1
|
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
|
2
|
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
|
3
|
de Dios N, Riedel R, Schanton M, Balestrini P, Pérez L, Pérez-Pérez A, Etcheverry T, Casale R, Farina M, Sánchez-Margalet V, Maymó J, Varone C. Placental apoptosis increased by hypoxia inducible factor-1 stabilization is counteracted by leptin†. Biol Reprod 2024; 111:708-722. [PMID: 38924703 DOI: 10.1093/biolre/ioae095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/16/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
During pregnancy, apoptosis is a physiological event critical in the remodeling and aging of the placenta. Increasing evidence has pointed toward the relevance of hypoxia as modulator of trophoblast cell death. Previous reports have shown that leptin, a placental cytokine, promotes cell survival in both cell culture and placental explant models. The aim of this work is to establish the role of leptin in apoptosis under hypoxic condition in trophoblast cells. In this study, we evaluated the effect of cobalt chloride, a hypoxia mimicking agent that stabilizes the expression of hypoxia-inducible factor-1 alpha, on Swan-71 and human placental explants. Hypoxia chamber was also used to generate 2% oxygen. Apoptosis was determined by the presence of apoptotic nucleus, fragmentation of DNA and Caspase-3 and PARP-1 cleavage. The pro-apoptotic proteins BAX, BID, BAD, and BAK and the anti-apoptotic effectors BCL-2, B-cell lymphoma-extra-large, and myeloid cell leukemia-1 were also analyzed. We found that hypoxia-inducible factor-1 alpha stabilization increased the appearance of apoptotic nucleus, fragmentation of DNA, and Caspase-3 and PARP-1 cleavage. Hypoxia mimicking conditions enhanced the expression of pro-apoptotic effectors BAX, BID, BAD, and BAK. Hypoxia-inducible factor-1 alpha stabilization also downregulated the level of BCL-2, B-cell lymphoma-extra-large, and myeloid cell leukemia-1. All these apoptotic parameters changes were reversed with leptin treatment. Moreover, we showed that leptin action on apoptosis modulation involves PI3K and MAPK signaling pathways. Obtained data demonstrate that hypoxia-inducible factor-1 alpha stabilization induces apoptosis in human placenta and leptin counteracts this effect, reinforcing its role as a survival cytokine.
Collapse
Affiliation(s)
- Nataly de Dios
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Rodrigo Riedel
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Malena Schanton
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Paula Balestrini
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Luciano Pérez
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Antonio Pérez-Pérez
- Departamento de Bioquímica Médica y Biología Molecular. Hospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Sevilla, España
| | - Tomás Etcheverry
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO, CONICET), Universidad de Buenos Aires. Facultad de Medicina, Buenos Aires, Argentina
| | - Roberto Casale
- Departamento Materno-Infantil, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Mariana Farina
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO, CONICET), Universidad de Buenos Aires. Facultad de Medicina, Buenos Aires, Argentina
| | - Víctor Sánchez-Margalet
- Departamento de Bioquímica Médica y Biología Molecular. Hospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Sevilla, España
| | - Julieta Maymó
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Cecilia Varone
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| |
Collapse
|
4
|
Elgazzaz M, Woodham PC, Maher J, Faulkner JL. Implications of pregnancy on cardiometabolic disease risk: preeclampsia and gestational diabetes. Am J Physiol Cell Physiol 2024; 327:C646-C660. [PMID: 39010840 PMCID: PMC11427017 DOI: 10.1152/ajpcell.00293.2024] [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: 05/02/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
Cardiometabolic disorders, such as obesity, insulin resistance, and hypertension, prior to and within pregnancy are increasing in prevalence worldwide. Pregnancy-associated cardiometabolic disease poses a great risk to the short- and long-term well-being of the mother and offspring. Hypertensive pregnancy, notably preeclampsia, as well as gestational diabetes are the major diseases of pregnancy growing in prevalence as a result of growing cardiometabolic disease prevalence. The mechanisms whereby obesity, diabetes, and other comorbidities lead to preeclampsia and gestational diabetes are incompletely understood and continually evolving in the literature. In addition, novel therapeutic avenues are currently being explored in these patients to offset cardiometabolic-induced adverse pregnancy outcomes in preeclamptic and gestational diabetes pregnancies. In this review, we discuss the emerging pathophysiological mechanisms of preeclampsia and gestational diabetes in the context of cardiometabolic risk as well as the most recent preclinical and clinical updates in the pathogenesis and treatment of these conditions.
Collapse
Affiliation(s)
- Mona Elgazzaz
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Padmashree C Woodham
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - James Maher
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Jessica L Faulkner
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
5
|
Walewska E, Makowczenko KG, Witek K, Laniecka E, Molcan T, Alvarez-Sanchez A, Kelsey G, Perez-Garcia V, Galvão AM. Fetal growth restriction and placental defects in obese mice are associated with impaired decidualisation: the role of increased leptin signalling modulators SOCS3 and PTPN2. Cell Mol Life Sci 2024; 81:329. [PMID: 39090270 PMCID: PMC11335253 DOI: 10.1007/s00018-024-05336-7] [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: 09/20/2023] [Revised: 04/05/2024] [Accepted: 06/28/2024] [Indexed: 08/04/2024]
Abstract
Decidualisation of the endometrium is a key event in early pregnancy, which enables embryo implantation. Importantly, the molecular processes impairing decidualisation in obese mothers are yet to be characterised. We hypothesise that impaired decidualisation in obese mice is mediated by the upregulation of leptin modulators, the suppressor of cytokine signalling 3 (SOCS3) and the protein tyrosine phosphatase non-receptor type 2 (PTPN2), together with the disruption of progesterone (P4)-signal transducer and activator of transcription (STAT3) signalling. After feeding mice with chow diet (CD) or high-fat diet (HFD) for 16 weeks, we confirmed the downregulation of P4 and oestradiol (E2) steroid receptors in decidua from embryonic day (E) 6.5 and decreased proliferation of stromal cells from HFD. In vitro decidualised mouse endometrial stromal cells (MESCs) and E6.5 deciduas from the HFD showed decreased expression of decidualisation markers, followed by the upregulation of SOCS3 and PTPN2 and decreased phosphorylation of STAT3. In vivo and in vitro leptin treatment of mice and MESCs mimicked the results observed in the obese model. The downregulation of Socs3 and Ptpn2 after siRNA transfection of MESCs from HFD mice restored the expression level of decidualisation markers. Finally, DIO mice placentas from E18.5 showed decreased labyrinth development and vascularisation and fetal growth restricted embryos. The present study revealed major defects in decidualisation in obese mice, characterised by altered uterine response to E2 and P4 steroid signalling. Importantly, altered hormonal response was associated with increased expression of leptin signalling modulators SOCS3 and PTPN2. Elevated levels of SOCS3 and PTPN2 were shown to molecularly affect decidualisation in obese mice, potentially disrupting the STAT3-PR regulatory molecular hub.
Collapse
Affiliation(s)
- Edyta Walewska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Karol G Makowczenko
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Krzysztof Witek
- Laboratory of Cell and Tissue Analysis and Imaging, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Elżbieta Laniecka
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Tomasz Molcan
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Andrea Alvarez-Sanchez
- Molecular Mechanisms of Placental Invasion, Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK.
| | - Vicente Perez-Garcia
- Molecular Mechanisms of Placental Invasion, Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera 3, 46012, Valencia, Spain.
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28049, Madrid, Spain.
| | - António M Galvão
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland.
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK.
- Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London, NW1 0TU, UK.
| |
Collapse
|
6
|
Lançoni SS, Albuquerque JP, Nakato AM, Souza de Nieto GCE, Tkac CM, Sigwalt MF, Nohama P, Souto LRT, Nassif PAN. Placental and Neonatal Serum Leptin Levels in Premature Infants After Phototherapy: Are They Determining Factors for the Safe Indication of Passive Exercises? J Multidiscip Healthc 2024; 17:3091-3100. [PMID: 38974370 PMCID: PMC11227856 DOI: 10.2147/jmdh.s458528] [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: 01/08/2024] [Accepted: 06/04/2024] [Indexed: 07/09/2024] Open
Abstract
Purpose To analyze leptin levels in placental tissue and premature infants undergoing phototherapy and to evaluate the potential for prescribing passive exercise after phototherapy in this population. Patients and Methods This analytical, longitudinal, prospective cohort study included 108 parturients and their respective premature infants. Variables examined included weight, gestational age, body mass index, sex, serum leptin levels in placental tissue, serum bilirubin levels, and reticulocyte count. Results When comparing each group to a leptin threshold, statistically significant differences were observed at all evaluated time points for placental leptin levels (p < 0.001). Additionally, reticulocyte count decreased in relation to rebound time (p < 0.004). No correlations were found between leptin/bilirubin levels, leptin/reticulocytes, onset of nutrition, and BMI/leptin levels. Conclusion The findings regarding leptin levels suggest that prescribing passive exercises to premature infants undergoing phototherapy may be feasible because this intervention did not increase leptin levels.
Collapse
Affiliation(s)
- Samira Said Lançoni
- Graduate Program in Principles of Surgery, Faculdade Evangélica Mackenzie do Paraná, Curitiba, Paraná, Brasil
- Graduate Program on Health Technology, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil
| | - Jocilene Pedroso Albuquerque
- Graduate Program in Principles of Surgery, Faculdade Evangélica Mackenzie do Paraná, Curitiba, Paraná, Brasil
- Hospital Universitário Evangélico Mackenzie, Curitiba, Paraná, Brasil
| | - Adriane Muller Nakato
- Graduate Program on Health Technology, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil
| | | | - Cláudio Marcelo Tkac
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil
| | - Marcos Fabiano Sigwalt
- Graduate Program in Principles of Surgery, Faculdade Evangélica Mackenzie do Paraná, Curitiba, Paraná, Brasil
- Hospital Universitário Evangélico Mackenzie, Curitiba, Paraná, Brasil
| | - Percy Nohama
- Graduate Program on Health Technology, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil
| | | | - Paulo Afonso Nunes Nassif
- Graduate Program in Principles of Surgery, Faculdade Evangélica Mackenzie do Paraná, Curitiba, Paraná, Brasil
- Hospital Universitário Evangélico Mackenzie, Curitiba, Paraná, Brasil
| |
Collapse
|
7
|
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
|
8
|
Cànaves-Gómez L, Fleischer A, Muncunill-Farreny J, Gimenez MP, Álvarez Ruiz De Larrinaga A, Sánchez Baron A, Codina Marcet M, De-La-Peña M, Morell-Garcia D, Peña Zarza J, Piñas Zebrian C, García Fernández S, Alonso A. Effect of Obstructive Sleep Apnea during Pregnancy on Fetal Development: Gene Expression Profile of Cord Blood. Int J Mol Sci 2024; 25:5537. [PMID: 38791576 PMCID: PMC11121783 DOI: 10.3390/ijms25105537] [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: 04/29/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Obstructive sleep apnea (OSA) is quite prevalent during pregnancy and is associated with adverse perinatal outcomes, but its potential influence on fetal development remains unclear. This study investigated maternal OSA impact on the fetus by analyzing gene expression profiles in whole cord blood (WCB). Ten women in the third trimester of pregnancy were included, five OSA and five non-OSA cases. WCB RNA expression was analyzed by microarray technology to identify differentially expressed genes (DEGs) under OSA conditions. After data normalization, 3238 genes showed significant differential expression under OSA conditions, with 2690 upregulated genes and 548 downregulated genes. Functional enrichment was conducted using gene set enrichment analysis (GSEA) applied to Gene Ontology annotations. Key biological processes involved in OSA were identified, including response to oxidative stress and hypoxia, apoptosis, insulin response and secretion, and placental development. Moreover, DEGs were confirmed through qPCR analyses in additional WCB samples (7 with OSA and 13 without OSA). This highlighted differential expression of several genes in OSA (EGR1, PFN1 and PRKAR1A), with distinct gene expression profiles observed during rapid eye movement (REM)-OSA in pregnancy (PFN1, UBA52, EGR1, STX4, MYC, JUNB, and MAPKAP). These findings suggest that OSA, particularly during REM sleep, may negatively impact various biological processes during fetal development.
Collapse
Affiliation(s)
- Laura Cànaves-Gómez
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain; (L.C.-G.); (A.F.); (J.M.-F.); (M.P.G.); (M.D.-L.-P.); (D.M.-G.); (J.P.Z.); (C.P.Z.); (S.G.F.)
| | - Aarne Fleischer
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain; (L.C.-G.); (A.F.); (J.M.-F.); (M.P.G.); (M.D.-L.-P.); (D.M.-G.); (J.P.Z.); (C.P.Z.); (S.G.F.)
- Genomic & Bioinformatics Platform, IdISBa, 07120 Palma de Mallorca, Spain
| | - Josep Muncunill-Farreny
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain; (L.C.-G.); (A.F.); (J.M.-F.); (M.P.G.); (M.D.-L.-P.); (D.M.-G.); (J.P.Z.); (C.P.Z.); (S.G.F.)
- Genomic & Bioinformatics Platform, IdISBa, 07120 Palma de Mallorca, Spain
| | - María Paloma Gimenez
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain; (L.C.-G.); (A.F.); (J.M.-F.); (M.P.G.); (M.D.-L.-P.); (D.M.-G.); (J.P.Z.); (C.P.Z.); (S.G.F.)
| | - Ainhoa Álvarez Ruiz De Larrinaga
- Hospital Universitario de Araba, 01009 Vitoria-Gasteiz, Spain;
- Departamento de Neurociencias, Instituto de Investigación Sanitaria Bioaraba, 01009 Vitoria-Gasteiz, Spain
| | | | - Mercedes Codina Marcet
- Servicio de Endocrinología, Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain;
| | - Mónica De-La-Peña
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain; (L.C.-G.); (A.F.); (J.M.-F.); (M.P.G.); (M.D.-L.-P.); (D.M.-G.); (J.P.Z.); (C.P.Z.); (S.G.F.)
- Servicio de Neumología, Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III (CIBERES), 28029 Madrid, Spain
| | - Daniel Morell-Garcia
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain; (L.C.-G.); (A.F.); (J.M.-F.); (M.P.G.); (M.D.-L.-P.); (D.M.-G.); (J.P.Z.); (C.P.Z.); (S.G.F.)
- Servicio de Análisis Clínicos, Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
| | - José Peña Zarza
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain; (L.C.-G.); (A.F.); (J.M.-F.); (M.P.G.); (M.D.-L.-P.); (D.M.-G.); (J.P.Z.); (C.P.Z.); (S.G.F.)
- Servicio de Pediatría, Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
| | - Concepción Piñas Zebrian
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain; (L.C.-G.); (A.F.); (J.M.-F.); (M.P.G.); (M.D.-L.-P.); (D.M.-G.); (J.P.Z.); (C.P.Z.); (S.G.F.)
- Servicio de Neumología, Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
| | - Susana García Fernández
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain; (L.C.-G.); (A.F.); (J.M.-F.); (M.P.G.); (M.D.-L.-P.); (D.M.-G.); (J.P.Z.); (C.P.Z.); (S.G.F.)
- Servicio de Neumología, Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
| | - Alberto Alonso
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain; (L.C.-G.); (A.F.); (J.M.-F.); (M.P.G.); (M.D.-L.-P.); (D.M.-G.); (J.P.Z.); (C.P.Z.); (S.G.F.)
- Servicio de Neumología, Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
9
|
Yu Y, Ma Q, Groth SW. Prepregnancy dieting and obstetrical and neonatal outcomes: Findings from a national surveillance project in the United States. Midwifery 2024; 132:103972. [PMID: 38493519 DOI: 10.1016/j.midw.2024.103972] [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: 04/17/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
PROBLEM AND BACKGROUND Women with overweight or obesity are recommended to lose weight before pregnancy. Dieting is one of the most used weight control strategies. However, the health implications of dieting before pregnancy remain unclear. AIM To evaluate the associations of dieting during the year before pregnancy with obstetrical and neonatal outcomes, including gestational weight gain (GWG), gestational diabetes, low birthweight, macrosomia, small-for-gestational-age infants (SGA), large-for-gestational-age infants (LGA), and preterm birth. METHODS This study analyzed data from the Pregnancy Risk Assessment Monitoring System (PRAMS), which is a surveillance project in the United States that collects data on maternal health before, during, and after pregnancy. Women who participated in PRAMS phase 7 with a prepregnancy body mass index ≥25 kg/m2 and a singleton birth were eligible. Statistical analyses included logistic regressions and post-hoc mediation analysis (Sobel Test). FINDINGS A total number of 51,399 women were included in the analysis. Women who self-reported prepregnancy dieting (42.8 %) had lower odds of SGA (adjusted odds ratio [aOR]: 0.87; 95 % CI: 0.79-0.97), and higher odds of excessive GWG vs adequate GWG (aOR: 1.42; 95 % CI: 1.32-1.52), gestational diabetes (aOR: 1.12; 95 % CI: 1.02-1.22), and LGA (aOR: 1.18; 95 % CI: 1.08-1.28). Furthermore, the association between prepregnancy dieting and LGA was mediated by excessive GWG (Sobel Test z-value = 5.72, p < 0.01). DISCUSSION AND CONCLUSION This analysis revealed that prepregnancy dieting was associated with several adverse consequences, including excessive GWG, gestational diabetes, and LGA infants. Findings contribute to an improved understanding of the perinatal implications of prepregnancy dieting.
Collapse
Affiliation(s)
- Yang Yu
- School of Nursing, University of Rochester, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| | - Qianheng Ma
- Department of Psychiatry and Behavioral Sciences, Stanford University, 401 Quarry Rd, Stanford, CA, 94305, USA
| | - Susan W Groth
- School of Nursing, University of Rochester, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| |
Collapse
|
10
|
Gopal SH, Donepudi R, Pammi M. Leptin deficiency, a potential mechanism for impaired fetal lung development in uteroplacental insufficiency? Pediatr Res 2024; 95:1410-1411. [PMID: 38263448 DOI: 10.1038/s41390-024-03038-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 12/29/2023] [Indexed: 01/25/2024]
Abstract
Uteroplacental insufficiency (UPI) is a major cause of fetal growth restriction (FGR). Leptin, an adipokine, has been shown to play a vital role in fetal organogenesis. There is evidence reporting leptin deficiency in preterm and growth-restricted fetuses. In this issue of Pediatric Research, Yuliana et al. report leptin expression and lung development in UPI-induced FGR rats. UPI-induced FGR rats expressed decreased lung leptin and had impaired lung development, as shown by decreased surface area and lung volume. They also found a significant association between lung radial alveolar count, serum leptin, von Willebrand factor, and specific metabolites on metabolomic analyses. Previous studies on leptin supplementation in vivo have been associated with improvement in lung maturation; supporting the evidence, that leptin improves lung growth and development in FGR and may have future therapeutic potential in the improvement of respiratory outcomes in these infants. Future studies to support evidence of this association in humans are warranted.
Collapse
Affiliation(s)
- Srirupa Hari Gopal
- Dept. of Pediatrics, Division of Neonatology, Baylor College of Medicine & Texas Children's Hospital, Houston, TX, USA
| | - Roopali Donepudi
- Dept. of Obstetrics & Gynecology, Maternal-Fetal Medicine, Baylor College of Medicine & Texas Children's Hospital, Houston, TX, USA
| | - Mohan Pammi
- Dept. of Pediatrics, Division of Neonatology, Baylor College of Medicine & Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
11
|
Chen J, Yang L, Chen Y, Yuan W, Chen Y, Liang H, Miao M, He G, Wang Z. Associations between maternal urinary kisspeptin in late pregnancy and decreased fetal growth: a pregnancy-birth cohort study. Front Endocrinol (Lausanne) 2024; 15:1257248. [PMID: 38318290 PMCID: PMC10839029 DOI: 10.3389/fendo.2024.1257248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024] Open
Abstract
Background Kisspeptin has been indicated to be a biomarker of fetal growth. Although some evidence suggested that maternal kisspeptin concentrations in early pregnancy were associated with increased fetal growth, studies are still limited and the effect of kisspeptin in late pregnancy remains unknown. This study aimed to investigate the associations between maternal kisspeptin in late pregnancy and fetal growth. Methods Based on the Shanghai-Minhang Birth Cohort study, 724 mother-neonate pairs were included in this study. We measured maternal kisspeptin concentrations in the urine samples collected in late pregnancy and neonatal anthropometric indices at birth. The associations between maternal kisspeptin and neonatal anthropometry were investigated using multiple linear regression models. Results Higher maternal urinary kisspeptin concentrations were associated with lower neonatal birth weight, head circumference, upper arm circumference, abdominal skinfold thickness, triceps skinfold thickness, and back skinfold thickness. The inverse associations were more pronounced for the highest kisspeptin levels versus the lowest. These patterns were consistent in analyses stratified by neonatal sex, with notably stable associations between maternal kisspeptin concentrations and skinfold thickness. Conclusion The present study suggested that maternal kisspeptin concentrations in late pregnancy might be inversely associated with fetal growth. The physiological mechanisms of maternal kisspeptin might differ from those in early pregnancy. Further studies are required to assess associations between maternal kisspeptin and energy homeostasis and explore the physiological roles of kisspeptin in late pregnancy.
Collapse
Affiliation(s)
- Jiaxian Chen
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Public Health, Fudan University, Shanghai, China
| | - Lan Yang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Public Health, Fudan University, Shanghai, China
| | - Yafei Chen
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Public Health, Fudan University, Shanghai, China
| | - Wei Yuan
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Yao Chen
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Hong Liang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Maohua Miao
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Gengsheng He
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai, China
| | - Ziliang Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| |
Collapse
|
12
|
Ozmen A, Nwabuobi C, Tang Z, Guo X, Larsen K, Guller S, Blas J, Moore M, Kayisli UA, Lockwood CJ, Guzeloglu-Kayisli O. Leptin-Mediated Induction of IL-6 Expression in Hofbauer Cells Contributes to Preeclampsia Pathogenesis. Int J Mol Sci 2023; 25:135. [PMID: 38203306 PMCID: PMC10778808 DOI: 10.3390/ijms25010135] [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: 10/19/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Leptin plays a crucial role in regulating energy homoeostasis, neuroendocrine function, metabolism, and immune and inflammatory responses. The adipose tissue is a main source of leptin, but during pregnancy, leptin is also secreted primarily by the placenta. Circulating leptin levels peak during the second trimester of human pregnancy and fall after labor. Several studies indicated a strong association between elevated placental leptin levels and preeclampsia (PE) pathogenesis and elevated serum interleukin-6 (IL-6) levels in PE patients. Therefore, we hypothesized that a local increase in placental leptin production induces IL-6 production in Hofbauer cells (HBCs) to contribute to PE-associated inflammation. We first investigated HBCs-specific IL-6 and leptin receptor (LEPR) expression and compared their immunoreactivity in PE vs. gestational age-matched control placentas. Subsequently, we examined the in vitro regulation of IL-6 as well as the phosphorylation levels of intracellular signaling proteins STAT3, STAT5, NF-κB, and ERK1/2 by increasing recombinant human leptin concentrations (10 to 1000 ng/mL) in primary cultured HBCs. Lastly, HBC cultures were incubated with leptin ± specific inhibitors of STAT3 or STAT5, or p65 NF-κB or ERK1/2 MAPK signaling cascades to determine relevant cascade(s) involved in leptin-mediated IL-6 regulation. Immunohistochemistry revealed ~three- and ~five-fold increases in IL-6 and LEPR expression, respectively, in HBCs from PE placentas. In vitro analysis indicated that leptin treatment in HBCs stimulate IL-6 in a concentration-dependent manner both at the transcriptional and secretory levels (p < 0.05). Moreover, leptin-treated HBC cultures displayed significantly increased phosphorylation levels of STAT5, p65 NF-κB, and ERK1/2 MAPK and pre-incubation of HBCs with a specific ERK1/2 MAPK inhibitor blocked leptin-induced IL-6 expression. Our in situ results show that HBCs contribute to the pathogenesis of PE by elevating IL-6 expression, and in vitro results indicate that induction of IL-6 expression in HBCs is primarily leptin-mediated. While HBCs display an anti-inflammatory phenotype in normal placentas, elevated levels of leptin may transform HBCs into a pro-inflammatory phenotype by activating ERK1/2 MAPK to augment IL-6 expression.
Collapse
Affiliation(s)
- Asli Ozmen
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Chinedu Nwabuobi
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Zhonghua Tang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA; (Z.T.); (S.G.)
| | - Xiaofang Guo
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Kellie Larsen
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Seth Guller
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA; (Z.T.); (S.G.)
| | - Jacqueline Blas
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Monica Moore
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Umit A. Kayisli
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Charles J. Lockwood
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| | - Ozlem Guzeloglu-Kayisli
- Department of Obstetrics & Gynecology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (A.O.); (C.N.); (X.G.); (K.L.); (J.B.); (M.M.); (U.A.K.); (C.J.L.)
| |
Collapse
|
13
|
Zeng S, Wu Y, Zhou M, Bai H, Liu X, Fan P. Association between genetic polymorphisms of leptin receptor and preeclampsia in Chinese women. J Matern Fetal Neonatal Med 2023; 36:2207708. [PMID: 37150847 DOI: 10.1080/14767058.2023.2207708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
OBJECTIVE Leptin signaling plays an important role in regulating metabolism and reproduction. In the present study, we investigated the relationship between polymorphisms of leptin receptor (LEPR) gene A223G and A668G and preeclampsia (PE) and evaluated influences of genotypes on clinical, metabolic, and oxidative stress indices in Chinese women. METHODS This is a case-control study including 322 patients with PE and 1295 healthy pregnant women. The two polymorphisms were genotyped by polymerase chain reaction-restriction fragments length polymorphism method. Clinical and biochemical parameters were analyzed. RESULTS The frequencies of the AA + AG genotypes (28.6% vs. 36.1%) and A allele (14.9% vs. 19.8%) of LEPR A223G polymorphism, and those of the AA + AG genotypes (17.7% vs. 24.6%) and A allele (9.0% vs. 12.9%) of LEPR A668G polymorphism were significantly lower in the PE group than those in the control group. The 223A and 668A alleles were protective factors against PE in the regression model, which included age and delivery body mass index as covariates (OR = 0.684, 95% CI: 0.506-0.926, p = .014; OR = 0.650, 95% CI: 0.456-0.927, p = .017, respectively). When the 668GG/223GG combined genotype served as the reference category, the 668A/223A combined allele had further enhanced the protective effect on PE (OR = 0.558, 95% CI: 0.374-0.833, p = .004). Patients possessing the LEPR 223A allele had higher total antioxidant capacity and lower oxidative stress index (p < .05), while those with the LEPR 668A allele had higher high-density lipoprotein cholesterol levels (p = .045) compared with those carrying the corresponding GG genotype. CONCLUSIONS The 223A and 668A alleles of LEPR polymorphisms are genetic protective factors for PE in Chinese women. The two alleles may exert a beneficial effect on oxidative stress and lipid metabolism in patients.
Collapse
Affiliation(s)
- Shuai Zeng
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yujie Wu
- Laboratory of Genetic Disease and Perinatal Medicine, Laboratory of the Key Perinatal Disease, Key Laboratory of Birth Defects and Related Disease of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Mi Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Huai Bai
- Laboratory of Genetic Disease and Perinatal Medicine, Laboratory of the Key Perinatal Disease, Key Laboratory of Birth Defects and Related Disease of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xinghui Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ping Fan
- Laboratory of Genetic Disease and Perinatal Medicine, Laboratory of the Key Perinatal Disease, Key Laboratory of Birth Defects and Related Disease of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Aisagbonhi O, Bui T, Nasamran CA, St Louis H, Pizzo D, Meads M, Mulholland M, Magallanes C, Lamale-Smith L, Laurent LC, Morey R, Jacobs MB, Fisch KM, Horii M. High placental expression of FLT1, LEP, PHYHIP and IL3RA - In persons of African ancestry with severe preeclampsia. Placenta 2023; 144:13-22. [PMID: 37949031 PMCID: PMC10843761 DOI: 10.1016/j.placenta.2023.10.008] [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: 07/05/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
INTRODUCTION Mortality from preeclampsia (PE) and PE-associated morbidities are 3-to 5-fold higher in persons of African ancestry than in those of Asian and European ancestries. METHODS To elucidate placental contribution to worse PE outcomes in African ancestry pregnancies, we performed bulk RNA sequencing on 50 placentas from persons with severe PE (sPE) of African (n = 9), Asian (n = 18) and European (n = 23) ancestries and 73 normotensive controls of African (n = 10), Asian (n = 15) and European (n = 48) ancestries. RESULTS Previously described canonical preeclampsia genes, involved in metabolism and hypoxia/angiogenesis including: LEP, HK2, FSTL3, FLT1, ENG, TMEM45A, ARHGEF4 and HTRA1 were upregulated sPE versus normotensive placentas across ancestries. LTF, NPR3 and PHYHIP were higher in African vs. Asian ancestry sPE placentas. Allograft rejection/adaptive immune response genes were upregulated in placentas from African but not in Asian or European ancestry sPE patients; IL3RA was of particular interest because the patient with the highest placental IL3RA expression, a person of African ancestry with sPE, developed postpartum cardiomyopathy, and was the only patient out of 123, that developed this condition. Interestingly, the sPE patients with the highest IL3RA expression among persons of Asian and European ancestries developed unexplained tachycardia peripartum, necessitating echocardiography in the European ancestry patient. The association between elevated placental IL3RA levels and unexplained tachycardia or peripartum cardiomyopathy was found to be significant in the 50 sPE patients (p = .0005). DISCUSSION High placental upregulation of both canonical preeclampsia and allograft rejection/adaptive immune response genes may contribute to worse PE outcomes in African ancestry sPE patients.
Collapse
Affiliation(s)
- Omonigho Aisagbonhi
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA.
| | - Tony Bui
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Chanond A Nasamran
- Center for Computational Biology and Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Hailee St Louis
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Donald Pizzo
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Morgan Meads
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Megan Mulholland
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Celestine Magallanes
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Leah Lamale-Smith
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Louise C Laurent
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Robert Morey
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Marni B Jacobs
- Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Kathleen M Fisch
- Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA; Center for Computational Biology and Bioinformatics, University of California, San Diego, La Jolla, CA, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Mariko Horii
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Center for Perinatal Discovery, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
15
|
Wang H, Liang W, Su W, Cui H, Wang H. Expression and significance of silent information regulator two homolog 1 in the placenta and plasma of patients with pre-eclampsia-a meta-analysis. Gynecol Endocrinol 2023; 39:2264983. [PMID: 37857342 DOI: 10.1080/09513590.2023.2264983] [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: 07/05/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023] Open
Abstract
OBJECTIVE This study aimed to collect, organize, and conduct a meta-analysis of the literature on the expression of silent information regulator two homolog 1 (SIRT1) in the placental tissue and plasma of patients with pre-eclampsia. METHODS The enrolled patients were divided into two groups: the pre-eclampsia group and the healthy group. This study summarized and analyzed the demographic characteristics of the two groups, including pregnancy age, gestational weeks, parity, gravidity, blood pressure, Body Mass Index, newborn weight, placental weight, and SIRT1 expression in placental tissue and maternal plasma. RESULTS Eleven studies were included in this research, with 586 cases in the pre-eclampsia group and 479 cases in the control group. Three research studies are reporting immunohistochemistry tests, among which the pre-eclampsia group had a positivity rate of 30.24% (62/205), while the control group had 58.02% (76/131); the two groups have a significant difference (p < 0.05). Two research studies reported the results of ELISA tests, with 107 cases in the pre-eclampsia group and 125 cases in the control group. A comparison of the SIRT1 test results showed a statistically significant difference between the two groups (p < 0.05). Pre-eclampsia group patients had lower gestational weeks, newborn birth weight, and placental weight compared to the healthy control group (all p < 0.05). However, systolic and diastolic blood pressures were higher in the pre-eclampsia group than in the control group (p < 0.05). CONCLUSION SIRT1 expression is downregulated in pre-eclampsia patients' plasma and placental tissue. Further research is needed to validate this conclusion.
Collapse
Affiliation(s)
- Hongling Wang
- Department of Ultrasound, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Wenwen Liang
- Department of Ultrasound, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Weihua Su
- Department of Obstetrics, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Hong Cui
- Department of Ultrasound, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Huifeng Wang
- Department of Ultrasound, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| |
Collapse
|
16
|
Halkjær SI, de Knegt VE, Kallemose T, Jensen JEB, Cortes D, Gluud LL, Wewer Albrechtsen NJ, Petersen AM. No effect of multi-strain probiotic supplementation on metabolic and inflammatory markers and newborn body composition in pregnant women with obesity: Results from a randomized, double-blind placebo-controlled study. Nutr Metab Cardiovasc Dis 2023; 33:2444-2454. [PMID: 37580231 DOI: 10.1016/j.numecd.2023.07.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/06/2023] [Accepted: 07/20/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND AND AIMS Modulation of the gut microbiome composition with probiotics may have beneficial metabolic effects in pregnant women with obesity. The aim was to investigate the effect of probiotic supplementation during pregnancy on metabolic and inflammatory markers and the body composition of the offspring. METHODS AND RESULTS A randomized double-blind trial in 50 pregnant women (pre-pregnancy BMI ≥30 and < 35 kg/m2) comparing multi-strain probiotics (Vivomixx®; 450 billion CFU/d) versus placebo from 14 to 20 weeks of gestation until delivery was carried out. Participants were followed with two predelivery visits at gestational week 27-30 and 36-37 and with one postdelivery visit. All visits included fasting blood samples (C-reactive protein (CRP), alanine aminotransferase (ALT), aspartate aminotransferase (AST), insulin, C-peptide, glucose, glucagon, and glucagon-like peptide-1 (GLP-1)). At delivery, umbilical cord blood samples were collected (GLP-1 and glucagon). At the postdelivery visit, a dual-energy X-ray absorptiometry (DXA) scan of the newborn was performed. Forty-nine of 50 participants completed the study until delivery, and 36 mother-offspring dyads underwent postdelivery examinations including a DXA scan. There were no significant differences in changes in measured biomarkers between the probiotic versus the placebo group. No differences were found in newborn body composition or GLP-1 and glucagon. GLP-1 measured in umbilical blood samples was positively correlated to fat percent in offspring from the probiotic group. CONCLUSION In this study of pregnant women with obesity and their newborns, there was no effect of probiotic supplementation in mothers or babies on metabolic or inflammatory biomarkers or on body composition of offspring. This study was registered at clinicaltrials.gov as NCT02508844.
Collapse
Affiliation(s)
- Sofie I Halkjær
- Gastrounit, Medical Section, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Victoria E de Knegt
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Thomas Kallemose
- Clinical Research Center, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Jens-Erik B Jensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Dina Cortes
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise L Gluud
- Gastrounit, Medical Section, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Andreas Munk Petersen
- Gastrounit, Medical Section, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Microbiology, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark.
| |
Collapse
|
17
|
de Jesus Nascimento AE, Santos LC, Santos BR, Santos EO, Cunha MCDSG, Snoeck PPDN, de Lavor MSL, Silva JF. Spatial and temporal expression profile of sex steroid receptors and antioxidant enzymes in the maternal-fetal interface of domestic cats. Theriogenology 2023; 210:234-243. [PMID: 37542738 DOI: 10.1016/j.theriogenology.2023.08.001] [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: 06/08/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
Sex steroids and antioxidant enzymes modulate uterine and placental physiology. Failures in the expression, signaling, and/or secretion of these mediators are associated with female infertility and gestational problems. However, there is no data on the expression profile of receptors for sex steroids and antioxidant enzymes in the maternal-fetal interface of domestic cats. Uterus and placenta samples from non-pregnant diestrus cats and cats in mid- and late pregnancy were used to analyze the protein and gene expression of the receptors for estrogen alpha (ERα), progesterone (PR), and androgen (AR) and the antioxidant enzymes superoxide dismutase 1 (SOD1), catalase, and glutathione peroxidase 1 (GPX1) by immunohistochemistry and qPCR. Higher uterine expression of ERα, Pr, and Sod1 was observed in the pregnant cats, especially in mid-pregnancy, compared to non-pregnant diestrus cats, as well as reduced endometrial catalase immunostaining. In the placenta, the mRNA expression of Erα, Pr, Ar, and Gpx1 was higher in late pregnancy in relation to mid-pregnancy. Moreover, weak or no placental expression was observed for catalase in mid- and late pregnancy, while strong immunostaining was observed for AR in trophoblasts and decidual cells in mid-pregnancy. The findings of this study demonstrated that pregnancy in female cats increases the uterine expression of sex steroid receptors and antioxidant enzymes, and that the placental expression of these mediators varies according to gestational age.
Collapse
Affiliation(s)
- Acácia Eduarda de Jesus Nascimento
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Luciano Cardoso Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Bianca Reis Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Emilly Oliveira Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Maria Clara da Silva Galrão Cunha
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Paola Pereira das Neves Snoeck
- Hospital Veterinário, Departamento de Ciências Agrarias e Ambientais, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Mário Sérgio Lima de Lavor
- Hospital Veterinário, Departamento de Ciências Agrarias e Ambientais, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Juneo Freitas Silva
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil.
| |
Collapse
|
18
|
ADAMS DM, BECKERS KF, FLANAGAN JP, GOMES VCL, LIU CC, SONES JL. Reversal of maternal obesity attenuates hypoxia and improves placental development in the preeclamptic-like BPH/5 mouse model. BIOCELL 2023; 47:2051-2058. [PMID: 37829603 PMCID: PMC10569287 DOI: 10.32604/biocell.2023.029644] [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: 02/28/2023] [Accepted: 07/06/2023] [Indexed: 10/14/2023]
Abstract
Background Women with obesity have higher risk of adverse pregnancy outcomes, including preeclampsia (PE). Late-gestational hypertension, aberrant fetoplacental development, and fetal growth restriction (FGR), hallmarks of PE, are observed spontaneously in BPH/5 mice. Similar to obese preeclamptic women, BPH/5 mice have higher visceral white adipose tissue (WAT) and circulating leptin. We hypothesized that attenuation of maternal obesity and serum leptin in pregnant BPH/5 mice will improve fetoplacental development by decreasing hypoxia markers and leptin expression at the maternal-fetal interface. Methods To test this hypothesis, BPH/5 mice were fed ad libitum (lib) and pair-fed (PF) to C57 ad lib controls beginning at embryonic day (e) 0.5. Hypoxia-related genes, hypoxia inducible factor (Hif) 1α, stem cell factor (Scf), heme oxygenase-1 (Ho-1), leptin (Lep), and leptin receptor (LepR) were assessed in e7.5 implantation sites. Results BPH/5 ad lib had 1.5 to 2-fold increase in Hif1α, Scf, and Ho-1 mRNA and a greater than 3-fold increase in leptin mRNA vs. C57 that was attenuated with PF. Exogenous leptin promoted Hif1α and Ho-1 mRNA expression in e7.5 decidua in vitro. While hypoxic conditions in vitro did not change decidual leptin mRNA. Furthermore, BPH/5 PF mice demonstrated improved fetal and placental outcomes later in gestation, with greater placental vascular area by e18.5 and attenuation of FGR. Conclusion In conclusion, pair-feeding BPH/5 mice beginning at conception may improve placental vasculature formation via decreased leptin and hypoxia-associated markers in this model. Future investigations are needed to better determine the effect of hypoxia and leptin on pregnancy outcomes in obese pregnant women.
Collapse
Affiliation(s)
- Daniella M. ADAMS
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Kalie F. BECKERS
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Juliet P. FLANAGAN
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Viviane C. L. GOMES
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Chin-Chi LIU
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Jenny L. SONES
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
19
|
Lis-Kuberka J, Pupek M, Orczyk-Pawiłowicz M. The Mother-Child Dyad Adipokine Pattern: A Review of Current Knowledge. Nutrients 2023; 15:4059. [PMID: 37764842 PMCID: PMC10535905 DOI: 10.3390/nu15184059] [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: 08/21/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
An important role in the network of interconnections between the mother and child is played by adipokines, which are adipose tissue hormones engaged in the regulation of metabolism. Alternations of maternal adipokines translate to the worsening of maternal insulin resistance as well as metabolic stress, altered placenta functions, and fetal development, which finally contribute to long-term metabolic unfavorable conditions. This paper is the first to summarize the current state of knowledge concerning the concentrations of individual adipokines in different biological fluids of maternal and cord plasma, newborn/infant plasma, milk, and the placenta, where it highlights the impact of adverse perinatal risk factors, including gestational diabetes mellitus, preeclampsia, intrauterine growth restriction, preterm delivery, and maternal obesity on the adipokine patterns in maternal-infant dyads. The importance of adipokine measurement and relationships in biological fluids during pregnancy and lactation is crucial for public health in the area of prevention of most diet-related metabolic diseases. The review highlights the huge knowledge gap in the field of hormones participating in the energy homeostasis and metabolic pathways during perinatal and postnatal periods in the mother-child dyad. An in-depth characterization is needed to confirm if the adverse outcomes of early developmental programming might be modulated via maternal lifestyle intervention.
Collapse
Affiliation(s)
- Jolanta Lis-Kuberka
- Department of Biochemistry and Immunochemistry, Division of Chemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| | | | - Magdalena Orczyk-Pawiłowicz
- Department of Biochemistry and Immunochemistry, Division of Chemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| |
Collapse
|
20
|
S C, G G, LA S, W N, P M, L A, A W, V F, P W, D G, T BT. Transcriptomic profiling reveals differential cellular response to copper oxide nanoparticles and polystyrene nanoplastics in perfused human placenta. ENVIRONMENT INTERNATIONAL 2023; 177:108015. [PMID: 37315489 DOI: 10.1016/j.envint.2023.108015] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/16/2023]
Abstract
The growing nanoparticulate pollution (e.g. engineered nanoparticles (NPs) or nanoplastics) has been shown to pose potential threats to human health. In particular, sensitive populations such as pregnant women and their unborn children need to be protected from harmful environmental exposures. However, developmental toxicity from prenatal exposure to pollution particles is not yet well studied despite evidence of particle accumulation in human placenta. Our study aimed to investigate how copper oxide NPs (CuO NPs; 10-20 nm) and polystyrene nanoplastics (PS NPs; 70 nm) impact on gene expression in ex vivo perfused human placental tissue. Whole genome microarray analysis revealed changes in global gene expression profile after 6 h of perfusion with sub-cytotoxic concentrations of CuO (10 µg/mL) and PS NPs (25 µg/mL). Pathway and gene ontology enrichment analysis of the differentially expressed genes suggested that CuO and PS NPs trigger distinct cellular response in placental tissue. While CuO NPs induced pathways related to angiogenesis, protein misfolding and heat shock responses, PS NPs affected the expression of genes related to inflammation and iron homeostasis. The observed effects on protein misfolding, cytokine signaling, and hormones were corroborated by western blot (accumulation of polyubiquitinated proteins) or qPCR analysis. Overall, the results of the present study revealed extensive and material-specific interference of CuO and PS NPs with placental gene expression from a single short-term exposure which deserves increasing attention. In addition, the placenta, which is often neglected in developmental toxicity studies, should be a key focus in the future safety assessment of NPs in pregnancy.
Collapse
Affiliation(s)
- Chortarea S
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland
| | - Gupta G
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland
| | - Saarimäki LA
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Netkueakul W
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland
| | - Manser P
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland
| | - Aengenheister L
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland; Human Biomonitoring Research Unit, Department of Precision Health, Luxembourg Institute of Health (LIH), 1 A-B, Rue Thomas Edison, L-1445 Strassen, Luxembourg
| | - Wichser A
- Laboratory for Advanced Analytical Technologies, Empa, Swiss Federal Laboratories for Materials, Science and Technology, Dübendorf, Switzerland
| | - Fortino V
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Wick P
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland
| | - Greco D
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Buerki-Thurnherr T
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland.
| |
Collapse
|
21
|
Wu Y, Kang F, Yang Y, Tao L, Chen Y, Li X. The protective effect of magnesium sulfate on placental inflammation via suppressing the NF-κB pathway in a preeclampsia-like rat model. Pregnancy Hypertens 2023; 31:4-13. [PMID: 36435037 DOI: 10.1016/j.preghy.2022.11.004] [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: 05/09/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
Abnormal placental inflammation has a role in the pathophysiology of preeclampsia. Magnesium sulfate (MgSO4) has anti-inflammatory properties and is a fetal neuroprotective agent. MgSO4 is often used to treat severe preeclampsia; however, the specificmechanisms of action underlyingthistherapeutic effect remain unclear. The objective of this study was to investigate the effects of MgSO4 (270 mg/kg) on placental inflammation in a rat model of lipopolysaccharide (LPS; 1.0 µg/kg)-induced preeclampsia. Compared to normal pregnant rats, LPS-treated pregnant rats had higher blood pressure, proteinuria, and expression of the anti-angiogenic factor sFlt-1 and the pro-inflammatory factors interleukin-1β (IL-1β) and IL-12 in placental tissue. LPS-treated pregnant rats had placental insufficiency, poor fetal outcomes, and significantly decreased expression of the anti-inflammatory factors apolipoprotein E (APOE) and IL-10 in placental tissue. MgSO4 treatment had favorable effects on maternal and fetal outcomes. MgSO4 treatment improved placental function by repressing an exaggerated inflammatory response in the placenta and promoting angiogenesis via the NF-κB pathway. These findings suggest MgSO4 has a potential role in the prevention of preeclampsia and in the treatment of mild and moderate preeclampsia.
Collapse
Affiliation(s)
- Yongyuan Wu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei 230032, Anhui, China
| | - Fen Kang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei 230032, Anhui, China
| | - Yuanyuan Yang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, No. 81 Meishan Road, Hefei 230032, Anhui, China
| | - Li Tao
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, No. 81 Meishan Road, Hefei 230032, Anhui, China
| | - Yueran Chen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, No. 81 Meishan Road, Hefei 230032, Anhui, China
| | - Xiaolan Li
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
22
|
Lodefalk M, Chelslín F, Patriksson Karlsson J, Hansson SR. Placental Changes and Neuropsychological Development in Children-A Systematic Review. Cells 2023; 12:cells12030435. [PMID: 36766778 PMCID: PMC9913696 DOI: 10.3390/cells12030435] [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: 12/19/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
Placental dysfunction may increase the offspring's later-life disease risk. The objective of this systematic review was to describe associations between pathological placental changes and neuropsychological outcomes in children after the neonatal period. The inclusion criteria were human studies; original research; direct placental variables; neuropsychological outcomes; and analysis between their associations. The exclusion criterion was the offspring's age-0-28 days or >19 years. The MEDLINE and EMBASE databases were last searched in May 2022. We utilized the ROBINS-I for the risk of bias assessment and performed a narrative synthesis. In total, 3252 studies were identified, out of which 16 were included (i.e., a total of 15,862 participants). Half of the studies were performed on children with neonatal complications, and 75% of the studies reported an association between a placental change and an outcome; however, following the completion of the funnel plots, a risk of publication bias was indicated. The largest study described a small association between placental size and a risk of psychiatric symptoms in boys only. Inconsistency between the studies limited the evidence in this review. In general, no strong evidence was found for an association between pathological placental changes and childhood neuropsychological outcomes after the neonatal period. However, the association between placental size and mental health in boys indicates a placental sexual dimorphism, thereby suggesting an increased vulnerability for male fetuses.
Collapse
Affiliation(s)
- Maria Lodefalk
- Department of Pediatrics, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
- University Health Care Research Centre, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
- Correspondence:
| | - Felix Chelslín
- Department of Pediatrics, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
| | - Johanna Patriksson Karlsson
- University Health Care Research Centre, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
| | - Stefan R. Hansson
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences Lund, Lund University, 221 00 Lund, Sweden
- Department of Obstetrics and Gynecology, Skåne University Hospital, 214 28 Malmö, Sweden
| |
Collapse
|
23
|
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
|
24
|
de Knegt VE, Hedley PL, Eltvedt AK, Placing S, Wøjdemann K, Shalmi AC, Rode L, Kanters JK, Sundberg K, Tabor A, Lausten-Thomsen U, Christiansen M. First-Trimester Maternal Serum Adiponectin/Leptin Ratio in Pre-Eclampsia and Fetal Growth. Life (Basel) 2023; 13:life13010130. [PMID: 36676079 PMCID: PMC9864486 DOI: 10.3390/life13010130] [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: 10/14/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
The serum adiponectin/leptin ratio (A/L ratio) is a surrogate marker of insulin sensitivity. Pre-eclampsia (PE) is associated with maternal metabolic syndrome and occasionally impaired fetal growth. We assessed whether the A/L ratio in first-trimester maternal serum was associated with PE and/or birth weight. Adiponectin and leptin were quantitated in first-trimester blood samples (gestational week 10+3−13+6) from 126 women who later developed PE with proteinuria (98 mild PE; 21 severe PE; 7 HELLP syndrome), and 297 controls, recruited from the Copenhagen First-Trimester Screening Study. The A/L ratio was reduced in PE pregnancies, median 0.17 (IQR: 0.12−0.27) compared with controls, median 0.32 (IQR: 0.19−0.62) (p < 0.001). A multiple logistic regression showed that PE was negatively associated with log A/L ratio independent of maternal BMI (odds ratio = 0.315, 95% CI = 0.191 to 0.519). Adiponectin (AUC = 0.632) and PAPP-A (AUC = 0.605) were negatively associated with PE, and leptin (AUC = 0.712) was positively associated with PE. However, the A/L ratio was a better predictor of PE (AUC = 0.737), albeit not clinically relevant as a single marker. No significant association was found between A/L ratio and clinical severity of pre-eclampsia or preterm birth. PE was associated with a significantly lower relative birth weight (p < 0.001). A significant negative correlation was found between relative birth weight and A/L ratio in controls (β = −0.165, p < 0.05) but not in PE pregnancies), independent of maternal BMI. After correction for maternal BMI, leptin was significantly associated with relative birth weight (β = 2.98, p < 0.05), while adiponectin was not significantly associated. Our findings suggest that an impairment of the A/L ratio (as seen in metabolic syndrome) in the first trimester is characteristic of PE, while aberrant fetal growth in PE is not dependent on insulin sensitivity, but rather on leptin-associated pathways.
Collapse
Affiliation(s)
- Victoria E. de Knegt
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark
- Department of Paediatrics, University Hospital Slagelse, 4200 Slagelse, Denmark
| | - Paula L. Hedley
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark
- Brazen Bio, Los Angeles, CA 90014, USA
| | - Anna K. Eltvedt
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark
| | - Sophie Placing
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark
| | - Karen Wøjdemann
- Department of Gynecology and Obstetrics, Bornholm Hospital, 3700 Rønne, Denmark
| | | | - Line Rode
- Department of Clinical Biochemistry, Copenhagen University Hospital Rigshospitalet, 2600 Glostrup, Denmark
| | - Jørgen K. Kanters
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Karin Sundberg
- Center of Fetal Medicine, Department of Obstetrics, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| | - Ann Tabor
- Center of Fetal Medicine, Department of Obstetrics, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ulrik Lausten-Thomsen
- Department of Neonatology, University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| | - Michael Christiansen
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Correspondence: ; Tel.: +45-326-836-57; Fax: +45-326-838-78
| |
Collapse
|
25
|
Cissé AH, Taine M, Tafflet M, de Lauzon‐Guillain B, Clément K, Khalfallah O, Davidovic L, Lioret S, Charles MA, Heude B. Cord blood leptin level and a common variant of its receptor as determinants of the BMI trajectory: The EDEN mother-child cohort. Pediatr Obes 2022; 17:e12955. [PMID: 35747935 PMCID: PMC9787343 DOI: 10.1111/ijpo.12955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/24/2022] [Accepted: 06/01/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cord blood leptin is an indicator of neonatal fat mass and could shape postnatal adiposity trajectories. Investigating genetic polymorphisms of the leptin receptor gene (LEPR) could help understand the mechanisms involved. OBJECTIVES We aimed to investigate the association of cord blood leptin level and the LEPR rs9436303 polymorphism, with body mass index (BMI) at adiposity peak (AP) and age at adiposity rebound (AR). METHODS In the EDEN cohort, BMI at AP and age at AR were estimated with polynomial mixed models, for 1713 and 1415 children, respectively. Multivariable linear regression models allowed for examining the associations of cord blood leptin level and LEPR rs9436303 genotype with BMI at AP and age at AR adjusted for potential confounders including birth size groups. We also tested interactions between cord blood leptin level and rs9436303 genotype. RESULTS Increased leptin level was associated with reduced BMI at AP and early age at AR (comparing the highest quintile of leptin level to the others). Rs9436303 G-allele carriage was associated with increased BMI at AP and later age at AR but did not modulate the association with leptin level. CONCLUSION These results illustrate the role of early life body composition and the intrauterine environment in the programming of adiposity in childhood.
Collapse
Affiliation(s)
- Aminata H. Cissé
- Centre for Research in Epidemiology and StatisticSUniversité de Paris‐cité, INSERM, INRAEParisFrance
| | - Marion Taine
- Centre for Research in Epidemiology and StatisticSUniversité de Paris‐cité, INSERM, INRAEParisFrance
| | - Muriel Tafflet
- Centre for Research in Epidemiology and StatisticSUniversité de Paris‐cité, INSERM, INRAEParisFrance
| | | | - Karine Clément
- NutriOmics Research Unit, Assistance Publique‐Hôpitaux de Paris, Pitié‐Salpêtrière Hopital, Nutrition Department ParisSorbonne Université, INSERMParisFrance
| | - Olfa Khalfallah
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, INSERM, Université Nice Côte d'Azur, UMR7275, UMR_SValbonneFrance
| | - Laetitia Davidovic
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, INSERM, Université Nice Côte d'Azur, UMR7275, UMR_SValbonneFrance
| | - Sandrine Lioret
- Centre for Research in Epidemiology and StatisticSUniversité de Paris‐cité, INSERM, INRAEParisFrance
| | - Marie A. Charles
- Centre for Research in Epidemiology and StatisticSUniversité de Paris‐cité, INSERM, INRAEParisFrance
| | - Barbara Heude
- Centre for Research in Epidemiology and StatisticSUniversité de Paris‐cité, INSERM, INRAEParisFrance
| |
Collapse
|
26
|
Danielli M, Thomas RC, Gillies CL, Hu J, Khunti K, Tan BK. Blood biomarkers to predict the onset of pre-eclampsia: A systematic review and meta-analysis. Heliyon 2022; 8:e11226. [PMID: 36387521 PMCID: PMC9649987 DOI: 10.1016/j.heliyon.2022.e11226] [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: 06/01/2022] [Revised: 09/29/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Pre-eclampsia is one of the most common pregnancy complications, and a major cause of fetal and maternal morbidity and mortality globally. Diagnosis currently takes place in the third trimester based on clinical symptoms. This systematic review and meta-analysis sought to determine the blood biomarkers that are associated with pre-eclampsia, and in particular, the biomarkers that could predict pre-eclampsia in early pregnancy. We searched the electronic databases (Medline, Embase, Cochrane Library) from inception up to March 2022. Prospective studies with 1000 or more participants that measured blood biomarkers to predict or diagnose pre-eclampsia have been included in this systematic review. Biomarkers' measurements were considered from the first up to the third trimester, but not during labor. Data concerning pre-eclampsia, biomarker measurements and study characteristics were extracted. Meta-analysis was performed when possible. We found a total of 43 studies (assessing 62 different biomarkers in 18,170 pregnancies, have been included in this systematic review, and a total of 6 studies (assessing 2 biomarkers have been included in the meta-analysis). Statistical analysis was performed for PlGF and sFlt-1. Mean difference in PlGF levels between pre-eclampsia and healthy pregnancies, appear to increase as the pregnancy progresses. Results of sFlt-1 meta-analysis were inconclusive. No significant publication bias was identified. This is the most comprehensive and up to date systematic review and meta-analysis on this important topic on blood biomarkers for the early prediction of pre-eclampsia. Further This research highlights the urgent needed for further discovery research to identify blood biomarkers that could predict the development of pre-eclampsia.
Collapse
Affiliation(s)
- Marianna Danielli
- Cardiovascular Sciences, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Roisin C. Thomas
- Cardiovascular Sciences, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Clare L. Gillies
- Diabetes Research Centre, Leicester General Hospital, Leicester, LE5 4PW, United Kingdom
| | - Jiamiao Hu
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, Fujian, China
| | - Kamlesh Khunti
- Diabetes Research Centre, Leicester General Hospital, Leicester, LE5 4PW, United Kingdom
- NIHR Applied Research Collaboration – East Midlands (ARC-EM), Leicester General Hospital, Leicester, LE5 4PW, United Kingdom
| | - Bee Kang Tan
- Cardiovascular Sciences, University of Leicester, Leicester, LE1 7RH, United Kingdom
- Diabetes Research Centre, Leicester General Hospital, Leicester, LE5 4PW, United Kingdom
| |
Collapse
|
27
|
Czamara D, Cruceanu C, Lahti-Pulkkinen M, Dieckmann L, Ködel M, Sauer S, Rex-Haffner M, Sammallahti S, Kajantie E, Laivuori H, Lahti J, Räikkönen K, Binder EB. Genome-Wide Copy Number Variant and High-Throughput Transcriptomics Analyses of Placental Tissues Underscore Persisting Child Susceptibility in At-Risk Pregnancies Cleared in Standard Genetic Testing. Int J Mol Sci 2022; 23:ijms231911448. [PMID: 36232765 PMCID: PMC9569583 DOI: 10.3390/ijms231911448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Several studies have shown that children from pregnancies with estimated first-trimester risk based on fetal nuchal translucency thickness and abnormal maternal serum pregnancy protein and hormone levels maintain a higher likelihood of adverse outcomes, even if initial testing for known genetic conditions is negative. We used the Finnish InTraUterine cohort (ITU), which is a comprehensively characterized perinatal cohort consisting of 943 mothers and their babies followed throughout pregnancy and 18 months postnatally, including mothers shortlisted for prenatal genetic testing but cleared for major aneuploidies (cases: n = 544, 57.7%) and control pregnancies (n = 399, 42.3%). Using genome-wide genotyping and RNA sequencing of first-trimester and term placental tissue, combined with medical information from registry data and maternal self-report data, we investigated potential negative medical outcomes and genetic susceptibility to disease and their correlates in placenta gene expression. Case mothers did not present with higher levels of depression, perceived stress, or anxiety during pregnancy. Case children were significantly diagnosed more often with congenital malformations of the circulatory system (4.12 (95% CI [1.22−13.93]) higher hazard) and presented with significantly more copy number duplications as compared to controls (burden analysis, based on all copy number variants (CNVs) with at most 10% frequency, 823 called duplications in 297 cases versus 626 called duplications in 277 controls, p = 0.01). Fifteen genes showed differential gene expression (FDR < 0.1) in association with congenital malformations in first-trimester but not term placenta. These were significantly enriched for genes associated with placental dysfunction. In spite of normal routine follow-up prenatal testing results in early pregnancy, case children presented with an increased likelihood of negative outcomes, which should prompt vigilance in follow-up during pregnancy and after birth.
Collapse
Affiliation(s)
- Darina Czamara
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany
| | - Cristiana Cruceanu
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Marius Lahti-Pulkkinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Finnish Institute for Health and Welfare, 00271 Helsinki, Finland
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Linda Dieckmann
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany
- International Max Planck Research School for Translational Psychiatry, 80804 Munich, Germany
| | - Maik Ködel
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany
| | - Susann Sauer
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany
| | - Monika Rex-Haffner
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany
| | - Sara Sammallahti
- Department of Obstetrics and Gynaecology, Helsinki University Hospital and University of Helsinki, 00014 Helsinki, Finland
| | - Eero Kajantie
- Finnish Institute for Health and Welfare, 00271 Helsinki, Finland
- Children’s Hospital, Helsinki University Hospital and University of Helsinki, 00014 Helsinki, Finland
- Faculty of Medicine, PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, 90014 Oulu, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Hannele Laivuori
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, 00014 Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital and Faculty of Medicine and Health Technology, Center for Child, Adolescent and Maternal Health Research, Tampere University, 33520 Tampere, Finland
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Elisabeth B. Binder
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, 80804 Munich, Germany
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Correspondence:
| |
Collapse
|
28
|
Veiga ECDA, Korkes HA, Salomão KB, Cavalli RC. Association of LEPTIN and other inflammatory markers with preeclampsia: A systematic review. Front Pharmacol 2022; 13:966400. [PMID: 36034841 PMCID: PMC9399808 DOI: 10.3389/fphar.2022.966400] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/08/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Preeclampsia is a serious pregnancy complication that affects 5%–10% of the obstetric population. Objective: To study inflammatory markers associated with preeclampsia. Search Strategy: Searches of articles on the topic published over a 10-year period (2009–2019) were performed in three databases (PubMed, Cochrane, and Embase) using the keywords preeclampsia and inflammatory markers. The PubMed search using 10 years and humans as filters retrieved 124 articles. Using an advanced search strategy, 0 articles were identified in Embase and 10 articles in Cochrane. After screening and eligibility assessment, 13 articles were included in the systematic review and meta-analysis. Meta-analysis and quality assessment of the studies were performed using the Review Manager 5.3 program. Results: For meta-analysis, women with preeclampsia were compared to control women, i.e., pregnancies without arterial hypertension. Leptin levels were significantly higher (p < 0.0002) in women with preeclampsia compared to controls. Total cholesterol was also significantly elevated in women with preeclampsia (p < 0.0001). There was no significant difference in HDL between groups, but women with preeclampsia had significantly increased LDL (p < 0.01). The same was observed for triglycerides, which were significantly increased in women with preeclampsia (p < 0.04) compared to controls. Analysis of TNF-alpha, an important inflammatory marker, showed higher levels in women with preeclampsia (p < 0.03) compared to controls. The same was observed for another important inflammatory marker, interleukin 6, which was significantly increased in women with preeclampsia (p < 0.0002). There was a significant increase of C-reactive protein in women with preeclampsia (p < 0.003) compared to controls. Conclusion: Women with preeclampsia have increased levels of inflammatory markers compared to control women.
Collapse
Affiliation(s)
- Eduardo Carvalho de Arruda Veiga
- Department of Obstetrics and Gynecology, University Hospital, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- *Correspondence: Eduardo Carvalho de Arruda Veiga,
| | - Henri Augusto Korkes
- Department of Obstetrics and Gynecology, University Hospital, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Karina Bezerra Salomão
- Department of Pediatrics, Ribeirão Preto Medical School University of São Paulo, Ribeirão Preto, Brazil
| | - Ricardo Carvalho Cavalli
- Department of Obstetrics and Gynecology, University Hospital, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
29
|
Fang Y, Fang D. Comprehensive analysis of placental gene-expression profiles and identification of EGFR-mediated autophagy and ferroptosis suppression in intrahepatic cholestasis of pregnancy. Gene 2022; 834:146594. [PMID: 35643225 DOI: 10.1016/j.gene.2022.146594] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/18/2022] [Accepted: 05/16/2022] [Indexed: 11/04/2022]
Abstract
Intrahepatic cholestasis of pregnancy (ICP) was the most common liver disease specific to pregnancy. The symptoms of ICP were maternal pruritus and increased bile acid level in serum which was related to preterm birth, fetal distress, meconium-stained amniotic fluid and stillbirth. However, the mechanism of ICP progression on fetal development remained obscure. Sequencing data of 2 normal placenta samples and 4 intrahepatic cholestasis samples during pregnancy was analyzed by GEO2R. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used for analysis of differentially expressed genes. MCODE - A plug-in of Cytoscape was used for molecular complex detection. STRING, Cytoscape, GeneMANIA, NetworkAnalyst, TransmiR, JASPAR, DGIdb and DrugBank were used in this study. Furthermore, histopathological and cell experiments were used to verify our results. Our study identified the key KEGG pathway and four MCODEs which were closely with ICP development, further, sorted by degree centrality, we showed top 30 genes from 7209 differential genes, such as TP53, SRC, EGFR, ESR1, IL10, CD8A, MAPK3, PTPRC, EGF, KIT, ITGAM, LEP and CSF2, etc. Moreover, these hub genes participated in JAK-STAT3 signaling pathway and STAT1/3 regulated these genes expression in a direct way or miRNA-mediated manner. Drug-target analysis about up-regulated genes among hub genes showed that these genes contained multiple drug action site. Furthermore, hub gene-EGFR was associated with destroyed autophagy and ferroptosis. In conclusion, our study analyzed key genes and pathways in ICP development. JAK-STAT3 pathway and EGFR might be a potential target for ICP therapy.
Collapse
Affiliation(s)
- Yan Fang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, People's Republic of China
| | - Dajun Fang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, People's Republic of China.
| |
Collapse
|
30
|
The Update of Fetal Growth Restriction Associated with Biomarkers. MATERNAL-FETAL MEDICINE 2022. [DOI: 10.1097/fm9.0000000000000156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
31
|
Leptin Levels of the Perinatal Period Shape Offspring's Weight Trajectories through the First Year of Age. Nutrients 2022; 14:nu14071451. [PMID: 35406063 PMCID: PMC9003253 DOI: 10.3390/nu14071451] [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: 02/23/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Leptin is a hormone regulating lifetime energy homeostasis and metabolism and its concentration is important starting from prenatal life. We aimed to investigate the association of perinatal leptin concentrations with growth trajectories during the first year of life. Methods: Prospective, longitudinal study, measuring leptin concentration in maternal plasma before delivery, cord blood (CB), and mature breast milk and correlating their impact on neonate’s bodyweight from birth to 1 year of age, in 16 full-term (FT), 16 preterm (PT), and 13 intrauterine growth-restricted (IUGR) neonates. Results: Maternal leptin concentrations were highest in the PT group, followed by IUGR and FT, with no statistical differences among groups (p = 0.213). CB leptin concentrations were significantly higher in FT compared with PT and IUGR neonates (PT vs. FT; IUGR vs. FT: p < 0.001). Maternal milk leptin concentrations were low, with no difference among groups. Maternal leptin and milk concentrations were negatively associated with all the neonates’ weight changes (p = 0.017 and p = 0.006), while the association with CB leptin was not significant (p = 0.051). Considering each subgroup individually, statistical analysis confirmed the previous results in PT and IUGR infants, with the highest value in the PT subgroup. In addition, this group’s results negatively correlated with CB leptin (p = 0.026) and showed the largest % weight increase. Conclusions: Leptin might play a role in neonatal growth trajectories, characterized by an inverse correlation with maternal plasma and milk. PT infants showed the highest correlation with hormone levels, regardless of source, seeming the most affected group by leptin guidance. Low leptin levels appeared to contribute to critical neonates’ ability to recover a correct body weight at 1 year. An eventual non-physiological “catch-up growth” should be monitored, and leptin perinatal levels may be an indicative tool. Further investigations are needed to strengthen the results.
Collapse
|
32
|
A study of the association between long non coding RNA (ULBP1 and MICA/B) expression and preeclampsia in Egypt. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
33
|
Xiao C, Wang Y, Fan Y. Bioinformatics Analysis Identifies Potential Related Genes in the Pathogenesis of Intrauterine Fetal Growth Retardation. Evol Bioinform Online 2022; 18:11769343221112780. [PMID: 35923419 PMCID: PMC9340335 DOI: 10.1177/11769343221112780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Intrauterine growth retardation (IUGR) affects approximately 10% to 15% of
all pregnancies worldwide. IUGR is not only associated with stillbirth and
newborn death, but also the delay of cognition in childhood and the
promotion of metabolic and vascular disorders in adulthood. Figuring out the
mechanism of IUGR is rather meaningful and valuable. Methods: Datasets related to IUGR were searched in the Gene Expression Omnibus
website. Principal component analysis (PCA) was used for normalization.
Differential expressed genes (DEGs) were screened out using the ggpot2 tool.
DEGs were used to conduct Gene Ontology (GO) terms, Kyoto Encyclopedia of
Genes and Genomes (KEGG) pathways enrichment analyses, and protein-protein
interaction (PPI) analysis. IUGR related genes were searched in the OMIM
website to look for the intersection with the DEGs. The DEGs were analyzed
for tissue-specific expression by the online resource BioGPS. The results
were displayed through volcano map, Venn map, box plot, heat map, and GSEA
enrichment plots drawn by R language packages. Results: Eleven DEGs were screened out of 2 datasets. One hundred ninety-five genes
related to IUGR in OMIM were retrieved. EGR2 was the only intersection gene
that was found in both groups. Genes associated with placental tissue
expression include COL17A1, HSD11B1, and LGALS14. Molecular functions of the
DEGs are related to the oxidoreductase activity. The following 4 signaling
pathways, reactome signaling by interleukins, reactome collagen degradation,
Naba secreted factors, and PID NFAT tfpathway, were enriched by GSEA. Two
critical modules comprising 5 up-regulated genes (LEP, PRL, TAC3, MMP14, and
ADAMTS4) and 4 down-regulated genes (TIMP4, FOS, CCK, and KISS1) were
identified by PPI analysis. Finally, we identified 6 genes (PRL, LGALS14,
EGR2, TAC3, LEP, and KISS1) that are potentially relevant to the
pathophysiology of IUGR. Conclusion: The candidate down-regulated genes LGALS14 and KISS1, as well as the
up-regulated genes PRL, EGR2, TAC3, and LEP, were found to be closely
related to IUGR by bioinformatics analysis. These hub genes are related to
hypoxia and oxidoreductase activities in placental development. We provide
useful and novel information to explore the potential mechanism of IUGR and
make efforts to the prevention of IUGR.
Collapse
Affiliation(s)
- Chao Xiao
- Department of Obstetrics and Gynecology, Zigong First People’s Hospital, Sichuan, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Sichuan, China
| | - Yao Wang
- Department of Obstetrics and Gynecology, Zigong First People’s Hospital, Sichuan, China
| | - Yuchao Fan
- Department of Anesthesiology, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
34
|
Wojczakowski W, Kimber-Trojnar Ż, Dziwisz F, Słodzińska M, Słodziński H, Leszczyńska-Gorzelak B. Preeclampsia and Cardiovascular Risk for Offspring. J Clin Med 2021; 10:jcm10143154. [PMID: 34300320 PMCID: PMC8306208 DOI: 10.3390/jcm10143154] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023] Open
Abstract
There is growing evidence of long-term cardiovascular sequelae in children after in utero exposure to preeclampsia. Maternal hypertension and/or placental ischaemia during pregnancy increase the risk of hypertension, stroke, diabetes, and cardiovascular disease (CVD) in the offspring later in life. The mechanisms associated with CVD seem to be a combination of genetic, molecular, and environmental factors which can be defined as fetal and postnatal programming. The aim of this paper is to discuss the relationship between pregnancy complicated by preeclampsia and possibility of CVD in the offspring. Unfortunately, due to its multifactorial nature, a clear dependency mechanism between preeclampsia and CVD is difficult to establish.
Collapse
Affiliation(s)
- Wiktor Wojczakowski
- Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland; (W.W.); (M.S.); (B.L.-G.)
| | - Żaneta Kimber-Trojnar
- Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland; (W.W.); (M.S.); (B.L.-G.)
- Correspondence: ; Tel.: +48-81-7244-769
| | - Filip Dziwisz
- Department of Interventional Cardiology and Cardiac Arrhythmias, Medical University of Lodz, 90-549 Łódź, Poland;
| | - Magdalena Słodzińska
- Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland; (W.W.); (M.S.); (B.L.-G.)
| | - Hubert Słodziński
- Institute of Medical Sciences, State School of Higher Education in Chełm, 22-100 Chełm, Poland;
| | - Bożena Leszczyńska-Gorzelak
- Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland; (W.W.); (M.S.); (B.L.-G.)
| |
Collapse
|