1
|
Negri F, Boeri L, Pozzi E, Raffo M, Ramadani R, Birolini G, Passarelli F, Corsini C, Belladelli F, Bertini A, Matloob R, d'Arma A, Eisenberg ML, Montorsi F, Salonia A. Low testosterone in primary infertile men is associated with a history of maternal obesity during pregnancy: Findings from a cross-sectional study. Andrology 2025. [PMID: 40366358 DOI: 10.1111/andr.70054] [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: 03/11/2025] [Revised: 04/05/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Maternal obesity increases offspring obesity risk, but limited data exist on its association with reproductive hormones in infertile males. OBJECTIVES To investigate the impact of maternal obesity during pregnancy on total testosterone levels in primary infertile men. MATERIALS AND METHODS Data from 418 men seeking medical help for primary infertility were analyzed. Demographics, clinical data, comorbidities, and history of maternal obesity during pregnancy were recorded. Testicular volume was assessed with Prader's orchidometer. Serum hormones, including calculated free testosterone, and semen parameters were collected. Low testosterone and low calculated free testosterone were defined as total testosterone <3 ng/mL and calculated free testosterone <120 pg/mL. Descriptive statistics and regression models evaluated associations between maternal obesity at pregnancy and both total testosterone and calculated free testosterone levels. RESULTS Median (interquartile range) age and body mass index were 36 (33‒40) years and 24.8 (23.1‒26.8) kg/m2. Men with low testosterone (n = 69) had higher body mass index, Charlson comorbidity index score, and follicle-stimulating hormone, but lower testicular volume, sex hormone-binding globulin, sperm concentration, motility, and normal morphology compared to men without low testosterone (all p < 0.04). Similarly, men with low calculated free testosterone (n = 226) showed higher sex hormone-binding globulin, lower estradiol, sperm concentration, and motility than those with normal calculated free testosterone values (all p < 0.02). Low testosterone (7.2% vs. 2.0%, p = 0.01) and low calculated free testosterone (4.4% vs. 1.0%, p = 0.03) were more frequently reported during maternal obesity. Multivariable logistic regression analysis showed that maternal obesity at pregnancy (odds ratio: 5.9), higher body mass index (odds ratio: 1.1), and lower testicular volume (odds ratio: 0.9) were independent predictors of low testosterone (all p < 0.01), adjusting for age. Multivariable linear regression analyses identified maternal obesity at pregnancy (β = ‒4.1) and lower testicular volume (β = 1.8) as independent predictors of calculated free testosterone (all p < 0.001), after accounting for age and body mass index. CONCLUSIONS Low testosterone is a frequent characteristic in primary infertile men. Although rare, maternal obesity at pregnancy is significantly associated with reduced total testosterone and calculated free testosterone levels. Further preventive strategies and close follow-up should be considered in this specific group.
Collapse
Affiliation(s)
- Fausto Negri
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Urology, School of Medicine, Stanford University, Stanford, California, USA
| | - Luca Boeri
- Department of Urology, IRCCS Fondazione Ca' Granda, Policlinico di Milano, Milan, Italy
| | - Edoardo Pozzi
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Massimiliano Raffo
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Riccardo Ramadani
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | | | - Federica Passarelli
- Department of Urology, IRCCS Fondazione Ca' Granda, Policlinico di Milano, Milan, Italy
| | - Christian Corsini
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Federico Belladelli
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandro Bertini
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Rayan Matloob
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessia d'Arma
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Michael L Eisenberg
- Department of Urology, School of Medicine, Stanford University, Stanford, California, USA
| | - Francesco Montorsi
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea Salonia
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
2
|
McMullan A, Zwierzynski JB, Jain N, Haneline LS, Shou W, Kua KL, Hota SK, Durbin MD. Role of Maternal Obesity in Offspring Cardiovascular Development and Congenital Heart Defects. J Am Heart Assoc 2025; 14:e039684. [PMID: 40314345 DOI: 10.1161/jaha.124.039684] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/21/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Congenital heart disease is a leading cause of death in newborns, yet many of its molecular mechanisms remain unknown. Both maternal obesity and diabetes increase the risk of congenital heart disease in offspring, with recent studies suggesting these conditions may have distinct teratogenic mechanisms. The global prevalence of obesity is rising, and while maternal obesity is a known risk factor for fetal congenital heart disease, the specific mechanisms are largely unexplored. METHODS AND RESULTS We used a murine model of diet-induced maternal obesity, without diabetes, to produce dams that were overweight but had normal blood glucose levels. Embryos were generated and their developing hearts analyzed. Transcriptome analysis was performed using single-nucleus and bulk RNA sequencing. Global and phospho-enriched proteome analysis was performed using tandem mass tag-mass spectroscopy. Immunobloting and histologic evaluation were also performed. Analysis revealed disrupted oxidative phosphorylation and reactive oxygen species formation, with reduced antioxidant capacity, evidenced by downregulation of genes Sod1 and Gp4x, and disrupted Hif1a signaling. Evidence of oxidative stress, cell death signaling, and alteration in Rho GTPase and actin cytoskeleton signaling was also observed. Genes involved in cardiac morphogenesis, including Hand2, were downregulated, and fewer mature cardiomyocytes were present. Histologic analysis confirmed increased cardiac defects in embryos exposed to maternal obesity. CONCLUSIONS These findings demonstrate that maternal obesity alone can result in cardiac defects through mechanisms similar to those associated with maternal hyperglycemia. This study provides valuable insight into the role of maternal obesity, a growing and modifiable risk factor, in the development of the most common birth defect, congenital heart disease.
Collapse
Affiliation(s)
- Ashleigh McMullan
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
| | | | - Nina Jain
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
| | - Laura S Haneline
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
| | - Weinian Shou
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
| | - Kok Lim Kua
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
- Center for Diabetes and Metabolic Disease Research Indiana University School of Medicine Indianapolis IN USA
| | - Swetansu K Hota
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
| | - Matthew D Durbin
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
| |
Collapse
|
3
|
Ghaffary EM, Bjørklund G, Bhat RS, Mirmosayyeb O. Adipokines in multiple sclerosis: Immune dysregulation, neuroinflammation, and therapeutic opportunities. Autoimmun Rev 2025; 24:103825. [PMID: 40311722 DOI: 10.1016/j.autrev.2025.103825] [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: 03/16/2025] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder of the central nervous system (CNS), characterized by demyelination, neuroinflammation, and the progressive accumulation of neurologic deficits. Adipose tissue secretes predominantly the bioactive molecules, known as adipokines, which have drawn considerable attention for their roles in modulating immune and metabolic pathways in people with MS (PwMS). Dysregulated adipokines, such as resistin, leptin, and chemerin, induce pro-inflammatory T-cell polarization while deteriorating Blood-Brain Barrier (BBB) integrity. Adiponectin, by contrast, has both immunomodulatory and neuroprotective functions. The opposing functionality highlights the biomarker and the therapeutic potential of adipokines. Preclinical and translational findings have shed light on the role of adipokines in the pathophysiology of MS by influencing T-cell, glial, and BBB functions. In clinical settings, the assessment of adipokines can function as an indicator of prognosis and diagnosis via distinct patterns of expression. In addition, alterations to adipokine profiles through lifestyle changes and pharmaceutical treatment may complement established disease-modifying treatments (DMTs). This study has highlighted the multifaceted role of adipokines in MS management, while further studies exploring the role of adipokine-mediated immunometabolic regulation are suggested.
Collapse
Affiliation(s)
- Elham Moases Ghaffary
- Division of Pharmacology and Pharmaceutical Sciences, University of Missouri-Kansas City School of Pharmacy, Kansas City, MO, USA
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway.
| | - Ramesa Shafi Bhat
- Biochemistry Department, Science College of King Saud University, Riyadh, Saudi Arabia
| | - Omid Mirmosayyeb
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
4
|
Ormindean CM, Ciortea R, Măluțan AM, Bucuri CE, Diculescu DM, Iuhas CI, Porumb CG, Ormindean V, Roman MP, Nati ID, Suciu V, Hăprean AE, Mihu D. Adipokines as Potential Biomarkers in Pregnancy: A Naturalistic Study of Adipokines in Pregnant Women and Newborns. Biomolecules 2025; 15:607. [PMID: 40427500 PMCID: PMC12109043 DOI: 10.3390/biom15050607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Maternal obesity is an escalating public health concern that adversely affects pregnancy outcomes. Adipokines play a key role in regulating metabolism and fetal development, but their dynamic changes during pregnancy remain inadequately understood. Objective: This study investigates maternal and fetal adipokine variations throughout pregnancy and their associations with maternal body mass index (BMI), abdominal wall thickness, and neonatal outcomes. Methods: A prospective case-control study was conducted involving 74 pregnant women categorized by BMI. Maternal blood samples were collected at mid-pregnancy and delivery, and additional analysis of umbilical-cord blood was performed. Clinical parameters such as BMI, abdominal wall thickness, and fetal growth metrics were also recorded. Results: Adiponectin levels were significantly lower in obese pregnancies, whereas leptin and visfatin levels increased with higher maternal BMI. Umbilical-cord blood leptin levels correlated positively with maternal BMI and neonatal birth weight, while ghrelin levels were reduced in neonates born to obese mothers. Significant adipokine fluctuations were observed between mid-pregnancy and delivery. Conclusions: Maternal obesity is associated with distinct alterations in adipokine profiles. These findings highlight the potential of maternal adipokines, given their links to maternal adiposity, as predictive biomarkers for adverse pregnancy outcomes and long-term metabolic risks in offspring. Further interventional research is warranted to evaluate targeted strategies aimed at improving perinatal metabolic health.
Collapse
Affiliation(s)
| | | | - Andrei Mihai Măluțan
- 2nd Department of Obstetrics and Gynaecology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (C.M.O.); (D.M.D.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Sun H, Zhao Q, Liang X, He Y, Li Y, Yu J, Ding J, Yu C. Effects of cold environment exposure on female reproductive health and its regulatory mechanisms. Front Genet 2025; 16:1570053. [PMID: 40270542 PMCID: PMC12014596 DOI: 10.3389/fgene.2025.1570053] [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: 02/02/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
Objective To investigate the effects of cold environment exposure on female reproductive capacity and explore its potential regulatory mechanisms. Methods Female mice were subjected to cold water immersion to simulate cold environment exposure. Weight changes during cold exposure were recorded. Serum levels of anti-Müllerian hormone (AMH), estradiol (E2), follicle-stimulating hormone (FSH), and luteinizing hormone (LH) were measured using enzyme-linked immunosorbent assay (ELISA). Ovarian and uterine tissues were collected via surgical procedures, and transcriptomic sequencing was performed to explore potential regulatory mechanisms. ELISA was used to assess the levels of inflammatory cytokines, including interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-18 (IL-18), and tumor necrosis factor-alpha (TNF-α) in peritoneal fluid. Furthermore, immunohistochemistry was used to detect the expression levels of IL-1, IL-6, and IL-18 in ovarian tissues, as well as IL-6 and IL-18 in uterine tissues. Results Compared with the control group, female mice exposed to cold environments exhibited a significant increase in body weight and elevated serum levels of AMH, E2, FSH, and LH. Transcriptomic sequencing of ovarian and uterine tissues indicated that differentially expressed genes were primarily enriched in inflammation-related pathways, including the cAMP signaling pathway, cytokine-cytokine receptor interaction, and PI3K-Akt signaling pathway. Additionally, levels of inflammatory cytokines in the peritoneal fluid, including IL-1β, IL-6, IL-18, and TNF-α, were significantly elevated. Immunohistochemical analysis showed that the expression levels of IL-1, IL-6, and IL-18 were markedly increased in ovarian tissue, while IL-6 and IL-18 expression levels were significantly elevated in uterine tissue. These differences were statistically significant (P < 0.05). Conclusion Cold environment exposure may induce inflammatory responses in the uterus and ovaries, contributing to the formation of an inflammatory microenvironment in the reproductive system. This process may lead to disruptions in sex hormone levels and ultimately impair female reproductive capacity.
Collapse
Affiliation(s)
- Hongyi Sun
- Basic Medicine School, Naval Medical University, Shanghai, China
| | - Qianqian Zhao
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiaolan Liang
- Basic Medicine School, Naval Medical University, Shanghai, China
| | - Yalun He
- Department of Physiotherapy of Traditional Chinese Medicine, Beidaihe Rehabilitation and Recuperation Center, The People’s Liberation Army Joint Logistic Support Force, Qinhuangdao, China
| | - Yangshuo Li
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jin Yu
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jie Ding
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chaoqin Yu
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
6
|
Meiirmanova Z, Mukhanbetzhanov N, Jarmukhanov Z, Vinogradova E, Kozhakhmetova S, Morenko M, Duisebayeva A, Poddighe D, Kushugulova A, Kozhakhmetov S. Alterations in Gut Microbiota of Infants Born to Mothers with Obesity. Biomedicines 2025; 13:838. [PMID: 40299456 PMCID: PMC12024737 DOI: 10.3390/biomedicines13040838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/30/2025] Open
Abstract
Background: The impact of maternal obesity on offspring health remains a major and pressing issue. We investigated its impact on the development of the infant gut microbiome during the first six months of life, examining the taxonomic composition, metabolic pathways, and antibiotic resistance genes. Methods: Twenty-four mother-infant pairs were divided into maternally obese (OB, BMI > 36) and normal weight (BM) groups. Shotgun metagenomic sequencing was performed on stool samples collected at birth and at 1, 3, and 6 months. A total of 12 maternal samples and 23 infant samples (n = 35) in the obese group and 12 maternal samples and 30 infant samples (n = 42) in the control group were sequenced. The analysis included taxonomic profiling (MetaPhlAn 4), metabolic pathway analysis (HUMAnN 3), and antibiotic resistance gene screening (CARD/ABRicate). Results: The OB group showed reduced alpha diversity in the first month (p ≤ 0.01) and an increased Firmicutes/Bacteroidetes ratio, peaking at 3 months (p ≤ 0.001). The metabolic profiling revealed enhanced carbohydrate breakdown (p ≤ 0.001) in the BM group and lipid biosynthesis (p ≤ 0.0001) in the OB group pathways. Strong correlations emerged between Lactobacillales and fatty acid biosynthesis (r = 0.7, p ≤ 0.0001) and between Firmicutes and lincosamide (r = 0.8, p ≤ 0.0001). Conclusions: The infants of obese mothers had significantly altered development of the infant gut microbiome, affecting both composition and metabolic potential. These changes may have long-term health consequences and suggest potential therapeutic targets for intervention.
Collapse
Affiliation(s)
- Zarina Meiirmanova
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Ave., Block S1, Astana Z05H0P9, Kazakhstan; (Z.M.); (N.M.); (Z.J.); (E.V.); (A.D.); (A.K.)
- Department of Children’s Diseases with Courses in Allergology, Hematology and Endocrinology, NJSC “Astana Medical University”, Astana Z01G6C5, Kazakhstan;
| | - Nurislam Mukhanbetzhanov
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Ave., Block S1, Astana Z05H0P9, Kazakhstan; (Z.M.); (N.M.); (Z.J.); (E.V.); (A.D.); (A.K.)
| | - Zharkyn Jarmukhanov
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Ave., Block S1, Astana Z05H0P9, Kazakhstan; (Z.M.); (N.M.); (Z.J.); (E.V.); (A.D.); (A.K.)
| | - Elizaveta Vinogradova
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Ave., Block S1, Astana Z05H0P9, Kazakhstan; (Z.M.); (N.M.); (Z.J.); (E.V.); (A.D.); (A.K.)
- Interdisciplinary Sports Research, Center for Genetics and Life Sciences, Sirius University of Science and Technology, 1 Olympic Ave., Sirius Federal Territory 354340, Russia
| | | | - Marina Morenko
- Department of Children’s Diseases with Courses in Allergology, Hematology and Endocrinology, NJSC “Astana Medical University”, Astana Z01G6C5, Kazakhstan;
| | - Arailym Duisebayeva
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Ave., Block S1, Astana Z05H0P9, Kazakhstan; (Z.M.); (N.M.); (Z.J.); (E.V.); (A.D.); (A.K.)
- Department of Children’s Diseases with Courses in Allergology, Hematology and Endocrinology, NJSC “Astana Medical University”, Astana Z01G6C5, Kazakhstan;
- Innovative Center ArtScience, Astana Z11F5A9, Kazakhstan
| | - Dimitri Poddighe
- College of Health Sciences, VinUniversity, Gia Lam District, Hanoi 10000, Vietnam;
| | - Almagul Kushugulova
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Ave., Block S1, Astana Z05H0P9, Kazakhstan; (Z.M.); (N.M.); (Z.J.); (E.V.); (A.D.); (A.K.)
- Kazakhstan Society of Human Microbiome Researchers, Astana Z05H0P9, Kazakhstan
| | - Samat Kozhakhmetov
- Laboratory of Microbiome, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Ave., Block S1, Astana Z05H0P9, Kazakhstan; (Z.M.); (N.M.); (Z.J.); (E.V.); (A.D.); (A.K.)
- Kazakhstan Society of Human Microbiome Researchers, Astana Z05H0P9, Kazakhstan
| |
Collapse
|
7
|
Mercado-López L, Muñoz Y, Farias C, Beyer MP, Carrasco-Gutiérrez R, Caicedo-Paz AV, Dagnino-Subiabre A, Espinosa A, Valenzuela R. High-Fat Diet in Perinatal Period Promotes Liver Steatosis and Low Desaturation Capacity of Polyunsaturated Fatty Acids in Dams: A Link with Anxiety-Like Behavior in Rats. Nutrients 2025; 17:1180. [PMID: 40218938 PMCID: PMC11990584 DOI: 10.3390/nu17071180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES This study investigates the effects of a high-fat diet (HFD) during pregnancy and lactation on maternal and offspring health, focusing on behavioral, metabolic, and fatty acid composition outcomes in a rat model. METHODS Twelve female Sprague-Dawley rats were fed either a control diet, CD (n = 6), or HFD (n = 6) for 12 weeks, encompassing mating, gestation, and lactation periods (18 weeks). Anxiety-like behavior, maternal behavior, depression-like behavior, and social play were studied. Post mortem, the liver function, hepatic steatosis, and fatty acid composition (erythrocytes, liver, adipose tissue) were evaluated. In regard to desaturase enzymes (Δ-6D and Δ-5D), liver activity, protein mass, and gene expression (RT-PCR) were analyzed. Additionally, gene expression of PPAR-α, ACOX, CPT1-α, SREBP-1c, ACC, and FAS was assessed. Statistical analysis was performed using Student's t-test, mean ± SD (p < 0.05). RESULTS The HFD significantly increased maternal weight and anxiety-like behavior while reducing social interactions exclusively in male offspring (p < 0.05). It also led to a significant decrease in the synthesis and content of n-3 PUFAs in the analyzed tissues, induced hepatic steatosis, and upregulated the expression of pro-lipogenic genes in the maternal liver. CONCLUSIONS These findings suggest that long-term HFD consumption alters tissue fatty acid composition, disrupts metabolic homeostasis, and contributes to behavioral changes, increasing anxiety-like behaviors in pregnant dams and reducing social interactions in male offspring. Overall, this study provides further insight into the detrimental effects of HFD consumption during the perinatal period.
Collapse
Affiliation(s)
- Lorena Mercado-López
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (L.M.-L.); (Y.M.); (C.F.); (M.P.B.)
- Direccion de Postgrado, Facultad Medicina, Universidad Andres Bello, Santiago 8370035, Chile
| | - Yasna Muñoz
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (L.M.-L.); (Y.M.); (C.F.); (M.P.B.)
- Escuela de Nutrición y Dietética, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Camila Farias
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (L.M.-L.); (Y.M.); (C.F.); (M.P.B.)
| | - María Paz Beyer
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (L.M.-L.); (Y.M.); (C.F.); (M.P.B.)
- Escuela de Nutrición y Dietética, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Robinson Carrasco-Gutiérrez
- Laboratory of Stress Neurobiology, Interdisciplinary Centre for Health Studies (CIESAL), Institute of Physiology, Universidad de Valparaíso, Valparaíso 2360102, Chile;
| | - Angie Vanessa Caicedo-Paz
- Escuela de Agronomía, Facultad de Ciencias Agronómicas y de los Alimentos, Pontificia Universidad Católica de Valparaíso, Quillota 2260000, Chile;
| | - Alexies Dagnino-Subiabre
- Laboratory of Stress Neurobiology, Interdisciplinary Centre for Health Studies (CIESAL), Institute of Physiology, Universidad de Valparaíso, Valparaíso 2360102, Chile;
| | - Alejandra Espinosa
- Escuela de Medicina, Campus San Felipe, Universidad de Valparaíso, San Felipe 2170000, Chile;
- Department of Medical Technology, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (L.M.-L.); (Y.M.); (C.F.); (M.P.B.)
| |
Collapse
|
8
|
Zhang Q, Xie S, Zhong Q, Zhang X, Luo L, Yang Q. Bacillus subtilis-Derived Surfactin Alleviates Offspring Intestinal Inflammatory Injuries Through Breast Milk. Nutrients 2025; 17:1009. [PMID: 40290006 PMCID: PMC11945067 DOI: 10.3390/nu17061009] [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: 01/21/2025] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Enteric and diarrheal diseases pose a significant threat to infant health, highlighting the importance of immune defenses in early life, especially maternal protection, in establishing a robust gastrointestinal environment. Surfactin, a bioactive peptide from Bacillus subtilis, has immunomodulatory properties, yet its influence on offspring via maternal gut interference is not fully understood. This study examines the effects of maternal surfactin consumption on breast milk's immunological properties and its consequent effects on neonatal intestinal health. METHODS Twenty-eight gravid mice were randomly categorized into two cohorts and were given surfactin or not in drinking water from one week after conception to 21 days postpartum. Cross-fostering experiments were conducted within 12 h after birth. Pups from the surfactin-supplemented dams were fostered and nursed by the control dams, while the pups from the control dams were nursed by the surfactin-supplemented dams. RESULTS The findings show that the pups from the surfactin-supplemented dams had increased body weight, improved intestinal morphology with longer villus and deeper crypts, the upregulation of genes related to mucins and antimicrobial peptides, and an increase in IgA+ and CD3+ T cells within the intestinal mucosa. Further, the cross-fostering experiments suggested that the pups nursed by the surfactin-supplemented dams gained more weight, had less intestinal damage, less inflammation, and lower oxidative stress levels induced by Salmonella typhimurium, indicating the immunological benefits of surfactin conveyed through breast milk. Additionally, the expression of pro-inflammatory factors, including nitric oxide, TNF-α, IL-1β, IL-6, MCP-1, and ROS, induced by LPS in the macrophages was significantly inhibited with milk from the surfactin-supplemented dam (MSD) treatment. Interestingly, the MSD treatment induced a shift in macrophage polarization from pro-inflammatory (M1-like) to anti-inflammatory (M2-like), evidenced by the decreased expression of IL-12p40 and iNOS and the increased expression of CD206, TGF-β, and Arg-1. In terms of mechanism, surfactin improved the contents of the anti-inflammatory factors IL-4, IL-10, and TGF-β in the breast milk. CONCLUSIONS This research contributes to understanding how maternal interference can modulate breast milk composition, influence infant gastrointestinal development and immunity, and provide nutritional strategy insights.
Collapse
Affiliation(s)
| | | | | | | | | | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary medicine, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China; (Q.Z.); (S.X.); (Q.Z.); (X.Z.); (L.L.)
| |
Collapse
|
9
|
Albrecht M, Reitis N, Pagenkemper M, Tallarek AC, Pietras L, Koops T, Zazara DE, Giannou AD, Garcia MG. Sexual and reproductive health in overweight and obesity: Aims and visions for integrated research approaches. J Reprod Immunol 2025; 168:104454. [PMID: 39983242 DOI: 10.1016/j.jri.2025.104454] [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: 07/31/2024] [Revised: 01/03/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025]
Abstract
Overweight and obesity increasingly affect women in their reproductive phase, during family planning, pregnancy, breastfeeding and the postpartum period. Overweight and obesity are associated with impaired sexual and reproductive health, including increased rates of infertility, pregnancy complications, and reduced breastfeeding rates. Furthermore, maternal overweight and obesity are associated with long-term negative health consequences for the child, such as an increased risk for respiratory and metabolic disease. With the Junior Research Center for Reproduction: Sexual and Reproductive Health in Overweight and Obesity (SRHOO Center), we aim to address the effects of overweight and obesity on sexual and reproductive health in an interdisciplinary approach, combining the areas of obstetrics and midwifery, sexual health research, basic research in feto-maternal medicine and glycoimmunology, pediatrics, as well as endocrinology, metabolism and bariatric surgery. Combining these areas of expertise, we seek (1) to understand the effects of overweight and obesity on sexual and reproductive health in different patient groups, focusing on their specific needs in order to provide appropriate counselling and access to healthcare; (2) to improve reproductive health in different groups of overweight and obese patients; and (3) to create interdisciplinary, comprehensive scientific and clinical training regarding sexual and reproductive health in overweight and obese patients. In this short introduction to the SRHOO Center, we provide information on its structure, aims and individual projects as well as its presumed long-term implications for clinical care and public health.
Collapse
Affiliation(s)
- Marie Albrecht
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Junior Research Center for Reproduction: Sexual and Reproductive Health in Overweight and Obesity (SRHOO), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina Reitis
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Junior Research Center for Reproduction: Sexual and Reproductive Health in Overweight and Obesity (SRHOO), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute for Health Services Research in Dermatology and Nursing (IVDP), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mirja Pagenkemper
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Junior Research Center for Reproduction: Sexual and Reproductive Health in Overweight and Obesity (SRHOO), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ann-Christin Tallarek
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Junior Research Center for Reproduction: Sexual and Reproductive Health in Overweight and Obesity (SRHOO), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Pietras
- Junior Research Center for Reproduction: Sexual and Reproductive Health in Overweight and Obesity (SRHOO), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute for Sex Research, Sexual Medicine and Forensic Psychiatry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thula Koops
- Junior Research Center for Reproduction: Sexual and Reproductive Health in Overweight and Obesity (SRHOO), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute for Sex Research, Sexual Medicine and Forensic Psychiatry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dimitra E Zazara
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Junior Research Center for Reproduction: Sexual and Reproductive Health in Overweight and Obesity (SRHOO), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; University Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anastasios D Giannou
- Junior Research Center for Reproduction: Sexual and Reproductive Health in Overweight and Obesity (SRHOO), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of General and Abdominal Surgery, Asklepios Hospital Barmbek, Hamburg, Germany; Semmelweis University Budapest, Asklepios Campus Hamburg, Hamburg, Germany; General Surgery, Liver, Pancreas and Intestinal Transplantation Unit, Hospital Universitario-Fundación Favaloro, Buenos Aires, Argentina
| | - Mariana G Garcia
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Junior Research Center for Reproduction: Sexual and Reproductive Health in Overweight and Obesity (SRHOO), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Glyco-HAM, a cooperation of University of Hamburg, Technology Platform Mass Spectrometry and University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
10
|
Amberntsson A, Bärebring L, Winkvist A, Lissner L, Brantsæter AL, Erlund I, Papadopoulou E, Augustin H. Maternal vitamin D status in relation to cardiometabolic risk factors in children from the Norwegian Environmental Biobank. PLoS One 2025; 20:e0318071. [PMID: 39999040 PMCID: PMC11856322 DOI: 10.1371/journal.pone.0318071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 01/09/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Maternal 25-hydroxyvitamin D (25OHD) status has been associated with birth weight and childhood growth. Further, maternal 25OHD status may also influence cardiometabolic outcomes in childhood. This study investigated the association between maternal 25OHD concentration in pregnancy and markers of cardiometabolic risk in 7-12-year-old children. METHODS Data were obtained from the Norwegian Environmental Biobank (NEB) including 244 mother-child pairs in the Norwegian Mother, Father and Child Cohort Study (MoBa) participating in NEB part I and II. Childhood outcomes investigated were z-scores of anthropometrics, blood lipids and hormones. Associations between maternal 25OHD and individual cardiometabolic risk factors in children were assessed by linear regression, adjusted for maternal pre-pregnancy BMI, maternal education, child's sex, age and BMI, and tested for interaction with pre-pregnancy BMI. RESULTS Per 10 nmol/L increase in maternal 25OHD, childhood adiponectin z-score increased by 0.067 standard deviations (p = 0.039). There were no associations between maternal 25OHD concentration and any other cardiometabolic risk factor in childhood. CONCLUSION The results indicate that higher maternal vitamin D status during pregnancy may be related to higher childhood adiponectin z-score, but not with any other cardiometabolic risk marker. Whether adiponectin could be one pathway linking vitamin D to cardiometabolic health remains to be determined.
Collapse
Affiliation(s)
- Anna Amberntsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Linnea Bärebring
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Winkvist
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lauren Lissner
- School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anne Lise Brantsæter
- Department of Food Safety, Division of Climate and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
- Centre for Sustainable Diets, Norwegian Institute of Public Health, Oslo, Norway
| | - Iris Erlund
- Department of Government Services, Finnish Institute for Health and Welfare, Helsinki, Finland
- Institute for Nutrition and Health Research, Helsinki, Finland
| | - Eleni Papadopoulou
- Division of Health Service, Global Health Cluster, Norwegian Institute of Public Health, Oslo, Norway
| | - Hanna Augustin
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
11
|
Arenas G, Barrera MJ, Contreras-Duarte S. The Impact of Maternal Chronic Inflammatory Conditions on Breast Milk Composition: Possible Influence on Offspring Metabolic Programming. Nutrients 2025; 17:387. [PMID: 39940245 PMCID: PMC11820913 DOI: 10.3390/nu17030387] [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: 12/26/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 02/14/2025] Open
Abstract
Breastfeeding is the best way to provide newborns with crucial nutrients and produce a unique bond between mother and child. Breast milk is rich in nutritious and non-nutritive bioactive components, such as immune cells, cytokines, chemokines, immunoglobulins, hormones, fatty acids, and other constituents. Maternal effects during gestation and lactation can alter these components, influencing offspring outcomes. Chronic inflammatory maternal conditions, such as obesity, diabetes, and hypertension, impact breast milk composition. Breast milk from obese mothers exhibits changes in fat content, cytokine levels, and hormonal concentrations, potentially affecting infant growth and health. Similarly, diabetes alters the composition of breast milk, impacting immune factors and metabolic markers. Other pro-inflammatory conditions, such as dyslipidemia and metabolic syndrome, have been barely studied. Thus, maternal obesity, diabetes, and altered tension parameters have been described as modifying the composition of breast milk in its macronutrients and other important biomolecules, likely affecting the offspring's weight. This review emphasizes the impact of chronic inflammatory conditions on breast milk composition and its potential implications for offspring development through the revision of full-access original articles.
Collapse
Affiliation(s)
- Gabriela Arenas
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510602, Chile;
| | - María José Barrera
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Santiago 7510157, Chile;
| | - Susana Contreras-Duarte
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Santiago 8420524, Chile
| |
Collapse
|
12
|
Reiss JD, Yang W, Chang AL, Long JZ, Marić I, Profit J, Sylvester KG, Stevenson DK, Aghaeepour N, Shaw GM. Transgenerational associations between newborn metabolic profiles and bronchopulmonary dysplasia in neonates born to mothers with an obese phenotype. Sci Rep 2025; 15:1144. [PMID: 39774255 PMCID: PMC11707363 DOI: 10.1038/s41598-025-85252-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025] Open
Abstract
Maternal obesity increases risk for bronchopulmonary dysplasia (BPD) by up to 42%. Identifying metabolic features that may contribute to the association between maternal pre-pregnancy body mass index (BMI) and BPD is critical in defining the molecular relationship between these conditions. We investigated the association between maternal obesity and BPD using newborn screen metabolites as an explanatory variable. We hypothesized that elevated pre-pregnancy BMI compared to a normal BMI referent group, is associated with increased circulating short and long-chain acylcarnitines and subsequent development of BPD. This was a retrospective study with linkage of maternal pre-pregnancy BMI, with newborn screen metabolites obtained from the California Newborn Screening Program and further linked with neonatal outcomes. Results demonstrated elevated levels of phenylalanine and proline associated with an increased risk for BPD (OR 5.3, 95% CI 1.2-23.8 and OR 5.4, 95% CI 1.3-22.3) in the obesity group compared to the referent group. Short- and long-chain acylcarnitines demonstrated a mildly increased risk for BPD in neonates of mothers with severe obesity compared to controls. The findings suggest that specific metabolites may influence the molecular conditioning that increases susceptibility to BPD.
Collapse
Affiliation(s)
- Jonathan D Reiss
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA.
| | - Wei Yang
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA
| | - Alan L Chang
- Stanford Department of Anesthesiology, Perioperative and Pain Medicine, 300 Pasteur Drive, Stanford, CA, USA
| | - Jonathan Z Long
- Department of Pathology and Stanford, Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University School of Medicine, Stanford, CA, USA
| | - Ivana Marić
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA
| | - Jochen Profit
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA
| | - Karl G Sylvester
- Stanford Department of Surgery, Division of Pediatric Surgery, 453 Quarry Rd, Palo Alto, CA, USA
| | - David K Stevenson
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA
| | - Nima Aghaeepour
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA
- Stanford Department of Anesthesiology, Perioperative and Pain Medicine, 300 Pasteur Drive, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Gary M Shaw
- Stanford Department of Pediatrics, Division of Neonatology, 453 Quarry Rd, Palo Alto, CA, USA
| |
Collapse
|
13
|
Du Y, Benny PA, Shao Y, Schlueter RJ, Gurary A, Lum-Jones A, Lassiter CB, AlAkwaa FM, Tiirikainen M, Towner D, Ward WS, Garmire LX. Multiomics analysis of umbilical cord hematopoietic stem cells from a multiethnic cohort of Hawaii reveals the intergenerational effect of maternal prepregnancy obesity and risks for cancers. Gigascience 2025; 14:giaf039. [PMID: 40388307 PMCID: PMC12087453 DOI: 10.1093/gigascience/giaf039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/11/2025] [Accepted: 03/07/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Maternal obesity is a health concern that may predispose newborns to a high risk of medical problems later in life. To understand the intergenerational effect of maternal obesity, we hypothesized that the maternal obesity effect is mediated by epigenetic changes in the CD34+/CD38-/Lin- hematopoietic stem cells (uHSCs) in the offspring. To investigate this, we conducted a DNA methylation centric multiomics study. We measured DNA methylation and gene expression of the CD34+/CD38-/Lin- uHSCs and metabolomics of the cord blood, all from a multiethnic cohort from Kapiolani Medical Center for Women and Children in Honolulu, Hawaii (n=72, collected between 2016 and 2018). RESULTS Differential methylation analysis unveiled a global hypermethylation pattern in the maternal prepregnancy obese group (BH adjusted P < 0.05), after adjusting for major clinical confounders. KEGG pathway enrichment, WGCNA, and PPI analyses revealed that hypermethylated CpG sites were involved in critical biological processes, including cell cycle, protein synthesis, immune signaling, and lipid metabolism. Utilizing Shannon entropy on uHSCs methylation, we discerned notably higher quiescence of uHSCs impacted by maternal obesity. Additionally, the integration of multiomics data-including methylation, gene expression, and metabolomics-provided further evidence of dysfunctions in adipogenesis, erythropoietin production, cell differentiation, and DNA repair, aligning with the findings at the epigenetic level. Furthermore, we trained a random forest classifier using the CpG sites in the genes of the top pathways associated with maternal obesity, and applied it to predict cancer versus adjacent normal sample labels in 14 Cancer Genome Atlas (TCGA) cancer types. Five of 14 cancers showed balanced accuracy of 0.6 or higher: LUSC (0.87), PAAD (0.83), KIRC (0.71), KIRP (0.63) and BRCA (0.60). CONCLUSIONS This study revealed the significant correlation between prepregnancy maternal obesity and multiomics-level molecular changes in the uHSCs of offspring, particularly at the DNA methylation level. These maternal-obesity-associated epigenetic markers in uHSCs may contribute to increased risks in certain cancers of the offspring. Larger and multicenter cohort validation studies are warranted to follow up the current single-site study.
Collapse
Affiliation(s)
- Yuheng Du
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Paula A Benny
- Department of Obstetrics and Gynecology, University of Hawaii, Honolulu, HI 96826, USA
| | - Yuchen Shao
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ryan J Schlueter
- Department of Obstetrics and Gynecology, University of Hawaii, Honolulu, HI 96826, USA
| | - Alexandra Gurary
- Department of Obstetrics and Gynecology, University of Hawaii, Honolulu, HI 96826, USA
| | - Annette Lum-Jones
- University of Hawaii Cancer Center, Population Sciences of the Pacific Program-Epidemiology, Honolulu, HI 96813, USA
| | - Cameron B Lassiter
- University of Hawaii Cancer Center, Population Sciences of the Pacific Program-Epidemiology, Honolulu, HI 96813, USA
| | - Fadhl M AlAkwaa
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maarit Tiirikainen
- University of Hawaii Cancer Center, Population Sciences of the Pacific Program-Epidemiology, Honolulu, HI 96813, USA
| | - Dena Towner
- Department of Obstetrics and Gynecology, University of Hawaii, Honolulu, HI 96826, USA
| | - W Steven Ward
- Department of Obstetrics and Gynecology, University of Hawaii, Honolulu, HI 96826, USA
| | - Lana X Garmire
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
14
|
Rodrigues FDS, Jantsch J, de Farias Fraga G, Dias VS, Pereira Medeiros C, Wickert F, Schroder N, Giovernardi M, Guedes RP. Cannabidiol partially rescues behavioral, neuroinflammatory and endocannabinoid dysfunctions stemming from maternal obesity in the adult offspring. Neuropharmacology 2025; 262:110196. [PMID: 39447736 DOI: 10.1016/j.neuropharm.2024.110196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/15/2024] [Accepted: 10/19/2024] [Indexed: 10/26/2024]
Abstract
Maternal obesity is known to increase the risk of psychiatric disorders, such as anxiety, depression, schizophrenia and autism spectrum disorder in the offspring. While preventive measures are well-documented, practical approaches for addressing the damages once they are already established are limited. We have recently demonstrated the interplay between maternal obesity and treatment with cannabidiol (CBD) on neuroinflammation and peripheral metabolic disturbances during adolescence, however, it is known that both factors tend to vary throughout life. Therefore, here we investigated the potential of CBD to mitigate these alterations in the adult offspring of obese dams. Female Wistar rats were fed a cafeteria diet for 12 weeks prior to mating, and during gestation and lactation. Offspring received CBD (50 mg/kg) for 3 weeks from the 70th day of life. Behavioral tests assessed anxiety-like manifestations and social behavior, while neuroinflammatory and endocannabinoid markers were evaluated in the hypothalamus, prefrontal cortex (PFC) and hippocampus, as well as the biochemical profile in the plasma. CBD treatment attenuated maternal obesity-induced anxiety-like and social behavioral alterations, restoring exacerbated astrocytic and microglial markers in the hypothalamus, PFC and hippocampus of the offspring, as well as endocannabinoid levels in the PFC, with notable sex differences. Additionally, CBD attenuated plasma glucose and lipopolysaccharides (LPS) concentrations in females. These findings underscore the persistent influence of maternal obesity on the offspring's health, encompassing metabolic irregularities and behavioral impairments, as well as the role of the endocannabinoid system in mediating these outcomes across the lifespan.
Collapse
Affiliation(s)
- Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jeferson Jantsch
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Gabriel de Farias Fraga
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Victor Silva Dias
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Camila Pereira Medeiros
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fernanda Wickert
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Nadja Schroder
- Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Marcia Giovernardi
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Rio Grande do Sul, Brazil
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170, Rio Grande do Sul, Brazil.
| |
Collapse
|
15
|
Du Y, Benny PA, Shao Y, Schlueter RJ, Gurary A, Lum-Jones A, Lassiter CB, AlAkwaa FM, Tiirikainen M, Towner D, Ward WS, Garmire LX. Multi-omics Analysis of Umbilical Cord Hematopoietic Stem Cells from a Multi-ethnic Cohort of Hawaii Reveals the Intergenerational Effect of Maternal Pre-Pregnancy Obesity and Risk Prediction for Cancers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.27.24310936. [PMID: 39108521 PMCID: PMC11302719 DOI: 10.1101/2024.07.27.24310936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Background Maternal obesity is a health concern that may predispose newborns to a high risk of medical problems later in life. To understand the intergenerational effect of maternal obesity, we hypothesized that the maternal obesity effect is mediated by epigenetic changes in the CD34+/CD38-/Lin- hematopoietic stem cells (uHSCs) in the offspring. Towards this, we conducted a DNA methylation centric multi-omics study. We measured the DNA methylation and gene expression in the CD34+/CD38-/Lin- uHSCs and metabolomics of the cord blood, all from a multi-ethnic cohort (n=72) from Kapiolani Medical Center for Women and Children in Honolulu, Hawaii (collected between 2016 and 2018). Results Differential methylation (DM) analysis unveiled a global hypermethylation pattern in the maternal pre-pregnancy obese group (BH adjusted p<0.05), after adjusting for major clinical confounders. KEGG pathway enrichment, WGCNA, and PPI analyses revealed hypermethylated CpG sites were involved in critical biological processes, including cell cycle, protein synthesis, immune signaling, and lipid metabolism. . Utilizing Shannon entropy on uHSCs methylation, we discerned notably higher quiescence of uHSCs impacted by maternal obesity. Additionally, the integration of multi-omics data-including methylation, gene expression, and metabolomics-provided further evidence of dysfunctions in adipogenesis, erythropoietin production, cell differentiation, and DNA repair, aligning with the findings at the epigenetic level. Furthermore, we trained a random forest classifier using the CpG sites in the genes of the top pathways associated with maternal obesity, and applied it to predict cancer vs. adjacent normal labels from samples in 14 Cancer Genome Atlas (TCGA) cancer types. Five of 14 cancers showed balanced accuracy of 0.6 or higher: LUSC (0.87), PAAD (0.83), KIRC (0.71), KIRP (0.63) and BRCA (0.60). Conclusions This study revealed the significant correlation between pre-pregnancy maternal obesity and multi-omics level molecular changes in the uHSCs of offspring, particularly in DNA methylation. Moreover, these maternal obesity epigenetic markers in uHSCs may predispose offspring to higher risks in certain cancers.
Collapse
Affiliation(s)
- Yuheng Du
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI
| | - Paula A. Benny
- Department of Obstetrics and Gynecology, University of Hawaii, Honolulu, HI
| | - Yuchen Shao
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI
| | - Ryan J. Schlueter
- Department of Obstetrics and Gynecology, University of Hawaii, Honolulu, HI
| | - Alexandra Gurary
- Department of Obstetrics and Gynecology, University of Hawaii, Honolulu, HI
| | - Annette Lum-Jones
- University of Hawaii Cancer Center, Population Sciences of the Pacific Program-Epidemiology, Honolulu, HI
| | - Cameron B Lassiter
- University of Hawaii Cancer Center, Population Sciences of the Pacific Program-Epidemiology, Honolulu, HI
| | | | - Maarit Tiirikainen
- University of Hawaii Cancer Center, Population Sciences of the Pacific Program-Epidemiology, Honolulu, HI
| | - Dena Towner
- Department of Obstetrics and Gynecology, University of Hawaii, Honolulu, HI
| | - W. Steven Ward
- Department of Obstetrics and Gynecology, University of Hawaii, Honolulu, HI
| | - Lana X Garmire
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI
| |
Collapse
|
16
|
Qin X, Zhang M, Chen S, Tang Y, Cui J, Ding G. Short-chain fatty acids in fetal development and metabolism. Trends Mol Med 2024:S1471-4914(24)00329-0. [PMID: 39694776 DOI: 10.1016/j.molmed.2024.11.014] [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: 08/31/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024]
Abstract
Short-chain fatty acids (SCFAs), primarily derived from gut microbiota, play a role in regulating fetal development; however, the mechanism remains unclear. Fetal SCFAs levels depends on maternal SCFAs transported via the placenta. Metabolic stress, particularly from diabetes and obesity, can disrupt maternal SCFAs levels, impairing fetal metabolic reprogramming. Dysregulated SCFAs may negatively impact the development of the fetal cardiovascular, nervous, and immune systems, potentially contributing to adverse outcomes in adulthood. This review focuses on recent advances regarding the role of maternal SCFAs in shaping the metabolic profile of offspring, especially in the context of various maternal metabolic disorders. Given that SCFAs may influence fetal development through the placenta-embryo axis, targeted SCFAs supplementation could be a promising strategy against developmental diseases associated with intrauterine risk factors.
Collapse
Affiliation(s)
- Xueyun Qin
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Mo Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Shiting Chen
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Yunhui Tang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Jiajun Cui
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Guolian Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| |
Collapse
|
17
|
Coathup V, Ashdown HF, Carson C, Santorelli G, Quigley MA. Associations between maternal body mass index and childhood infections in UK primary care: findings from the Born in Bradford birth cohort study. Arch Dis Child 2024; 110:59-66. [PMID: 39332843 DOI: 10.1136/archdischild-2024-326951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 09/17/2024] [Indexed: 09/29/2024]
Abstract
OBJECTIVE To explore associations between maternal body mass index (BMI) in early pregnancy and childhood infections. DESIGN Birth cohort study linked to primary care records. SETTING Bradford, UK. PARTICIPANTS Live singleton births within the Born in Bradford cohort study between 2007 and 2011. EXPOSURES Maternal BMI in early pregnancy. MAIN OUTCOME MEASURES The total number of infections between birth and ~14 years of age with subgroup analysis by infection type and age. RESULTS A total of 9037 mothers and 9540 children were included in the main analysis. 45% of women were of Pakistani ethnicity and 6417 women (56%) were overweight or obese. There was an overall trend for an increasing infection rate with increasing maternal BMI. In adjusted models, only those with obesity grade 2-3 had offspring with significantly higher rates of infection during the first year of life (RR 1.12 (95% CI 1.05 to 1.20)) compared with women of healthy weight. However, by age 5 to <15 years, children born to overweight women (RR 1.09 (95% CI 1.02 to 1.16)), obese grade 1 women (RR 1.18 (95% CI 1.09 to 1.28)) or obese grade 2 women (RR 1.31 (95% CI 1.16 to 1.48)) all had significantly higher rates of infection compared with those born to healthy weight mothers. Respiratory tract and skin/soft tissue infections made up the majority of excess infections. CONCLUSIONS Maternal BMI was positively associated with rates of offspring infection in this study cohort, and suggests that we should be supporting women to achieve a healthy weight for pregnancy. Future research should investigate whether this is replicated in other populations, whether there is a causal association and the potential mechanisms and areas for intervention.
Collapse
Affiliation(s)
- Victoria Coathup
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Helen Frances Ashdown
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Claire Carson
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Gillian Santorelli
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Trust, Bradford, UK
| | - Maria A Quigley
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
18
|
Zhao D, Chai S, Yuan N, Sun J, Zhao X, Zhang X. Triglyceride-glycaemic index: Insights into predicting fetal macrosomia and its interaction with gestational diabetes mellitus: A cohort study of Chinese pregnant women. Eur J Clin Invest 2024; 54:e14300. [PMID: 39136403 DOI: 10.1111/eci.14300] [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: 02/22/2024] [Accepted: 07/28/2024] [Indexed: 11/20/2024]
Abstract
OBJECTIVE This study investigates the association between a new insulin resistance indicator, the triglyceride-glucose (TyG) index, and the risk of macrosomia. DESIGN This is a prospective cohort study. METHODS This study included 1332 women who delivered at Peking University International Hospital between October 2017 and August 2019. Participants were divided equally into three groups based on the TyG index. Logistic regression and restricted cubic spline (RCS) analyses were used to evaluate the relationship between the TyG index and macrosomia and conducted subgroup analyses. The TyG index's ability to predict macrosomia was assessed using the receiver operating characteristic (ROC) curve. RESULTS Multivariable logistic regression analysis revealed that the TyG index is an independent risk factor for macrosomia (Odds ratio [OR] 1.84, 95% confidence interval [CI] 1.02-3.30, p < .05). RCS analysis indicates that the risk of macrosomia increases with the rise of the TyG index (p for nonlinearity <.001) when the TyG index is >6.53. Subgroup analysis showed a synergistic additive interaction between the TyG index and gestational diabetes mellitus (GDM) of macrosomia. The area under the ROC curve for the predictive model was 0.733 (95% CI 0.684, 0.781), with a sensitivity of 76.4% and specificity of 66.9%. Incorporating the TyG index alongside traditional risk factors notably enhances macrosomia prediction (p < .05). CONCLUSIONS The TyG index independently predicts macrosomia, and exhibits an additive interaction with GDM in its occurrence. Integrating the TyG index with traditional risk factors improves the prediction of macrosomia. TRIAL REGISTRY Clinical trials. gov [NCT02966405].
Collapse
Affiliation(s)
- Dan Zhao
- Department of Endocrinology, Peking University International Hospital, Beijing, China
| | - Sanbao Chai
- Department of Endocrinology, Peking University International Hospital, Beijing, China
| | - Ning Yuan
- Department of Endocrinology, Peking University International Hospital, Beijing, China
| | - Jianbin Sun
- Department of Endocrinology, Peking University International Hospital, Beijing, China
| | - Xin Zhao
- Department of Endocrinology, Peking University International Hospital, Beijing, China
| | - Xiaomei Zhang
- Department of Endocrinology, Peking University International Hospital, Beijing, China
| |
Collapse
|
19
|
Barrea L, Camastra S, Garelli S, Guglielmi V, Manco M, Velluzzi F, Barazzoni R, Verde L, Muscogiuri G. Position statement of Italian Society of Obesity (SIO): Gestational Obesity. Eat Weight Disord 2024; 29:61. [PMID: 39331227 PMCID: PMC11436444 DOI: 10.1007/s40519-024-01688-y] [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: 06/02/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
PURPOSE Gestational obesity (GO) presents a multifaceted challenge to maternal and fetal health, with an escalating prevalence and far-reaching consequences extending beyond pregnancy. This perspective statement by the Italian Society of Obesity (SIO) provides current insights into the diagnosis, maternal and fetal impacts, and treatment strategies for managing this pressing condition. METHODS This article provides a comprehensive review of the maternal and fetal effects of GO and provides suggestions on strategies for management. Comprehensive review was carried out using the MEDLINE/PubMed, CINAHL, EMBASE, and Cochrane Library databases. RESULTS The diagnosis of GO primarily relies on pre-pregnancy body mass index (BMI), although standardized criteria remain contentious. Anthropometric measures and body composition assessments offer valuable insights into the metabolic implications of GO. Women with GO are predisposed to several health complications, which are attributed to mechanisms such as inflammation and insulin resistance. Offspring of women with GO face heightened risks of perinatal complications and long-term metabolic disorders, indicating intergenerational transmission of obesity-related effects. While nutritional interventions are a cornerstone of management, their efficacy in mitigating complications warrants further investigation. Additionally, while pharmacological interventions have been explored in other contexts, evidence on their safety and efficacy specifically for GO remains lacking, necessitating further investigation. CONCLUSION GO significantly impacts maternal and fetal health, contributing to both immediate and long-term complications. Effective management requires a multifaceted approach, including precise diagnostic criteria, personalized nutritional interventions, and potential pharmacological treatments. These findings underscore the need for individualized care strategies and further research to optimize outcomes for mothers and their offspring are needed. Enhanced understanding and management of GO can help mitigate its intergenerational effects, improving public health outcomes. LEVEL OF EVIDENCE Level V narrative review.
Collapse
Affiliation(s)
- Luigi Barrea
- Dipartimento Di Benessere, Nutrizione E Sport, Centro Direzionale, Università Telematica Pegaso, Via Porzio, Isola F2, 80143, Naples, Italy
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Stefania Camastra
- Department of Clinical and Experimental Medicine, University of Pisa, 56126, Pisa, Italy
| | - Silvia Garelli
- Division of Endocrinology and Diabetes Prevention and Care, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Valeria Guglielmi
- Unit of Internal Medicine and Obesity Center, Department of Systems Medicine, Policlinico Tor Vergata, University of Rome Tor Vergata, Rome, Italy
| | - Melania Manco
- Predictive and Preventive Medicine Research Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Fernanda Velluzzi
- Obesity Unit, Department of Medical Sciences and Public Health, University Hospital of Cagliari, Cagliari, Italy
| | - Rocco Barazzoni
- Department of Internal Medicine, Trieste University Hospital, Trieste, Italy
| | - Ludovica Verde
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Public Health, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Giovanna Muscogiuri
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italia.
- Cattedra Unesco "Educazione alla Salute e Allo Sviluppo Sostenibile", Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, Italia.
| |
Collapse
|
20
|
Mihai M, Vladut S, Sonia-Teodora L, Laura Mihaela S, Victoria N, Irina Elena M, Claudiu M. Correlation between Overweight, Obesity, Gestational Diabetes Mellitus, Adipokines (Adipolin and Adiponectin), and Adverse Pregnancy Outcomes: A Pilot Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1544. [PMID: 39336585 PMCID: PMC11434542 DOI: 10.3390/medicina60091544] [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: 07/14/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Background: The prevalence of overweight (OW), obesity (OB), and gestational diabetes mellitus (GDM) has been increasing worldwide in recent years. Adipolin is a new adipokine with reduced circulating levels in obesity and type 2 diabetes mellitus (T2DM). Objectives: Our prospective case-control study aimed to evaluate the maternal serum levels of adipolin and adiponectin, metabolic parameters, and anthropometric characteristics at the time of oral glucose tolerance test (OGTT) in pregnant women with a pre-pregnancy body mass index (BMI) ≥ 25 Kg/m2 and correlate them with newborn adipolin, adiponectin levels, and anthropometric characteristics of the newborns, and secondly to evaluate pregnancy outcomes. Material and Methods: After the OGTT results, we had 44 OW/OB pregnant women with GDM, 30 OW/OB pregnant women without GDM, and 92 lean healthy (LH) pregnant women. Data were analyzed by ANOVA and correlation tests, with a p-value < 0.05 considered significant. Results: We found no differences between adipolin values of the OW/OB pregnant women with GDM and the LH group (p > 0.99), OW/OB without GDM and the LH group (p = 0.56), and between OW/OB groups (p = 0.57). OW/OB pregnant women with GDM had a higher rate of gestational hypertension compared with the LH group (p < 0.0001). Newborns from OW/OB pregnant women with GDM were more frequently diagnosed with jaundice (p = 0.02), and they required more frequent admission to the neonatal intensive care unit (NICU) for treatment of respiratory distress (p = 0.01) compared with newborns from LH mothers. Conclusions: Our study revealed that the serum levels of adipolin in the second trimester among the group of OW/OB pregnant women with GDM, matched for age and BMI with OW/OB pregnant women without GDM, were not significantly different. This suggests that adipolin may not play an essential role in the occurrence of GDM in these patients. Despite good glycemic control during pregnancy, OW/OB pregnant women with GDM and their newborns tend to have more complications (gestational hypertension, jaundice, NICU admission) than LH pregnant women and their newborns, highlighting the importance of weight control before pregnancy.
Collapse
Affiliation(s)
- Muntean Mihai
- Departament of Obstetrics and Gynecology 2, University of Medicine Pharmacy Science and Technology George Emil Palade of Târgu Mureș, 540142 Târgu Mureș, Romania; (M.M.); (M.C.)
| | - Săsăran Vladut
- Departament of Obstetrics and Gynecology 2, University of Medicine Pharmacy Science and Technology George Emil Palade of Târgu Mureș, 540142 Târgu Mureș, Romania; (M.M.); (M.C.)
| | - Luca Sonia-Teodora
- Departament of Obstetrics and Gynecology 2, University of Medicine Pharmacy Science and Technology George Emil Palade of Târgu Mureș, 540142 Târgu Mureș, Romania; (M.M.); (M.C.)
| | - Suciu Laura Mihaela
- Departament of Neonatology, University of Medicine Pharmacy Science and Technology George Emil Palade of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Nyulas Victoria
- Departament of Informatics and Medical Biostatistics, University of Medicine Pharmacy Science and Technology George Emil Palade of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | | | - Mărginean Claudiu
- Departament of Obstetrics and Gynecology 2, University of Medicine Pharmacy Science and Technology George Emil Palade of Târgu Mureș, 540142 Târgu Mureș, Romania; (M.M.); (M.C.)
| |
Collapse
|
21
|
Li J, Wang M, Zhou H, Jin Z, Yin H, Yang S. The role of pyroptosis in the occurrence and development of pregnancy-related diseases. Front Immunol 2024; 15:1400977. [PMID: 39351226 PMCID: PMC11439708 DOI: 10.3389/fimmu.2024.1400977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
Pyroptosis is a form of programmed cell death that is crucial in the development of various diseases, including autoimmune diseases, atherosclerotic diseases, cancer, and pregnancy complications. In recent years, it has gained significant attention in national and international research due to its association with inflammatory immune overactivation and its involvement in pregnancy complications such as miscarriage and preeclampsia (PE). The mechanisms discussed include the canonical pyroptosis pathway of gasdermin activation and pore formation (caspase-1-dependent pyroptosis) and the non-canonical pyroptosis pathway (cysteoaspartic enzymes other than caspase-1). These pathways work on various cellular and factorial levels to influence normal pregnancy. This review aims to summarize and analyze the pyroptosis pathways associated with abnormal pregnancies and pregnancy complications. The objective is to enhance pregnancy outcomes by identifying various targets to prevent the onset of pyroptosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Shuli Yang
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin
University, Changchun, Jilin, China
| |
Collapse
|
22
|
Baker YE, Teale G, Vasilevski V, Mekonnen A, Sweet L. Obesity in women giving birth in Victoria, 2010-2019: a retrospective cohort study. Med J Aust 2024; 221:162-168. [PMID: 39010287 DOI: 10.5694/mja2.52387] [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: 05/02/2023] [Accepted: 11/29/2023] [Indexed: 07/17/2024]
Abstract
OBJECTIVE To assess the prevalence of obesity in pregnant women in Victoria, 2010-2019. STUDY DESIGN Retrospective cohort study; analysis of Victorian Perinatal Data Collection data. SETTING, PARTICIPANTS Women who gave birth in seventeen Victorian Department of Health areas (eight metropolitan, nine regional), 2010-2019. MAIN OUTCOME MEASURES Proportions of births to women with obesity (body mass index ≥ 30 kg/m2), by Department of Health area and year. RESULTS A total of 710 364 births with records that included the mothers' BMI were recorded in Victoria during 2010-2019. The proportion of births to women with obesity rose from 19.6% (95% confidence interval [CI], 19.3-19.9%) in 2010 to 21.5% (95% CI, 21.2-21.8%) in 2019; the proportion of births to women with normal weight declined from 49.0% (95% CI, 48.6-49.4%) to 46.8% (95% CI, 46.4-47.1%). In metropolitan areas, the proportion of births to women with obesity rose from 17.7% (95% CI, 17.7-17.8%) to 19.4% (95% CI, 19.3-19.4%); in regional areas, it increased from 25.0% (95% CI, 25.0-25.1%) to 29.1% (95% CI, 29.0-29.2%). The increase in prevalence of obesity was greater among women living in the lowest socio-economic standing (Index of Relative Socio-Economic Disadvantage) quintile than for those residing in the quintile of least disadvantage (adjusted rate ratio, 2.16; 95% CI, 2.12-2.20). CONCLUSION The proportion of births to Victorian women with obesity rose during 2010-2019, particularly in regional areas. Ensuring that regional health services are adequately resourced to meet the needs of the increasing number of women at risk of obesity during pregnancy is vital.
Collapse
Affiliation(s)
| | | | - Vidanka Vasilevski
- Centre for Quality and Patient Safety Research, Institute for Health Transformation, Deakin University, Melbourne, VIC
| | | | - Linda Sweet
- Centre for Quality and Patient Safety Research, Institute for Health Transformation, Deakin University, Melbourne, VIC
| |
Collapse
|
23
|
Leonard KM, Schmiedecke SS, Talley RL, Damicis JR, Walton RB, Burd I, Napolitano PG, Ieronimakis N. Maternal obesity alters fetal neuroinflammation in a murine model of preterm birth. AJOG GLOBAL REPORTS 2024; 4:100361. [PMID: 39072339 PMCID: PMC11278798 DOI: 10.1016/j.xagr.2024.100361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Preterm birth from intrauterine infection is a leading cause of neonatal neurologic morbidity. Likewise, maternal obesity is associated with intra-amniotic infection and inflammation. Whether maternal obesity is a risk factor for fetal brain injury that occurs with premature birth remains unknown. This study hypothesized that maternal obesity intensifies fetal neuroinflammation in the setting of premature delivery. OBJECTIVE This study aimed to examine the influence of maternal obesity on perinatal neuroinflammatory responses that arise with preterm birth using a murine model. STUDY DESIGN Dams with obesity were generated via a high-fat diet that was maintained throughout pregnancy. In parallel, dams without obesity (normal) received a control diet. All dams were paired with males on normal diet. Pregnant dams were randomized to receive an intrauterine administration of bacterial endotoxin (lipopolysaccharide) or the vehicle (phosphate-buffered saline) on embryo day 15.5 of what is typically a 19- to 21-day gestation. Fetal brains were harvested 6 hours after intrauterine administrations, and the expressions of key inflammatory cytokines (Il1b, Il6, and Tnf) and panels of metabolic, immune, and inflammatory genes were analyzed. RESULTS With the phosphate-buffered saline, there was no difference in gene expression related to maternal obesity. There were substantial differences in Il6 and immune/inflammatory expression profiles in fetal brains from dams with obesity vs normal dams that received lipopolysaccharide. Few differences were observed among the metabolic genes examined under these conditions. The gene expression pattern associated with maternal obesity correlated with pathways related to white matter injury. CONCLUSION The expression of neuroinflammatory markers instigated by bacterial endotoxin via intrauterine lipopolysaccharide was greater in embryo brains obtained from dams with obesity. Expression profiles suggest that in combination with intrauterine inflammation, maternal obesity may increase the risk of fetal white matter injury. Further investigation is warranted to understand the relationship between maternal health and neurologic outcomes associated with prematurity.
Collapse
Affiliation(s)
- Katherine M. Leonard
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Madigan Army Medical Center, Tacoma, WA (Leonard, Schmiedecke, Walton, and Ieronimakis)
| | - Stacey S. Schmiedecke
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Madigan Army Medical Center, Tacoma, WA (Leonard, Schmiedecke, Walton, and Ieronimakis)
| | - Rebecca L. Talley
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA (Talley, Damicis, and Ieronimakis)
| | - Jennifer R. Damicis
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA (Talley, Damicis, and Ieronimakis)
| | - Robert B. Walton
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Madigan Army Medical Center, Tacoma, WA (Leonard, Schmiedecke, Walton, and Ieronimakis)
| | - Irina Burd
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland, Baltimore, MD (Burd)
| | - Peter G. Napolitano
- Department of Obstetrics and Gynecology, University of Washington Medical Center, Seattle, WA (Napolitano)
| | - Nicholas Ieronimakis
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Madigan Army Medical Center, Tacoma, WA (Leonard, Schmiedecke, Walton, and Ieronimakis)
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA (Talley, Damicis, and Ieronimakis)
| |
Collapse
|
24
|
Mealy G, Brennan K, Killeen SL, Kilbane M, Yelverton C, Saldova R, Groeger D, VanSinderen D, Cotter PD, Doyle SL, McAuliffe FM. Impact of previous pregnancy and BMI on cellular and serum immune activity from early to late pregnancy. Sci Rep 2024; 14:16055. [PMID: 38992196 PMCID: PMC11239859 DOI: 10.1038/s41598-024-66651-4] [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: 01/26/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024] Open
Abstract
Immunological adaptions during pregnancy play a crucial role in healthy fetal development. Aberrant immune modifications however contribute to adverse pregnancy outcomes, which may be driven by maternal factors such as previous pregnancies and BMI. This secondary analysis of the MicrobeMom2 RCT investigates the changes to maternal inflammatory biomarkers derived from serum and stimulated peripheral blood mononuclear cells (PBMCs) during pregnancy, and the effects of previous pregnancies (parity) and BMI on maternal immune responses. Changes in immune and metabolic biomarkers from early (11-15 weeks' gestation) to late (28-32 weeks' gestation) pregnancy were compared using paired t-tests. Participants were then split by parity (nulliparous, parous) and BMI (BMI < 25, BMI > = 25), and the relationship between parity and BMI with immune biomarker levels was examined using independent t-tests, paired t-tests, ANCOVA, and linear regression. Equivalent non-parametric tests were used for skewed data. Recruited women (n = 72) were on average 31.17 (SD ± 4.53) years of age and 25.11 (SD ± 3.82) BMI (kg/m2). Of these, 51 (70.8%) had a previous term pregnancy. Throughout gestation, PBMC cytokines displayed contrasting trends to serum, with a dampening of immune responses noted in PBMCs, and enhanced production of cytokines observed in the serum. Significant decreases in PBMC derived TNF-α, IL-10 and IFN-γ were seen from early to late pregnancy. Serum C3, IL-17A, IL-6, TNF-α, CD163, GDF-15 and leptin increased throughout gestation. First pregnancy was associated with higher levels of leptin in late pregnancy, while parous women showed significant decreases in PBMC derived TNF-α, IL10, and IFN-γ with gestation. Differences in levels of C3, IL-17A, TNF-α, GDF-15 and leptin were observed across BMI groups. Overall, serum-derived cytokines exhibit contrasting levels to those derived from stimulated PBMCs. Maternal immune responses undergo significant changes from early to late pregnancy, which are influenced by parity and BMI. These differences aid our understanding as to why first-time mothers are at greater risk of placental disease such as pre-eclampsia and fetal growth restriction.
Collapse
Affiliation(s)
- Grace Mealy
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin 2, Ireland
| | - Kiva Brennan
- Department of Clinical Medicine, Trinity College Institute of Neuroscience, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Sarah Louise Killeen
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin 2, Ireland
| | - Mark Kilbane
- Department of Clinical Chemistry, St Vincent's University Hospital, Dublin, Ireland
| | - Cara Yelverton
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin 2, Ireland
| | - Radka Saldova
- The National Institute for Bioprocessing, Research, and Training (NIBRT), Dublin, Ireland
- UCD School of Medicine, College of Health and Agricultural Science (CHAS), University College Dublin (UCD), Dublin, Ireland
| | - David Groeger
- PrecisionBiotics Group Ltd (Novozymes), Cork Airport Business Park, Kinsale Road, Cork, Ireland
| | - Douwe VanSinderen
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Paul D Cotter
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Moorepark, Teagasc Food Research Centre, Fermoy, Cork, Ireland
| | - Sarah L Doyle
- Department of Clinical Medicine, Trinity College Institute of Neuroscience, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Fionnuala M McAuliffe
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin 2, Ireland.
| |
Collapse
|
25
|
Ropa J, Van't Hof W. The fulfilled promise and unmet potential of umbilical cord blood. Curr Opin Hematol 2024; 31:168-174. [PMID: 38602152 DOI: 10.1097/moh.0000000000000817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
PURPOSE OF REVIEW Here, we review classic and emerging uses of umbilical cord blood and highlight strategies to improve its utility, focusing on selection of the appropriate units and cell types for the intended applications. RECENT LITERATURE Recent studies have shown advancements in cord blood cell utility in a variety of cellular therapies and have made strides in elucidating manners to select the best units for therapy and target new ways to improve the various cell subpopulations for their respective applications. SUMMARY Umbilical cord blood is a proven source of cells for hematopoietic cell transplantation and research and is an important potential source for additional cellular therapies. However, cord blood utility is limited by low "doses" of potent cells that can be obtained from individual units, a limitation that is specific to cord blood as a donor source. In addition to traditional CD34 + progenitor cells, cord blood lymphocytes are being pursued as therapeutic entities with their own unique properties and characteristics. Thus, selection of ideal units depends on the intended therapeutic entity and target, and identification of differential potency parameters is critical to drive effective banking strategies accommodating successful clinical use of cord blood in broader cell therapy settings.
Collapse
Affiliation(s)
- James Ropa
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana
| | | |
Collapse
|
26
|
Harris RA, Crandell J, Taylor JY, Santos HP. Childhood Racism and Cardiometabolic Risk in Latina Mothers Across the First Postpartum Year. Psychosom Med 2024; 86:531-540. [PMID: 38573031 PMCID: PMC11230847 DOI: 10.1097/psy.0000000000001306] [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] [Indexed: 04/05/2024]
Abstract
OBJECTIVE Immigrant Latinas, particularly of Mexican descent, initially achieve healthy perinatal outcomes. Although this advantage wears off across generations in the United States (US), the early life psychosocial mechanisms that may initiate a cascade of biological vulnerabilities remain elusive. The current investigation aimed to understand the extent to which childhood experiences of racism may contribute to elevated levels of C-reactive protein (CRP), an early indicator of cardiometabolic risk, during the first postpartum year. METHODS Latinas from the Community and Child Health Network ( N = 457) retrospectively reported experiences of childhood racism and childhood country of residence via structured questionnaires. Interviewers collected CRP bloodspots and height and weight measurements for body mass index at 6 months and 1 year postpartum. RESULTS Latinas who grew up in the US experienced a steeper increase of CRP levels across the first postpartum year ( β = 0.131, p = .009) and had higher CRP levels 1 year postpartum than Latinas who grew up in Latin America. Based on Bayesian path analyses, Latinas who grew up in the US reported higher levels of childhood racism than Latinas who immigrated after childhood ( β = 0.27; 95% credible interval = 0.16-0.37). In turn, childhood racism mediated the relationship between country of childhood residence and elevated CRP at 6 months and 1 year postpartum, even after adjusting for sociodemographic and behavioral covariates. After adjusting for body mass index, mediational relationships became nonsignificant. CONCLUSIONS This study is an important first step toward understanding how childhood racism may contribute to postmigratory health patterns among Latinas, particularly cardiometabolic risk 1 year after childbirth.
Collapse
Affiliation(s)
- Rebeca Alvarado Harris
- School of Nursing, University of North Carolina at Chapel Hill, North Carolina, United States
| | - Jamie Crandell
- Department of Biostatistics, University of North Carolina at Chapel Hill, North Carolina, United States
| | - Jacquelyn Y. Taylor
- Center for Research on People of Color, Columbia University School of Nursing, New York, United States
| | - Hudson P Santos
- The University of Miami School of Nursing and Health Studies, Florida, United States
| |
Collapse
|
27
|
Pang W, Zhang B, Zhang J, Chen T, Han Q, Yang Z. Effects of maternal advanced lipoxidation end products diet on the glycolipid metabolism and gut microbiota in offspring mice. Front Nutr 2024; 11:1421848. [PMID: 38962449 PMCID: PMC11220281 DOI: 10.3389/fnut.2024.1421848] [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: 04/23/2024] [Accepted: 05/30/2024] [Indexed: 07/05/2024] Open
Abstract
Introduction Dietary advanced lipoxidation end products (ALEs), which are abundant in heat-processed foods, could induce lipid metabolism disorders. However, limited studies have examined the relationship between maternal ALEs diet and offspring health. Methods To investigate the transgenerational effects of ALEs, a cross-generation mouse model was developed. The C57BL/6J mice were fed with dietary ALEs during preconception, pregnancy and lactation. Then, the changes of glycolipid metabolism and gut microbiota of the offspring mice were analyzed. Results Maternal ALEs diet not only affected the metabolic homeostasis of dams, but also induced hepatic glycolipid accumulation, abnormal liver function, and disturbance of metabolism parameters in offspring. Furthermore, maternal ALEs diet significantly upregulated the expression of TLR4, TRIF and TNF-α proteins through the AMPK/mTOR/PPARα signaling pathway, leading to dysfunctional glycolipid metabolism in offspring. In addition, 16S rRNA analysis showed that maternal ALEs diet was capable of altered microbiota composition of offspring, and increased the Firmicutes/Bacteroidetes ratio. Discussion This study has for the first time demonstrated the transgenerational effects of maternal ALEs diet on the glycolipid metabolism and gut microbiota in offspring mice, and may help to better understand the adverse effects of dietary ALEs.
Collapse
Affiliation(s)
- Wenwen Pang
- School of Medicine, Nankai University, Tianjin, China
| | - Bowei Zhang
- School of Medicine, Nankai University, Tianjin, China
| | - Junshi Zhang
- Department of Hematology, Oncology Center, Tianjin Union Medical Center, Nankai University, Tianjin, China
| | - Tianyi Chen
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiurong Han
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhen Yang
- Department of Clinical Laboratory, Tianjin Union Medical Center, Nankai University, Tianjin, China
| |
Collapse
|
28
|
Chawla S, Laptook AR, Smith EA, Tan S, Natarajan G, Wyckoff MH, Greenberg RG, Ambalavanan N, Bell EF, Van Meurs KP, Hintz SR, Vohr BR, Werner EF, Das A, Shankaran S. Association of maternal pre-pregnancy or first trimester body mass index with neurodevelopmental impairment or death in extremely low gestational age neonates. J Perinatol 2024; 44:802-810. [PMID: 38396053 PMCID: PMC11742262 DOI: 10.1038/s41372-024-01905-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 01/19/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024]
Abstract
OBJECTIVE To compare the rates of death or survival with severe neurodevelopmental impairment (sNDI) at 2 years among extremely preterm infants in relation to pre-pregnancy or first-trimester maternal body mass index (BMI). METHODS This retrospective cohort study included extremely preterm infants (gestational age 220/7-266/7 weeks). The study was conducted at National Institute of Child Health and Human Development Neonatal Research Network sites. The primary outcome was death or sNDI at 2 years. RESULTS Data on the primary outcome were available for 1208 children. Death or sNDI was not different among the three groups: 54.9% in normal, 56.1% in overweight, and 53.4% in obese group (p = 0.39). There was no significant difference in mortality, sNDI, moderate/severe cerebral palsy, Bayley Scales of Infant Development (BSID)-III cognitive composite score <70, BSID-III language composite score <70 in adjusted models. CONCLUSION Neurodevelopmental outcome was not significantly associated with maternal pre-pregnancy BMI among extreme preterm infants.
Collapse
Affiliation(s)
- Sanjay Chawla
- Department of Pediatrics, Central Michigan University, Wayne State University, Children's Hospital of Michigan, Detroit, MI, USA.
| | - Abbot R Laptook
- Department of Pediatrics, Women and Infants Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | | | | | - Girija Natarajan
- Department of Pediatrics, Central Michigan University, Wayne State University, Children's Hospital of Michigan, Detroit, MI, USA
| | - Myra H Wyckoff
- Department of Pediatrics, UT Southwestern Medical Center at Dallas, Dallas, TX, USA
| | | | | | - Edward F Bell
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Krisa P Van Meurs
- Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Susan R Hintz
- Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Betty R Vohr
- Department of Pediatrics, Women and Infants Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Erika F Werner
- Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, MA, USA
| | - Abhik Das
- Social, Statistical, and Environmental Sciences Unit, RTI International, Rockville, MA, USA
| | | |
Collapse
|
29
|
Lackovic M, Nikolic D, Milicic B, Dimitrijevic D, Jovanovic I, Radosavljevic S, Mihajlovic S. Pre-Pregnancy Obesity and Infants' Motor Development within the First Twelve Months of Life: Who Is Expected to Be the Ultimate Carrier of the Obesity Burden? Nutrients 2024; 16:1260. [PMID: 38732507 PMCID: PMC11085635 DOI: 10.3390/nu16091260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/28/2024] [Accepted: 04/11/2024] [Indexed: 05/13/2024] Open
Abstract
INTRODUCTION Pre-pregnancy obesity is a significant public health concern with profound implications for maternal and child health. The burgeoning evidence suggests that maternal obesity prior to conception is intricately linked with an increased risk of gestational complications, as well as with adverse neonatal outcomes. Furthermore, the long and short-term health of offspring, including the risk of early motor development impairment, obesity, and metabolic syndrome in childhood and adulthood, may be adversely affected as well. Addressing pre-pregnancy obesity is critical for improving overall maternal and child health outcomes, and therefore, the aim of this study was to evaluate the connections linking pre-pregnancy obesity with infants' motor development within the first twelve months of infants' lives. MATERIAL AND METHODS This study included 200 mother-infant pairs divided into two groups based on their pre-pregnancy body mass index values. To assess infants' early motor development, we used the Alberta Infant Motor Scale (AIMS) and evaluated the parameters of infants' early motor development at the ages of three, six, nine, and twelve months. RESULTS Pre-pregnancy overweight/obesity was significantly associated with excessive gestational weight gain (p < 0.001), fetal macrosomia (p = 0.022), and a family history of diabetes and cardiovascular diseases (p = 0.048 and p = 0.041, respectively), as well as with all observed parameters of early motor development at the ages of three, six, nine, and twelve months: AIMS 3 months total (p < 0.001), AIMS 6 months total (p < 0.001), AIMS 9 months total (p < 0.001), and AIMS 12 months total (p < 0.001). Furthermore, pre-pregnancy overweight/obesity was a significant predictor for AIMS 6 months total (p = 0.043) and AIMS 6 months supination (p = 0.017). CONCLUSIONS Pre-pregnancy obesity is a critical determinant of pregnancy outcomes and offspring early motor development, with possible far-reaching implications for children's long-term well-being. Addressing this issue requires a comprehensive approach that includes preconception weight management, targeted interventions during the pregnancy and postpartum periods, and ongoing research to better understand the underlying mechanisms and develop effective strategies for prevention and management.
Collapse
Affiliation(s)
- Milan Lackovic
- Department of Obstetrics and Gynecology, University Hospital “Dragisa Misovic”, Heroja Milana Tepica 1, 11000 Belgrade, Serbia;
| | - Dejan Nikolic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (D.N.); (D.D.)
- Department of Physical Medicine and Rehabilitation, University Children’s Hospital, 11000 Belgrade, Serbia
| | - Biljana Milicic
- Department of Medical Statistics, Faculty of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Dejan Dimitrijevic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (D.N.); (D.D.)
- Clinic for Gynecology and Obstetrics “Narodni Front”, 11000 Belgrade, Serbia;
| | - Ivona Jovanovic
- Clinic for Gynecology and Obstetrics “Narodni Front”, 11000 Belgrade, Serbia;
| | - Sofija Radosavljevic
- Department of Radiology, University Hospital “Dragisa Misovic”, Heroja Milana Tepica 1, 11000 Belgrade, Serbia
| | - Sladjana Mihajlovic
- Department of Obstetrics and Gynecology, University Hospital “Dragisa Misovic”, Heroja Milana Tepica 1, 11000 Belgrade, Serbia;
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (D.N.); (D.D.)
| |
Collapse
|
30
|
Shang Y, Wang X, Su S, Ji F, Shao D, Duan C, Chen T, Liang C, Zhang D, Lu H. Identifying of immune-associated genes for assessing the obesity-associated risk to the offspring in maternal obesity: A bioinformatics and machine learning. CNS Neurosci Ther 2024; 30:e14700. [PMID: 38544384 PMCID: PMC10973700 DOI: 10.1111/cns.14700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND Perinatal exposure to maternal obesity predisposes offspring to develop obesity later in life. Immune dysregulation in the hypothalamus, the brain center governing energy homeostasis, is pivotal in obesity development. This study aimed to identify key candidate genes associated with the risk of offspring obesity in maternal obesity. METHODS We obtained obesity-related datasets from the Gene Expression Omnibus (GEO) database. GSE135830 comprises gene expression data from the hypothalamus of mouse offspring in a maternal obesity model induced by a high-fat diet model (maternal high-fat diet (mHFD) group and maternal chow (mChow) group), while GSE127056 consists of hypothalamus microarray data from young adult mice with obesity (high-fat diet (HFD) and Chow groups). We identified differentially expressed genes (DEGs) and module genes using Limma and weighted gene co-expression network analysis (WGCNA), conducted functional enrichment analysis, and employed a machine learning algorithm (least absolute shrinkage and selection operator (LASSO) regression) to pinpoint candidate hub genes for diagnosing obesity-associated risk in offspring of maternal obesity. We constructed a nomogram receiver operating characteristic (ROC) curve to evaluate the diagnostic value. Additionally, we analyzed immune cell infiltration to investigate immune cell dysregulation in maternal obesity. Furthermore, we verified the expression of the candidate hub genes both in vivo and in vitro. RESULTS The GSE135830 dataset revealed 2868 DEGs between the mHFD offspring and the mChow group and 2627 WGCNA module genes related to maternal obesity. The overlap of DEGs and module genes in the offspring with maternal obesity in GSE135830 primarily enriched in neurodevelopment and immune regulation. In the GSE127056 dataset, 133 DEGs were identified in the hypothalamus of HFD-induced adult obese individuals. A total of 13 genes intersected between the GSE127056 adult obesity DEGs and the GSE135830 maternal obesity module genes that were primarily enriched in neurodevelopment and the immune response. Following machine learning, two candidate hub genes were chosen for nomogram construction. Diagnostic value evaluation by ROC analysis determined Sytl4 and Kncn2 as hub genes for maternal obesity in the offspring. A gene regulatory network with transcription factor-miRNA interactions was established. Dysregulated immune cells were observed in the hypothalamus of offspring with maternal obesity. Expression of Sytl4 and Kncn2 was validated in a mouse model of hypothalamic inflammation and a palmitic acid-stimulated microglial inflammation model. CONCLUSION Two candidate hub genes (Sytl4 and Kcnc2) were identified and a nomogram was developed to predict obesity risk in offspring with maternal obesity. These findings offer potential diagnostic candidate genes for identifying obesity-associated risks in the offspring of obese mothers.
Collapse
Affiliation(s)
- Yanxing Shang
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
| | - Xueqin Wang
- Department of Endocrinology, Affiliated Hospital 2Nantong UniversityNantongChina
| | - Sixuan Su
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
- Department of Pathogen Biology, Medical CollegeNantong UniversityNantongChina
| | - Feng Ji
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
| | - Donghai Shao
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
| | - Chengwei Duan
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
| | - Tianpeng Chen
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
| | - Caixia Liang
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
| | - Dongmei Zhang
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Nantong Clinical Medical College of Kangda College of Nanjing Medical UniversityNantongChina
- Nantong Municipal Key Laboratory of Metabolic Immunology and Disease MicroenvironmentNantong First People's HospitalNantongChina
- Department of Pathogen Biology, Medical CollegeNantong UniversityNantongChina
| | - Hongjian Lu
- Medical Research Center, Affiliated Hospital 2Nantong UniversityNantongChina
- Jiangsu Provincial Medical Key Discipline (Laboratory) Cultivation Unit, Medical Research CenterNantong First People's HospitalNantongChina
- Department of Rehabilitation Medicine, Affiliated Hospital 2Nantong UniversityNantongChina
| |
Collapse
|
31
|
Adamczak L, Mantaj U, Sibiak R, Gutaj P, Wender-Ozegowska E. Physical activity, gestational weight gain in obese patients with early gestational diabetes and the perinatal outcome - a randomised-controlled trial. BMC Pregnancy Childbirth 2024; 24:104. [PMID: 38308265 PMCID: PMC10836025 DOI: 10.1186/s12884-024-06296-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 01/28/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Excessive gestational weight gain, especially among women with gestational diabetes, is associated with several adverse perinatal outcomes. Our study aimed to analyse the impact of the use of pedometers to supervise physical activity on maternal health and the obstetric outcomes of pregnant women with obesity and early gestational diabetes. METHODS 124 pregnant patients were enrolled in the presented research. INCLUSION CRITERIA singleton pregnancy, age > 18 years, gestational diabetes diagnosed in the first half of pregnancy (< 20th week of pregnancy), obesity according to the American Endocrine Society criteria. Each patient was advised to take at least 5000 steps daily. Patients were randomly assigned to pedometers (N = 62), and were recommended to monitor daily the number of steps. The group without pedometers (N = 62) was not observed. Visit (V1) was scheduled between the 28th and 32nd gestational week (GW), and visit (V2) occurred between the 37th and 39th GW. Anthropometric measurements and blood samples were collected from all patients at each appointment. Foetal and maternal outcomes were analysed at the end of the study. RESULTS In the group supervised by pedometers, there were significantly fewer newborns with macrosomia (p = 0,03). Only 45% of patients satisfied the recommended physical activity guidelines. Patients who walked more than 5000 steps per day had significantly higher body weight at baseline (p = 0,005), but weight gain was significantly lower than in the group that did not exceed 5000 steps per day (p < 0,001). The perinatal outcome in the group of patients performing more than 5000 steps did not demonstrate significant differences with when compared to less active group. ROC curve for weight gain above the guidelines indicated a statistically substantial cut-off point for this group at the level of 4210 steps/day (p = 0.00001). CONCLUSIONS Monitoring the activity of pregnant patients with gestational diabetes and obesity by pedometers did not have a significantly impact on their metabolic control and weight gain. However, it contributed to less macrosomia. Furthermore, physical activity over 5,000 steps per day positively affects weight loss, as well as contributes to improved obstetric and neonatal outcomes.
Collapse
Affiliation(s)
- Lukasz Adamczak
- Department of Reproduction, Chair of Fetomaternal Medicine, Poznan University of Medical Sciences, Poznan, Poland.
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland.
| | - Urszula Mantaj
- Department of Reproduction, Chair of Fetomaternal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Rafał Sibiak
- Department of Reproduction, Chair of Fetomaternal Medicine, Poznan University of Medical Sciences, Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Paweł Gutaj
- Department of Reproduction, Chair of Fetomaternal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Ewa Wender-Ozegowska
- Department of Reproduction, Chair of Fetomaternal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
32
|
Papadakis S, Thompson JR, Feczko E, Miranda-Dominguez O, Dunn GA, Selby M, Mitchell AJ, Sullivan EL, Fair DA. Perinatal Western-style diet exposure associated with decreased microglial counts throughout the arcuate nucleus of the hypothalamus in Japanese macaques. J Neurophysiol 2024; 131:241-260. [PMID: 38197176 PMCID: PMC11286309 DOI: 10.1152/jn.00213.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 12/13/2023] [Accepted: 01/03/2024] [Indexed: 01/11/2024] Open
Abstract
Perinatal exposure to a high-fat, high-sugar Western-style diet (WSD) is associated with altered neural circuitry in the melanocortin system. This association may have an underlying inflammatory component, as consumption of a WSD during pregnancy can lead to an elevated inflammatory environment. Our group previously demonstrated that prenatal WSD exposure was associated with increased markers of inflammation in the placenta and fetal hypothalamus in Japanese macaques. In this follow-up study, we sought to determine whether this heightened inflammatory state persisted into the postnatal period, as prenatal exposure to inflammation has been shown to reprogram offspring immune function and long-term neuroinflammation would present a potential means for prolonged disruptions to microglia-mediated neuronal circuit formation. Neuroinflammation was approximated in 1-yr-old offspring by counting resident microglia and peripherally derived macrophages in the region of the hypothalamus examined in the fetal study, the arcuate nucleus (ARC). Microglia and macrophages were immunofluorescently stained with their shared marker, ionized calcium-binding adapter molecule 1 (Iba1), and quantified in 11 regions along the rostral-caudal axis of the ARC. A mixed-effects model revealed main effects of perinatal diet (P = 0.011) and spatial location (P = 0.003) on Iba1-stained cell count. Perinatal WSD exposure was associated with a slight decrease in the number of Iba1-stained cells, and cells were more densely located in the center of the ARC. These findings suggest that the heightened inflammatory state experienced in utero does not persist postnatally. This inflammatory response trajectory could have important implications for understanding how neurodevelopmental disorders progress.NEW & NOTEWORTHY Prenatal Western-style diet exposure is associated with increased microglial activity in utero. However, we found a potentially neuroprotective reduction in microglia count during early postnatal development. This trajectory could inform the timing of disruptions to microglia-mediated neuronal circuit formation. Additionally, this is the first study in juvenile macaques to characterize the distribution of microglia along the rostral-caudal axis of the arcuate nucleus of the hypothalamus. Nearby neuronal populations may be greater targets during inflammatory insults.
Collapse
Affiliation(s)
- Samantha Papadakis
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, United States
- Department of Psychiatry, Oregon Health & Science University, Portland, Oregon, United States
| | - Jacqueline R Thompson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - Eric Feczko
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, United States
- Masonic Institute for the Developing Brain, University of Minnesota Medical School, Minneapolis, Minnesota, United States
| | - Oscar Miranda-Dominguez
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, United States
- Masonic Institute for the Developing Brain, University of Minnesota Medical School, Minneapolis, Minnesota, United States
| | - Geoffrey A Dunn
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| | - Matthew Selby
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| | - A J Mitchell
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, United States
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - Elinor L Sullivan
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, United States
- Department of Psychiatry, Oregon Health & Science University, Portland, Oregon, United States
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - Damien A Fair
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, United States
- Masonic Institute for the Developing Brain, University of Minnesota Medical School, Minneapolis, Minnesota, United States
- Institute of Child Development, College of Education and Human Development, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
33
|
Jo S, Alejandro EU. RISING STARS: Mechanistic insights into maternal-fetal cross talk and islet beta-cell development. J Endocrinol 2023; 259:e230069. [PMID: 37855321 PMCID: PMC10692651 DOI: 10.1530/joe-23-0069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/18/2023] [Indexed: 10/20/2023]
Abstract
The metabolic health trajectory of an individual is shaped as early as prepregnancy, during pregnancy, and lactation period. Both maternal nutrition and metabolic health status are critical factors in the programming of offspring toward an increased propensity to developing type 2 diabetes in adulthood. Pancreatic beta-cells, part of the endocrine islets, which are nutrient-sensitive tissues important for glucose metabolism, are primed early in life (the first 1000 days in humans) with limited plasticity later in life. This suggests the high importance of the developmental window of programming in utero and early in life. This review will focus on how changes to the maternal milieu increase offspring's susceptibility to diabetes through changes in pancreatic beta-cell mass and function and discuss potential mechanisms by which placental-driven nutrient availability, hormones, exosomes, and immune alterations that may impact beta-cell development in utero, thereby affecting susceptibility to type 2 diabetes in adulthood.
Collapse
Affiliation(s)
- Seokwon Jo
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Emilyn U Alejandro
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
34
|
Wayland JL, Doll JR, Lawson MJ, Stankiewicz TE, Oates JR, Sawada K, Damen MSMA, Alarcon PC, Haslam DB, Trout AT, DeFranco EA, Klepper CM, Woo JG, Moreno-Fernandez ME, Mouzaki M, Divanovic S. Thermoneutral Housing Enables Studies of Vertical Transmission of Obesogenic Diet-Driven Metabolic Diseases. Nutrients 2023; 15:4958. [PMID: 38068816 PMCID: PMC10708424 DOI: 10.3390/nu15234958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Vertical transmission of obesity is a critical contributor to the unabated obesity pandemic and the associated surge in metabolic diseases. Existing experimental models insufficiently recapitulate "human-like" obesity phenotypes, limiting the discovery of how severe obesity in pregnancy instructs vertical transmission of obesity. Here, via utility of thermoneutral housing and obesogenic diet feeding coupled to syngeneic mating of WT obese female and lean male mice on a C57BL/6 background, we present a tractable, more "human-like" approach to specifically investigate how maternal obesity contributes to offspring health. Using this model, we found that maternal obesity decreased neonatal survival, increased offspring adiposity, and accelerated offspring predisposition to obesity and metabolic disease. We also show that severe maternal obesity was sufficient to skew offspring microbiome and create a proinflammatory gestational environment that correlated with inflammatory changes in the offspring in utero and adulthood. Analysis of a human birth cohort study of mothers with and without obesity and their infants was consistent with mouse study findings of maternal inflammation and offspring weight gain propensity. Together, our results show that dietary induction of obesity in female mice coupled to thermoneutral housing can be used for future mechanistic interrogations of obesity and metabolic disease in pregnancy and vertical transmission of pathogenic traits.
Collapse
Affiliation(s)
- Jennifer L. Wayland
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jessica R. Doll
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Matthew J. Lawson
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Traci E. Stankiewicz
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jarren R. Oates
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Keisuke Sawada
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michelle S. M. A. Damen
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Pablo C. Alarcon
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - David B. Haslam
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Andrew T. Trout
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Department of Radiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Emily A. DeFranco
- Department of Obstetrics and Gynecology, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Corie M. Klepper
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jessica G. Woo
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Maria E. Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Marialena Mouzaki
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Senad Divanovic
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
35
|
Mazur D, Satora M, Rekowska AK, Kabała Z, Łomża A, Kimber-Trojnar Ż, Leszczyńska-Gorzelak B. Influence of Breastfeeding on the State of Meta-Inflammation in Obesity-A Narrative Review. Curr Issues Mol Biol 2023; 45:9003-9018. [PMID: 37998742 PMCID: PMC10670570 DOI: 10.3390/cimb45110565] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
Obesity has become an emerging health issue worldwide that continues to grow in females of reproductive age as well. Obesity, as a multisystem and chronic disease, is associated with metabolic inflammation, which is defined as chronic low-grade systemic inflammation mediated by, i.a., adipose tissue macrophages. Lactation has been proven to have a beneficial influence on maternal health and could help restore metabolic balance, especially in the state of maternal obesity. In this review, we aimed to analyze the influence of breastfeeding on chronic low-grade meta-inflammation caused by obesity. We performed a comprehensive literature review using the PubMed, Science Direct, and Google Scholar electronic databases. For this purpose, we searched for "metabolic inflammation"; "meta-inflammation"; "obesity"; "breastfeeding"; "fetal programming"; "energy metabolism"; "postpartum"; "immunity"; "immune system"; and "inflammation" keyword combinations. While the clinical impact of breastfeeding on maternal and offspring health is currently well known, we decided to gain insight into more specific metabolic effects of adiposity, lipid, and glucose homeostasis, and immunological effects caused by the activity of cytokines, macrophages, and other immune system cells. Further research on the immunological and metabolic effects of breastfeeding in obese patients is key to understanding and potentially developing obesity therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - Żaneta Kimber-Trojnar
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-059 Lublin, Poland; (D.M.); (M.S.); (A.K.R.); (Z.K.); (A.Ł.); (B.L.-G.)
| | | |
Collapse
|
36
|
Jiménez-Osorio AS, Carreón-Torres E, Correa-Solís E, Ángel-García J, Arias-Rico J, Jiménez-Garza O, Morales-Castillejos L, Díaz-Zuleta HA, Baltazar-Tellez RM, Sánchez-Padilla ML, Flores-Chávez OR, Estrada-Luna D. Inflammation and Oxidative Stress Induced by Obesity, Gestational Diabetes, and Preeclampsia in Pregnancy: Role of High-Density Lipoproteins as Vectors for Bioactive Compounds. Antioxidants (Basel) 2023; 12:1894. [PMID: 37891973 PMCID: PMC10604737 DOI: 10.3390/antiox12101894] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/05/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Inflammation and oxidative stress are essential components in a myriad of pathogenic entities that lead to metabolic and chronic diseases. Moreover, inflammation in its different phases is necessary for the initiation and maintenance of a healthy pregnancy. Therefore, an equilibrium between a necessary/pathologic level of inflammation and oxidative stress during pregnancy is needed to avoid disease development. High-density lipoproteins (HDL) are important for a healthy pregnancy and a good neonatal outcome. Their role in fetal development during challenging situations is vital for maintaining the equilibrium. However, in certain conditions, such as obesity, diabetes, and other cardiovascular diseases, it has been observed that HDL loses its protective properties, becoming dysfunctional. Bioactive compounds have been widely studied as mediators of inflammation and oxidative stress in different diseases, but their mechanisms of action are still unknown. Nonetheless, these agents, which are obtained from functional foods, increase the concentration of HDL, TRC, and antioxidant activity. Therefore, this review first summarizes several mechanisms of HDL participation in the equilibrium between inflammation and oxidative stress. Second, it gives an insight into how HDL may act as a vector for bioactive compounds. Third, it describes the relationships between the inflammation process in pregnancy and HDL activity. Consequently, different databases were used, including MEDLINE, PubMed, and Scopus, where scientific articles published in the English language up to 2023 were identified.
Collapse
Affiliation(s)
- Angélica Saraí Jiménez-Osorio
- Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado Hida go, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (A.S.J.-O.); (J.Á.-G.); (J.A.-R.); (O.J.-G.); (L.M.-C.); (R.M.B.-T.); (M.L.S.-P.); (O.R.F.-C.)
| | - Elizabeth Carreón-Torres
- Department of Molecular Biology, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico;
| | - Emmanuel Correa-Solís
- Instituto de Farmacobiología, Universidad de la Cañada, Carretera Teotitlán-San Antonio Nanahuatipán Km 1.7 s/n., Paraje Titlacuatitla, Teotitlán de Flores Magón 68540, Oaxaca, Mexico;
| | - Julieta Ángel-García
- Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado Hida go, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (A.S.J.-O.); (J.Á.-G.); (J.A.-R.); (O.J.-G.); (L.M.-C.); (R.M.B.-T.); (M.L.S.-P.); (O.R.F.-C.)
| | - José Arias-Rico
- Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado Hida go, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (A.S.J.-O.); (J.Á.-G.); (J.A.-R.); (O.J.-G.); (L.M.-C.); (R.M.B.-T.); (M.L.S.-P.); (O.R.F.-C.)
| | - Octavio Jiménez-Garza
- Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado Hida go, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (A.S.J.-O.); (J.Á.-G.); (J.A.-R.); (O.J.-G.); (L.M.-C.); (R.M.B.-T.); (M.L.S.-P.); (O.R.F.-C.)
| | - Lizbeth Morales-Castillejos
- Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado Hida go, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (A.S.J.-O.); (J.Á.-G.); (J.A.-R.); (O.J.-G.); (L.M.-C.); (R.M.B.-T.); (M.L.S.-P.); (O.R.F.-C.)
| | - Hugo Alexander Díaz-Zuleta
- Facultad de Ciencias de la Salud, Universidad de Ciencias Aplicadas y Ambientales, Cl. 222 #54-21, Bogotá 111166, Colombia;
| | - Rosa María Baltazar-Tellez
- Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado Hida go, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (A.S.J.-O.); (J.Á.-G.); (J.A.-R.); (O.J.-G.); (L.M.-C.); (R.M.B.-T.); (M.L.S.-P.); (O.R.F.-C.)
| | - María Luisa Sánchez-Padilla
- Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado Hida go, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (A.S.J.-O.); (J.Á.-G.); (J.A.-R.); (O.J.-G.); (L.M.-C.); (R.M.B.-T.); (M.L.S.-P.); (O.R.F.-C.)
| | - Olga Rocío Flores-Chávez
- Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado Hida go, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (A.S.J.-O.); (J.Á.-G.); (J.A.-R.); (O.J.-G.); (L.M.-C.); (R.M.B.-T.); (M.L.S.-P.); (O.R.F.-C.)
| | - Diego Estrada-Luna
- Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado Hida go, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (A.S.J.-O.); (J.Á.-G.); (J.A.-R.); (O.J.-G.); (L.M.-C.); (R.M.B.-T.); (M.L.S.-P.); (O.R.F.-C.)
| |
Collapse
|
37
|
Wang X, Tong J, Liang C, Wang X, Ma Y, Tao S, Liu M, Wang Y, Liu J, Yan S, Gao G, Wu X, Huang K, Cao Y, Tao F. Trimester-specific effects of maternal exposure to single and mixed metals on cord serum inflammatory cytokines levels: A prospective birth cohort study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 895:165086. [PMID: 37379910 DOI: 10.1016/j.scitotenv.2023.165086] [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: 05/09/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Cord blood inflammatory cytokines are vital in early-life programming. An increasing number of studies concern the effect of maternal exposure to different metal elements during pregnancy on inflammatory cytokines, but limited studies have explored the association between maternal exposure to mixed metals and cord blood inflammatory cytokine levels. METHODS We measured serum concentrations of vanadium (V), copper (Cu), arsenic (As), cadmium (Cd), and barium (Ba) in the first, second, and third trimesters and eight cord serum inflammatory cytokines (IFN-γ, IL-1β, IL-6, IL-8, IL-10, IL-12p70, IL-17A, and TNF-α) in 1436 mother-child dyads from the Ma'anshan Birth Cohort. Generalized linear models and Bayesian kernel machine regression (BKMR) were performed to assess the association of single and mixed metal exposure during each trimester with cord serum inflammatory cytokine levels, respectively. RESULTS Regarding metal exposure in the first trimester, V was positively associated with TNF-α (β = 0.33, 95 % CI: 0.13, 0.53); Cu was positively associated with IL-8 (β = 0.23, 95 % CI: 0.07, 0.39); Ba was positively associated with IFN-γ and IL-6; As was negatively associated with IFN-γ and IL-17A; and Cd was negatively associated with IFN-γ, IL-1β, IL-12p70, IL-17A, and TNF-α. BKMR revealed that exposure to metal mixtures in the first trimester was positively associated with IL-8 and TNF-α but negatively associated with IL-17A. Moreover, V contributed the most to these associations. Interaction effects were observed between Cd and As and between Cd and Cu with IL-8, and between Cd and V with IL-17A. Among males, As decreased inflammatory cytokines; among females, Cu increased inflammatory cytokine levels, whereas Cd decreased inflammatory cytokine concentrations. CONCLUSIONS Maternal exposure to metal mixtures in the first trimester interfered with cord serum inflammatory cytokine levels. The associations of maternal exposure to As, Cu and Cd with inflammatory cytokines showed sex differences. Further studies are warranted to support the findings and explore the mechanism of the susceptibility window and sex-specific disparity.
Collapse
Affiliation(s)
- Xing Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Juan Tong
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Chunmei Liang
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; National Health Commission Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University; Hefei 230032, Anhui, China
| | - Xueqing Wang
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; National Health Commission Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University; Hefei 230032, Anhui, China
| | - Yufan Ma
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Shuman Tao
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; National Health Commission Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University; Hefei 230032, Anhui, China
| | - Meng Liu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Yafei Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Jia Liu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Shuangqin Yan
- Ma'anshan Maternal and Child Health Care Hospital, Ma'anshan 243011, Anhui, China
| | - Guopeng Gao
- Ma'anshan Maternal and Child Health Care Hospital, Ma'anshan 243011, Anhui, China
| | - Xiaoyan Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; National Health Commission Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University; Hefei 230032, Anhui, China
| | - Kun Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; National Health Commission Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University; Hefei 230032, Anhui, China
| | - Yunxia Cao
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; National Health Commission Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; National Health Commission Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University; Hefei 230032, Anhui, China.
| |
Collapse
|
38
|
Capobianco E, Pirrone I. Paternal programming of fetoplacental and offspring metabolic disorders. Placenta 2023; 141:71-77. [PMID: 37355440 DOI: 10.1016/j.placenta.2023.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/08/2023] [Accepted: 06/11/2023] [Indexed: 06/26/2023]
Abstract
The alarming increase in the prevalence of metabolic pathologies is of worldwide concern and has been linked not only to genetic factors but also to a large number of non-genetic factors. In recent years, there has been increasing interest in the study of the programming of metabolic diseases, such as type 2 diabetes mellitus (T2DM) and obesity, by paternal exposure, a paradigm termed "Paternal Origins of Health and Disease" (POHaD). This term derives from the better known "Developmental Origins of Health and Disease" (DOHaD), which focuses on the involvement of the maternal intrauterine environment and complications during pregnancy associated with the health and disease of the offspring. Studies on paternal programming have documented environmentally induced epigenetic modifications in the male germline and in seminal plasma, which lead to intergenerational and transgenerational phenotypes, evident already during fetoplacental development. Studies with animal models at both ends of the nutritional spectrum (undernutrition or overnutrition) have been performed to understand the possible mechanisms and signaling pathways leading to the programming of metabolic disorders by exploring epigenetic changes throughout the life of the offspring. The aim of this review was to address the evidence of the programming of fetoplacental developmental alterations and metabolic pathologies in the offspring of males with metabolic disorders and unhealthy exposures, highlighting the mechanisms involved in such programming and looking for paternal interventions to reduce negative health outcomes in the offspring.
Collapse
Affiliation(s)
- Evangelina Capobianco
- Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina.
| | - Irune Pirrone
- Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| |
Collapse
|
39
|
Prado Spalm FH, Cuervo Sánchez ML, Furland NE, Vallés AS. Lipid peroxidation and neuroinflammation: A possible link between maternal fructose intake and delay of acquisition of neonatal reflexes in Wistar female rats. Dev Neurobiol 2023; 83:167-183. [PMID: 37435772 DOI: 10.1002/dneu.22921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/20/2023] [Accepted: 06/24/2023] [Indexed: 07/13/2023]
Abstract
Fructose is a common sweetener found in the daily diet supplemented to many processed and ultra-processed foods and beverages. Consumption of fructose-sweetened beverages has drastically increased in the last decades and is widely associated with metabolic disease, systemic pro-inflammatory status, and adverse transgenerational effects. To date, the impact of maternal fructose intake in brain function of the offspring is less explored. Therefore, the aim of this study was first, to investigate adverse effects in developmental milestones of the progeny of mothers with metabolic syndrome (MetS), induced by ad libitum consumption of a 20% fructose solution, and second to identify possible molecular changes in the nervous system of the newborns associated with maternal fructose intake. Wistar rats were randomly separated into two groups with access to water or fructose (20% w/v in water) for 10 weeks. After MetS was confirmed, dams were mated with control males and continued drinking water or fructose solution during gestation. At postnatal day (PN) 1, a subgroup of offspring of each sex was sacrificed and brains were dissected for oxidative stress and inflammatory status analysis. Changes in the developmental milestones due to maternal fructose consumption were studied (PN3-PN21) in another subgroup of offspring. Sexually dimorphic effects were found on the progeny's acquisition of neurodevelopmental milestones, in brain lipid peroxidation, neuroinflammation, and antioxidative defensive response. Our results suggest that dams' MetS, induced by fructose intake, disrupts brain redox homeostasis in female offspring and affects sensorimotor brain circuitry which may have a translational value for studying neurodevelopmental diseases.
Collapse
Affiliation(s)
- Facundo H Prado Spalm
- Nutrition and Neurodevelopmental Laboratory, INIBIBB-CONICET-UNS, Bahía Blanca, Argentina
| | - Marié L Cuervo Sánchez
- Nutrition and Neurodevelopmental Laboratory, INIBIBB-CONICET-UNS, Bahía Blanca, Argentina
| | - Natalia E Furland
- Nutrition and Neurodevelopmental Laboratory, INIBIBB-CONICET-UNS, Bahía Blanca, Argentina
| | - Ana S Vallés
- Nutrition and Neurodevelopmental Laboratory, INIBIBB-CONICET-UNS, Bahía Blanca, Argentina
| |
Collapse
|
40
|
Zheng S, Yin J, Yue H, Li L. Maternal high-fat diet increases the susceptibility of offspring to colorectal cancer via the activation of intestinal inflammation. Front Nutr 2023; 10:1191206. [PMID: 37252240 PMCID: PMC10213637 DOI: 10.3389/fnut.2023.1191206] [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: 03/21/2023] [Accepted: 04/27/2023] [Indexed: 05/31/2023] Open
Abstract
A high-fat diet plays a key role in the pathogenesis of colorectal cancer, and this effect on the gut can also occur in the offspring of mothers with a high-fat diet. In this review, we discuss the role of a high-fat diet in the pathogenesis of colorectal cancer and summarize the effects of a maternal high-fat diet on the activation of inflammation and development of colorectal cancer in offspring. Studies have found that a maternal high-fat diet primarily induces an inflammatory response in the colorectal tissue of both the mother herself and the offspring during pregnancy. This leads to the accumulation of inflammatory cells in the colorectal tissue and the release of inflammatory cytokines, which further activate the NF-κb and related inflammatory signaling pathways. Research suggests that high levels of lipids and inflammatory factors from mothers with a high-fat diet are passed to the offspring through the transplacental route, which induces colorectal inflammation, impairs the intestinal microecological structure and the intestinal barrier, and interferes with intestinal development in the offspring. This in turn activates the NF-κb and related signaling pathways, which further aggravates intestinal inflammation. This process of continuous inflammatory stimulation and repair may promote the uncontrolled proliferation of colorectal mucosal cells in the offspring, thus increasing their susceptibility to colorectal cancer.
Collapse
Affiliation(s)
- Shimin Zheng
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jianbin Yin
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Orthopedics, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Hui Yue
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Lifu Li
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
41
|
Naomi R, Teoh SH, Embong H, Balan SS, Othman F, Bahari H, Yazid MD. The Role of Oxidative Stress and Inflammation in Obesity and Its Impact on Cognitive Impairments-A Narrative Review. Antioxidants (Basel) 2023; 12:antiox12051071. [PMID: 37237937 DOI: 10.3390/antiox12051071] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Obesity is a chronic low-grade inflammatory condition that induces the generation of oxidative stress and inflammation. This oxidative stress and inflammation stimulate brain atrophy and some morphological changes in the brain that eventually result in cognitive impairments. However, there is no exact study that has summarized the role of oxidative stress and inflammation in obesity and its impact on cognitive impairments. Thus, the objective of this review is to recapitulate the current role of oxidative stress and inflammation in cognitive decline based on in vivo evidence. A comprehensive search was performed in Nature, Medline and Ovid, ScienceDirect, and PubMed, and the search was limited to the past 10 years of publication. From the search, we identified 27 articles to be further reviewed. The outcome of this study indicates that a greater amount of fat stored in individual adipocytes in obesity induces the formation of reactive oxygen species and inflammation. This will lead to the generation of oxidative stress, which may cause morphological changes in the brain, suppress the endogenous antioxidant system, and promote neuroinflammation and, eventually, neuronal apoptosis. This will impair the normal function of the brain and specific regions that are involved in learning, as well as memory. This shows that obesity has a strong positive correlation with cognitive impairments. Hence, this review summarizes the mechanism of oxidative stress and inflammation that induce memory loss based on animal model evidence. In conclusion, this review may serve as an insight into therapeutic development focusing on oxidative stress and inflammatory pathways to manage an obesity-induced cognitive decline in the future.
Collapse
Affiliation(s)
- Ruth Naomi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Soo Huat Teoh
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas 13200, Malaysia
| | - Hashim Embong
- Department of Emergency Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Santhra Segaran Balan
- Department of Diagnostic and Allied Health Sciences, Faculty of Health and Life Sciences, Management and Science University, Shah Alam 40100, Malaysia
| | - Fezah Othman
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Hasnah Bahari
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
42
|
Naomi R, Rusli RNM, Othman F, Balan SS, Abidin AZ, Embong H, Teoh SH, Jasni AS, Jumidil SH, Matraf KSYB, Zakaria ZA, Bahari H, Yazid MD. Elateriospermum tapos Yogurt Supplement in Maternal Obese Dams during Pregnancy Modulates the Body Composition of F1 Generation. Nutrients 2023; 15:1258. [PMID: 36904258 PMCID: PMC10005445 DOI: 10.3390/nu15051258] [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: 01/21/2023] [Revised: 03/01/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Maternal obesity is a key predictor of childhood obesity and a determining factor for a child's body composition. Thus, any form of maternal nutrition during the gestational period plays a vital role in influencing the growth of the fetus. Elateriospermum tapos (E. tapos) yogurt has been found to comprise many bioactive compounds such as tannins, saponins, α-linolenic acid, and 5'-methoxy-bilobate with apocynoside I that could cross the placenta and exhibit an anti-obesity effect. As such, this study aimed to investigate the role of maternal E. tapos yogurt supplementation on offspring body composition. In this study, 48 female Sprague Dawley (SD) rats were induced with obesity using a high-fat diet (HFD) and were allowed to breed. Upon confirmation of pregnancy, treatment was initiated with E. tapos yogurt on the obese dams up to postnatal day 21. The weaning offspring were then designated into six groups according to their dam's group (n = 8) as follows; normal food and saline (NS), HFD and saline (HS), HFD and yogurt (HY), HFD and 5 mg/kg of E. tapos yogurt (HYT5), HFD and 50 mg/kg of E. tapos yogurt (HYT50), and HFD and 500 mg/kg of E. tapos yogurt (HYT500). The body weight of the offspring was accessed every 3 days up to PND 21. All the offspring were euthanized on PND 21 for tissue harvesting and blood sample collection. The results showed that both male and female offspring of obese dams treated with E. tapos yogurt showed growth patterns similar to NS and reduced levels of triglycerides (TG), cholesterol, LDL, non-HDL, and leptin. Liver enzymes such as ALT, ALP, AST, GGT, and globulin, and renal markers such as sodium, potassium, chloride, urea, and creatinine levels significantly reduced (p < 0.05) in the offspring of E. tapos yogurt-treated obese dams with the normal histological architecture of the liver, kidney, colon, RpWAT, and visceral tissue that is comparable to NS. In toto, E. tapos yogurt supplementation of obese dams exerted an anti-obesity effect by preventing intergenerational obesity by reversing HFD-induced damage in the fat tissue of the offspring.
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
| | - Fezah Othman
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Santhra Segaran Balan
- Department of Diagnostic and Allied Health Sciences, Faculty of Health and Health Sciences, Management and Science University, Shah Alam 40100, Malaysia
| | - Azrina Zainal Abidin
- Department of Diagnostic and Allied Health Sciences, Faculty of Health and Health Sciences, Management and Science University, Shah Alam 40100, Malaysia
| | - Hashim Embong
- Department of Emergency Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Soo Huat Teoh
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang 13200, Malaysia
| | - Azmiza Syawani Jasni
- Department of Medical Microbiology & Parasitology, Faculty of Medicine & Health Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Siti Hadizah Jumidil
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Khaled Salem Yaslam Ba Matraf
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Zainul Amiruddin Zakaria
- Borneo Research on Algesia, Inflammation and Neurodegeneration (BRAIN) Group, Faculty of Medicine and Health Sciences, Sabah Universiti Malaysia, Jalan UMS, Kota Kinabalu 88400, Malaysia
| | - Hasnah Bahari
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
43
|
Yong W, Wang J, Leng Y, Li L, Wang H. Role of Obesity in Female Reproduction. Int J Med Sci 2023; 20:366-375. [PMID: 36860674 PMCID: PMC9969507 DOI: 10.7150/ijms.80189] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/19/2023] [Indexed: 02/05/2023] Open
Abstract
Contemporary scientists need no "p value" and "relative risk" statistics to be exquisitely aware of the increasing prevalence of obesity and complications posed by obesity. It is now well recognized that obesity is strongly associated with type 2 diabetes, hypertension, vascular disease, tumors and reproductive disorders. Obese women show lower levels of gonadotropin hormones, reduced fecundity, higher miscarriage rates and poorer outcomes of in vitro fertilization, revealing that obesity affects female reproduction. In addition, adipose tissue contains special immune cells and obesity-induced inflammation is a chronic, low-grade inflammatory response. Herein, we mainly review detrimental influences of obesity in the complete process of female reproduction, including hypothalamic-pituitary-ovarian axis, oocyte maturation, embryo and fetal development. In the latter part, we view obesity-induced inflammation and discuss related epigenetic impact on female reproduction.
Collapse
Affiliation(s)
- Wei Yong
- Center Laboratory of the Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Jiajia Wang
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yan Leng
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Lijie Li
- Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Han Wang
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China.,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|