Shi W, Li W. Autophagy-related biomarkers in preeclampsia: the underlying mechanism, correlation to the immune microenvironment and drug screening. BMC Pregnancy Childbirth 2024; 24:1. [PMID: 38166707 PMCID: PMC10759589 DOI: 10.1186/s12884-023-06211-2] [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: 10/11/2023] [Accepted: 12/17/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Preeclampsia is a life-threatening disease of pregnancy that lacks effective pharmaceuticals which can target its pathogenesis. Since preeclampsia involves complex pathological processes, including autophagy, this study aims to explore autophagy-related mechanisms of preeclampsia and to screen potential drugs. METHODS Firstly, the datasets GSE75010, GSE24129, GSE66273, and autophagic genes lists were downloaded from public databases. Then, a weighted gene co-expression network analysis (WGCNA) was applied to filter autophagic-related hub genes of preeclampsia. The differential expression levels of the hub genes were validated with datasets GSE24129 and GSE66273. Next, the GO and KEGG enrichment, protein-protein interacting (PPI) network, as well as the downstream pathways was analyzed via the starBase, STRING and Cytoscape to determine the functions and regulatory network of the hub genes. Additionally, the immune microenvironment of preeclampsia was investigated by the CIBERSORTX database. Finally, three herb ingredients, berberine, baicalein, and luteolin were screened by molecular docking in comparison to pravastatin, metformin, and aspirin, to predict potential drugs for treating preeclampsia. RESULTS A total of 54 autophagy-related genes were filtered by WGCNA. After filtering with |GS| > 0.5 and |MM| > 0.8, three hub genes, namely PKM, LEP, and HK2, were identified and validated. Among these genes, PKM and LEP were overexpressed in women older than 35 years old ( p<0.05; p<0.05); the expression of PKM, LEP, and HK2 differed remarkably in women with different BMI (all p<0.05); PKM overexpressed in women with hypertension (p<0.05). The regulatory network of hub genes demonstrated that they were mainly enriched in metabolic pathways, including the AMPK signaling pathway, glucagon signaling pathway, adipocytokine signaling pathway, and central carbon metabolism. Then, immune microenvironment analysis turned out that M2 macrophages were reduced in preeclampsia women (p<0.0001) and were negatively correlated with the expression of PKM (r=-0.2, p<0.05), LEP (r=-0.4, p<0.0001), and HK2 (r=-0.3, p<0.001). Lastly, molecular docking showed baicalein and luteolin could bind intimately to hub genes. CONCLUSION PKM, LEP, and HK2 could be promising biomarkers for preeclampsia, which might regulate the pathogenesis of preeclampsia via metabolism pathways and immune microenvironment. Baicalein and luteolin could be potential therapeutics for preeclampsia.
Collapse
Affiliation(s)
- Rui Wan
- Department of Obstetrics, Huanghe Sanmenxia Hospital Affiliated to Henan University of Science and Technology, Sanmenxia, China
| | - Peng Yao
- Department of Clinical Laboratory, Huanghe Sanmenxia Hospital Affiliated to Henan University of Science and Technology, Sanmenxia, China
| | - Yuxiu Wang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Lele Zhang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Wei Guo
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Meiying Du
- Health Examination Center, Shanghai Children's Medical Center Hainan Branch, Shanghai Jiao Tong University School of Medicine, Sanya, China
| | - Yu Wang
- College of Chemical Engineering, Shijiazhuang University, Shijiazhuang, China.
| | - Wenbiao Shi
- Department of Obstetrics, Qujing Maternal and Child Health Hospital, Qujing, China.
| | - Weili Li
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd, Shanghai, China.
| |
Collapse
|
27
|
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
|
28
|
Everett AB, Garvey WT, Fernandez JR, Habegger K, Harper LM, Battarbee AN, Martin SL, Moore BA, Fouts AE, Bahorski J, Chandler-Laney PC. Leptin resistance in children with in utero exposure to maternal obesity and gestational diabetes. Pediatr Obes 2023; 18:e13081. [PMID: 37859518 PMCID: PMC10841866 DOI: 10.1111/ijpo.13081] [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: 05/03/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Leptin resistance occurs with obesity, but it is unknown if individuals at risk for obesity develop leptin resistance prior to obesity. OBJECTIVE Investigate whether leptin resistance is independent of weight status in children at risk for obesity due to intrauterine exposure to maternal obesity or gestational diabetes mellitus (GDM). METHODS Mother-child dyads (N = 179) were grouped by maternal pregnancy weight and GDM status: (1) normal weight, no GDM; (2) overweight/obesity, no GDM; (3) overweight/obesity with GDM. Children (4-10 years) were further stratified by current body mass index (BMI) <85th or ≥85th percentile. Leptin resistance of children and mothers was calculated as fasting leptin/fat mass index. Two-way ANOVA was used to assess whether leptin concentrations and leptin resistance differed by current weight status or in utero exposure group, after adjusting for race, sex and Tanner stage. RESULTS Children with a BMI ≥85th percentile had more leptin resistance than those with a BMI <85th percentile (p < 0.001), but leptin resistance did not differ by in utero exposure. Similarly, leptin resistance in women was associated with weight status and not prior GDM. CONCLUSIONS Results suggest that leptin concentrations are associated with obesity but not risk for obesity based on in utero exposure to maternal obesity or GDM.
Collapse
Affiliation(s)
- Alysha B. Everett
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - W. Timothy Garvey
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jose R. Fernandez
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kirk Habegger
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lorie M. Harper
- Department of Women’s Health, Division of Maternal-Fetal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Ashley N. Battarbee
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Samantha L. Martin
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Women’s Reproductive Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Bethany A. Moore
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Amelia E. Fouts
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jessica Bahorski
- School of Nursing, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Paula C. Chandler-Laney
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
29
|
Enebe JT, Enebe NO, Nwagha TU, Meka IA, Nwankwo ME, Izuka EO, Egede JO, Ugwu IA, Okoro NI, Okoye HC, Iyoke CA. Serum leptin levels and relationship with maternal weight gain at term among obese and non-obese pregnant women in Enugu, Nigeria: a comparative cross-sectional study. J Int Med Res 2023; 51:3000605231213265. [PMID: 38017364 PMCID: PMC10686020 DOI: 10.1177/03000605231213265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 10/23/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVE To determine and compare the mean maternal serum leptin levels, the prevalence of high serum leptin levels and mean gestational weight gain at term among obese and non-obese pregnant women in Enugu, Nigeria. METHODS This cross-sectional comparative study enrolled obese and non-obese pregnant women. The serum leptin levels of the women were determined using an enzyme-linked immunosorbent assay kit. Anthropometric and sociodemographic data were obtained and compared. Mean weight gain during pregnancy was determined. RESULTS A total of 170 pregnant women were included in the study. The mean ± SD serum leptin level (99.39 ± 50.2 ng/ml) and the prevalence of hyperleptinaemia (81 of 85 patients; 95.3%) among the obese pregnant women at term were significantly higher than those of the non-obese pregnant women (48.98 ± 30.35 ng/ml/65 of 85 patients; 76.5%). The mean percentage weight gain was significantly higher in the non-obese women compared with the obese women at term. The predictors of high maternal serum leptin level at term among the participants were the employment status and levels of education of the participants. CONCLUSION Maternal serum leptin level, maternal weight gain and prevalence of hyperleptinaemia at term were significantly higher in the obese compared with the non-obese pregnant women.
Collapse
Affiliation(s)
- Joseph Tochukwu Enebe
- Department of Obstetrics and Gynaecology, Enugu State University of Science and Technology, College of Medicine/Teaching Hospital, Parklane, Enugu, Nigeria
| | - Nympha Onyinye Enebe
- Department of Community Medicine, University of Nigeria Teaching Hospital (UNTH), Ituku Ozalla, Enugu, Nigeria
| | - Theresa Ukamaka Nwagha
- Department of Haematology and Immunology, University of Nigeria Teaching Hospital (UNTH), Ituku Ozalla, Enugu, Nigeria
- Department of Haematology and Immunology, College of Medicine, Ituku Ozalla Campus, Enugu, Nigeria
| | - Ijeoma Angela Meka
- Department of Chemical Pathology, College of Medicine, University of Nigeria Teaching Hospital (UNTH), Ituku Ozalla, Enugu, Nigeria
| | - Malackay Ezenwaeze Nwankwo
- Department of Obstetrics and Gynaecology, Enugu State University of Science and Technology, College of Medicine/Teaching Hospital, Parklane, Enugu, Nigeria
| | - Emmanuel Obiora Izuka
- Department of Obstetrics and Gynaecology, University of Nigeria Teaching Hospital (UNTH), Ituku Ozalla, Enugu, Nigeria
| | - John Okafor Egede
- Department of Obstetrics and Gynaecology, Alex Ekwueme Federal University Teaching Hospital, Abakaliki, Nigeria
| | - Innocent Anayochukwu Ugwu
- Department of Obstetrics and Gynaecology, Enugu State University of Science and Technology, College of Medicine/Teaching Hospital, Parklane, Enugu, Nigeria
| | - Ngozi Ijeoma Okoro
- Department of Chemical Pathology, College of Medicine, Enugu State University of Science and Technology (ESUT), Enugu, Nigeria
| | - Helen Chioma Okoye
- Department of Haematology and Immunology, University of Nigeria Teaching Hospital (UNTH), Ituku Ozalla, Enugu, Nigeria
| | - Chukwuemeka Anthony Iyoke
- Department of Obstetrics and Gynaecology, College of Medicine, University of Nigeria Ituku/Ozalla Campus, Enugu State, Nigeria
| |
Collapse
|
30
|
Basak S, Varma S, Duttaroy AK. Modulation of fetoplacental growth, development and reproductive function by endocrine disrupters. Front Endocrinol (Lausanne) 2023; 14:1215353. [PMID: 37854189 PMCID: PMC10579913 DOI: 10.3389/fendo.2023.1215353] [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: 05/01/2023] [Accepted: 09/14/2023] [Indexed: 10/20/2023] Open
Abstract
Maternal endocrine homeostasis is vital to a successful pregnancy, regulated by several hormones such as human chorionic gonadotropin, estrogen, leptin, glucocorticoid, insulin, prostaglandin, and others. Endocrine stress during pregnancy can modulate nutrient availability from mother to fetus, alter fetoplacental growth and reproductive functions. Endocrine disrupters such as bisphenols (BPs) and phthalates are exposed in our daily life's highest volume. Therefore, they are extensively scrutinized for their effects on metabolism, steroidogenesis, insulin signaling, and inflammation involving obesity, diabetes, and the reproductive system. BPs have their structural similarity to 17-β estradiol and their ability to bind as an agonist or antagonist to estrogen receptors to elicit an adverse response to the function of the endocrine and reproductive system. While adults can negate the adverse effects of these endocrine-disrupting chemicals (EDCs), fetuses do not equip themselves with enzymatic machinery to catabolize their conjugates. Therefore, EDC exposure makes the fetoplacental developmental window vulnerable to programming in utero. On the one hand prenatal BPs and phthalates exposure can impair the structure and function of the ovary and uterus, resulting in placental vascular defects, inappropriate placental expression of angiogenic growth factors due to altered hypothalamic response, expression of nutrient transporters, and epigenetic changes associated with maternal endocrine stress. On the other, their exposure during pregnancy can affect the offspring's metabolic, endocrine and reproductive functions by altering fetoplacental programming. This review highlights the latest development in maternal metabolic and endocrine modulations from exposure to estrogenic mimic chemicals on subcellular and transgenerational changes in placental development and its effects on fetal growth, size, and metabolic & reproductive functions.
Collapse
Affiliation(s)
- Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Saikanth Varma
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
31
|
Chen K, Geng H, Liu J, Ye C. Alteration in gut mycobiota of patients with polycystic ovary syndrome. Microbiol Spectr 2023; 11:e0236023. [PMID: 37702484 PMCID: PMC10580825 DOI: 10.1128/spectrum.02360-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/13/2023] [Indexed: 09/14/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a serious disease characterized by high androgen, insulin resistance (IR), hyperglycemia, and obesity, leading to infertility. The gut mycobiota has been reported to evolve in metabolic diseases including obesity, hyperglycemia, and fatty liver. However, little is known about the gut mycobiota and PCOS. In the current study, we recruited 17 PCOS patients and 17 age-matched healthy controls for community structure and functional analysis of the gut mycobiota. The results showed that PCOS patients have reduced diversity and richness of the gut microbiota compared with healthy controls. β-Diversity analysis showed that the community structure of the gut microbiota of patients with PCOS was significantly different from healthy controls. At the phylum level, PCOS patients have reduced Basidiomycota and increased Ascomycota compared with healthy controls. At the family level, the higher relative abundance of Saccharomycetaceae and lower Trichosporonaceae and Ascomycota_unclassified were detected in PCOS patients than in healthy controls. At the genus level, different microbial compositions were also observed between PCOS patients and healthy controls. In addition, PICRUSt2 showed that patients with PCOS have different microbial functions in the gut compared with healthy controls. LEfSe indicated that Saccharomyces and Lentinula were enriched in the fecal samples of PCOS patients, while Aspergillus was depleted compared with healthy controls. Our finding indicates that PCOS patients have different community structures and functions of the gut mycobiota, which provides new insight into PCOS pathogenesis and intervention. IMPORTANCE It was found that intestinal fungi as well as serum metabolites in PCOS patients were significantly different from those in healthy subjects. However, no studies have been done to show exactly which fungus interacts with which bacteria in humans or which fungus acts alone. As fungal research progresses, it will be possible to fill this gap.
Collapse
Affiliation(s)
- Ke Chen
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Huafeng Geng
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Junbao Liu
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Cong Ye
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
32
|
Gonçalves CCRA, Feitosa BM, Cavalcante BV, Lima ALGDSB, de Souza CM, Joventino LB, Cavalcante MB. Obesity and recurrent miscarriage: The interconnections between adipose tissue and the immune system. Am J Reprod Immunol 2023; 90:e13757. [PMID: 37641378 DOI: 10.1111/aji.13757] [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: 05/08/2023] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 08/31/2023] Open
Abstract
Currently, obesity is considered a global public health problem. It is the main risk factor for noncommunicable diseases and reproductive complications, such as recurrent miscarriage (RM). RM affects approximately 1% of couples of reproductive age, and recent studies suggest that its prevalence is increasing. Immunological abnormalities may be responsible for a significant number of cases of unexplained RM. Obesity is recognized as a chronic low-grade inflammatory condition. The accumulation of fat in obese adipose tissue promotes changes in the local and systemic immune response. Adipokines, exosomes, micro-RNAs, lipids, and other factors released or secreted by adipose tissue are responsible for the interconnection between obesity and the immune system. Obesity-induced dysregulation of the innate and acquired immune response is also involved in the immunopathology of pregnancy loss in patients with unexplained RM. Therefore, understanding the communication pathways between maternal adipose tissue and the immune response in women living with obesity and RM is an important objective. Thus, diagnostic tools and new immunomodulatory therapies may be proposed for the management of patients with concurrent obesity and RM.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marcelo Borges Cavalcante
- Medical School, Universidade de Fortaleza (UNIFOR), Fortaleza, CE, Brazil
- Postgraduate Program in Medical Sciences, Universidade de Fortaleza (UNIFOR), Fortaleza, CE, Brazil
- CONCEPTUS - Reproductive Medicine, Fortaleza, Brazil
| |
Collapse
|
33
|
Guadix P, Corrales I, Vilariño-García T, Rodríguez-Chacón C, Sánchez-Jiménez F, Jiménez-Cortegana C, Dueñas JL, Sánchez-Margalet V, Pérez-Pérez A. Expression of nutrient transporters in placentas affected by gestational diabetes: role of leptin. Front Endocrinol (Lausanne) 2023; 14:1172831. [PMID: 37497352 PMCID: PMC10366688 DOI: 10.3389/fendo.2023.1172831] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/14/2023] [Indexed: 07/28/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is the most frequent pathophysiological state of pregnancy, which in many cases produces fetuses with macrosomia, requiring increased nutrient transport in the placenta. Recent studies by our group have demonstrated that leptin is a key hormone in placental physiology, and its expression is increased in placentas affected by GDM. However, the effect of leptin on placental nutrient transport, such as transport of glucose, amino acids, and lipids, is not fully understood. Thus, we aimed to review literature on the leptin effect involved in placental nutrient transport as well as activated leptin signaling pathways involved in the expression of placental transporters, which may contribute to an increase in placental nutrient transport in human pregnancies complicated by GDM. Leptin appears to be a relevant key hormone that regulates placental transport, and this regulation is altered in pathophysiological conditions such as gestational diabetes. Adaptations in the placental capacity to transport glucose, amino acids, and lipids may underlie both under- or overgrowth of the fetus when maternal nutrient and hormone levels are altered due to changes in maternal nutrition or metabolic disease. Implementing new strategies to modulate placental transport may improve maternal health and prove effective in normalizing fetal growth in cases of intrauterine growth restriction and fetal overgrowth. However, further studies are needed to confirm this hypothesis.
Collapse
Affiliation(s)
- Pilar Guadix
- Obstetrics and Gynecology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, Seville, Spain
| | - Isabel Corrales
- Obstetrics and Gynecology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, Seville, Spain
| | - Teresa Vilariño-García
- Clinical Biochemistry Service, Virgen del Rocio University Hospital, School of Medicine, University of Seville, Seville, Spain
| | - Carmen Rodríguez-Chacón
- Clinical Biochemistry Service, Virgen Macarena University Hospital and Department of Medical Biochemistry and Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Flora Sánchez-Jiménez
- Clinical Biochemistry Service, Virgen Macarena University Hospital and Department of Medical Biochemistry and Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Carlos Jiménez-Cortegana
- Clinical Biochemistry Service, Virgen Macarena University Hospital and Department of Medical Biochemistry and Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - José L. Dueñas
- Obstetrics and Gynecology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, Seville, Spain
| | - Víctor Sánchez-Margalet
- Clinical Biochemistry Service, Virgen Macarena University Hospital and Department of Medical Biochemistry and Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Antonio Pérez-Pérez
- Clinical Biochemistry Service, Virgen Macarena University Hospital and Department of Medical Biochemistry and Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| |
Collapse
|
34
|
Poniedziałek-Czajkowska E, Mierzyński R, Leszczyńska-Gorzelak B. Preeclampsia and Obesity-The Preventive Role of Exercise. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1267. [PMID: 36674022 PMCID: PMC9859423 DOI: 10.3390/ijerph20021267] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/05/2023] [Accepted: 01/08/2023] [Indexed: 06/10/2023]
Abstract
Obesity is now recognized as a worldwide epidemic. An inadequate diet and reduced physical activity are acknowledged as the leading causes of excess body weight. Despite growing evidence that obesity is a risk factor for unsuccessful pregnancies, almost half of all women who become pregnant today are overweight or obese. Common complications of pregnancy in this group of women are preeclampsia and gestational hypertension. These conditions are also observed more frequently in women with excessive weight gain during pregnancy. Preeclampsia is one of the most serious pregnancy complications with an unpredictable course, which in its most severe forms, threatens the life and health of the mother and her baby. The early identification of the risk factors for preeclampsia development, including obesity, allows for the implementation of prophylaxis and a reduction in maternal and fetal complications risk. Additionally, preeclampsia and obesity are the recognized risk factors for developing cardiovascular disease in later life, so prophylaxis and treating obesity are paramount for their prevention. Thus, a proper diet and physical activity might play an essential role in the prophylaxis of preeclampsia in this group of women. Limiting weight gain during pregnancy and modifying the metabolic risk factors with regular physical exercise creates favorable metabolic conditions for pregnancy development and benefits the elements of the pathogenetic sequence for preeclampsia development. In addition, it is inexpensive, readily available and, in the absence of contraindications to its performance, safe for the mother and fetus. However, for this form of prevention to be effective, it should be applied early in pregnancy and, for overweight and obese women, proposed as an essential part of planning pregnancy. This paper aims to present the mechanisms of the development of hypertension in pregnancy in obese women and the importance of exercise in its prevention.
Collapse
|
35
|
Wang Y, Bai X, Guo X, Gao X, Chen Y, Li H, Fan W, Han C. Bioinformatics analysis combined with clinical sample screening reveals that leptin may be a biomarker of preeclampsia. Front Physiol 2023; 13:1031950. [PMID: 36685185 PMCID: PMC9846503 DOI: 10.3389/fphys.2022.1031950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction: Preeclampsia (PE) is a gestational hypertensive disease with unclear pathogenesis. This study aimed to identify the genes that play an important role in determining the pathogenesis of PE using bioinformatics analysis and fundamental researches. Materials and methods: Datasets from the Gene Expression Omnibus (GEO) database were used to screen for differentially expressed genes (DEGs). The NCBI, SangerBox, and other databases were used to analyze the functions of the DEGs. Targetscan7, miRWalk, ENCORI, DIANA TOOLS, CircBank databases, and the Cytoscape tool were used to construct the lncRNA/circRNA-miRNA- LEP network. SRAMP, RPISeq, RBPsuite, and catRPAID were used to analyze the RNA modifications of LEP. Immune cell infiltration was analyzed using the dataset GSE75010. Placental tissues from normal pregnant women and PE patients were collected, screened for gene expression using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blotting. The results were further verified in HTR-8/SVneo cell line hypoxia model and PE mouse model. Results: Our analyses revealed that LEP was significantly upregulated in eight datasets. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses indicated that LEP was involved in the JAK/STAT signaling pathway, angiogenesis, and placental development. Immune cell infiltration analysis showed that M1 and M2 macrophages differed between normal pregnancies and those in PE patients. A competing endogenous RNA (ceRNA) network was constructed, and proteins interacting with LEP were identified. RNA modification sites of LEP were also identified. Finally, the overexpression of LEP in PE was confirmed in clinical samples, HTR-8/SVneo cell line and PE mouse model. Conclusion: Our results indicate that LEP overexpression is associated with PE and may be a potential diagnostic marker and therapeutic target.
Collapse
Affiliation(s)
- Yajuan Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuening Bai
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Guo
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoli Gao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuanyuan Chen
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huanrong Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenjun Fan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Cha Han
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China,*Correspondence: Cha Han,
| |
Collapse
|
36
|
Fan M, Dong L, Meng Y, Wang Y, Zhen J, Qiu J. Leptin Promotes HTR-8/SVneo Cell Invasion via the Crosstalk between MTA1/WNT and PI3K/AKT Pathways. DISEASE MARKERS 2022; 2022:7052176. [PMID: 36457544 PMCID: PMC9708374 DOI: 10.1155/2022/7052176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 10/16/2022] [Accepted: 11/08/2022] [Indexed: 08/31/2023]
Abstract
The process of placental invasion is essential for a successful pregnancy. Leptin is involved in trophoblast invasiveness, and its dysregulation is connected with a series of diseases, including preeclampsia. However, the knowledge of the precise mechanisms in leptin-induced trophoblast invasiveness is still limited. According to the present research, transwell assay suggested that leptin is a dose- and time-dependent regulator in inducing HTR-8/SVneo cell invasion. Western blot analysis and immunofluorescence staining revealed that leptin-induced MMP9 expression is essential in the invasion process of HTR-8/SVneo cells. Mechanistically, we demonstrated that leptin activated β-catenin via the crosstalk between the MTA1/WNT and PI3K/AKT pathways. Besides, we showed that downregulating the key molecules in the signaling pathways by siRNA can inhibit leptin-induced MMP9 expression and further suppress invasion of HTR-8/SVneo cells. In conclusion, our study revealed a new regulatory mechanism of leptin-induced HTR-8/SVneo cell invasiveness and will provide novel insights into the causes and potential therapeutic targets for diseases related to dysregulation of trophoblast invasion in the future.
Collapse
Affiliation(s)
- Minghua Fan
- Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Lihua Dong
- Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yanping Meng
- Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Yao Wang
- Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Junhui Zhen
- Department of Pathology, School of Medicine, Shandong University, Jinan, 250021 Shandong, China
| | - Jianqing Qiu
- Department of Obstetrics and Gynecology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| |
Collapse
|
37
|
Kristiansen O, Roland MC, Zucknick M, Reine TM, Kolset SO, Henriksen T, Lekva T, Michelsen T. Maternal body mass index and placental weight: a role for fetal insulin, maternal insulin and leptin. J Endocrinol Invest 2022; 45:2105-2121. [PMID: 35781790 PMCID: PMC9525437 DOI: 10.1007/s40618-022-01842-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 06/11/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE Placental weight (PW) has been found to mediate the main effect of maternal BMI on fetal size. Still, the BMI-PW association is poorly understood. Therefore, we aimed to explore potential explanatory variables, including gestational weight gain (GWG), early- and late-pregnancy circulating levels of maternal glucose, insulin, leptin, adiponectin, triglycerides, LDL-C, and HDL-C, and fetal insulin. METHODS We included two studies of pregnant women from Oslo University Hospital, Norway: the prospective STORK (n = 263) and the cross-sectional 4-vessel method study (4-vessel; n = 165). We used multiple linear regression for data analyses. A non-linear BMI-PW association was observed, which leveled off from BMI25. Therefore, BMI <25 and ≥25 were analyzed separately (n = 170/122 and 93/43 for STORK/4-vessel). Confounding variables included maternal age, parity, and gestational age. RESULTS PW increased significantly per kg m-2 only among BMI <25 (univariate model's std.β[p] = 0.233 [0.002] vs. 0.074[0.48]/0.296[0.001] vs. -0.030[0.85] for BMI <25 vs. ≥25 in STORK/4-vessel). Maternal early- but not late-pregnancy insulin and term fetal insulin were associated with PW. The estimated effect of early pregnancy insulin was similar between the BMI groups but statistically significant only among BMI <25 (std.β[p] = 0.182[0.016] vs. 0.203[0.07] for BMI <25 vs. ≥25). Late pregnancy leptin was inversely associated with PW with a 1.3/1.7-fold greater effect among BMI ≥25 than BMI <25 in the STORK/4-vessel. CONCLUSIONS The BMI-PW association was non-linear: an association was observed for BMI <25 but not for BMI ≥25. Leptin may be involved in the non-linear association through a placental-adipose tissue interplay. Maternal early pregnancy insulin and fetal insulin at term were associated with PW.
Collapse
Affiliation(s)
- O Kristiansen
- Department of Obstetrics, Division of Obstetrics and Gynecology, Rikshospitalet, Oslo University Hospital, 0424, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318, Oslo, Norway.
- Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317, Oslo, Norway.
| | - M C Roland
- Department of Obstetrics, Division of Obstetrics and Gynecology, Rikshospitalet, Oslo University Hospital, 0424, Oslo, Norway
- Norwegian Research Centre for Women's Health, Rikshospitalet, Oslo University Hospital, 0424, Oslo, Norway
| | - M Zucknick
- Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - T M Reine
- Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317, Oslo, Norway
- Institute of Cancer Genetics and Informatics, Radiumhospitalet, Oslo University Hospital, 0424, Oslo, Norway
| | - S O Kolset
- Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317, Oslo, Norway
| | - T Henriksen
- Department of Obstetrics, Division of Obstetrics and Gynecology, Rikshospitalet, Oslo University Hospital, 0424, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318, Oslo, Norway
| | - T Lekva
- Research Institute of Internal Medicine, Rikshospitalet, Oslo University Hospital, 0424, Oslo, Norway
| | - T Michelsen
- Department of Obstetrics, Division of Obstetrics and Gynecology, Rikshospitalet, Oslo University Hospital, 0424, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318, Oslo, Norway
| |
Collapse
|
38
|
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: 4] [Impact Index Per Article: 1.3] [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
|
39
|
Rogers BM, Stephens JM, Sones JL. Linking Inflammatory Adipose Tissue to Placental Abnormalities in Obese Preeclamptic Pregnancies. Physiol Genomics 2022; 54:319-324. [PMID: 35816649 PMCID: PMC9377783 DOI: 10.1152/physiolgenomics.00041.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Preeclampsia (PE), a pregnancy-specific disorder, is characterized by maternal hypertension and proteinuria or another accompanying sign/symptom of multi-organ dysfunction. Maternal symptoms resolve with delivery of the baby and, importantly, the placenta. Therefore, the placenta plays a causal role in PE. However, the precise cause of abnormal placental development and/or function is unknown. Women with obesity have an increased risk of developing PE that is potentially related to the increased inflammation that accompanies increased maternal adiposity. Furthermore, inflammatory adipokines, i.e., leptin, have been linked to the development of systemic inflammation, hypertension, and other adverse outcomes associated with PE. Rodent models that recapitulate key pathophysiologic features of the maternal and fetal syndrome have been used translationally to study PE. This review covers inflammatory adipokines, immune cells and impaired placental development associated with PE in women and in rodent models of PE that utilize functional genomics to test causation.
Collapse
Affiliation(s)
- Brianna M Rogers
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States.,School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Jacqueline M Stephens
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Jenny L Sones
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States.,School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
40
|
Alston MC, Redman LM, Sones JL. An Overview of Obesity, Cholesterol, and Systemic Inflammation in Preeclampsia. Nutrients 2022; 14:2087. [PMID: 35631228 PMCID: PMC9143481 DOI: 10.3390/nu14102087] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/12/2022] [Accepted: 05/14/2022] [Indexed: 02/06/2023] Open
Abstract
Preeclampsia (PE), an inflammatory state during pregnancy, is a significant cause of maternal and fetal morbidity and mortality. Adverse outcomes associated with PE include hypertension, proteinuria, uterine/placental abnormalities, fetal growth restriction, and pre-term birth. Women with obesity have an increased risk of developing PE likely due to impaired placental development from altered metabolic homeostasis. Inflammatory cytokines from maternal adipose tissue and circulating cholesterol have been linked to systemic inflammation, hypertension, and other adverse outcomes associated with PE. This review will summarize the current knowledge on the role of nutrients, obesity, and cholesterol signaling in PE with an emphasis on findings from preclinical models.
Collapse
Affiliation(s)
- Morgan C. Alston
- Departments of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA;
- Reproductive Endocrinology and Women’s Health Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA;
| | - Leanne M. Redman
- Reproductive Endocrinology and Women’s Health Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA;
| | - Jennifer L. Sones
- Departments of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA;
- Reproductive Endocrinology and Women’s Health Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA;
| |
Collapse
|
41
|
Hayashi D, Noguchi E, Maruo K, Hara M, Nakayama SF, Takada H. Maternal BMI and allergy in children until 3 years of age (JECS). THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2022; 1:43-50. [PMID: 37780583 PMCID: PMC10510001 DOI: 10.1016/j.jacig.2022.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/16/2022] [Accepted: 02/20/2022] [Indexed: 10/03/2023]
Abstract
Background Maternal prepregnancy body mass index (BMI) may influence allergic diseases in the children who are the product of those pregnancies. Objective The purpose of our study was to investigate the association between mothers' prepregnancy BMI and the risk of physician-diagnosed asthma, food allergy (FA), and atopic dermatitis (AD) in their children during the first 3 years of life. Methods Data on mothers' prepregnancy BMI and physician-diagnosed asthma, FA, and AD in their children until the age of 3 years were obtained from the Japan Environment and Children's Study, a nationwide birth cohort study that has recruited 103,099 pregnant women between 2011 and 2014. Logistic regression analysis was used to analyze the results. Results We analyzed 67,204 mother-child pairs with available information on physician-diagnosed allergic diseases. The risk of asthma was significantly higher in children born to overweight mothers (adjusted OR [aOR] =1.17 [95% CI = 1.07-1.28]) and obese mothers (aOR = 1.28 [95% CI = 1.08-1.50]), whereas the risk of FA, cow's milk allergy, and egg allergy decreased significantly in children born to overweight mothers (aOR = 0.84 [95% CI = 0.76-0.92]; aOR = 0.78 [95% CI = 0.64-0.93]; and aOR = 0.83 [95% CI = 0.74-0.94]) and obese mothers (aOR = 0.81 [95% CI = 0.67-0.97]; aOR = 0.58 [95% CI = 0.36-0.87]; and aOR = 0.73 [95% CI = 0.56-0.93]) compared with in children born to normal weight mothers, respectively. Associations between AD and maternal BMI were not detected. Conclusion Our study showed that an increase in mothers' prepregnancy BMI was associated with an increase in asthma prevalence and a decrease in FA prevalence in their children. Further studies are needed to reveal the mechanisms associated with maternal BMI and pediatric allergic diseases.
Collapse
Affiliation(s)
- Daisuke Hayashi
- Japan Environment and Children’s Study Program Office, National Institute for Environmental Studies, Tsukuba, Japan
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Pediatrics, Tsukuba Medical Center Hospital, Tsukuba, Japan
| | - Emiko Noguchi
- Japan Environment and Children’s Study Program Office, National Institute for Environmental Studies, Tsukuba, Japan
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kazushi Maruo
- Department of Biostatistics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Monami Hara
- Japan Environment and Children’s Study Program Office, National Institute for Environmental Studies, Tsukuba, Japan
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba, Japan
| | - Shoji F. Nakayama
- Japan Environment and Children’s Study Program Office, National Institute for Environmental Studies, Tsukuba, Japan
| | - Hidetoshi Takada
- Department of Child Health, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba, Japan
| |
Collapse
|
42
|
The WWOX/HIF1A Axis Downregulation Alters Glucose Metabolism and Predispose to Metabolic Disorders. Int J Mol Sci 2022; 23:ijms23063326. [PMID: 35328751 PMCID: PMC8955937 DOI: 10.3390/ijms23063326] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/10/2022] [Accepted: 03/17/2022] [Indexed: 02/01/2023] Open
Abstract
Recent reports indicate that the hypoxia-induced factor (HIF1α) and the Warburg effect play an initiating role in glucotoxicity, which underlies disorders in metabolic diseases. WWOX has been identified as a HIF1α regulator. WWOX downregulation leads to an increased expression of HIF1α target genes encoding glucose transporters and glycolysis’ enzymes. It has been proven in the normoglycemic mice cells and in gestational diabetes patients. The aim of the study was to determine WWOX’s role in glucose metabolism regulation in hyperglycemia and hypoxia to confirm its importance in the development of metabolic disorders. For this purpose, the WWOX gene was silenced in human normal fibroblasts, and then cells were cultured under different sugar and oxygen levels. Thereafter, it was investigated how WWOX silencing alters the genes and proteins expression profile of glucose transporters and glycolysis pathway enzymes, and their activity. In normoxia normoglycemia, higher glycolysis genes expression, their activity, and the lactate concentration were observed in WWOX KO fibroblasts in comparison to control cells. In normoxia hyperglycemia, it was observed a decrease of insulin-dependent glucose uptake and a further increase of lactate. It likely intensifies hyperglycemia condition, which deepen the glucose toxic effect. Then, in hypoxia hyperglycemia, WWOX KO caused weaker glucose uptake and elevated lactate production. In conclusion, the WWOX/HIF1A axis downregulation alters glucose metabolism and probably predispose to metabolic disorders.
Collapse
|
43
|
Cowman W, Scroggins SM, Hamilton WS, Karras AE, Bowdler NC, Devor EJ, Santillan MK, Santillan DA. Association between plasma leptin and cesarean section after induction of labor: a case control study. BMC Pregnancy Childbirth 2022; 22:29. [PMID: 35031012 PMCID: PMC8759283 DOI: 10.1186/s12884-021-04372-6] [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: 05/26/2021] [Accepted: 12/30/2021] [Indexed: 11/17/2022] Open
Abstract
Background Obesity in pregnancy is common, with more than 50% of pregnant women being overweight or obese. Obesity has been identified as an independent predictor of dysfunctional labor and is associated with increased risk of failed induction of labor resulting in cesarean section. Leptin, an adipokine, is secreted from adipose tissue under the control of the obesity gene. Concentrations of leptin increase with increasing percent body fat due to elevated leptin production from the adipose tissue of obese individuals. Interestingly, the placenta is also a major source of leptin production during pregnancy. Leptin has regulatory effects on neuronal tissue, vascular smooth muscle, and nonvascular smooth muscle systems. It has also been demonstrated that leptin has an inhibitory effect on myometrial contractility with both intensity and frequency of contractions decreased. These findings suggest that leptin may play an important role in dysfunctional labor and be associated with the outcome of induction of labor at term. Our aim is to determine whether maternal plasma leptin concentration is indicative of the outcome of induction of labor at term. We hypothesize that elevated maternal plasma leptin levels are associated with a failed term induction of labor resulting in a cesarean delivery. Methods In this case-control study, leptin was measured in 3rd trimester plasma samples. To analyze labor outcomes, 174 women were selected based on having undergone an induction of labor (IOL), (115 women with successful IOL and 59 women with a failed IOL). Plasma samples and clinical information were obtained from the UI Maternal Fetal Tissue Bank (IRB# 200910784). Maternal plasma leptin and total protein concentrations were measured using commercially available assays. Bivariate analyses and logistic regression models were constructed using regression identified clinically significant confounding variables. All variables were tested at significance level of 0.05. Results Women with failed IOL had higher maternal plasma leptin values (0.5 vs 0.3 pg, P = 0.01). These women were more likely to have obesity (mean BMI 32 vs 27 kg/m2, P = 0.0002) as well as require multiple induction methods (93% vs 73%, p = 0.008). Logistic regression showed Bishop score (OR 1.5, p < 0.001), BMI (OR 0.92, P < 0.001), preeclampsia (OR 0.12, P = 0.010), use of multiple methods of induction (OR 0.22, P = 0.008) and leptin (OR 0.42, P = 0.017) were significantly associated with IOL outcome. Specifically, after controlling for BMI, Bishop Score, and preeclampsia, leptin was still predictive of a failed IOL with an odds ratio of 0.47 (P = 0.046). Finally, using leptin as a predictor for fetal outcomes, leptin was also associated with of fetal intolerance of labor, with an odds ratio of 2.3 (P = 0.027). This association remained but failed to meet statistical significance when controlling for successful (IOL) (OR 1.5, P = 0.50). Conclusions Maternal plasma leptin may be a useful tool for determining which women are likely to have a failed induction of labor and for counseling women about undertaking an induction of labor versus proceeding with cesarean delivery.
Collapse
Affiliation(s)
- Whitney Cowman
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA.,Present Address: Department of Obstetrics & Gynecology, Iowa Methodist Medical Center, 1200 Pleasant Street, Des Moines, IA, 50309, USA
| | - Sabrina M Scroggins
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA
| | - Wendy S Hamilton
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA
| | - Alexandra E Karras
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA
| | - Noelle C Bowdler
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA
| | - Eric J Devor
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA
| | - Mark K Santillan
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA
| | - Donna A Santillan
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA.
| |
Collapse
|
44
|
Bruno A, Ferrante G, Di Vincenzo S, Pace E, La Grutta S. Leptin in the Respiratory Tract: Is There a Role in SARS-CoV-2 Infection? Front Physiol 2022; 12:776963. [PMID: 35002761 PMCID: PMC8727443 DOI: 10.3389/fphys.2021.776963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/26/2021] [Indexed: 12/23/2022] Open
Abstract
Leptin is a pleiotropic adipocytokine involved in several physiologic functions, with a known role in innate and adaptive immunity as well as in tissue homeostasis. Long- and short-isoforms of leptin receptors are widely expressed in many peripheral tissues and organs, such as the respiratory tract. Similar to leptin, microbiota affects the immune system and may interfere with lung health through the bidirectional crosstalk called the “gut-lung axis.” Obesity leads to impaired protective immunity and altered susceptibility to pulmonary infections, as those by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although it is known that leptin and microbiota link metabolism and lung health, their role within the SARS-CoV2 coronavirus disease 2019 (COVID-19) deserves further investigations. This review aimed to summarize the available evidence about: (i) the role of leptin in immune modulation; (ii) the role of gut microbiota within the gut-lung axis in modulating leptin sensitivity; and (iii) the role of leptin in the pathophysiology of COVID-19.
Collapse
Affiliation(s)
- Andreina Bruno
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Giuliana Ferrante
- Pediatric Division, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy
| | - Serena Di Vincenzo
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Elisabetta Pace
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Stefania La Grutta
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| |
Collapse
|
45
|
Stefaniak M, Dmoch-Gajzlerska E. Maternal Serum and Cord Blood Leptin Concentrations at Delivery in Normal Pregnancies and in Pregnancies Complicated by Intrauterine Growth Restriction. Obes Facts 2022; 15:62-69. [PMID: 34872096 PMCID: PMC8820166 DOI: 10.1159/000519609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 09/07/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Leptin is a polypeptide hormone, and in pregnancy, it is secreted by the placenta and maternal and fetal adipose tissues. Normal leptin production is a factor responsible for uncomplicated gestation, embryo development, and fetal growth. The study compared maternal serum and cord blood leptin concentrations at delivery in normal pregnancies and in pregnancies complicated by intrauterine growth restriction (IUGR). METHODS The study was performed in 25 pregnant women with isolated IUGR and in 194 pregnant women without any complications. Leptin concentrations in maternal serum and in cord blood samples collected at delivery were measured by ELISA and subsequently analyzed by maternal body mass index (BMI), mode of delivery, and infant gender and birth weight. For comparative analyses of normally distributed variables, parametric tests were used, that is, the Student t test and a one-way ANOVA. The nonparametric Mann-Whitney test was used when the distribution was not normal. The Pearson correlation coefficient was calculated to assess the correlation between normally distributed variables (p < 0.05). RESULTS In pregnancies complicated by IUGR, the mean maternal serum leptin concentration at delivery was significantly higher (52.73 ± 30.49 ng/mL) than in normal pregnancies (37.17 ± 28.07 ng/mL) (p = 0.01). The mean cord blood leptin concentration in pregnancies complicated by IUGR was 7.97 ± 4.46 ng/mL and significantly lower than in normal pregnancies (14.78 ± 15.97 ng/mL) (p = 0.04). In normal pregnancies, but not in pregnancies complicated by IUGR, a statistically significant correlation was established between maternal serum leptin concentrations and maternal BMI at delivery (r = 0.22; p = 0.00). No statistically significant correlation was found between cord blood leptin concentrations and maternal BMI in either study subjects or controls. In normal pregnancies, but not in pregnancies complicated by IUGR, a strong correlation was observed between cord blood leptin concentrations and birth weight (r = 0.23; p = 0.00). CONCLUSIONS Elevated maternal blood leptin concentrations in pregnancies complicated by IUGR may indicate a significant adverse effect of elevated leptin on fetal growth. The differences in leptin concentrations, measured in maternal serum and in cord blood, between the study subjects and controls suggest that deregulated leptin levels may increase the risk of obstetric complications associated with placental insufficiency.
Collapse
|
46
|
Metabolic-endocrine disruption due to preterm birth impacts growth, body composition, and neonatal outcome. Pediatr Res 2022; 91:1350-1360. [PMID: 34040160 PMCID: PMC9197767 DOI: 10.1038/s41390-021-01566-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 02/05/2023]
Abstract
Despite optimized nutrition, preterm-born infants grow slowly and tend to over-accrete body fat. We hypothesize that the premature dissociation of the maternal-placental-fetal unit disrupts the maintenance of physiological endocrine function in the fetus, which has severe consequences for postnatal development. This review highlights the endocrine interactions of the maternal-placental-fetal unit and the early perinatal period in both preterm and term infants. We report on hormonal levels (including tissue, thyroid, adrenal, pancreatic, pituitary, and placental hormones) and nutritional supply and their impact on infant body composition. The data suggest that the premature dissociation of the maternal-placental-fetal unit leads to a clinical picture similar to panhypopituitarism. Further, we describe how the premature withdrawal of the maternal-placental unit, neonatal morbidities, and perinatal stress can cause differences in the levels of growth-promoting hormones, particularly insulin-like growth factors (IGF). In combination with the endocrine disruption that occurs following dissociation of the maternal-placental-fetal unit, the premature adaptation to the extrauterine environment leads to early and fast accretion of fat mass in an immature body. In addition, we report on interventional studies that have aimed to compensate for hormonal deficiencies in infants born preterm through IGF therapy, resulting in improved neonatal morbidity and growth. IMPACT: Preterm birth prematurely dissociates the maternal-placental-fetal unit and disrupts the metabolic-endocrine maintenance of the immature fetus with serious consequences for growth, body composition, and neonatal outcomes. The preterm metabolic-endocrine disruption induces symptoms resembling anterior pituitary failure (panhypopituitarism) with low levels of IGF-1, excessive postnatal fat mass accretion, poor longitudinal growth, and failure to thrive. Appropriate gestational age-adapted nutrition alone seems insufficient for the achievement of optimal growth of preterm infants. Preliminary results from interventional studies show promising effects of early IGF-1 supplementation on postnatal development and neonatal outcomes.
Collapse
|
47
|
Molin J, Vanky E, Løvvik TS, Dehlin E, Bixo M. Gestational weight gain, appetite regulating hormones, and metformin treatment in polycystic ovary syndrome: A longitudinal, placebo-controlled study. BJOG 2021; 129:1112-1121. [PMID: 34865304 DOI: 10.1111/1471-0528.17042] [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] [Received: 06/23/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To explore mechanisms that modulate gestational weight gain (GWG) in women with polycystic ovary syndrome (PCOS) and healthy controls. DESIGN Sub-sample of randomised controlled trials (PCOS) combined with a prospective cohort (controls). SETTING Eleven Norwegian, Swedish, and Icelandic hospitals. POPULATION Pregnant women with PCOS treated with metformin (PCOS-M, n = 36) or placebo (PCOS-P, n = 37), and healthy pregnant women (HC, n = 15). METHODS Serum levels of the appetite regulating hormones leptin, ghrelin, allopregnanolone, and soluble leptin receptor (sOB-R) were determined in the first and third trimesters. MAIN OUTCOME MEASURES Excessive GWG (eGWG) relative to body mass index according to Institute of Medicine (IOM) guideline. Serum leptin/sOB-R ratio, or free-leptin-index (FLI), as biomarker of leptin sensitivity. Serum ghrelin and allopregnanolone levels. RESULTS The overall prevalence of eGWG was 44% (38/86). Women with eGWG had higher first and third trimester FLI (P < 0.001), and lower third trimester allopregnanolone levels (P = 0.003) versus women with non-eGWG. The prevalence of eGWG was lower in PCOS-M versus PCOS-P (28% versus 62%, odds ratio = 0.4, 95% CI 0.2-0.8, P = 0.005). FLI decreased during pregnancy in PCOS-M (P = 0.01), but remained unaltered in PCOS-P and HC. Ghrelin and allopregnanolone levels were comparable in PCOS-M, PCOS-P and HC throughout pregnancy. CONCLUSION Excessive GWG is associated with enhanced leptin resistance, and attenuated physiological increase in serum allopregnanolone levels during pregnancy. Metformin reduces the risk for eGWG and improves leptin sensitivity in pregnant women with PCOS.
Collapse
Affiliation(s)
- Johanna Molin
- Department of Clinical Sciences, Obstetrics and Gynaecology, Umeå University, Umeå, Sweden
| | - Eszter Vanky
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Obstetrics and Gynaecology, St Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Tone S Løvvik
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Obstetrics and Gynaecology, St Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Eva Dehlin
- Department of Clinical Sciences, Obstetrics and Gynaecology, Umeå University, Umeå, Sweden
| | - Marie Bixo
- Department of Clinical Sciences, Obstetrics and Gynaecology, Umeå University, Umeå, Sweden
| |
Collapse
|
48
|
Obeidat RA, Abdo N, Sakee B, Alghazo S, Jbarah OF, Hazaimeh EA, Albeitawi S. Maternal and fetal serum leptin levels and their association with maternal and fetal variables and labor: A cross-sectional study. Ann Med Surg (Lond) 2021; 72:103050. [PMID: 34815864 PMCID: PMC8591461 DOI: 10.1016/j.amsu.2021.103050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/06/2021] [Accepted: 11/09/2021] [Indexed: 12/24/2022] Open
Abstract
Background Leptin is a polypeptide hormone that may be implicated in the pathogenesis of various disorders during pregnancy. We sought to determine serum leptin levels among pregnant women and their fetuses and to investigate their association with fetal and maternal variables. Method 452 pregnant women who attended to labor ward between January 2020 and August 2020 were included in the study. Serum leptin concentrations were measured using enzyme-linked immunosorbent assay method. Mann-Whitney U test and Spearman's correlation test were used for statistical analysis. A multivariate linear regression analysis was then performed. Significance level was considered at alpha <0.05. Results The median maternal and fetal serum leptin levels were 6.42 [4.16-8.51] ng/mL and 2.9 [1.03-5.36] ng/mL respectively. There was no significant correlation between maternal and fetal serum leptin levels (p = 0.064). Maternal serum leptin levels correlated positively with maternal body mass index (BMI) (r = 0.117, p = 0.005). Besides, maternal serum leptin levels were significantly higher in nulliparous women (7.57 [4.45-9.30] ng/mL vs. 6.22 [4.02-8.30] ng/mL, p = 0.037) and in women who were in active labor (6.83 [4.39-8.92] ng/mL vs. 6.25 [4.04-8.30] ng/mL, p = 0.047). Fetal serum leptin levels were significantly higher in large for gestational age (LGA) fetuses (4.81 [2.13-7.22] ng/mL vs. 2.80 [0.96-5.16] ng/mL, p = 0.003) and in fetuses with preterm premature ruptures of membranes (PPROM) (5.23 [2.42-8.07] ng/mL vs. 2.86 [1.00-5.23] ng/mL, p = 0.021). Conclusion Maternal serum leptin levels were influenced by maternal BMI, parity and labor. Fetal serum leptin levels were higher among LGA fetuses and in fetuses with PPROM.
Collapse
Affiliation(s)
- Rawan A Obeidat
- Department of Obstetrics and Gynecology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Nour Abdo
- Department of Public Health and Family Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Baraa Sakee
- Department of Obstetrics and Gynecology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Shahed Alghazo
- Department of Obstetrics and Gynecology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F Jbarah
- Department of Neuroscience, Neurosurgery Division, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Ethar A Hazaimeh
- Department of Neuroscience, Neurology Division, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Soha Albeitawi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| |
Collapse
|
49
|
Huang Q, Hao S, You J, Yao X, Li Z, Schilling J, Thyparambil S, Liao WL, Zhou X, Mo L, Ladella S, Davies-Balch SR, Zhao H, Fan D, Whitin JC, Cohen HJ, McElhinney DB, Wong RJ, Shaw GM, Stevenson DK, Sylvester KG, Ling XB. Early-pregnancy prediction of risk for pre-eclampsia using maternal blood leptin/ceramide ratio: discovery and confirmation. BMJ Open 2021; 11:e050963. [PMID: 34824115 PMCID: PMC8627403 DOI: 10.1136/bmjopen-2021-050963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE This study aimed to develop a blood test for the prediction of pre-eclampsia (PE) early in gestation. We hypothesised that the longitudinal measurements of circulating adipokines and sphingolipids in maternal serum over the course of pregnancy could identify novel prognostic biomarkers that are predictive of impending event of PE early in gestation. STUDY DESIGN Retrospective discovery and longitudinal confirmation. SETTING Maternity units from two US hospitals. PARTICIPANTS Six previously published studies of placental tissue (78 PE and 95 non-PE) were compiled for genomic discovery, maternal sera from 15 women (7 non-PE and 8 PE) enrolled at ProMedDx were used for sphingolipidomic discovery, and maternal sera from 40 women (20 non-PE and 20 PE) enrolled at Stanford University were used for longitudinal observation. OUTCOME MEASURES Biomarker candidates from discovery were longitudinally confirmed and compared in parallel to the ratio of placental growth factor (PlGF) and soluble fms-like tyrosine kinase (sFlt-1) using the same cohort. The datasets were generated by enzyme-linked immunosorbent and liquid chromatography-tandem mass spectrometric assays. RESULTS Our discovery integrating genomic and sphingolipidomic analysis identified leptin (Lep) and ceramide (Cer) (d18:1/25:0) as novel biomarkers for early gestational assessment of PE. Our longitudinal observation revealed a marked elevation of Lep/Cer (d18:1/25:0) ratio in maternal serum at a median of 23 weeks' gestation among women with impending PE as compared with women with uncomplicated pregnancy. The Lep/Cer (d18:1/25:0) ratio significantly outperformed the established sFlt-1/PlGF ratio in predicting impending event of PE with superior sensitivity (85% vs 20%) and area under curve (0.92 vs 0.52) from 5 to 25 weeks of gestation. CONCLUSIONS Our study demonstrated the longitudinal measurement of maternal Lep/Cer (d18:1/25:0) ratio allows the non-invasive assessment of PE to identify pregnancy at high risk in early gestation, outperforming the established sFlt-1/PlGF ratio test.
Collapse
Affiliation(s)
| | - Shiying Hao
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Palo Alto, California, USA
| | - Jin You
- Department of Bioengineering, University of California Riverside, Riverside, California, USA
| | | | - Zhen Li
- Department of Surgery, Stanford University, Stanford, California, USA
- Binhai Industrial Technology Research Institute, Zhejiang University, Tianjin, China
- School of Electrical Engineering, Southeast University, Nanjing, China
| | | | | | | | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Lihong Mo
- Department of Obstetrics and Gynecology, University of California San Francisco, Fresno, California, USA
| | - Subhashini Ladella
- Department of Obstetrics and Gynecology, University of California San Francisco, Fresno, California, USA
| | | | - Hangyi Zhao
- Department of Mathematics, Stanford University, Stanford, California, USA
| | - David Fan
- Department of Statistics and Applied Probability, University of California Santa Barbara, Santa Barbara, California, USA
| | - John C Whitin
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Harvey J Cohen
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Doff B McElhinney
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Palo Alto, California, USA
| | - Ronald J Wong
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Gary M Shaw
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - David K Stevenson
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Karl G Sylvester
- Department of Surgery, Stanford University, Stanford, California, USA
| | - Xuefeng B Ling
- Department of Surgery, Stanford University, Stanford, California, USA
| |
Collapse
|
50
|
Savard C, Lemieux S, Plante AS, Gagnon M, Leblanc N, Veilleux A, Tchernof A, Morisset AS. Longitudinal changes in circulating concentrations of inflammatory markers throughout pregnancy: are there associations with diet and weight status? Appl Physiol Nutr Metab 2021; 47:287-295. [PMID: 34767478 DOI: 10.1139/apnm-2021-0395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The natural inflammation occurring during pregnancy can, under certain conditions, be associated with adverse pregnancy outcomes. This study aimed to: 1) quantify changes in circulating concentrations of leptin, adiponectin, interleukin-6 (IL-6) and C-reactive protein (CRP) across trimesters of pregnancy, according to pre-pregnancy body mass index (ppBMI); and 2) examine the trimester-specific associations between the inflammatory markers' concentrations, a Mediterranean diet score (MDS) and the dietary inflammatory index (DII). We measured leptin, adiponectin and IL-6 by ELISA, and CRP by high-sensitivity immunonephelometry, in blood samples from 79 pregnant women (age: 32.1 ± 3.7 years; ppBMI: 25.7 ± 5.8 kg/m2). Three web-based 24h recalls were completed at each trimester and used to compute the MDS and the DII. CRP concentrations remained stable across trimesters, whereas concentrations of leptin and IL-6 increased, and adiponectin concentrations decreased (p<0.001). Changes in leptin and adiponectin concentrations also differed according to ppBMI categories (p<0.05). As for the dietary scores, the only significant association was observed in the second trimester between leptin concentrations and the MDS (r=-0.26, p<0.05). In conclusion, ppBMI and the progression of pregnancy itself probably supplant the potential associations between diet and the inflammation occurring during that period. Novelty: • Circulating leptin and IL-6 concentrations increased across trimesters whereas CRP was stable, and adiponectin decreased. • Variations in circulating leptin and adiponectin concentrations differed by ppBMI categories. • Very few associations were observed between dietary scores and inflammatory markers.
Collapse
Affiliation(s)
- Claudia Savard
- Laval University, 4440, School of Nutrition, Quebec, Quebec, Canada;
| | - Simone Lemieux
- Laval University, School of Nutrition, INAF, Pavillon des services, 2440 Boulevard Hochelaga, Quebec, Quebec, Canada, G1V 0A6;
| | | | - Marianne Gagnon
- Laval University, 4440, School of Nutrition, Quebec, Quebec, Canada.,CHU de Québec-Université Laval, 36896, Endocrinology and Nephrology Unit, Quebec, Quebec, Canada.,Laval University, 4440, Institute of Nutrition and Functional Foods, Quebec, Quebec, Canada;
| | - Nadine Leblanc
- Laval University, Department of Food Science and Nutrition, Quebec, Quebec, Canada;
| | - Alain Veilleux
- Laval University, 4440, School of Nutrition, Quebec, Quebec, Canada;
| | - André Tchernof
- Laval University, Laval University Medical Center, Quebec, Quebec, Canada.,Laval University, Institute of Nutrition and Functional Foods (INAF), Quebec, Quebec, Canada;
| | | |
Collapse
|