1
|
Muraglia M, Faienza MF, Tardugno R, Clodoveo ML, Matias De la Cruz C, Bermúdez FG, Munizaga MG, Valencia L, Corbo F, Orellana-Manzano A. Breastfeeding: science and knowledge in pediatric obesity prevention. Front Med (Lausanne) 2024; 11:1430395. [PMID: 39399112 PMCID: PMC11466875 DOI: 10.3389/fmed.2024.1430395] [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: 05/09/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
The increasing prevalence of childhood obesity worldwide is a significant concern due to its link to severe health issues in adulthood, such as non-communicable diseases (NCDs). To address this issue, this review evaluates the effectiveness of various preventive measures for childhood obesity, focusing on maternal nutrition and breastfeeding. The study underscores the criticality of the periconceptional period, where the diets of both parents can influence epigenetic modifications that impact the child's metabolic pathways and obesity risks. Breastfeeding is a potent protective mechanism against early-onset obesity, significantly enhancing the infant's metabolic and immune health by modifying DNA methylation and gene expression. Furthermore, the perspective underscores the significance of the Mediterranean diet during the periconceptional period and lactation. This diet can effectively prevent gestational complications and improve breast milk quality, fostering optimal infant development. Recognizing that obesity results from genetic, epigenetic, environmental, and social factors, the paper advocates for a comprehensive, multidisciplinary approach from the earliest stages of life. This approach champions a balanced maternal diet, exclusive breastfeeding, and timely introduction to complementary foods. In conclusion, addressing pediatric obesity requires a multifaceted strategy emphasizing improving prenatal and postnatal nutrition. Further research is necessary to understand the epigenetic mechanisms influenced by nutrition and their long-term effects on children's health. This will help refine interventions that curb the obesity epidemic among future generations.
Collapse
Affiliation(s)
- Marilena Muraglia
- Department of Pharmacy - Drug Science, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “A. Moro”, Bari, Italy
| | - Roberta Tardugno
- Department of Pharmacy - Drug Science, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Maria Lisa Clodoveo
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari-Aldo Moro, Bari, Italy
| | - Carmen Matias De la Cruz
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Fátima German Bermúdez
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - María Gabriela Munizaga
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Luz Valencia
- Licenciatura en Nutrición y Dietética, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Filomena Corbo
- Department of Pharmacy - Drug Science, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Andrea Orellana-Manzano
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| |
Collapse
|
2
|
Hertiš Petek T, Marčun Varda N. Childhood Cardiovascular Health, Obesity, and Some Related Disorders: Insights into Chronic Inflammation and Oxidative Stress. Int J Mol Sci 2024; 25:9706. [PMID: 39273654 PMCID: PMC11396019 DOI: 10.3390/ijms25179706] [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: 06/04/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Childhood obesity and associated metabolic abnormalities have become pressing public health concerns worldwide, significantly impacting cardiovascular health. Metabolic syndrome, characterized by a cluster of metabolic abnormalities including central obesity, altered glucose metabolism, dyslipidemia, and arterial hypertension, has emerged as a critical precursor to cardiovascular disease. Chronic systemic inflammation and oxidative stress seem to play pivotal roles in the pathogenesis of childhood obesity-related disorders such as early atherosclerosis. A significant distinction between the objective components of cardiovascular health metrics, including body mass index, blood pressure, cholesterol, and fasting glucose levels, and the definition of metabolic syndrome is evident in the identification of obesity. Whereas cardiovascular health metrics predominantly rely on body mass index percentiles to assess obesity, metabolic syndrome criteria prioritize waist circumference, specifically targeting individuals with a measurement ≥90th percentile. This discrepancy emphasizes the need for a nuanced approach in assessing the risks associated with obesity and underscores the importance of considering multiple factors when evaluating cardiovascular risk in children. By recognizing the complex interplay between various health metrics, obesity and metabolic syndrome criteria, clinicians can more accurately identify individuals at risk and tailor interventions accordingly to mitigate cardiovascular disease in children with obesity.
Collapse
Affiliation(s)
- Tjaša Hertiš Petek
- Department of Paediatrics, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia
| | - Nataša Marčun Varda
- Department of Paediatrics, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| |
Collapse
|
3
|
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
|
4
|
Selinger CP. Risk of developing IBD in high-risk individuals: the need to study the exposome more. Gut 2024:gutjnl-2024-333347. [PMID: 39089859 DOI: 10.1136/gutjnl-2024-333347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
|
5
|
Hu YC, Huang TC, Huang LW, Cheng HL, Hsieh BS, Chang KL. S-Equol Ameliorates Menopausal Osteoarthritis in Rats through Reducing Oxidative Stress and Cartilage Degradation. Nutrients 2024; 16:2364. [PMID: 39064807 PMCID: PMC11280421 DOI: 10.3390/nu16142364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease leading to articular cartilage destruction. Menopausal and postmenopausal women are susceptible to both OA and osteoporosis. S-equol, a soy isoflavone-derived molecule, is known to reduce osteoporosis in estrogen-deficient mice, but its role in OA remains unknown. This study aimed to explore the effect of S-equol on different degrees of menopausal OA in female Sprague-Dawley (SD) rats induced by estrogen deficiency caused by bilateral ovariectomy (OVX) combined with intra-articular injection of mono-iodoacetate (MIA). Knee joint histopathological change; serum biomarkers of bone turnover, including N-terminal propeptide of type I procollagen (PINP), C-terminal telopeptide of type I collagen (CTX-I) and N-terminal telopeptide of type I collagen (NTX-I); the cartilage degradation biomarkers hyaluronic acid (HA) and N-terminal propeptide of type II procollagen (PIINP); and the matrix-degrading enzymes matrix metalloproteinases (MMP)-1, MMP-3 and MMP-13, as well as the oxidative stress-inducing molecules nitric oxide (NO) and hydrogen peroxide (H2O2), were assessed for evaluation of OA progression after S-equol supplementation for 8 weeks. The results showed that OVX without or with MIA injection induced various severity levels of menopausal OA by increasing pathological damage, oxidative stress, and cartilage matrix degradation to various degrees. Moreover, S-equol supplementation could significantly reduce these increased biomarkers in different severity levels of OA. This indicates that S-equol can lessen menopausal OA progression by reducing oxidative stress and the matrix-degrading enzymes involved in cartilage degradation.
Collapse
Affiliation(s)
- Yu-Chen Hu
- Department of Biochemistry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (Y.-C.H.); (T.-C.H.); (B.-S.H.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Tzu-Ching Huang
- Department of Biochemistry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (Y.-C.H.); (T.-C.H.); (B.-S.H.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Li-Wen Huang
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
| | - Hsiao-Ling Cheng
- Department of Pharmacy, Kaohsiung Municipal Min-Sheng Hospital, Kaohsiung 802511, Taiwan;
| | - Bau-Shan Hsieh
- Department of Biochemistry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (Y.-C.H.); (T.-C.H.); (B.-S.H.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Kee-Lung Chang
- Department of Biochemistry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (Y.-C.H.); (T.-C.H.); (B.-S.H.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| |
Collapse
|
6
|
Chico-Barba G, Sámano R, Martínez-Rojano H, Morales-Hernández RM, Barrientos-Galeana E, Luna-Hidalgo A, Kaufer-Horwitz M, Obrador GT, Villa-Romero AR. Total Gestational Weight Gain Is Explained by Leptin and Body Fat, Regardless of Pre-Pregnancy Body Mass Index and Other Adipokines, in Mexican Adolescents. Nutrients 2024; 16:2147. [PMID: 38999894 PMCID: PMC11242962 DOI: 10.3390/nu16132147] [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: 06/12/2024] [Revised: 06/30/2024] [Accepted: 07/03/2024] [Indexed: 07/14/2024] Open
Abstract
Pre-pregnancy body mass index (pBMI) is a predictor of gestational weight gain (GWG). However, other factors, such as adipokines and inflammation markers, may also be associated with GWG. The aim of the study was to determine the association of leptin, adiponectin, irisin, and C-reactive protein, with GWG in adolescents. A longitudinal study was conducted from 2018 to 2023 in adolescents with a clinically healthy pregnancy. The assessments included sociodemographic and clinical data, pBMI, percent of body fat, serum concentrations of leptin, adiponectin, irisin, and high-sensitivity C-reactive protein (hsCRP), and total GWG adequacy. Cox regression models were performed, the outcome variables were inadequate and excessive GWG. In 198 participants, being overweight/obesity was marginally associated with a protective effect against inadequate GWG (HR = 0.44, 95%CI = 0.18-1.06), regardless of maternal characteristics and adipokines. Leptin (HR = 1.014, 95%CI = 1.008-1.021), and body fat percent (HR = 1.11, 95%CI = 1.05-1.17) were associated with a higher risk of excessive GWG, independent of other maternal variables such as pBMI, while adiponectin was associated with a lower risk. These findings suggest that, in Mexican adolescents, adipose tissue and its adipokines during pregnancy may play a more significant role in the final GWG than body weight.
Collapse
Affiliation(s)
- Gabriela Chico-Barba
- Programa de Maestría y Doctorado en Ciencias Médicas, Odontológicas y de la Salud, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | - Reyna Sámano
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | - Hugo Martínez-Rojano
- Sección de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | | | - Edgar Barrientos-Galeana
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | - Andrea Luna-Hidalgo
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | - Martha Kaufer-Horwitz
- Dirección de Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | | | - Antonio Rafael Villa-Romero
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
7
|
Zhu SL, Zhang HT, Du YY, Jiang Y, Wang SS, Ding WC, Feng L. Histological Features of Uterine Myometrial Dysfunction: Possible Involvement of Localized Inflammation. Curr Med Sci 2024; 44:633-641. [PMID: 38789820 DOI: 10.1007/s11596-024-2873-3] [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: 01/25/2024] [Accepted: 04/16/2024] [Indexed: 05/26/2024]
Abstract
OBJECTIVE The latest perspective suggests that elevated levels of inflammation and cytokines are implicated in atonic postpartum hemorrhage. Lipopolysaccharide (LPS) has been widely used to induce inflammation in animal models. Therefore, this study aimed to induce uterine inflammation using LPS to investigate whether local inflammation triggers dysfunction and atrophy in the myometrium, as well as the potential underlying molecular mechanisms involved. METHODS In vivo, an animal model was established by intraperitoneal injection of 300 μg/ kg LPS in rats on gestational day 21. Hematoxylin-eosin (H&E) staining and Masson staining were employed to determine morphological changes in the rat uterine smooth muscle. Enzyme-linked immunosorbent assay (ELISA) was used to detect inflammatory cytokines. Immunohistochemistry, tissue fluorescence, and Western blotting were conducted to assess the expression levels of the uterine contraction-related proteins Toll-like receptor 4 (TLR4) and the nuclear factor kappa-B (NF-κB) signaling pathway. In vitro, human uterine smooth muscle cells (HUtSMCs) were exposed to 2 μg/mL LPS to further elucidate the involvement of the TLR4/NF-κB signaling pathway in LPS-mediated inflammation. RESULTS In this study, LPS induced uterine myometrial dysfunction in rats, leading to a disorganized arrangement, a significant increase in collagen fiber deposition, and widespread infiltration of inflammatory cells. In both in vivo animal models and in vitro HUtSMCs, LPS elevated IL-6, IL-1β, and TNF-α levels while concurrently suppressing the expression of connexin 43 (Cx43) and oxytocin receptor (OXTR). Mechanistically, the LPS-treated group exhibited TLR4 activation, and the phosphorylation levels of p65 and IκBα were notably increased. CONCLUSION LPS triggered the TLR4/NF-κB signaling pathway, inducing an inflammatory response in the myometrium and leading to uterine myometrial dysfunction and uterine atony.
Collapse
Affiliation(s)
- Sheng-Lan Zhu
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui-Ting Zhang
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuan-Yuan Du
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yi Jiang
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shao-Shuai Wang
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen-Cheng Ding
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Ling Feng
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
8
|
Celik D, Campisi M, Cannella L, Pavanello S. The effect of low birth weight as an intrauterine exposure on the early onset of sarcopenia through possible molecular pathways. J Cachexia Sarcopenia Muscle 2024; 15:770-780. [PMID: 38553412 PMCID: PMC11154781 DOI: 10.1002/jcsm.13455] [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: 08/31/2023] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 06/07/2024] Open
Abstract
Sarcopenia, a musculoskeletal disease characterized by the progressive loss of skeletal muscle mass, strength, and physical performance, presents significant challenges to global public health due to its adverse effects on mobility, morbidity, mortality, and healthcare costs. This comprehensive review explores the intricate connections between sarcopenia and low birth weight (LBW), emphasizing the developmental origins of health and disease (DOHaD) hypothesis, inflammatory processes (inflammaging), mitochondrial dysfunction, circadian rhythm disruptions, epigenetic mechanisms, and genetic variations revealed through genome-wide studies (GWAS). A systematic search strategy was developed using PubMed to identify relevant English-language publications on sarcopenia, LBW, DOHaD, inflammaging, mitochondrial dysfunction, circadian disruption, epigenetic mechanisms, and GWAS. The publications consist of 46.2% reviews, 21.2% cohort studies, 4.8% systematic reviews, 1.9% cross-sectional studies, 13.4% animal studies, 4.8% genome-wide studies, 5.8% epigenome-wide studies, and 1.9% book chapters. The review identified key factors contributing to sarcopenia development, including the DOHaD hypothesis, LBW impact on muscle mass, inflammaging, mitochondrial dysfunction, the influence of clock genes, the role of epigenetic mechanisms, and genetic variations revealed through GWAS. The DOHaD theory suggests that LBW induces epigenetic alterations during foetal development, impacting long-term health outcomes, including the early onset of sarcopenia. LBW correlates with reduced muscle mass, grip strength, and lean body mass in adulthood, increasing the risk of sarcopenia. Chronic inflammation (inflammaging) and mitochondrial dysfunction contribute to sarcopenia, with LBW linked to increased oxidative stress and dysfunction. Disrupted circadian rhythms, regulated by genes such as BMAL1 and CLOCK, are associated with both LBW and sarcopenia, impacting lipid metabolism, muscle mass, and the ageing process. Early-life exposures, including LBW, induce epigenetic modifications like DNA methylation (DNAm) and histone changes, playing a pivotal role in sarcopenia development. Genome-wide studies have identified candidate genes and variants associated with lean body mass, muscle weakness, and sarcopenia, providing insights into genetic factors contributing to the disorder. LBW emerges as a potential early predictor of sarcopenia development, reflecting the impact of intrauterine exposures on long-term health outcomes. Understanding the complex interplay between LBW with inflammaging, mitochondrial dysfunction, circadian disruption, and epigenetic factors is essential for elucidating the pathogenesis of sarcopenia and developing targeted interventions. Future research on GWAS and the underlying mechanisms of LBW-associated sarcopenia is warranted to inform preventive strategies and improve public health outcomes.
Collapse
Affiliation(s)
- Dilek Celik
- Department of Pharmceutical and Pharmacological SciencesUniversity of PaduaPaduaItaly
| | - Manuela Campisi
- Department of Cardiac Thoracic Vascular Sciences and Public HealthUniversity of PaduaPaduaItaly
| | - Luana Cannella
- Department of Cardiac Thoracic Vascular Sciences and Public HealthUniversity of PaduaPaduaItaly
| | - Sofia Pavanello
- Department of Cardiac Thoracic Vascular Sciences and Public HealthUniversity of PaduaPaduaItaly
- University Hospital of PadovaPaduaItaly
| |
Collapse
|
9
|
Hauta-Alus HH, Rosendahl J, Holmlund-Suila EM, Valkama SM, Enlund-Cerullo M, Nurhonen M, Kajantie E, Mäkitie O, Andersson S. Low-grade inflammation from prenatal period to age 6-8 years in a Vitamin D trial. Pediatr Res 2024; 95:1578-1586. [PMID: 38225452 PMCID: PMC11126391 DOI: 10.1038/s41390-024-03019-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 11/29/2023] [Accepted: 12/26/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND Low-grade systemic inflammation measured as high sensitivity C-reactive protein (hs-CRP) has been associated with non-communicable disease risk. We assessed whether prenatal inflammation and early-childhood vitamin D are associated with inflammation until age 6-8. METHODS We analyzed blood hs-CRP and 25-hydroxy vitamin D [25(OH)D] in pregnancy, at birth from umbilical cord blood (UCB), from offspring at ages 1, 2, and 6-8 years in the Vitamin D Intervention in Infants (VIDI) study. VIDI was a randomized-controlled trial of vitamin D supplementation of 10 μg/day or 30 μg/day from age 2 weeks until 2 years in 975 infants recruited in 2013-14, with follow-up at age 6-8 in 2019-21 (n = 283). RESULTS Pregnancy hs-CRP was associated with UCB hs-CRP (r = 0.18, p < 0.001) but not independently with childhood hs-CRP (Estimate [95% CI] 0.04 [<-0.00, 0.09]). Higher UCB hs-CRP was associated independently with higher hs-CRP until 6-8 years (0.20 [0.12, 0.29]). Infant vitamin D dose had no effect on longitudinal hs-CRP (6-8 years, 0.11 [-0.04, 0.25]). Childhood 25(OH)D were associated positively with hs-CRP until age 6-8 (0.01 [>0.00, 0.01]). CONCLUSION Our results indicate that in children, inflammation, assessed by hs-CRP, persists from birth until 6-8 years. We observed positive associations between 25(OH)D and hs-CRP in vitamin D-sufficient children. IMPACT High sensitivity C-reactive protein (hs-CRP) concentrations tract from birth to age 8 years Our novel finding suggests a long-lasting pro-inflammatory phenotype in the child Higher vitamin D concentration - but not dose - is associated with higher childhood hs-CRP Chronic disease risk related to inflammation may in part originate from the prenatal period or early childhood Further studies are needed to investigate the effects of inflammation on long-term clinical health outcomes.
Collapse
Affiliation(s)
- Helena H Hauta-Alus
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism (CAMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Population Health unit, National Institute for Health and Welfare (THL), Helsinki, Finland.
- PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.
| | - Jenni Rosendahl
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism (CAMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Elisa M Holmlund-Suila
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism (CAMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Saara M Valkama
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism (CAMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Maria Enlund-Cerullo
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism (CAMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Markku Nurhonen
- Population Health unit, National Institute for Health and Welfare (THL), Helsinki, Finland
| | - Eero Kajantie
- Population Health unit, National Institute for Health and Welfare (THL), Helsinki, Finland
- PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Outi Mäkitie
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Sture Andersson
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
10
|
Snoek KM, van de Woestijne N, Ritfeld VEEG, Klaassen RA, Versendaal H, Galjaard S, Willemsen SP, Laven JSE, Steegers-Theunissen RPM, Schoenmakers S. Preconception maternal gastric bypass surgery and the impact on fetal growth parameters. Surg Obes Relat Dis 2024; 20:128-137. [PMID: 37805294 DOI: 10.1016/j.soard.2023.08.015] [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: 02/22/2023] [Revised: 07/27/2023] [Accepted: 08/28/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND Bariatric surgery is increasingly performed in women of reproductive age. As bariatric surgery will result in postoperative rapid catabolic weight loss which potentially leads to fetal malnutrition and directly related impaired intra-uterine growth, it is advised to postpone pregnancy for at least 12-18 months after surgery. OBJECTIVES To investigate the consequences of preconception gastric bypass surgery (pGB) on fetal growth parameters and maternal pregnancy outcome. SETTING Maasstad Hospital, The Netherlands, general hospital and Erasmus Medical Center, The Netherlands, university hospital. METHODS We included 97 pGB pregnancies (Maasstad hospital) and 440 non-bariatric pregnancies (Rotterdam Periconception cohort, Erasmus Medical Center). Longitudinal second and third trimester fetal growth parameters (head circumference, biparietal diameter, femur length, abdominal circumference, estimated fetal weight) were analyzed using linear mixed models, adjusting for covariates and possible confounders. Fetal growth and birthweight in pGB pregnancies were compared to non-bariatric pregnancies and Dutch reference curves. Maternal pregnancy outcome in the pGB group was compared to non-bariatric pregnancies. RESULTS All fetal growth parameters of pGB pregnancies were significantly decreased at 20 weeks' gestation (P < .001) and throughout the remaining part of pregnancy (P < .05) compared with non-bariatric pregnancies (crude and adjusted models). In our cohort, gestational weight gain was not significantly associated with birthweight corrected for gestational age. Birthweight was significantly lower in pGB pregnancies (estimate -241 grams [95% CI, -342.7 to -140.0]) with a 2-fold increased risk of small-for-gestational-age (SGA) (adjusted odds ratio 2.053 [95% CI, 1.058 to 3.872]). Compared to the non-bariatric pregnancies, we found no significant differences in maternal pregnancy outcome. CONCLUSIONS PGB is associated with overall reduced fetal growth trajectories and a 2-fold increased risk of SGA, without significant adverse consequences for maternal pregnancy outcome. We recommend close monitoring of fetal growth after pGB.
Collapse
Affiliation(s)
- Katinka M Snoek
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Nadia van de Woestijne
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | | - René A Klaassen
- Department of Surgery, Maasstad Hospital, Rotterdam, The Netherlands
| | - Hans Versendaal
- Department of Obstetrics and Gynecology, Maasstad Hospital, Rotterdam, The Netherlands
| | - Sander Galjaard
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Sten P Willemsen
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands; Department of Biostatistics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Joop S E Laven
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | | - Sam Schoenmakers
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
11
|
Wiley KS, Kwon D, Knorr DA, Fox MM. Regulatory T-cell phenotypes in prenatal psychological distress. Brain Behav Immun 2024; 116:62-69. [PMID: 38016492 PMCID: PMC11402516 DOI: 10.1016/j.bbi.2023.11.033] [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: 06/05/2023] [Revised: 11/18/2023] [Accepted: 11/23/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Experiencing symptoms of psychological distress during pregnancy is common and has been linked to dysregulated immune functioning. In this context, immunoregulatory function is especially relevant because of its crucial role in establishment and maintenance of healthy pregnancy. However, little research has examined associations between women's prenatal psychological distress and immunoregulatory biomarkers. We investigated how symptoms of depression, anxiety, and stress relate to circulating levels of regulatory T-cells (Tregs). MATERIALS AND METHODS Pregnant Latina women were assessed at around 12 weeks of pregnancy (N = 82). These assessments included blood draws and self-report questionnaires assessing symptoms of depression, state anxiety, pregnancy-related anxiety, and perceived stress. Flow cytometry on PBMCs was used to quantify circulating Tregs, defined as CD3+CD4+CD25hiCD127loFoxP3+, and subpopulations positive for one of the following intra- or extracellular markers, CD45RA, CTLA-4, Helios, PD-1, TIM-3, and TIGIT. We collected 82 samples at 12 weeks. Multivariable linear regressions tested for associations between symptoms of psychological distress and Treg concentrations, adjusted for gestational age. RESULTS State anxiety symptoms at 12 weeks were negatively associated with parent Treg cell levels (b = -4.02, p = 0.023) and subpopulations Helios+ (b = -3.29, p = 0.019) and TIM3+ (b = -3.17, p = 0.008). Perceived stress was negatively associated with the PD-1+ subpopulation at 12 weeks (b = -4.02, p = 0.023). Depression was not related to Tregs or the subpopulations. CONCLUSION Our observation that symptoms of anxiety and stress are related to tolerogenic immunology suggests a possible biomechanism explaining correlations of maternal mood disorders with adverse outcomes for mothers and offspring.
Collapse
Affiliation(s)
- Kyle S Wiley
- Department of Anthropology, University of California, Los Angeles, United States; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, United States.
| | - Dayoon Kwon
- Department of Epidemiology, UCLA Fielding School of Public Health, University of California, Los Angeles, United States
| | - Delaney A Knorr
- Department of Anthropology, University of California, Los Angeles, United States
| | - Molly M Fox
- Department of Anthropology, University of California, Los Angeles, United States; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, United States
| |
Collapse
|
12
|
Zhu J, Li Z, Deng Y, Lan L, Yang J. Comprehensive reference intervals for white blood cell counts during pregnancy. BMC Pregnancy Childbirth 2024; 24:35. [PMID: 38182972 PMCID: PMC10768452 DOI: 10.1186/s12884-023-06227-8] [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: 09/01/2023] [Accepted: 12/26/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND White blood cell (WBC) count increases during pregnancy, necessitating reliable reference intervals for assessing infections and pregnancy-related complications. This study aimed to establish comprehensive reference intervals for WBC counts during pregnancy. METHODS The analysis included 17,737 pregnant women, with weekly WBC count measurements from pre-pregnancy to postpartum. A threshold linear regression model determined reference intervals, while Harris and Boyd's test partitioned the intervals. RESULTS WBC count exhibited a significant increase during pregnancy, characterized by a rapid rise before 7 weeks of gestation, followed by a plateau. Neutrophils primarily drove this increase, showing a similar pattern. The threshold regression model and Harris and Boyd's test supported partitioned reference intervals for WBC counts: 4.0-10.0 × 10^9/L for < = 2 weeks, 4.7-11.9 × 10^9/L for 3-5 weeks, and 5.7-14.4 × 10^9/L for > = 6 weeks of gestation. These reference intervals identified pregnant women with high WBC counts, who had a higher incidence of pregnancy-related complications including placenta previa, oligohydramnios, secondary uterine inertia, and intrauterine growth restriction. CONCLUSION This study establishes comprehensive reference intervals for WBC counts during pregnancy. Monitoring WBC counts is clinically relevant, as elevated levels are associated with an increased risk of infection and pregnancy-related complications.
Collapse
Affiliation(s)
- Jinxiu Zhu
- Longgang Maternity and Child Institute of Shantou University Medical College, Shenzhen, Guangdong, 518172, China
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Zexin Li
- Longgang Maternity and Child Institute of Shantou University Medical College, Shenzhen, Guangdong, 518172, China
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Yuguo Deng
- Department of Obstetrics, Longgang District Maternity and Child Healthcare Hospital of Shenzhen City, Shenzhen, Guangdong, 518172, China
- Longgang Maternity and Child Institute of Shantou University Medical College, Shenzhen, Guangdong, 518172, China
| | - Liting Lan
- Longgang Maternity and Child Institute of Shantou University Medical College, Shenzhen, Guangdong, 518172, China
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Jinying Yang
- Department of Obstetrics, Longgang District Maternity and Child Healthcare Hospital of Shenzhen City, Shenzhen, Guangdong, 518172, China.
- Longgang Maternity and Child Institute of Shantou University Medical College, Shenzhen, Guangdong, 518172, China.
| |
Collapse
|
13
|
Lubrano C, Parisi F, Coco C, Marelli E, Burello E, Cetin I. Associations between Maternal Nutritional Status, Hemodynamic Parameters, and Delivery Outcomes in Low-Risk Pregnancies: A Prospective Observational Study. Nutrients 2024; 16:183. [PMID: 38257076 PMCID: PMC10819587 DOI: 10.3390/nu16020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/01/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Maternal nutritional status represents a pivotal predictor of pregnancy outcome. This prospective observational study investigates the associations between maternal characteristics and nutritional habits at term, hemodynamic parameters, and pregnancy outcomes. Healthy women with singleton uncomplicated pregnancies were enrolled at 36-41 gestational weeks. At enrollment, a nutritional score (0-10) was calculated in order to quantify maternal adherence to a healthy diet and lifestyle. Maternal hemodynamic parameters were assessed by using the Ultrasonic Cardiac Output Monitor (USCOM), including cardiac output (CO), systemic vascular resistance (SVR) and Smith-Madigan inotropy index (SMII). Pregnancy outcomes were recorded at delivery. Associations between maternal characteristics and nutritional score, hemodynamic parameters, and pregnancy outcomes were investigated by using multi-adjusted generalized linear models. In total, 143 pregnancies were enrolled. Pregestational body mass index (BMI) was positively associated with SVR, and negatively associated with CO and SMII. Additionally, a positive association was detected between the nutritional score and SMII. Finally, CO was positively associated with birth and placental weight, while RVS showed a negative association with birth and placental weight. This study shows that maternal derangements in nutritional status and habits are associated with a compromised hemodynamic profile at term, with additional impacts on intrauterine growth.
Collapse
Affiliation(s)
- Chiara Lubrano
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (C.L.); (C.C.); (E.M.); (E.B.); (I.C.)
- Department of Woman, Mother and Child, Luigi Sacco and Vittore Buzzi Children Hospitals, ASST Fatebenefratelli-Sacco, Università degli Studi di Milano, 20154 Milan, Italy
| | - Francesca Parisi
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (C.L.); (C.C.); (E.M.); (E.B.); (I.C.)
- Department of Mother, Child and Neonate, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Chiara Coco
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (C.L.); (C.C.); (E.M.); (E.B.); (I.C.)
- Department of Woman, Mother and Child, Luigi Sacco and Vittore Buzzi Children Hospitals, ASST Fatebenefratelli-Sacco, Università degli Studi di Milano, 20154 Milan, Italy
| | - Elisabetta Marelli
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (C.L.); (C.C.); (E.M.); (E.B.); (I.C.)
- Department of Woman, Mother and Child, Luigi Sacco and Vittore Buzzi Children Hospitals, ASST Fatebenefratelli-Sacco, Università degli Studi di Milano, 20154 Milan, Italy
| | - Eleonora Burello
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (C.L.); (C.C.); (E.M.); (E.B.); (I.C.)
- Department of Woman, Mother and Child, Luigi Sacco and Vittore Buzzi Children Hospitals, ASST Fatebenefratelli-Sacco, Università degli Studi di Milano, 20154 Milan, Italy
| | - Irene Cetin
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (C.L.); (C.C.); (E.M.); (E.B.); (I.C.)
- Department of Mother, Child and Neonate, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
14
|
Ostojic SM, Ratgeber L, Betlehem J, Acs P. Molecular nutrition in life course perspective: Pinpointing metabolic pathways to target during periconception. MATERNAL & CHILD NUTRITION 2024; 20 Suppl 2:e13474. [PMID: 36794361 PMCID: PMC10765360 DOI: 10.1111/mcn.13474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/27/2022] [Accepted: 01/05/2023] [Indexed: 02/17/2023]
Abstract
Lifecourse nutrition encompasses nourishment from early development into parenthood. From preconception and pregnancy to childhood, late adolescence, and reproductive years, life course nutrition explores links between dietary exposures and health outcomes in current and future generations from a public health perspective, usually addressing lifestyle behaviours, reproductive well-being and maternal-child health strategies. However, nutritional factors that play a role in conceiving and sustaining new life might also require a molecular perspective and recognition of critical interactions between specific nutrients and relevant biochemical pathways. The present perspective summarises evidence about the links between diet during periconception and next-generation health and outlines the main metabolic networks involved in nutritional biology of this sensitive time frame.
Collapse
Affiliation(s)
- Sergej M. Ostojic
- Department of Nutrition and Public HealthUniversity of AgderKristiansandNorway
- Faculty of Health SciencesUniversity of PécsPécsHungary
- Applied Bioenergetcis Lab, Faculty of Sport and Physical EducationUniversity of Novi SadNovi SadSerbia
| | | | | | - Pongrac Acs
- Faculty of Health SciencesUniversity of PécsPécsHungary
| |
Collapse
|
15
|
Matuszak O, Banach W, Pogorzały B, Muszyński J, Mengesha SH, Bogdański P, Skrypnik D. The Long-Term Effect of Maternal Obesity on the Cardiovascular Health of the Offspring-Systematic Review. Curr Probl Cardiol 2024; 49:102062. [PMID: 37652110 DOI: 10.1016/j.cpcardiol.2023.102062] [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: 08/26/2023] [Accepted: 08/26/2023] [Indexed: 09/02/2023]
Abstract
Maternal obesity may affect offspring's cardiovascular health. Our literature search using PubMed, Web of Sciences included original English research and Google Scholar articles published over the past ten years, culminating in 96 articles in this topic. A mother's obesity during pregnancy has a negative impact on the cardiovascular risk for their offspring. Dependence was observed in relation to hypertension, coronary artery disease, stroke, and heart failure. The adverse impact of an abnormal diet in pregnant mice on heart hypertrophy was observed, and was also confirmed in human research. Pregnant women with obesity were at greater risk of having a child with innate heart disease than pregnant women with normal mass. To conclude: mother's obesity has a negative impact on the long-term cardiovascular consequences for their offspring, increasing their risk of high blood pressure, coronary heart disease, stroke and heart failure. It also increases the probability of heart hypertrophy and innate heart defects.
Collapse
Affiliation(s)
- Oskar Matuszak
- Faculty of Medicine, Poznań University of Medical Sciences, Poznań, Poland; Student Scientific Association of Lifestyle Medicine, Poznań University of Medical Sciences, Poznań, Poland
| | - Weronika Banach
- Faculty of Medicine, Poznań University of Medical Sciences, Poznań, Poland; Student Scientific Association of Lifestyle Medicine, Poznań University of Medical Sciences, Poznań, Poland
| | - Bartosz Pogorzały
- Department of Internal Medicine and Cardiology, District Hospital, Juraszów St. 7-19, Poznań, Poland
| | - Józef Muszyński
- Faculty of Medicine, Poznań University of Medical Sciences, Poznań, Poland; Student Scientific Association of Lifestyle Medicine, Poznań University of Medical Sciences, Poznań, Poland
| | - Solyana Hailemelekot Mengesha
- Faculty of Medicine, Poznań University of Medical Sciences, Poznań, Poland; Student Scientific Association of Lifestyle Medicine, Poznań University of Medical Sciences, Poznań, Poland
| | - Paweł Bogdański
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences; Poznań, Poland
| | - Damian Skrypnik
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences; Poznań, Poland.
| |
Collapse
|
16
|
Usman TO, Chhetri G, Yeh H, Dong HH. Beta-cell compensation and gestational diabetes. J Biol Chem 2023; 299:105405. [PMID: 38229396 PMCID: PMC10694657 DOI: 10.1016/j.jbc.2023.105405] [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: 08/28/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 01/18/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is characterized by glucose intolerance in pregnant women without a previous diagnosis of diabetes. While the etiology of GDM remains elusive, the close association of GDM with increased maternal adiposity and advanced gestational age implicates insulin resistance as a culpable factor for the pathogenesis of GDM. Pregnancy is accompanied by the physiological induction of insulin resistance in the mother secondary to maternal weight gain. This effect serves to spare blood glucose for the fetus. To overcome insulin resistance, maternal β-cells are conditioned to release more insulin into the blood. Such an adaptive response, termed β-cell compensation, is essential for maintaining normal maternal metabolism. β-cell compensation culminates in the expansion of β-cell mass and augmentation of β-cell function, accounting for increased insulin synthesis and secretion. As a result, a vast majority of mothers are protected from developing GDM during pregnancy. In at-risk pregnant women, β-cells fail to compensate for maternal insulin resistance, contributing to insulin insufficiency and GDM. However, gestational β-cell compensation ensues in early pregnancy, prior to the establishment of insulin resistance in late pregnancy. How β-cells compensate for pregnancy and what causes β-cell failure in GDM are subjects of investigation. In this mini-review, we will provide clinical and preclinical evidence that β-cell compensation is pivotal for overriding maternal insulin resistance to protect against GDM. We will highlight key molecules whose functions are critical for integrating gestational hormones to β-cell compensation for pregnancy. We will provide mechanistic insights into β-cell decompensation in the etiology of GDM.
Collapse
Affiliation(s)
- Taofeek O Usman
- Division of Endocrinology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Goma Chhetri
- Division of Endocrinology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hsuan Yeh
- Division of Endocrinology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - H Henry Dong
- Division of Endocrinology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
17
|
Thangaraj SV, Zeng L, Pennathur S, Lea R, Sinclair KD, Bellingham M, Evans NP, Auchus R, Padmanabhan V. Developmental programming: Impact of preconceptional and gestational exposure to a real-life environmental chemical mixture on maternal steroid, cytokine and oxidative stress milieus in sheep. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 900:165674. [PMID: 37495149 PMCID: PMC10568064 DOI: 10.1016/j.scitotenv.2023.165674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Gestational exposure to environmental chemicals (ECs) is associated with adverse, sex-specific offspring health effects of global concern. As the maternal steroid, cytokine and oxidative stress milieus can have critical effects on pregnancy outcomes and the programming of diseases in offspring, it is important to study the impact of real-life EC exposure, i.e., chronic low levels of mixtures of ECs on these milieus. Sheep exposed to biosolids, derived from human waste, is an impactful model representing the ECs humans are exposed to in real-life. Offspring of sheep grazed on biosolids-treated pasture are characterized by reproductive and metabolic disruptions. OBJECTIVE To determine if biosolids exposure disrupts the maternal steroid, cytokine and oxidative stress milieus, in a fetal sex-specific manner. METHODS Ewes were maintained before mating and through gestation on pastures fertilized with biosolids (BTP), or inorganic fertilizer (Control). From maternal plasma collected mid-gestation, 19 steroids, 14 cytokines, 6 oxidative stress markers were quantified. Unpaired t-test and ANOVA were used to test for differences between control and BTP groups (n = 15/group) and between groups based on fetal sex, respectively. Correlation between the different markers was assessed by Spearman correlation. RESULTS Concentrations of the mineralocorticoids - deoxycorticosterone, corticosterone, the glucocorticoids - deoxycortisol, cortisol, cortisone, the sex steroids - androstenedione, dehydroepiandrosterone, 16-OH-progesterone and reactive oxygen metabolites were higher in the BTP ewes compared to Controls, while the proinflammatory cytokines IL-1β and IL-17A and anti-inflammatory IL-36RA were decreased in the BTP group. BTP ewes with a female fetus had lower levels of IP-10. DISCUSSION These findings suggest that pre-conceptional and gestational exposure to ECs in biosolids increases steroids, reactive oxygen metabolites and disrupts cytokines in maternal circulation, likely contributors to the aberrant phenotypic outcomes seen in offspring of BTP sheep - a translationally relevant precocial model.
Collapse
Affiliation(s)
- S V Thangaraj
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - L Zeng
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - S Pennathur
- Departments of Medicine and Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - R Lea
- Schools of Biosciences and Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - K D Sinclair
- Schools of Biosciences and Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - M Bellingham
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - N P Evans
- School of Biodiversity One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - R Auchus
- Departments of Pharmacology & Internal medicine, Division of Metabolism, Endocrinology, & Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - V Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Guo F, Mao S, Long Y, Zhou B, Gao L, Huang H. The Influences of Perinatal Androgenic Exposure on Cardiovascular and Metabolic Disease of Offspring of PCOS. Reprod Sci 2023; 30:3179-3189. [PMID: 37380913 DOI: 10.1007/s43032-023-01286-w] [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: 01/11/2023] [Accepted: 06/08/2023] [Indexed: 06/30/2023]
Abstract
Hyperandrogenism is an endocrine disorder affecting a large population of reproductive-aged women, thus proportionally high number of fetuses are subjected to prenatal androgenic exposure (PNA). The short-term stimulations at critical ontogenetic stages can wield lasting influences on the health. The most commonly diagnosed conditions in reproductive age women is polycystic ovary syndrome (PCOS). PNA may affect the growth and development of many systems in the whole body and disrupts the normal metabolic trajectory in the offspring of PCOS, contributing to the prevalence of cardiovascular and metabolic diseases (CVMD), including myocardial hypertrophy, hypertension, hyperinsulinemia, insulin resistance, hyperglycemia, obesity, and dyslipidemia, which are the leading causes of hospitalizations in young PCOS offspring. In this review, we focus on the effects of prenatal androgenic exposure on the cardiovascular and metabolic diseases in offspring, discuss the possible pathogenesis respectively, and summarize potential management strategies to improve metabolic health of PCOS offspring. It is expected that the incidence of CVMD and the medical burden will be reduced in the future.
Collapse
Affiliation(s)
- Fei Guo
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Suqing Mao
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Yuhang Long
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Bokang Zhou
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Ling Gao
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Hefeng Huang
- Department of Reproduction and Development, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.
| |
Collapse
|
19
|
Kwok J, Khanolainen DP, Speyer LG, Murray AL, Torppa MP, Auyeung B. Examining Maternal Cardiometabolic Markers in Pregnancy on Child Emotional and Behavior Trajectories: Using Growth Curve Models on a Cohort Study. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:614-622. [PMID: 37881536 PMCID: PMC10593919 DOI: 10.1016/j.bpsgos.2023.08.004] [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: 11/30/2022] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 10/27/2023] Open
Abstract
Background Poor maternal cardiometabolic health in pregnancy is associated with negative effects on child health outcomes, but there is limited literature on child and adolescent socioemotional outcomes. The study aimed to investigate associations between maternal cardiometabolic markers during pregnancy with child and adolescent socioemotional trajectories. Methods Growth curve models were run to examine how maternal cardiometabolic markers in pregnancy affected child socioemotional trajectories from ages 4 to 16. Models were adjusted for all pregnancy trimesters and maternal, child, and socioeconomic covariates. This study used the Avon Longitudinal Study of Parents and Children (United Kingdom) cohort. Participants consisted of mother-child pairs (N = 15,133). Maternal predictors of fasting glucose, triglycerides, high-density lipoprotein, low-density lipoprotein, and body mass index were taken from each pregnancy trimester (T1, T2, T3). Child outcomes included emotional problems, conduct problems, and hyperactivity problems from the Strengths and Difficulties Questionnaire. Results Fully adjusted models showed significant associations between elevated T1 fasting glucose and increased conduct problems, higher T1 body mass index and increased hyperactivity problems, lowered T1 high-density lipoprotein and decreased hyperactivity problems, and elevated T2 triglycerides and increased hyperactivity problems. Conclusions Maternal cardiometabolic risk is associated with conduct and hyperactivity outcomes from ages 4 to 16. This study suggests that maternal markers of fasting glucose, low-density lipoprotein, high-density lipoprotein, and triglycerides during pregnancy could be added as supplements for clinical measures of risk when predicting child and adolescent socioemotional trajectories.
Collapse
Affiliation(s)
- Janell Kwok
- School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | | | - Lydia G. Speyer
- School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, United Kingdom
- Department of Psychology, Lancaster University, Lancaster, United Kingdom
| | - Aja L. Murray
- School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Minna P. Torppa
- Department of Teacher Education, University of Jyväskylä, Jyväskylä, Finland
| | - Bonnie Auyeung
- School of Philosophy, Psychology and Language Sciences, The University of Edinburgh, Edinburgh, United Kingdom
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
20
|
Kolac UK, Donmez Yalcin G, Karayel R, Yalcin A. The role of protein kinase R in placental inflammation, mtUPR and apoptosis. Placenta 2023; 139:200-211. [PMID: 37463546 DOI: 10.1016/j.placenta.2023.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023]
Abstract
INTRODUCTION Placental inflammation is implicated in the pathophysiology of many pregnancy complications, including fetal growth restriction, preeclampsia, gestational diabetes, and choriocarcinoma. Mitochondrial dysfunction, one of the outcomes of placental inflammation, is characterized by loss of membrane potential, accumulation of oxygen radicals, mitochondrial protein folding defects, and disturbances in mitochondrial dynamics. Protein kinase R (PKR) is stimulated by double-stranded RNA and bacterial endotoxins in the presence of pathogens and is a critical immune response enzyme. PKR is also correlated with the cell death response during endoplasmic reticulum stress. In this study, we aim to investigate the effects of PKR activity stimulated by lipopolysaccharide (LPS), and double-stranded RNA analog (Poly I:C) on mitochondrial unfolded protein response (mtUPR), mitochondrial membrane potential, apoptosis, and oxidative stress in placental trophoblasts. METHODS We applied LPS and Poly I:C to BeWo cells to induce PKR activation. In addition, cells were treated with 2-aminopurine (2-AP) to inhibit the kinase activity of PKR. Protein levels of ATP-dependent Clp protease proteolytic subunit (CLPP) and heat shock protein 60 (HSP60) were determined after treatments. Apoptotic markers were detected by real-time PCR and flow cytometry. PKR-induced reactive oxygen radicals (ROS) accumulation and mitochondrial membrane potential change were assessed by flow cytometry. RESULTS It was determined that PKR activation-induced apoptosis in BeWo cells by reducing the levels of mtUPR proteins (CLPP and HSP60) and caused a decrease in mitochondrial membrane potential. PKR inhibition was sufficient for decreases in apoptotic markers and caused a reduction in the ratio of depolarized and ROS (+) cells. DISCUSSION Our results showed that LPS and Poly I:C administration stimulated PKR in BeWo cells in vitro. Furthermore, PKR activation is correlated with the levels of proteins involved in mitochondrial homeostasis and apoptosis. Our findings will contribute to understanding the role of PKR activation in placental inflammation and related diseases.
Collapse
Affiliation(s)
- Umut Kerem Kolac
- Department of Medical Biology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin, Turkey
| | - Gizem Donmez Yalcin
- Department of Medical Biology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin, Turkey
| | - Ramazan Karayel
- Department of Medical Biology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin, Turkey
| | - Abdullah Yalcin
- Department of Medical Biology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin, Turkey.
| |
Collapse
|
21
|
Souza MDDDC, Ferreira LB, Dos Santos LC. Dietary Inflammatory Index during pregnancy is associated with birth weight and child anthropometry up to 10 years old: A systematic review and meta-analysis. Nutr Res 2023; 114:81-97. [PMID: 37209507 DOI: 10.1016/j.nutres.2023.04.009] [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: 02/01/2023] [Revised: 04/23/2023] [Accepted: 04/23/2023] [Indexed: 05/22/2023]
Abstract
Based on previous studies, we hypothesized that the dietary inflammatory potential in pregnant women might influence maternal and child health. The objective of this work is to review the literature on the association of Dietary Inflammatory Index (DII) during pregnancy with early and late maternal and child health outcomes. We searched Cochrane, Embase, PubMed, Scopus, Web of Science, and the Virtual Health Library. Observational studies on DII in the gestational period that met the objective of this review were selected. There was a double-blind evaluation of 185 studies, of which 16 were included in narrative synthesis and 9 in meta-analysis. High methodological quality, longitudinal studies (87.5%) and the Food Frequency Questionnaire for DII evaluation (68.8%) prevailed. Outcomes studied were gestational diabetes mellitus (n = 5), gestational age at delivery (n = 7), type of delivery (n = 3), gestational weight gain or pregestational body mass index (n = 11), and anthropometry at birth (n = 8) and of the child up to age 10 years (n = 4). Higher maternal DII was associated with increased risk of small-for-gestational-age babies (odds radio, 1.15; 95% confidence interval, 1.08-1.21; I2, 29%; P = .24) and low birth weight (<2.500 g) (odds ratio, 1.16; 95% confidence interval, 1.06-1.26; I2, 56%; P = .10). The association between higher maternal DII and higher risk of obesity in late childhood is also suggestive. Thus, maternal diet may be a modifiable factor of inflammation in pregnancy associated with health outcomes of the offspring.
Collapse
Affiliation(s)
- Mariane Dias Duarte de Carvalho Souza
- Nutrition Department, Nursing School, Federal University of Minas Gerais, Professor Alfredo Balena Avenue, 190 - Santa Efigênia, Belo Horizonte - MG, Brazil.
| | - Larissa Bueno Ferreira
- Nutrition Department, Faculty of Health Sciences, University of Brasilia, Darcy Ribeiro University Campus, Asa Norte, Brasília - DF, Brazil
| | - Luana Caroline Dos Santos
- Nutrition Department, Nursing School, Federal University of Minas Gerais, Professor Alfredo Balena Avenue, 190 - Santa Efigênia, Belo Horizonte - MG, Brazil
| |
Collapse
|
22
|
Zavatta A, Parisi F, Mandò C, Scaccabarozzi C, Savasi VM, Cetin I. Role of Inflammaging on the Reproductive Function and Pregnancy. Clin Rev Allergy Immunol 2023; 64:145-160. [PMID: 35031955 PMCID: PMC8760119 DOI: 10.1007/s12016-021-08907-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2021] [Indexed: 12/14/2022]
Abstract
During female lifetime and pregnancy, inflammation and cellular senescence are implicated in physiological processes, from ovulation and menstruation, to placental homeostasis and delivery. Several lifestyles, nutritional, and environmental insults, as well as long-lasting pregestational inflammatory diseases may lead to detrimental effects in promoting and sustaining a chronic excessive inflammatory response and inflammaging, which finally contribute to the decay of fertility and pregnancy outcome, with a negative effect on placental function, fetal development, and future health risk profile in the offspring. Maladaptation to pregnancy and obstetric disease may in turn increase maternal inflammaging in a feedback loop, speeding up aging processes and outbreak of chronic diseases. Maternal inflammaging may also impact, through transgenerational effects, on future adult health. Hence, efficacious interventions should be implemented by physicians and healthcare professionals involved in prevention activities to reduce the modifiable factors contributing to the inflammaging process in order to improve public health.
Collapse
Affiliation(s)
- Alice Zavatta
- Department of Woman Mother and Neonate 'V. Buzzi' Children Hospital, ASST Fatebenefratelli Sacco, 20154, Milan, Italy
- Department of Woman Mother and Neonate 'L. Sacco' Hospital, ASST Fatebenefratelli Sacco, 20157, Milan, Italy
| | - Francesca Parisi
- Department of Woman Mother and Neonate 'V. Buzzi' Children Hospital, ASST Fatebenefratelli Sacco, 20154, Milan, Italy
| | - Chiara Mandò
- Department of Biomedical and Clinical Sciences, "Luigi Sacco", University of Milan, 20157, Milan, Italy
| | - Chiara Scaccabarozzi
- Department of Woman Mother and Neonate 'L. Sacco' Hospital, ASST Fatebenefratelli Sacco, 20157, Milan, Italy
| | - Valeria M Savasi
- Department of Biomedical and Clinical Sciences, "Luigi Sacco", University of Milan, 20157, Milan, Italy
- Department of Woman Mother and Neonate 'L. Sacco' Hospital, ASST Fatebenefratelli Sacco, 20157, Milan, Italy
| | - Irene Cetin
- Department of Woman Mother and Neonate 'V. Buzzi' Children Hospital, ASST Fatebenefratelli Sacco, 20154, Milan, Italy.
- Department of Biomedical and Clinical Sciences, "Luigi Sacco", University of Milan, 20157, Milan, Italy.
| |
Collapse
|
23
|
Naomi R, Rusli RNM, Huat TS, Embong H, Bahari H, Kamaruzzaman MA. Early Intervention of Elateriospermum tapos Yoghurt in Obese Dams Mitigates Intergenerational Cognitive Deficits and Thigmotactic Behaviour in Male Offspring via the Modulation of Metabolic Profile. Nutrients 2023; 15:nu15061523. [PMID: 36986254 PMCID: PMC10052004 DOI: 10.3390/nu15061523] [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: 02/28/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Maternal obesity is an intergenerational vicious cycle and one of the primary causes of cognitive deficits and high anxiety levels in offspring, which often manifest independently of sex. It is proven that curbing the intergenerational inheritance of obesity through early intervention during the gestation period has a positive outcome on the body composition, cognitive function, and anxiety level of the offspring. A recent discovery shows that the consumption of Elateriospermum tapos (E. tapos) seed extract modulates body mass and ameliorates stress hormones in obese dams, while a probiotic bacterial strain can cross the placenta and boost a child's memory. Thus, we speculate that probiotics are the best medium to integrate plant extract (E. tapos extract) to access the effect on the child's cognition. Thus, this study aimed to investigate the early intervention of E. tapos yoghurt in obese dams in the cognition and anxiety levels of male offspring. In this study, 40 female rats were fed with a high-fat diet (HFD) to induce obesity before pregnancy, while another 8 rats were fed with standard rat pellets for 16 weeks. Upon successful copulation, treatment was initiated for the obese dams up to the postnatal day (PND) 21. The groups included normal chow and saline (NS), HFD and saline (HS), HFD and yoghurt (HY), HFD and 5 mg/kg E. tapos yoghurt (HYT5), HFD and 50 mg/kg E. tapos yoghurt (HYT50), and HFD and 500 mg/kg E. tapos yoghurt (HYT500). All rats were euthanised on PND 21, and the body mass index (BMI), Lee index, and waist circumference were measured for the male offspring. Hippocampal-dependent memory tests and open field tests were conducted to access for cognition and anxiety status. Fasting blood glucose (FBG), total fat (%), insulin, leptin, lipid profile, and antioxidant parameter on serum and hypothalamus (FRAP and GSH) were accessed on PND 21. The result shows male offspring of 50 mg/kg-supplemented obese dams have comparable total fat (%), lipid profile, insulin level, FBG level, plasma insulin level, recognition index, low anxiety level, and improved hypothalamic FRAP and GSH levels to the normal group. In conclusion, this study highlights that the effect of early intervention of our novel formulation of E. tapos yoghurt in obese dams alleviates cognitive deficits and anxiety in male offspring by modulating metabolic profiles at the dose of 50 mg/kg.
Collapse
Affiliation(s)
- Ruth Naomi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Rusydatul Nabila Mahmad Rusli
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Teoh Soo Huat
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang 13200, Malaysia
| | - Hashim Embong
- Department of Emergency Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| | - Hasnah Bahari
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Mohd Amir Kamaruzzaman
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
24
|
Xin Y, Sun X, Ren L, Chen G, Chen Y, Ni Y, He B. Maternal preconceptional inflammation transgenerationally alters metabolic and behavioral phenotypes in offspring. Life Sci 2023; 321:121577. [PMID: 36933826 DOI: 10.1016/j.lfs.2023.121577] [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: 02/13/2023] [Revised: 03/10/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023]
Abstract
AIMS Evidence is accumulating that maternal inflammation induces phenotypic changes in the next generation. However, whether maternal preconceptional inflammation alters metabolic and behavioral phenotypes in offspring remains poorly understood. MAIN METHODS Female mice were injected with either lipopolysaccharide or saline to establish the inflammatory model and then allowed to mate with normal males. Offspring from both control and inflammatory dams were subsequently given chow diet and water ad libitum, without any challenge, for metabolic and behavioral tests. KEY FINDINGS Male offspring derived from inflammatory mothers (Inf-F1) maintained on the chow diet developed impaired glucose tolerance and hepatic ectopic fat deposition. Hepatic transcriptome sequencing showed the largest gene changes related to the metabolic pathway. Moreover, Inf-F1 mice exhibited anxiety- and depressive-like behaviors and were accompanied by higher serum corticosterone concentration and lower glucocorticoid receptor abundance in the hippocampus. SIGNIFICANCE The results expand the current knowledge of developmental programming of health and disease to include maternal preconceptional health and provide a basis for understanding metabolic and behavioral alterations in offspring linked to maternal inflammation.
Collapse
Affiliation(s)
- Yining Xin
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xiaoxiao Sun
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Li Ren
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Guo Chen
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yingqi Chen
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yingdong Ni
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Bin He
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
25
|
Relationship between large and small for gestational age and hospital readmission after postpartum discharge: a population-based, data-linkage study. Eur J Pediatr 2023; 182:2245-2252. [PMID: 36869901 DOI: 10.1007/s00431-023-04908-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/17/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023]
Abstract
This study aims to determine the association of small for gestational age (SGA) and large for gestational age (LGA) at birth with hospital readmission after postpartum discharge for up to 28 days of delivery. This is a population-based, data-linkage study using the French National Uniform Hospital Discharge Database. "Healthy" singleton term infants born between January 1st, 2017, and November 30th, 2018, in the French South region were included. SGA and LGA were defined as birth weight < 10th and > 90th percentiles, respectively, according to sex and gestational age. A multivariable regression analysis was performed. Among 67,359 included infants, 2441 (3.6%) were readmitted, and 61% of them were hospitalized within 14 days postpartum. Hospitalized infants were more likely to be LGA at birth (10.3% vs. 8.6% in non-hospitalized infants, p < 0.01); the proportion of SGA infants did not differ between both groups. Compared to appropriate birth weight for GA (AGA) infants, LGA infants were more often hospitalized for infectious diseases (57.7% vs. 51.3%, p = 0.05). After regression analysis, LGA infants had a 20% higher odds of being hospitalized than those born AGA (aOR (95%CI) = 1.21 (1.06-1.39)), while aOR (95%CI) for SGA was 1.11 (0.96-1.28). CONCLUSION In contrast to SGA, LGA was associated with hospital readmission during the first month of life. Follow-up protocols that include LGA should be evaluated. WHAT IS KNOWN • Newborns are at high risk of hospital readmission during the postpartum period. • However, the influence of appropriateness for gestational age at birth, i.e. being born small for gestational age (SGA) or large for gestational age (LGA), has been little evaluated. WHAT IS NEW • In contrast to SGA born infants, we found that infants born LGA were at high risk of hospital admission and the main cause was infectious diseases. • This population should be considered at risk of early adverse outcomes and should require attentive medical follow-up after postpartum discharge.
Collapse
|
26
|
Characterization of Maternal Circulating MicroRNAs in Obese Pregnancies and Gestational Diabetes Mellitus. Antioxidants (Basel) 2023; 12:antiox12020515. [PMID: 36830073 PMCID: PMC9952647 DOI: 10.3390/antiox12020515] [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: 01/30/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Maternal obesity (MO) is expanding worldwide, contributing to the onset of Gestational Diabetes Mellitus (GDM). MO and GDM are associated with adverse maternal and foetal outcomes, with short- and long-term complications. Growing evidence suggests that MO and GDM are characterized by epigenetic alterations contributing to the pathogenesis of metabolic diseases. In this pilot study, plasma microRNAs (miRNAs) of obese pregnant women with/without GDM were profiled at delivery. Nineteen women with spontaneous singleton pregnancies delivering by elective Caesarean section were enrolled: seven normal-weight (NW), six obese without comorbidities (OB/GDM(-)), and six obese with GDM (OB/GDM(+)). miRNA profiling with miRCURY LNA PCR Panel allowed the analysis of the 179 most expressed circulating miRNAs in humans. Data acquisition and statistics (GeneGlobe and SPSS software) and Pathway Enrichment Analysis (PEA) were performed. Data analysis highlighted patterns of significantly differentially expressed miRNAs between groups: OB/GDM(-) vs. NW: n = 4 miRNAs, OB/GDM(+) vs. NW: n = 1, and OB/GDM(+) vs. OB/GDM(-): n = 14. For each comparison, PEA revealed pathways associated with oxidative stress and inflammation, as well as with nutrients and hormones metabolism. Indeed, miRNAs analysis may help to shed light on the complex epigenetic network regulating metabolic pathways in both the mother and the foeto-placental unit. Future investigations are needed to deepen the pregnancy epigenetic landscape in MO and GDM.
Collapse
|
27
|
Saros L, Vahlberg T, Koivuniemi E, Houttu N, Niinikoski H, Tertti K, Laitinen K. Fish Oil And/Or Probiotics Intervention in Overweight/Obese Pregnant Women and Overweight Risk in 24-Month-Old Children. J Pediatr Gastroenterol Nutr 2023; 76:218-226. [PMID: 36705702 PMCID: PMC9848211 DOI: 10.1097/mpg.0000000000003659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/18/2022] [Indexed: 01/28/2023]
Abstract
OBJECTIVES To evaluate whether a fish oil and/or probiotics intervention in pregnant women with overweight or obesity would influence the tendency of their 24-month-old children to become overweight and alter their body fat percentage. METHODS Women (n = 439) were double-blindly randomized into 4 intervention groups: fish oil+placebo, probiotics+placebo, probiotics+fish oil, and placebo+placebo (fish oil: 1.9 g docosahexaenoic acid and 0.22 g eicosapentaenoic acid, probiotics: Lacticaseibacillus rhamnosus HN001 and Bifidobacterium animalis ssp. lactis 420, 1010 colony-forming units each). The intervention lasted from early pregnancy until 6 months postpartum. Children's (n = 330) growth data (height, weight, head circumference), a secondary outcome of the trial, were evaluated at birth, 3, 6, 12, and 24 months of age and compared to Finnish growth charts. Body fat percentage was measured with air displacement plethysmography (24 months). Logistic regression and general linear models were used to analyze the data. RESULTS Probiotics+placebo [weight-for-height% adj. Odds ratio (OR) = 0.36, 95% confidence interval (CI) = 0.14-0.95] and probiotics+fish oil [weight-for-age standard deviation score (SD-score) adj. OR = 0.22, 95% CI = 0.07-0.71] associated with lower overweight odds in 24-month-old children compared to placebo+placebo. Results remained essentially the same, when probiotics' main effect (combined probiotics+placebo and probiotics+fish oil) was estimated; that is, lower overweight odds (weight-for-height% adj. OR = 0.48, 95% CI = 0.25-0.95 and weight-for-age SD-score adj. OR = 0.42, 95% CI = 0.20-0.88) compared to non-probiotics. No fish oil main effect (combined fish oil+placebo and probiotics+fish oil) was seen. The intervention did not influence body fat percentage. CONCLUSIONS The administration of probiotics solely and in combination with fish oil during pregnancy to women with overweight or obesity lowered the overweight odds of their 24-month-old children.
Collapse
Affiliation(s)
- Lotta Saros
- From the Institute of Biomedicine, Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Tero Vahlberg
- the Institute of Clinical Medicine and Biostatistics, University of Turku, Turku, Finland
| | - Ella Koivuniemi
- From the Institute of Biomedicine, Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Noora Houttu
- From the Institute of Biomedicine, Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Harri Niinikoski
- From the Institute of Biomedicine, Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
- the Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Kristiina Tertti
- the Department of Obstetrics and Gynecology, Turku University Hospital and University of Turku, Turku, Finland
| | - Kirsi Laitinen
- From the Institute of Biomedicine, Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
- Functional Foods Forum, University of Turku, Turku, Finland
| |
Collapse
|
28
|
Relationships between maternal body mass index and child cognitive outcomes at 3 years of age are buffered by specific early environments in a prospective Canadian birth cohort. J Dev Orig Health Dis 2023; 14:42-52. [PMID: 35481433 DOI: 10.1017/s2040174422000228] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Fetal and child development are shaped by early life exposures, including maternal health states, nutrition and educational and home environments. We aimed to determine if suboptimal pre-pregnancy maternal body mass index (BMI; underweight, overweight, obese) would associate with poorer cognitive outcomes in children, and whether early life nutritional, educational and home environments modify these relationships. Self-reported data were obtained from mother-infant dyads from the pan-Canadian prospective Maternal-Infant Research on Environmental Chemicals cohort. Relationships between potential risk factors (pre-pregnancy maternal BMI, breastfeeding practices and Home Observation Measurement of the Environment [HOME] score) and child cognitive development at age three (Weschler's Preschool and Primary Scale of Intelligence, Third Edition scale and its subcategories) were each evaluated using analysis of variance, multivariable regression models and moderating analyses. Amongst the 528 mother-child dyads, increasing maternal pre-pregnancy BMI was negatively associated with scores for child full-scale IQ (β [95% CI]; -2.01 [-3.43, -0.59], p = 0.006), verbal composite (-1.93 [-3.33, -0.53], p = 0.007), and information scale (-0.41 [-0.70, -0.14], p = 0.003) scores. Higher maternal education level or HOME score attenuated the negative association between maternal pre-pregnancy BMI and child cognitive outcome by 30%-41% and 7%-22%, respectively, and accounted for approximately 5%-10% greater variation in male children's cognitive scores compared to females. Maternal education and higher quality home environment buffer the negative effect of elevated maternal pre-pregnancy BMI on child cognitive outcomes. Findings suggest that relationships between maternal, social and environmental factors must be considered to reveal pathways that shape risk for, and resiliency against, suboptimal cognitive outcomes in early life.
Collapse
|
29
|
Puisto R, Turta O, Rautava S, Isolauri E. Early life exposures and development of allergic disease in infants with familial risk: Results from ongoing probiotic intervention trials. Acta Paediatr 2023; 112:115-121. [PMID: 35989564 DOI: 10.1111/apa.16518] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/03/2022] [Accepted: 08/15/2022] [Indexed: 12/15/2022]
Abstract
AIM We search revision of risk determinants of the ongoing allergy epidemic. METHODS Children numbering 433 born to mothers with allergic disease or sensitisation were selected from the three ongoing probiotic intervention trials for this case-control study. Children who developed atopic eczema or food allergy, had positive skinprick test results or had been prescribed inhaled corticosteroids by the age of 2 years were identified as cases (n = 231), while children without allergic manifestations were the healthy controls (n = 202). The data on early environmental exposures were collected from prospectively documented study records. The statistical analyses were adjusted for potential confounders. RESULTS Determinants associated with the increased risk of atopic eczema were lower maternal prepregnancy BMI (aOR 0.15, 95% CI: 0.037-0.54) and maternal intrapartum antibiotic treatment (aOR 2.21, 95% CI 1.20-4.10), the latter also linked to obstructive respiratory symptoms (aOR 3.87, 95% CI 1.07-14.06). The risk of allergic sensitisation was associated with lower maternal prepegnancy BMI (aOR 0.18, 95% CI 0.43-0.79) and intrapartum antibiotic treatment (aOR 2.13, 95% CI 1.07-4.22). CONCLUSION Based on our demonstrations, interventions such as personalised diets, can be optimised for specific subgroups and definite risk periods.
Collapse
Affiliation(s)
- Reetta Puisto
- Department of Pediatrics, Faculty of Medicine, University of Turku, Turku, Finland
| | - Olli Turta
- Department of Pediatrics, Faculty of Medicine, University of Turku, Turku, Finland.,Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Samuli Rautava
- Department of Pediatrics, Faculty of Medicine, University of Turku, Turku, Finland.,Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Erika Isolauri
- Department of Pediatrics, Faculty of Medicine, University of Turku, Turku, Finland.,Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
30
|
Fernandes GM, Sasaki LMP, Jardim-Santos GP, Schulte HL, Motta F, da Silva ÂP, de Carvalho AO, Pereira YR, Alves CDO, de Araújo Júnior DA, Mendonça-Silva DL, Costa KN, de Castro MEC, Lauand L, Nery RDR, Tristão R, Kurizky PS, Nóbrega ODT, Espindola LS, de Castro LCG, Alpoim PN, Godoi LC, Dusse LMSA, Coelho-dos-Reis JGA, do Amaral LR, Gomes MDS, Bertarini PLL, Brito-de-Sousa JP, da Costa-Rocha IA, Campi-Azevedo AC, Peruhype-Magalhães V, Teixeira-Carvalho A, Zaconeta AM, Soares AADSM, Valim V, Gomes CM, de Albuquerque CP, Martins-Filho OA, da Mota LMH. Panoramic snapshot of serum soluble mediator interplay in pregnant women with convalescent COVID-19: an exploratory study. Front Immunol 2023; 14:1176898. [PMID: 37122732 PMCID: PMC10130456 DOI: 10.3389/fimmu.2023.1176898] [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: 03/01/2023] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction SARS-CoV-2 infection during pregnancy can induce changes in the maternal immune response, with effects on pregnancy outcome and offspring. This is a cross-sectional observational study designed to characterize the immunological status of pregnant women with convalescent COVID-19 at distinct pregnancy trimesters. The study focused on providing a clear snapshot of the interplay among serum soluble mediators. Methods A sample of 141 pregnant women from all prenatal periods (1st, 2nd and 3rd trimesters) comprised patients with convalescent SARS-CoV-2 infection at 3-20 weeks after symptoms onset (COVID, n=89) and a control group of pre-pandemic non-infected pregnant women (HC, n=52). Chemokine, pro-inflammatory/regulatory cytokine and growth factor levels were quantified by a high-throughput microbeads array. Results In the HC group, most serum soluble mediators progressively decreased towards the 2nd and 3rd trimesters of pregnancy, while higher chemokine, cytokine and growth factor levels were observed in the COVID patient group. Serum soluble mediator signatures and heatmap analysis pointed out that the major increase observed in the COVID group related to pro-inflammatory cytokines (IL-6, TNF-α, IL-12, IFN-γ and IL-17). A larger set of biomarkers displayed an increased COVID/HC ratio towards the 2nd (3x increase) and the 3rd (3x to 15x increase) trimesters. Integrative network analysis demonstrated that HC pregnancy evolves with decreasing connectivity between pairs of serum soluble mediators towards the 3rd trimester. Although the COVID group exhibited a similar profile, the number of connections was remarkably lower throughout the pregnancy. Meanwhile, IL-1Ra, IL-10 and GM-CSF presented a preserved number of correlations (≥5 strong correlations in HC and COVID), IL-17, FGF-basic and VEGF lost connectivity throughout the pregnancy. IL-6 and CXCL8 were included in a set of acquired attributes, named COVID-selective (≥5 strong correlations in COVID and <5 in HC) observed at the 3rd pregnancy trimester. Discussion and conclusion From an overall perspective, a pronounced increase in serum levels of soluble mediators with decreased network interplay between them demonstrated an imbalanced immune response in convalescent COVID-19 infection during pregnancy that may contribute to the management of, or indeed recovery from, late complications in the post-symptomatic phase of the SARS-CoV-2 infection in pregnant women.
Collapse
Affiliation(s)
- Geraldo Magela Fernandes
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
- *Correspondence: Geraldo Magela Fernandes, ; Olindo Assis Martins-Filho,
| | - Lizandra Moura Paravidine Sasaki
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
- Hospital Universitário de Brasília, Universidade de Brasília (UnB), Brasília, Brazil
| | | | - Heidi Luise Schulte
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
| | - Felipe Motta
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
| | | | | | | | | | | | | | | | - Maria Eduarda Canellas de Castro
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
- Hospital Universitário de Brasília, Universidade de Brasília (UnB), Brasília, Brazil
| | - Lucas Lauand
- Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
| | | | - Rosana Tristão
- Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
| | - Patricia Shu Kurizky
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
- Hospital Universitário de Brasília, Universidade de Brasília (UnB), Brasília, Brazil
| | | | - Laila Salmen Espindola
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
| | - Luiz Cláudio Gonçalves de Castro
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
- Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
| | | | - Lara Carvalho Godoi
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Laurence Rodrigues do Amaral
- Laboratório de Bioinformática e Análises Moleculares, Universidade Federal de Uberlândia, Patos de Minas, Brazil
| | - Matheus de Souza Gomes
- Laboratório de Bioinformática e Análises Moleculares, Universidade Federal de Uberlândia, Patos de Minas, Brazil
| | - Pedro Luiz Lima Bertarini
- Laboratório de Bioinformática e Análises Moleculares, Universidade Federal de Uberlândia, Patos de Minas, Brazil
| | | | | | | | | | | | | | - Alexandre Anderson de Sousa Munhoz Soares
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
- Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
| | - Valéria Valim
- Hospital Universitário Cassiano Antônio Moraes, Universidade Federal do Espírito Santo (HUCAM-UFES), Vitória, Brazil
- Programa de Pós-Graduação em Saúde Coletiva (PPGSC), Centro de Ciências Médicas, Universidade Federal do Espírito Santo, Vitória, Brazil
| | - Ciro Martins Gomes
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
- Faculdade de Medicina, Universidade de Brasília (UnB), Brasília, Brazil
- Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília (UnB), Brasília, Brazil
| | - Cleandro Pires de Albuquerque
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
- Hospital Universitário de Brasília, Universidade de Brasília (UnB), Brasília, Brazil
| | - Olindo Assis Martins-Filho
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Brazil
- *Correspondence: Geraldo Magela Fernandes, ; Olindo Assis Martins-Filho,
| | - Licia Maria Henrique da Mota
- Programa de Pós-Graduação em Ciências Médicas, Universidade de Brasília (UnB), Brasília, Brazil
- Hospital Universitário de Brasília, Universidade de Brasília (UnB), Brasília, Brazil
- Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília (UnB), Brasília, Brazil
| |
Collapse
|
31
|
Role of Adipose Tissue microRNAs in the Onset of Metabolic Diseases and Implications in the Context of the DOHaD. Cells 2022; 11:cells11233711. [PMID: 36496971 PMCID: PMC9739499 DOI: 10.3390/cells11233711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
The worldwide epidemic of obesity is associated with numerous comorbid conditions, including metabolic diseases such as insulin resistance and diabetes, in particular. The situation is likely to worsen, as the increase in obesity rates among children will probably lead to an earlier onset and more severe course for metabolic diseases. The origin of this earlier development of obesity may lie in both behavior (changes in nutrition, physical activity, etc.) and in children's history, as it appears to be at least partly programmed by the fetal/neonatal environment. The concept of the developmental origin of health and diseases (DOHaD), involving both organogenesis and epigenetic mechanisms, encompasses such programming. Epigenetic mechanisms include the action of microRNAs, which seem to play an important role in adipocyte functions. Interestingly, microRNAs seem to play a particular role in propagating local insulin resistance to other key organs, thereby inducing global insulin resistance and type 2 diabetes. This propagation involves the active secretion of exosomes containing microRNAs by adipocytes and adipose tissue-resident macrophages, as well as long-distance communication targeting the muscles and liver, for example. Circulating microRNAs may also be useful as biomarkers for the identification of populations at risk of subsequently developing obesity and metabolic diseases.
Collapse
|
32
|
Ferrari L, Iodice S, Cantone L, Solazzo G, Dioni L, Hoxha M, Vicenzi M, Mozzoni P, Bergamaschi E, Persico N, Bollati V. Extracellular vesicles and their miRNA contents counterbalance the pro-inflammatory effect of air pollution during physiological pregnancy: A focus on Syncytin-1 positive vesicles. ENVIRONMENT INTERNATIONAL 2022; 169:107502. [PMID: 36095930 DOI: 10.1016/j.envint.2022.107502] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 06/15/2023]
Abstract
The impact of exposure to respirable particulate matter (PM) during pregnancy is a growing concern, as several studies have associated increased risks of adverse pregnancy and birth outcomes, and impaired intrauterine growth with air pollution. The molecular mechanisms responsible for such effects are still under debate. Extracellular vesicles (EVs), which travel in body fluids and transfer microRNAs (miRNAs) between tissues (e.g., pulmonary environment and placenta), might play an important role in PM-induced risk. We sought to determine whether the levels of PM with aerodynamic diameters of ≤10 µm (PM10) and ≤2.5 µm (PM2.5) are associated with changes in plasmatic EV release and EV-miRNA content by investigating 518 women enrolled in the INSIDE study during the first trimester of pregnancy. In all models, we included both the 90-day averages of PM (long-term effects) and the differences between the daily estimate of PM and the 90-day average (short-term effects). Short-term PM10 and PM2.5 were associated with increased concentrations of all seven EV types that we assayed (positive for human antigen leukocyte G (HLA-G), Syncytin-1 (Sync-1), CD14, CD105, CD62e, CD61, or CD25 determinants), while long-term PM10 showed a trend towards decreased EV concentrations. Increased Sync-1 + EV levels were associated with the plasmatic decrease of sVCAM-1, but not of sICAM-1, which are circulating biomarkers of endothelial dysfunction. Thirteen EV-miRNAs were downregulated in response to long-term PM10 and PM2.5 variations, while seven were upregulated (p-value < 0.05, false discovery rate p-value (qFDR) < 0.1). Only one EV-miRNA (hsa-miR-221-3p) was downregulated after short-term variations. The identified PM-modulated EV-miRNAs exhibited putative roles in inflammation, gestational hypertension, and pre-eclampsia, as highlighted by miRNA target analysis. Our findings strongly support the hypothesis that EVs have an important role in modulating PM exposure effects during pregnancy, possibly through their miRNA cargo.
Collapse
Affiliation(s)
- Luca Ferrari
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy; Department of Preventive Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Simona Iodice
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Laura Cantone
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Giulia Solazzo
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Laura Dioni
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Mirjam Hoxha
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Marco Vicenzi
- Cardiovascular Disease Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Paola Mozzoni
- Department of Medicine and Surgery, Università degli Studi di Parma, Parma, Italy
| | - Enrico Bergamaschi
- Department of Public Health Sciences and Paediatrics, Università degli Studi di Torino, Torino, Italy
| | - Nicola Persico
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy; Department of Obstetrics and Gynecology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Bollati
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy; Department of Preventive Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
33
|
Shook LL, Fourman LT, Edlow AG. Immune Responses to SARS-CoV-2 in Pregnancy: Implications for the Health of the Next Generation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1465-1473. [PMID: 36192115 PMCID: PMC9536183 DOI: 10.4049/jimmunol.2200414] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/08/2022] [Indexed: 11/06/2022]
Abstract
Widespread SARS-CoV-2 infection among pregnant individuals has led to a generation of fetuses exposed in utero, but the long-term impact of such exposure remains unknown. Although fetal infection is rare, children born to mothers with SARS-CoV-2 infection may be at increased risk for adverse neurodevelopmental and cardiometabolic outcomes. Fetal programming effects are likely to be mediated at least in part by maternal immune activation. In this review, we discuss recent evidence regarding the effects of prenatal SARS-CoV-2 infection on the maternal, placental, and fetal immune response, as well as the implications for the long-term health of offspring. Extrapolating from what is known about the impact of maternal immune activation in other contexts (e.g., obesity, HIV, influenza), we review the potential for neurodevelopmental and cardiometabolic morbidity in offspring. Based on available data suggesting potential increased neurodevelopmental risk, we highlight the importance of establishing large cohorts to monitor offspring born to SARS-CoV-2-positive mothers for neurodevelopmental and cardiometabolic sequelae.
Collapse
Affiliation(s)
- Lydia L Shook
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA; and
| | - Lindsay T Fourman
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, MA;
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA; and
| |
Collapse
|
34
|
Wang Y, Wang K, Du M, Khandpur N, Rossato SL, Lo CH, VanEvery H, Kim DY, Zhang FF, Chavarro JE, Sun Q, Huttenhower C, Song M, Nguyen LH, Chan AT. Maternal consumption of ultra-processed foods and subsequent risk of offspring overweight or obesity: results from three prospective cohort studies. BMJ 2022; 379:e071767. [PMID: 36198411 PMCID: PMC9533299 DOI: 10.1136/bmj-2022-071767] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/18/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To assess whether maternal ultra-processed food intake during peripregnancy and during the child rearing period is associated with offspring risk of overweight or obesity during childhood and adolescence. DESIGN Population based prospective cohort study. SETTING The Nurses' Health Study II (NHSII) and the Growing Up Today Study (GUTS I and II) in the United States. PARTICIPANTS 19 958 mother-child (45% boys, aged 7-17 years at study enrollment) pairs with a median follow-up of 4 years (interquartile range 2-5 years) until age 18 or the onset of overweight or obesity, including a subsample of 2925 mother-child pairs with information on peripregnancy diet. MAIN OUTCOME MEASURES Multivariable adjusted, log binomial models with generalized estimating equations and an exchangeable correlation structure were used to account for correlations between siblings and to estimate the relative risk of offspring overweight or obesity defined by the International Obesity Task Force. RESULTS 2471 (12.4%) offspring developed overweight or obesity in the full analytic cohort. After adjusting for established maternal risk factors and offspring's ultra-processed food intake, physical activity, and sedentary time, maternal consumption of ultra-processed foods during the child rearing period was associated with overweight or obesity in offspring, with a 26% higher risk in the group with the highest maternal ultra-processed food consumption (group 5) versus the lowest consumption group (group 1; relative risk 1.26, 95% confidence interval 1.08 to 1.47, P for trend<0.001). In the subsample with information on peripregnancy diet, while rates were higher, peripregnancy ultra-processed food intake was not significantly associated with an increased risk of offspring overweight or obesity (n=845 (28.9%); group 5 v group 1: relative risk 1.17, 95% confidence interval 0.89 to 1.53, P fortrend=0.07). These associations were not modified by age, sex, birth weight, and gestational age of offspring or maternal body weight. CONCLUSIONS Maternal consumption of ultra-processed food during the child rearing period was associated with an increased risk of overweight or obesity in offspring, independent of maternal and offspring lifestyle risk factors. Further study is needed to confirm these findings and to understand the underlying biological mechanisms and environmental determinants. These data support the importance of refining dietary recommendations and the development of programs to improve nutrition for women of reproductive age to promote offspring health.
Collapse
Affiliation(s)
- Yiqing Wang
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kai Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Mengxi Du
- Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Neha Khandpur
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
- Center for Epidemiological Studies in Health and Nutrition, Faculty of Public Health, University of São Paulo, São Paulo, Brazil
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sinara Laurini Rossato
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Institute of Geography, Graduation course of Collective Health, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Chun-Han Lo
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Hannah VanEvery
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Daniel Y Kim
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Fang Fang Zhang
- Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
- Department of Public Health and Community Medicine, School of Medicine, Tufts University, Boston, MA, USA
| | - Jorge E Chavarro
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Qi Sun
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mingyang Song
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Long H Nguyen
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
35
|
Cao N, Lan C, Chen C, Xu Z, Luo H, Zheng S, Gong X, Ren H, Li Z, Qu S, Yu C, Yang J, Jose PA, Chen Y, Wu G, Hu C, Yu J, Zeng C. Prenatal Lipopolysaccharides Exposure Induces Transgenerational Inheritance of Hypertension. Circulation 2022; 146:1082-1095. [PMID: 36004643 PMCID: PMC9529859 DOI: 10.1161/circulationaha.122.059891] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/26/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND Adverse environmental exposure during the prenatal period can lead to diseases in the offspring, including hypertension. Whether or not the hypertensive phenotype can be transgenerationally transmitted is not known. METHODS Pregnant Sprague Dawley rats were intraperitoneally injected with lipopolysaccharide (LPS) on gestation days 6, 8, 10, and 12 to generate the prenatal LPS exposure model. Blood pressure was monitored by both telemetry and tail-cuff method. RNA sequencing was performed to analyze transcriptome alteration in the kidney of the third generation. Tempol and spironolactone were used to test the potential preventative and therapeutic effect of targeting reactive oxygen species and mineralocorticoid receptor signaling, respectively. Molecular biological experiments were performed to illustrate the mechanism of epigenetic and transcription regulation. RESULTS Prenatal LPS exposure can impair the ability to excrete a salt load and induce hypertension from the first to the third generations, with the fourth and fifth generations, inducing salt-sensitive hypertension. Compared with control pups, the transcriptome in the kidney of the hypertensive third-generation prenatal LPS-exposed offspring have upregulation of the Ras-related C3 botulinum toxin substrate 1 (Rac1) gene and activation of mineralocorticoid receptor signaling. Furthermore, we found that LPS exposure during pregnancy triggered oxidative stress that upregulated KDM3B (histone lysine demethylase 3B) in the oocytes of first-generation female rats, leading to an inheritable low level of H3K9me2 (histone H3 lysine 9 dimethylation), resulting in the transgenerational upregulation of Rac1. Based on these findings, we treated the LPS-exposed pregnant rats with the reactive oxygen species scavenger, tempol, which successfully prevented hypertension in the first-generation offspring and the transgenerational inheritance of hypertension. CONCLUSIONS These findings show that adverse prenatal exposure induces transgenerational hypertension through an epigenetic-regulated mechanism and identify potentially preventive and therapeutic strategies for hypertension.
Collapse
Affiliation(s)
- Nian Cao
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, P.R. China
| | - Cong Lan
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Caiyu Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Zaicheng Xu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Hao Luo
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Xue Gong
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, P.R. China
| | - Hongmei Ren
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Zhuxin Li
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Shuang Qu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Cheng Yu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Jining Yang
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, The Third Military Medical University, Chongqing, P.R. China
| | - Pedro A. Jose
- Division of Renal Diseases and Hypertension, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Yundai Chen
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, P.R. China
| | - Gengze Wu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Cuimei Hu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Junyi Yu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, The Third Military Medical University, Chongqing, P. R. China
- Heart Center of Fujian Province, Union Hospital, Fujian Medical University, Fuzhou, P.R. China
- Department of Cardiology, Chongqing General Hospital, Chongqing, P. R. China
- Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, P. R. China
| |
Collapse
|
36
|
Gurbuz O, Yorgancı A, Ozgu-Erdinc AS, Tasci Y. First trimester screening of serum advanced glycation end products levels of pregnant women who have risk factors for gestational diabetes and their obstetric outcomes: a preliminary case-control study. J OBSTET GYNAECOL 2022; 42:3048-3054. [PMID: 35653797 DOI: 10.1080/01443615.2022.2081796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Advanced glycation end-products (AGE) are complex compounds formed by nonenzymatic glycosylation of proteins, nucleic acids, and lipids with glucose in the blood. We aimed to investigate whether there was a difference in first-trimester serum AGE levels of pregnant women with and without risk factors for gestational diabetes mellitus (GDM) and their obstetric outcomes. There were 44 women in study group who have risk factors for GDM and 44 as controls. Demographic features, serum AGE levels, adverse perinatal and neonatal outcomes were compared between groups. Five patients (11.4%) in the study group and one patient (2.3%) in the control group were diagnosed as GDM (p = .2). The serum AGE values were not statistically different between the study and control groups. There were no statistical differences between groups in terms of adverse perinatal and neonatal outcomes. However, in the group with adverse perinatal outcome (n = 25), AGE values were higher than the control group. The results of our preliminary study suggested that high-risk women for GDM did not have increased serum levels of AGE in the first trimester. Nevertheless, a high first-trimester serum AGE level was found to be associated with adverse perinatal outcomes. IMPACT STATEMENTWhat is already known on this subject? Advanced glycation end products (AGE) are markers that are associated with diabetes and its complications. For pregnant women, a high third trimester serum AGEs levels were found in women who had gestational diabetes.What do the results of this study add? The results of our study revealed that first trimester screening of serum AGE levels of women who had risk factors for gestational diabetes was not discriminate. Nevertheless, a high first trimester serum AGE levels was associated with adverse perinatal outcome.What are the implications of these findings for clinical practice and/or further research? Whether reducing exogenous sources of AGE (western-style diet, smoking) before pregnancy will be associated with better pregnancy outcomes should be investigated in future studies.
Collapse
Affiliation(s)
- Ozge Gurbuz
- Clinics of Obstetrics and Gynecology, Ministry of Health, Gaziantep Şehitkamil State Hospital, Gaziantep, Turkey
| | - Ayçağ Yorgancı
- Department of Obstetrics and Gynecology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - A Seval Ozgu-Erdinc
- Department of Obstetrics and Gynecology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Yasemin Tasci
- School of Medicine, Department of Obstetrics and Gynecology, Kütahya Health Sciences University, Kütahya, Turkey
| |
Collapse
|
37
|
Muacevic A, Adler JR, Adetokunbo S, Omokhodion O, Fasokun M, Akingbule AS, Martins C, Fakorede M, Ogundipe T, Filani O. Increasing Pre-pregnancy Body Mass Index and Pregnancy Outcomes in the United States. Cureus 2022; 14:e28695. [PMID: 36196279 PMCID: PMC9525097 DOI: 10.7759/cureus.28695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2022] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION As many Americans are becoming overweight or obese, increased body mass index (BMI) is fast becoming normalized. There is a need for more research that highlights the association between pre-pregnancy obesity and adverse pregnancy outcomes. AIM To determine the association between increasing pre-pregnancy BMI and adverse pregnancy outcomes. METHODS We utilized the United States Vital Statistics records to collate data on all childbirths in the United States between 2015 and 2019. We determined the association between increasing pre-pregnancy BMI and adverse pregnancy outcomes using multivariate analysis. Neonatal outcomes measures include the five-minute Apgar score, neonatal unit admission, neonates receiving assisted ventilation > six hours, neonatal antibiotics use, and neonatal seizures. Maternal outcomes include cesarean section rate, mothers requiring blood transfusion, unplanned hysterectomy, and intensive care unit admission. In addition, we controlled for maternal parameters such as race/ethnicity, age, insurance type, and pre-existing conditions such as chronic hypertension and prediabetes. Other covariates include paternal race, age and education level, gestational diabetes mellitus, induction of labor, weight gain during pregnancy, gestational age at delivery, and delivery weight. RESULTS We studied 15,627,572 deliveries in the US Vital Statistics records between 2015 and 2019. Among these women, 3.36% were underweight, 43.19% were with a normal BMI, 26.34% were overweight, 14.73% were in the obese class I, 7.23% were in the obese class II, and 5.14% were in the obese class III. Increasing pre-pregnancy BMI was associated with significant adverse outcomes across all measures of maternal and neonatal outcomes. CONCLUSION A strong association exists between increasing pre-pregnancy BMI and adverse maternal and neonatal outcomes. The higher risk of adverse pregnancy outcomes among overweight and obese women remained even after controlling for other traditional risk factors of adverse maternal and neonatal outcomes.
Collapse
|
38
|
Berti C, Elahi S, Catalano P, Bhutta ZA, Krawinkel MB, Parisi F, Agostoni C, Cetin I, Hanson M. Obesity, Pregnancy and the Social Contract with Today's Adolescents. Nutrients 2022; 14:3550. [PMID: 36079808 PMCID: PMC9459961 DOI: 10.3390/nu14173550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 12/16/2022] Open
Abstract
Adolescent health and well-being are of great concern worldwide, and adolescents encounter particular challenges, vulnerabilities and constraints. The dual challenges of adolescent parenthood and obesity are of public health relevance because of the life-altering health and socioeconomic effects on both the parents and the offspring. Prevention and treatment strategies at the individual and population levels have not been successful in the long term, suggesting that adolescent pregnancy and obesity cannot be managed by more of the same. Here, we view adolescent obese pregnancy through the lens of the social contract with youth. The disruption of this contract is faced by today's adolescents, with work, social and economic dilemmas which perpetuate socioeconomic and health inequities across generations. The lack of employment, education and social opportunities, together with obesogenic settings, increase vulnerability and exposure to lifelong health risks, affecting their offspring's life chances too. To break such vicious circles of disadvantage and achieve sustainable solutions in real-world settings, strong efforts on the part of policymakers, healthcare providers and the community must be oriented towards guaranteeing equity and healthy nutrition and environments for today's adolescents. The involvement of adolescents themselves in developing such programs is paramount, not only so that they feel a sense of agency but also to better meet their real life needs.
Collapse
Affiliation(s)
- Cristiana Berti
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pediatric Unit, 20122 Milan, Italy
| | | | - Patrick Catalano
- Mother Infant Research Institute, Tufts University School of Medicine, Boston 02111, MA, USA
| | - Zulfiqar A. Bhutta
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Center of Excellence in Women and Child Health, The Aga Khan University, Karachi 74800, Pakistan
| | - Michael B. Krawinkel
- Institute of Nutritional Sciences—International Nutrition, Justus-Liebig-University, 35392 Giessen, Germany
| | - Francesca Parisi
- Department of Woman, Mother and Neonate, “V. Buzzi” Children Hospital, ASST Fatebenefratelli Sacco, 20154 Milan, Italy
| | - Carlo Agostoni
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pediatric Unit, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Irene Cetin
- Department of Woman, Mother and Neonate, “V. Buzzi” Children Hospital, ASST Fatebenefratelli Sacco, 20154 Milan, Italy
- Department of Biomedical and Clinical Sciences, School of Medicine, University of Milan, 20154 Milan, Italy
| | - Mark Hanson
- Institute of Developmental Sciences and NIHR Biomedical Research Centre, University of Southampton and University Hospital Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
39
|
Evsevieva M, Sergeeva O, Mazurakova A, Koklesova L, Prokhorenko-Kolomoytseva I, Shchetinin E, Birkenbihl C, Costigliola V, Kubatka P, Golubnitschaja O. Pre-pregnancy check-up of maternal vascular status and associated phenotype is crucial for the health of mother and offspring. EPMA J 2022; 13:351-366. [PMID: 36061831 PMCID: PMC9437153 DOI: 10.1007/s13167-022-00294-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/04/2022] [Indexed: 12/15/2022]
Abstract
Abstract Cardiovascular disease remains the leading cause of disease burden globally with far-reaching consequences including enormous socio-economic burden to healthcare and society at large. Cardiovascular health is decisive for reproductive function, healthy pregnancy and postpartum. During pregnancy, maternal cardiovascular system is exposed to highly increased haemodynamic stress that significantly impacts health status of the mother and offspring. Resulting from sub-optimal maternal health conditions overlooked in pre-pregnancy time, progressive abnormalities can be expected during pregnancy and postpartum. Contextually, there are two main concepts to follow in the framework of predictive, preventive and personalised medicine, namely to develop: 1. advanced screening of sub-optimal health conditions in young populations to predict and prevent individual health risks prior to planned pregnancies 2. in-depth companion diagnostics during pregnancy to predict and prevent long-lasting postpartum health risks of the mother and offspring. Data collected in the current study demonstrate group-specific complications to health of the mother and offspring and clinical relevance of the related phenotyping in pre-pregnant mothers. Diagnostic approach proposed in this study revealed its great clinical utility demonstrating important synergies between cardiovascular maladaptation and connective tissue dysfunction. Co-diagnosed pre-pregnancy low BMI of the mother, connective tissue dysfunction, increased stiffness of peripheral vessels and decreased blood pressure are considered a highly specific maternal phenotype useful for innovative screening programmes in young populations to predict and prevent severe risks to health of the mother and offspring. This crucial discovery brings together systemic effects characteristic, for example, for individuals with Flammer syndrome predisposed to the phenotype-specific primary vascular dysregulation, pregnancy-associated risks, normal tension glaucoma, ischemic stroke at young age, impaired wound healing and associated disorders. Proposed maternal phenotyping is crucial to predict and effectively protect both the mother and offspring against health-to-disease transition. Pre-pregnancy check-up focused on sub-optimal health and utilising here described phenotypes is pivotal for advanced health policy. Plain English abstract Cardiovascular health is decisive for reproductive function and healthy pregnancy. During pregnancy, maternal cardiovascular system may demonstrate health-to-disease transition relevant for the affected mother and offspring. Overlooked in pre-pregnancy time, progressive abnormalities can be expected during pregnancy and lifelong. Here we co-diagnosed maternal pre-pregnancy low bodyweight with systemic effects which may increase risks of pregnancy, eye and heart disorders and ischemic stroke at young age, amongst others. Innovative screening programmes focused on sub-optimal health in young populations to predict and to mitigate individual health risks prior to pregnancy is an essential innovation for health policy proposed.
Collapse
Affiliation(s)
- Maria Evsevieva
- Stavropol State Medical University, Stavropol, Russian Federation
| | - Oksana Sergeeva
- Stavropol State Medical University, Stavropol, Russian Federation
| | - Alena Mazurakova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | | | | | | | | | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| |
Collapse
|
40
|
Cirulli F, De Simone R, Musillo C, Ajmone-Cat MA, Berry A. Inflammatory Signatures of Maternal Obesity as Risk Factors for Neurodevelopmental Disorders: Role of Maternal Microbiota and Nutritional Intervention Strategies. Nutrients 2022; 14:nu14153150. [PMID: 35956326 PMCID: PMC9370669 DOI: 10.3390/nu14153150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/18/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Obesity is a main risk factor for the onset and the precipitation of many non-communicable diseases. This condition, which is associated with low-grade chronic systemic inflammation, is of main concern during pregnancy leading to very serious consequences for the new generations. In addition to the prominent role played by the adipose tissue, dysbiosis of the maternal gut may also sustain the obesity-related inflammatory milieu contributing to create an overall suboptimal intrauterine environment. Such a condition here generically defined as “inflamed womb” may hold long-term detrimental effects on fetal brain development, increasing the vulnerability to mental disorders. In this review, we will examine the hypothesis that maternal obesity-related gut dysbiosis and the associated inflammation might specifically target fetal brain microglia, the resident brain immune macrophages, altering neurodevelopmental trajectories in a sex-dependent fashion. We will also review some of the most promising nutritional strategies capable to prevent or counteract the effects of maternal obesity through the modulation of inflammation and oxidative stress or by targeting the maternal microbiota.
Collapse
Affiliation(s)
- Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; or
- Correspondence: (F.C.); (A.B.)
| | - Roberta De Simone
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.D.S.); (M.A.A.-C.)
| | - Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; or
- PhD Program in Behavioral Neuroscience, Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Antonietta Ajmone-Cat
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.D.S.); (M.A.A.-C.)
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; or
- Correspondence: (F.C.); (A.B.)
| |
Collapse
|
41
|
Zhang B, Gong X, Han B, Chu M, Gong C, Yang J, Chen L, Wang J, Bai Z, Zhang Y. Ambient PM 2.5 exposures and systemic inflammation in women with early pregnancy. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 829:154564. [PMID: 35302014 DOI: 10.1016/j.scitotenv.2022.154564] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/21/2022] [Accepted: 03/10/2022] [Indexed: 06/14/2023]
Abstract
The association between ambient fine particulate matter (PM2.5) and systemic inflammation in women with early pregnancy is unclear. This study estimated the effects of PM2.5 exposures on inflammatory biomarkers in women with normal early pregnancy (NEP) or clinically recognized early pregnancy loss (CREPL). Serum interleukin-1beta (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) were measured in 228 early pregnant women recruited in Tianjin, China. Maternal PM2.5 exposures at lag 0 through lag 30 before blood collection were estimated using temporally-adjusted land use regression models. Daily exposures to ambient PM10, NO2, SO2, CO and 8-hours maximum ozone were estimated using city-level concentrations. Single-day lag effects at lag 0 through lag 7 were estimated using multivariable linear regression models. Distributed lag effects and cumulative effects over the preceding seven days and 30 days were estimated using distributed lag non-linear models. Serum IL-1β (8.0% increase at lag 3), IL-6 (33.9% increase at lag 5) and TNF-α (12.7% increase at lag 5) in early pregnant women were significantly increased with an interquartile range increase in PM2.5 exposures adjusted for temporal confounders and demographic characteristics. These effects were robust in several two-pollutant models. Distributed lag effects over the preceding 30 days also showed that the three cytokines were significantly increased with PM2.5 on some lag days. Among all cumulative effects of PM2.5 on the three cytokines in all subjects or in the two groups, only IL-6 was significantly increased in CREPL women over the preceding seven days and 30 days. No significant cumulative effect of PM2.5 was observed in NEP women. In conclusion, exposure to ambient PM2.5 may induce systemic inflammation in women in the first trimester of pregnancy. Whether the PM2.5-related cumulative increase in maternal IL-6 is involved in the pathogenic mechanisms of early pregnancy loss needs to be identified in future research.
Collapse
Affiliation(s)
- Bumei Zhang
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xian Gong
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Bin Han
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China
| | - Mengyu Chu
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Chen Gong
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Junnan Yang
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Li Chen
- School of Geographic and Environmental Sciences, Tianjin Normal University, Tianjin, China
| | - Jianmei Wang
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhipeng Bai
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China; Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, USA
| | - Yujuan Zhang
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin, China; State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, China.
| |
Collapse
|
42
|
Panchal SK, John OD, Mathai ML, Brown L. Anthocyanins in Chronic Diseases: The Power of Purple. Nutrients 2022; 14:2161. [PMID: 35631301 PMCID: PMC9142943 DOI: 10.3390/nu14102161] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 02/06/2023] Open
Abstract
Anthocyanins are mainly purple-coloured phenolic compounds of plant origin that as secondary metabolites are important in plant survival. Understanding their health benefits in humans requires sourcing these unstable compounds in sufficient quantities at a reasonable cost, which has led to improved methods of extraction. Dark-coloured fruits, cereals and vegetables are current sources of these compounds. The range of potential sustainable sources is much larger and includes non-commercialised native plants from around the world and agri-waste containing anthocyanins. In the last 5 years, there have been significant advances in developing the therapeutic potential of anthocyanins in chronic human diseases. Anthocyanins exert their beneficial effects through improvements in gut microbiota, oxidative stress and inflammation, and modulation of neuropeptides such as insulin-like growth factor-1. Their health benefits in humans include reduced cognitive decline; protection of organs such as the liver, as well as the cardiovascular system, gastrointestinal tract and kidneys; improvements in bone health and obesity; and regulation of glucose and lipid metabolism. This review summarises some of the sources of anthocyanins and their mechanisms and benefits in the treatment of chronic human diseases.
Collapse
Affiliation(s)
- Sunil K. Panchal
- School of Science, Western Sydney University, Penrith, NSW 2753, Australia;
- Global Centre for Land-Based Innovation, Western Sydney University, Penrith, NSW 2753, Australia
| | - Oliver D. John
- Faculty of Science and Natural Resources, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia; or
| | - Michael L. Mathai
- Institute of Health and Sport, College of Health and Biomedicine, Victoria University, Melbourne, VIC 3021, Australia;
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
| | - Lindsay Brown
- School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia
| |
Collapse
|
43
|
Mandò C, Abati S, Anelli GM, Favero C, Serati A, Dioni L, Zambon M, Albetti B, Bollati V, Cetin I. Epigenetic Profiling in the Saliva of Obese Pregnant Women. Nutrients 2022; 14:2122. [PMID: 35631263 PMCID: PMC9146705 DOI: 10.3390/nu14102122] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 02/06/2023] Open
Abstract
Maternal obesity is associated with inflammation and oxidative stress, strongly impacting the intrauterine environment with detrimental consequences for both mother and offspring. The saliva is a non-invasive biofluid reflecting both local and systemic health status. This observational study aimed to profile the epigenetic signature in the saliva of Obese (OB) and Normal-Weight (NW) pregnant women. Sixteen NW and sixteen OB Caucasian women with singleton spontaneous pregnancies were enrolled. microRNAs were quantified by the OpenArray Platform. The promoter region methylation of Suppressor of Cytokine Signaling 3 (SOCS3) and Transforming Growth Factor Beta 1 (TGF-Beta1) was assessed by pyrosequencing. There were 754 microRNAs evaluated: 20 microRNAs resulted in being differentially expressed between OB and NW. microRNA pathway enrichment analysis showed a significant association with the TGF-Beta signaling pathway (miTALOS) and with fatty acids biosynthesis/metabolism, lysine degradation, and ECM-receptor interaction pathways (DIANA-miRPath). Both SOCS3 and TGF-Beta1 were significantly down-methylated in OB vs. NW. These results help to clarify impaired mechanisms involved in obesity and pave the way for the understanding of specific damaged pathways. The characterization of the epigenetic profile in saliva of pregnant women can represent a promising tool for the identification of obesity-related altered mechanisms and of possible biomarkers for early diagnosis and treatment of pregnancy-adverse conditions.
Collapse
Affiliation(s)
- Chiara Mandò
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (G.M.A.); (A.S.); (I.C.)
| | - Silvio Abati
- Department of Dentistry, University Vita-Salute San Raffaele, 20132 Milan, Italy;
| | - Gaia Maria Anelli
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (G.M.A.); (A.S.); (I.C.)
| | - Chiara Favero
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (C.F.); (L.D.); (B.A.); (V.B.)
| | - Anaïs Serati
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (G.M.A.); (A.S.); (I.C.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20054 Segrate, Italy
| | - Laura Dioni
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (C.F.); (L.D.); (B.A.); (V.B.)
| | - Marta Zambon
- Department of Woman, Mother and Child, Luigi Sacco and Vittore Buzzi Children Hospital, ASST Fatebenefratelli-Sacco, 20154 Milan, Italy;
| | - Benedetta Albetti
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (C.F.); (L.D.); (B.A.); (V.B.)
| | - Valentina Bollati
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy; (C.F.); (L.D.); (B.A.); (V.B.)
- Occupational Health Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Irene Cetin
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (G.M.A.); (A.S.); (I.C.)
- Department of Woman, Mother and Child, Luigi Sacco and Vittore Buzzi Children Hospital, ASST Fatebenefratelli-Sacco, 20154 Milan, Italy;
| |
Collapse
|
44
|
Hertiš Petek T, Petek T, Močnik M, Marčun Varda N. Systemic Inflammation, Oxidative Stress and Cardiovascular Health in Children and Adolescents: A Systematic Review. Antioxidants (Basel) 2022; 11:894. [PMID: 35624760 PMCID: PMC9137597 DOI: 10.3390/antiox11050894] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/16/2022] [Accepted: 04/24/2022] [Indexed: 12/02/2022] Open
Abstract
Recent studies indicate that cerebrovascular diseases and processes of atherosclerosis originate in the childhood era and are largely influenced by chronic inflammation. Some features of vascular dysfunction in adulthood may even be programmed prenatally via genetic influences and an unfavorable intrauterine milieu. Oxidative stress, defined by an imbalance between the production and generation of reactive oxygen species (ROS) in cells and tissues and the capability of an organism to scavenge these molecules via antioxidant mechanisms, has been linked to adverse cardiovascular health in adults, yet has not been systematically reviewed in the pediatric population. We performed a systematic search as per the PRISMA guidelines in PubMed/Medline and Cochrane Reviews and detected, in total, 1228 potentially eligible pediatric articles on systemic inflammation, oxidative stress, antioxidant use, cardiovascular disease and endothelial dysfunction. The abstracts and full-text manuscripts of these were screened for inclusion and exclusion criteria, and a total of 160 articles were included. The results indicate that systemic inflammation and oxidative stress influence cardiovascular health in many chronic pediatric conditions, including hypertension, obesity, diabetes mellitus types 1 and 2, chronic kidney disease, hyperlipidemia and obstructive sleep apnea. Exercise and diet may diminish ROS formation and enhance the total serum antioxidant capacity. Antioxidant supplementation may, in selected conditions, contribute to the diminution of the oxidative state and improve endothelial function; yet, in many areas, studies provide unsatisfactory results.
Collapse
Affiliation(s)
- Tjaša Hertiš Petek
- Department of Paediatrics, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia; (T.H.P.); (M.M.); (N.M.V.)
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Tadej Petek
- Department of Paediatrics, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia; (T.H.P.); (M.M.); (N.M.V.)
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Mirjam Močnik
- Department of Paediatrics, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia; (T.H.P.); (M.M.); (N.M.V.)
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Nataša Marčun Varda
- Department of Paediatrics, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia; (T.H.P.); (M.M.); (N.M.V.)
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| |
Collapse
|
45
|
Goeckenjan M, Strowitzki T. Stoffwechsel der Mutter vor und bei Schwangerschaftseintritt. GYNAKOLOGISCHE ENDOKRINOLOGIE 2022. [DOI: 10.1007/s10304-022-00440-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
46
|
Sugino KY, Mandala A, Janssen RC, Gurung S, Trammell M, Day MW, Brush RS, Papin JF, Dyer DW, Agbaga MP, Friedman JE, Castillo-Castrejon M, Jonscher KR, Myers DA. Western diet-induced shifts in the maternal microbiome are associated with altered microRNA expression in baboon placenta and fetal liver. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2022; 3:945768. [PMID: 36935840 PMCID: PMC10012127 DOI: 10.3389/fcdhc.2022.945768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Maternal consumption of a high-fat, Western-style diet (WD) disrupts the maternal/infant microbiome and contributes to developmental programming of the immune system and nonalcoholic fatty liver disease (NAFLD) in the offspring. Epigenetic changes, including non-coding miRNAs in the fetus and/or placenta may also underlie this risk. We previously showed that obese nonhuman primates fed a WD during pregnancy results in the loss of beneficial maternal gut microbes and dysregulation of cellular metabolism and mitochondrial dysfunction in the fetal liver, leading to a perturbed postnatal immune response with accelerated NAFLD in juvenile offspring. Here, we investigated associations between WD-induced maternal metabolic and microbiome changes, in the absence of obesity, and miRNA and gene expression changes in the placenta and fetal liver. After ~8-11 months of WD feeding, dams were similar in body weight but exhibited mild, systemic inflammation (elevated CRP and neutrophil count) and dyslipidemia (increased triglycerides and cholesterol) compared with dams fed a control diet. The maternal gut microbiome was mainly comprised of Lactobacillales and Clostridiales, with significantly decreased alpha diversity (P = 0.0163) in WD-fed dams but no community-wide differences (P = 0.26). At 0.9 gestation, mRNA expression of IL6 and TNF in maternal WD (mWD) exposed placentas trended higher, while increased triglycerides, expression of pro-inflammatory CCR2, and histological evidence for fibrosis were found in mWD-exposed fetal livers. In the mWD-exposed fetus, hepatic expression levels of miR-204-5p and miR-145-3p were significantly downregulated, whereas in mWD-exposed placentas, miR-182-5p and miR-183-5p were significantly decreased. Notably, miR-1285-3p expression in the liver and miR-183-5p in the placenta were significantly associated with inflammation and lipid synthesis pathway genes, respectively. Blautia and Ruminococcus were significantly associated with miR-122-5p in liver, while Coriobacteriaceae and Prevotellaceae were strongly associated with miR-1285-3p in the placenta; both miRNAs are implicated in pathways mediating postnatal growth and obesity. Our findings demonstrate that mWD shifts the maternal microbiome, lipid metabolism, and inflammation prior to obesity and are associated with epigenetic changes in the placenta and fetal liver. These changes may underlie inflammation, oxidative stress, and fibrosis patterns that drive NAFLD and metabolic disease risk in the next generation.
Collapse
Affiliation(s)
- Kameron Y. Sugino
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Ashok Mandala
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Rachel C. Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Sunam Gurung
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - MaJoi Trammell
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Michael W. Day
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Richard S. Brush
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - James F. Papin
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - David W. Dyer
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Martin-Paul Agbaga
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Marisol Castillo-Castrejon
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Karen R. Jonscher
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- CORRESPONDENCE: Karen R. Jonscher,
| | - Dean A. Myers
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
47
|
Lisboa PC, Miranda RA, Souza LL, Moura EG. Can breastfeeding affect the rest of our life? Neuropharmacology 2021; 200:108821. [PMID: 34610290 DOI: 10.1016/j.neuropharm.2021.108821] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/17/2021] [Accepted: 09/30/2021] [Indexed: 12/31/2022]
Abstract
The breastfeeding period is one of the most important critical windows in our development, since milk, our first food after birth, contains several compounds, such as macronutrients, micronutrients, antibodies, growth factors and hormones that benefit human health. Indeed, nutritional, and environmental alterations during lactation, change the composition of breast milk and induce alterations in the child's development, such as obesity, leading to the metabolic dysfunctions, cardiovascular diseases and neurobehavioral disorders. This review is based on experimental animal models, most of them in rodents, and summarizes the impact of an adequate breast milk supply in view of the developmental origins of health and disease (DOHaD) concept, which has been proposed by researchers in the areas of epidemiology and basic science from around the world. Here, experimental advances in understanding the programming during breastfeeding were compiled with the purpose of generating knowledge about the genesis of chronic noncommunicable diseases and to guide the development of public policies to deal with and prevent the problems arising from this phenomenon. This review article is part of the special issue on "Cross talk between periphery and brain".
Collapse
Affiliation(s)
- Patricia C Lisboa
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Rosiane A Miranda
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luana L Souza
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Egberto G Moura
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
48
|
Primary HSV-2 Infection in Early Pregnancy Results in Transplacental Viral Transmission and Dose-Dependent Adverse Pregnancy Outcomes in a Novel Mouse Model. Viruses 2021; 13:v13101929. [PMID: 34696359 PMCID: PMC8538385 DOI: 10.3390/v13101929] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 12/30/2022] Open
Abstract
Herpes simplex virus type 2 (HSV-2) infection affects 24 million births annually and is associated with adverse pregnancy outcomes, including neonatal herpes; however, the mechanisms underlying in utero transmission of HSV-2 are largely unknown. We examined the effects of primary HSV-2 infection during early pregnancy on gestational outcomes in a novel, clinically relevant mouse model. Pregnant C57BL/6 mice were infected intravaginally with 102–105 pfu/mL HSV-2 on gestation day (gd) 4.5. Controls were infected, nonpregnant, diestrus-staged mice and pregnant, uninfected mice. Compared to nonpregnant mice, pregnant mice were 100-fold more susceptible to HSV-2 infection. Three days post-inoculation (gd7.5), viral DNA was present in implantation sites, but pregnancy outcomes were largely unaffected by infection. Eight days post-inoculation (gd12.5), HSV-2 DNA persisted in placental tissues, resulting in inflammation and hemorrhage. Fetal and placental weights were reduced and fetal loss was observed with high viral doses. HSV-2 DNA and increased expression of pro-inflammatory mediators were detected in fetal tissues at gd12.5, signifying viral transmission and fetal infection, even with low viral doses. This mouse model shows a dose-dependent effect of primary HSV-2 infection on pregnancy outcomes and suggests that fetal loss may occur due to placental inflammation, thus providing valuable insight into in utero transmission of HSV-2.
Collapse
|
49
|
Adherence to the Mediterranean diet during pregnancy is associated with lower odds of excessive gestational weight gain and postpartum weight retention: results of the Mother-Infant Study Cohort. Br J Nutr 2021; 128:1401-1412. [PMID: 34294166 DOI: 10.1017/s0007114521002762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
During the first 1000 d of life, gestational weight gain (GWG) and postpartum weight retention (PPWR) are considered critical determinants of nutritional status. This study examined the effect of adherence to the Mediterranean diet (MD) during pregnancy on GWG and PPWR at 2 and 6 months among women in the United Arab Emirates (UAE), using data from the Mother-Infant Study Cohort. The latter is a prospective study, for which pregnant women were recruited (n 243) during their third trimester and were followed up for 18 months. Data on socio-demographic characteristics and anthropometric measurements were obtained. An eighty-six-item FFQ was used to examine dietary intake during pregnancy. Adherence to the MD was assessed using the alternate MD (aMED) and the Lebanese MD (LMD). Adherence to the MD, PPWR2 (2 months) and PPWR6 (6 months) were considered high if participants belonged to the third tertile of the respective measures. Results indicated that 57·5 % of participants had excessive GWG while 50·7 % and 45 % retained ≥ 5 kg at 2 and 6 months postpartum, respectively. After adjustment, adherence to both MD scores was associated with lower odds of excessive GWG (aMED, OR:0·41, 95 % CI:0·18, 0·93; LMD, OR:0·40, 95 % CI: 0·16, 0·98). Adherence to MD was also associated with PPWR2 (aMED: OR: 0·23, 95 % CI: 0·06, 0·88) and PPWR6 (aMED OR:0·26; 95 % CI:0·08-0·86; LMD, OR:0·32; 95 % CI: 0·1, 0·98). The findings of this study showed that adherence to the MD may reduce GWG and PPWR and, hence, underscored the importance of promoting the MD for better health of the mother and infant.
Collapse
|