1
|
Yoneshiro T, Matsushita M, Sakai J, Saito M. Brown fat thermogenesis and cold adaptation in humans. J Physiol Anthropol 2025; 44:11. [PMID: 40259336 PMCID: PMC12010580 DOI: 10.1186/s40101-025-00391-w] [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: 11/25/2024] [Accepted: 03/30/2025] [Indexed: 04/23/2025] Open
Abstract
Brown adipose tissue (BAT) is a site of non-shivering thermogenesis (NST) in mammals. Since the rediscovery of BAT in adult humans, there has been a remarkable advance in human BAT researches, revealing the significant roles of this thermogenic tissue in cold-induced NST and cold adaptation. Cold stress influences BAT in various time spans: acute cold exposure promptly activates BAT to induce NST, which contributes to immediate maintenance of body temperature. Prolonged cold exposure recruits BAT, resulting in increased capacity of NST and improved cold tolerance. Such BAT adaptation not only occurs in the exposed individual but also is passed on to the next generation, probably via the paternal lineage. As such, BAT plays a role in acute, chronic, and transgenerational adaptation to cold environment in humans.
Collapse
Affiliation(s)
- Takeshi Yoneshiro
- Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980 - 8575, Japan.
| | - Mami Matsushita
- Department of Nutrition, School of Nursing and Nutrition, Tenshi College, Sapporo, Hokkaido, 065 - 0013, Japan
| | - Juro Sakai
- Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980 - 8575, Japan
| | - Masayuki Saito
- Department of Nutrition, School of Nursing and Nutrition, Tenshi College, Sapporo, Hokkaido, 065 - 0013, Japan
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060 - 0818, Japan
| |
Collapse
|
2
|
Li S, Zhou L, Ren J, Liu J, Zhang Q, Xiao X. The role of cecal metabolites and DNA methylation in deciphering the effects of maternal genistein intake on white fat browning in adult female offspring. Food Funct 2025; 16:3090-3100. [PMID: 40146209 DOI: 10.1039/d4fo04761c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Maternal diets during pregnancy and lactation are critical determinants that regulate the metabolic homeostasis in offspring. Our previous research demonstrated that maternal genistein (GEN) intervention ameliorated the dysregulation of glucolipid metabolism induced by intrauterine overnutrition in adult offspring, accompanied by changes in the composition of gut microbiota; however, the underlying mechanisms remain unclear. Here, we used a maternal overnutrition model induced by excess energy intake before and throughout pregnancy and lactation, with maternal GEN administered during the same period. The female offspring were raised on a standard chow diet until sacrificed at 24 weeks. The mRNA levels of browning markers were quantified in inguinal subcutaneous adipose tissues, followed by methylation analysis via the MassArray method. Cecal contents were collected for untargeted metabolomic analysis and a target quantitative analysis of methionine cycle metabolites. Spearman correlation analyses were used to assess whether cecal metabolites are involved in the methylation of browning-related genes and influence their expression. The results showed that maternal GEN supplementation reversed the downregulation of browning markers caused by perinatal high-fat diets in adult female offspring, consistent with a reduction in their methylation levels. Subsequently, we also found that maternal GEN consumption altered cecal metabolite profiles in offspring, promoting the production of bile acids, potent regulators of glucolipid metabolism, and reducing metabolites involved in the methionine cycle, key methyl donors for the methylation process. Furthermore, the abundances of these metabolites were significantly correlated with the methylation and expression levels of browning markers. Overall, this discovery suggested that maternal GEN intake decreased the methylation level of browning markers and induced browning in white adipose tissue of offspring, which correlated with alterations in cecal metabolites. We provide a novel theoretical basis for GEN as a promising nutritional supplement to break the vicious cycle of maternal metabolic disturbances being transmitted to offspring.
Collapse
Affiliation(s)
- Shunhua Li
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Liyuan Zhou
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jing Ren
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Jieying Liu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
3
|
Sinha S, Ahmad R, Chowdhury K, Islam S, Mehta M, Haque M. Childhood Obesity: A Narrative Review. Cureus 2025; 17:e82233. [PMID: 40231296 PMCID: PMC11995813 DOI: 10.7759/cureus.82233] [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: 03/21/2025] [Accepted: 04/14/2025] [Indexed: 04/16/2025] Open
Abstract
Obesity among children has emerged as a worldwide health issue due to childhood obesity becoming a pandemic, and it is often linked to various illnesses, fatal outcomes, and disability in adulthood. Obesity has become an epidemic issue in both developed and developing countries, particularly among youngsters. The most common factors contributing to non-communicable diseases (NCDs) are unhealthy eating habits, desk-bound games, avoidance of physical activity-requiring activities, smoking, alcohol usage, and other added items. All these factors increase NCDs, including obesity, resulting in various morbidities and early death. Additionally, childhood obesity has psychological, emotional, cognitive, societal, and communicative effects. For example, it raises the possibility of issues related to physical appearance, self-esteem, confidence level, feelings of isolation, social disengagement, stigma, depression, and a sense of inequality. Children who consume more energy-dense, high-fat, low-fiber-containing food than they need usually store the excess as body fat. Standardizing indicators and terminology for obesity-related metrics is critical for better understanding the comparability of obesity prevalence and program effectiveness within and between countries. The underlying variables must be altered to reduce or avoid harm to the target organ in children. As a result, reducing childhood obesity is a considerable public health goal for the benefit of society and the long-term well-being of individuals.
Collapse
Affiliation(s)
- Susmita Sinha
- Physiology, Enam Medical College and Hospital, Dhaka, BGD
| | - Rahnuma Ahmad
- Physiology, Medical College for Women and Hospital, Dhaka, BGD
| | - Kona Chowdhury
- Pediatrics, Enam Medical College and Hospital, Dhaka, BGD
| | - Shamima Islam
- Forensic Medicine, Enam Medical College and Hospital, Dhaka, BGD
| | - Miral Mehta
- Pedodontics and Preventive Dentistry, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| | - Mainul Haque
- Pharmacology and Therapeutics, National Defence University of Malaysia, Kuala Lumpur, MYS
- Research, Karnavati School of Dentistry, Karnavati University, Gandhinagar, IND
| |
Collapse
|
4
|
Massalha M, Iskander R, Hassan H, Spiegel E, Erez O, Nachum Z. Gestational diabetes mellitus - more than the eye can see - a warning sign for future maternal health with transgenerational impact. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2025; 6:1527076. [PMID: 40235646 PMCID: PMC11997571 DOI: 10.3389/fcdhc.2025.1527076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/06/2025] [Indexed: 04/17/2025]
Abstract
Gestational diabetes mellitus (GDM) is regarded by many as maternal maladaptation to physiological insulin resistance during the second half of pregnancy. However, recent evidence indicates that alterations in carbohydrate metabolism can already be detected in early pregnancy. This observation, the increasing prevalence of GDM, and the significant short and long-term implications for the mother and offspring call for reevaluation of the conceptual paradigm of GDM as a syndrome. This review will present evidence for the syndromic nature of GDM and the controversies regarding screening, diagnosis, management, and treatment.
Collapse
Affiliation(s)
- Manal Massalha
- Department of Obstetrics and Gynecology, Emek Medical Center, Afula, Israel
- Rappaport Faculty of Medicine, Technion, Institute of technology, Haifa, Israel
| | - Rula Iskander
- Department of Obstetrics and Gynecology, Emek Medical Center, Afula, Israel
| | - Haya Hassan
- Department of Obstetrics and Gynecology, Emek Medical Center, Afula, Israel
| | - Etty Spiegel
- Department of Obstetrics and Gynecology, Emek Medical Center, Afula, Israel
| | - Offer Erez
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Beer Sheva, Israel
- Faculty of Medicine, Ben Gurion University of the Negev, Beer Sheva, Israel
- Department of Obstetrics and Gynecology, Hutzel Women’s Hospital, Wayne State University, Detroit, MI, United States
| | - Zohar Nachum
- Department of Obstetrics and Gynecology, Emek Medical Center, Afula, Israel
- Rappaport Faculty of Medicine, Technion, Institute of technology, Haifa, Israel
| |
Collapse
|
5
|
Aladwan S, Issa R, Al. Safadi W, Alnsour L, Al‐Halaseh LK. Perceptions and Management of Pregnancy-Related Skin Changes: A Cross-Sectional Study on Knowledge, Practices, and Use of Skincare Product. J Cosmet Dermatol 2025; 24:e70132. [PMID: 40145230 PMCID: PMC11948173 DOI: 10.1111/jocd.70132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/24/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Hormonal, metabolic, and immunologic factors may cause several skin changes during pregnancy. Therefore, it is important for pregnant women to be aware of these expected changes in their skin appearance in order to prevent unwanted effects and to choose the appropriate preventive or treatment measures via trustable sources of information. OBJECTIVE This study would highlight the most common normal and abnormal physiological skin changes mothers usually complain about before or after pregnancy. The most commonly used skin care products, their sources of knowledge, information, perception, and experiences on these problems and products were also considered. In addition, patient satisfaction levels and their sources and types of knowledge were also investigated. METHODS Across sectional survey was distributed among women who met the inclusion criteria and were citizens in the Hashimate Kingdom of Jordan. This survey was composed of a number of questions used for investigating participant's socio-demographic characteristics, during and post-pregnancy characteristics and medications used, comparison of skin related complains reported by these women, in addition to their use of skin care products. Women's knowledge, perception, and experience regarding their use of skin care products were also included. RESULTS Of the 337 participants in this study, 6.5% and 6.8% of women were using thyroid medications and antihypertensive agents. An increase of around 3% in pregnancy-related skin changes, such as hyperpigmentation, hair loss, cellulite, and wrinkles, was predominant among women. Despite the prevalence of hyperpigmentation, only 4% and 17% of women used depigmentation and sunblock products. Moreover, women declare that their information about the use of skin care products was mainly via social media or self-experience. A low rate of consultation with dermatologists and the reliance on self-diagnosis or non-professional advice were shown. CONCLUSION This study suggests a lack of awareness about effective preventive measures for skin-related disorders commonly occurring during pregnancy, potentially exacerbated by reliance on unverified sources of information, such as social media. Therefore, incorporating education about skin changes into routine prenatal care could empower women to make informed decisions and reduce the stigma associated with these changes.
Collapse
Affiliation(s)
- Safwan Aladwan
- Department of Cosmetic Science, Faculty of Allied Medical SciencesAl‐Ahliyya Amman UniversityAmmanJordan
| | - Reem Issa
- Faculty of PharmacyMiddle East UniversityAmmanJordan
| | - Wala'a Al. Safadi
- Biopharmaceutics and Clinical Pharmacy Department, Pharmacological and Diagnostics Research Centre, Faculty of PharmacyAl‐Ahliyya Amman UniversityAmmanJordan
| | - Lilian Alnsour
- Department of Pharmacology and TherapeuticsCollege of Medicine and Health Sciences, United Arab Emirates UniversityAl AinUAE
| | - Lidia Kamal Al‐Halaseh
- Department of Pharmaceutical Chemistry, Faculty of PharmacyMutah UniversityAl‐KarakJordan
| |
Collapse
|
6
|
Bar A, Moran R, Mendelsohn-Cohen N, Korem Kohanim Y, Mayo A, Toledano Y, Alon U. Pregnancy and postpartum dynamics revealed by millions of lab tests. SCIENCE ADVANCES 2025; 11:eadr7922. [PMID: 40138427 PMCID: PMC11939066 DOI: 10.1126/sciadv.adr7922] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Pregnancy and delivery involve dynamic alterations in many physiological systems. However, the physiological dynamics during pregnancy and after delivery have not been systematically analyzed at high temporal resolution in a large human population. Here, we present the dynamics of 76 lab tests based on a cross-sectional analysis of 44 million measurements from over 300,000 pregnancies. We analyzed each test at weekly intervals from 20 weeks preconception to 80 weeks postpartum, providing detailed temporal profiles. About half of the tests take 3 months to a year to return to baseline postpartum, highlighting the physiological load of childbirth. The precision of the data revealed effects of preconception supplements, overshoots after delivery and intricate temporal responses to changes in blood volume and renal filtration rate. Pregnancy complications-gestational diabetes, preeclampsia, and postpartum hemorrhage-showed distinct dynamical changes. These results provide a comprehensive dynamic portrait of the systems physiology of pregnancy.
Collapse
Affiliation(s)
- Alon Bar
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ron Moran
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Netta Mendelsohn-Cohen
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yael Korem Kohanim
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yoel Toledano
- Division of Maternal Fetal Medicine, Helen Schneider Women’s Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
7
|
Inoue Y, Yokoyama M, Inoue S, Imai M, Onji H, Yano A, Uchikura Y, Matsubara Y, Matsubara K, Hamada H, Tomita H, Iwama N, Watanabe Z, Ishikuro M, Obara T, Metoki H, Ota C, Kuriyama S, Arima T, Yaegashi N, Saito M, Sugiyama T. Association between paternal physique and obesity in children at the age of 3 years: the Japan Environment and Children's Study. J Dev Orig Health Dis 2025; 16:e17. [PMID: 40116036 DOI: 10.1017/s2040174424000473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Obesity during development has been reported to be a determinant factor in the future development of non-communicable diseases (NCDs). Parental obesity is suggested to be a predictor of children's obesity, and it is important to consider parental factors to prevent NCDs in the progeny. Previously, we showed that paternal height had a stronger association with infant birth weight than paternal body mass index (BMI) in the Japanese population. However, only a few studies have examined the association between paternal physique and postnatal obesity. This study aimed to investigate the association between parental physique and obesity in children at the age of 3. This study used fixed data on 33,291 parent-child pairs from the Japan Environment and Children's Study, an ongoing national birth cohort study. The association between paternal physique (BMI and height) and children's obesity at the age of 3 was examined using multivariate logistic regression analysis. The higher the paternal BMI quartiles, the higher the odds ratio for obesity in male and female children at 3 years of age (P < 0.0001). However, paternal height quartiles were not associated with male or female obesity. These results differ from the association between paternal physique and infant birth weight, and it is possible that prenatal epigenetic and environmental factors of paternal origin were responsible for the differences between these two studies. The association between paternal BMI and obesity in children at the age of 3 suggests that paternal factors may be involved in the development of NCDs in future progeny.
Collapse
Affiliation(s)
- Yui Inoue
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Maki Yokoyama
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Shota Inoue
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Matome Imai
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Hiroshi Onji
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Akiko Yano
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Yuka Uchikura
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Yuko Matsubara
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Keiichi Matsubara
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Hirotaka Hamada
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hasumi Tomita
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Noriyuki Iwama
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Zen Watanabe
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Mami Ishikuro
- Division of Molecular Epidemiology, Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Division of Molecular Epidemiology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Taku Obara
- Division of Molecular Epidemiology, Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Division of Molecular Epidemiology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hirohito Metoki
- Division of Public Health, Hygiene and Epidemiology, Tohoku Medical Pharmaceutical University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Chiharu Ota
- Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Shinichi Kuriyama
- Division of Molecular Epidemiology, Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Division of Molecular Epidemiology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- International Research Institute of Disaster Science, Tohoku University, Sendai, Miyagi, Japan
| | - Takahiro Arima
- Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Nobuo Yaegashi
- Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Masatoshi Saito
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Maternal and Fetal Therapeutics, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Takashi Sugiyama
- Department of Obstetrics and Gynecology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| |
Collapse
|
8
|
Mittal R, Prasad K, Lemos JRN, Arevalo G, Hirani K. Unveiling Gestational Diabetes: An Overview of Pathophysiology and Management. Int J Mol Sci 2025; 26:2320. [PMID: 40076938 PMCID: PMC11900321 DOI: 10.3390/ijms26052320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/14/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Gestational diabetes mellitus (GDM) is characterized by an inadequate pancreatic β-cell response to pregnancy-induced insulin resistance, resulting in hyperglycemia. The pathophysiology involves reduced incretin hormone secretion and signaling, specifically decreased glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), impairing insulinotropic effects. Pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), impair insulin receptor substrate-1 (IRS-1) phosphorylation, disrupting insulin-mediated glucose uptake. β-cell dysfunction in GDM is associated with decreased pancreatic duodenal homeobox 1 (PDX1) expression, increased endoplasmic reticulum stress markers (CHOP, GRP78), and mitochondrial dysfunction leading to impaired ATP production and reduced glucose-stimulated insulin secretion. Excessive gestational weight gain exacerbates insulin resistance through hyperleptinemia, which downregulates insulin receptor expression via JAK/STAT signaling. Additionally, hypoadiponectinemia decreases AMP-activated protein kinase (AMPK) activation in skeletal muscle, impairing GLUT4 translocation. Placental hormones such as human placental lactogen (hPL) induce lipolysis, increasing circulating free fatty acids which activate protein kinase C, inhibiting insulin signaling. Placental 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) overactivity elevates cortisol levels, which activate glucocorticoid receptors to further reduce insulin sensitivity. GDM diagnostic thresholds (≥92 mg/dL fasting, ≥153 mg/dL post-load) are lower than type 2 diabetes to prevent fetal hyperinsulinemia and macrosomia. Management strategies focus on lifestyle modifications, including dietary carbohydrate restriction and exercise. Pharmacological interventions, such as insulin or metformin, aim to restore AMPK signaling and reduce hepatic glucose output. Emerging therapies, such as glucagon-like peptide-1 receptor (GLP-1R) agonists, show potential in improving glycemic control and reducing inflammation. A mechanistic understanding of GDM pathophysiology is essential for developing targeted therapeutic strategies to prevent both adverse pregnancy outcomes and the progression to overt diabetes in affected women.
Collapse
Affiliation(s)
| | | | | | | | - Khemraj Hirani
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (K.P.); (J.R.N.L.); (G.A.)
| |
Collapse
|
9
|
Yu W, Miao H, Gong Y. Lymphocyte Subsets and Cytokine Changes in Women With Gestational Diabetes Mellitus: A Systematic Review. J Diabetes Res 2025; 2025:3494697. [PMID: 40225013 PMCID: PMC11986944 DOI: 10.1155/jdr/3494697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 01/28/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction: Gestational diabetes mellitus (GDM) is a major health concern during pregnancy, affecting both the mother and the baby. Immune system alterations, particularly changes in lymphocyte subsets and cytokine profiles, have been associated with the pathophysiology of various metabolic disorders, including diabetes. This study is aimed at systematically reviewing the literature on the changes in lymphocyte subsets and cytokines in GDM. Methods: In this systematic review, we applied specific criteria to select observational studies (such as case-controls, cross-sectionals, or cohorts) that focused on pregnant women. We performed an extensive search across electronic databases, including Web of Science, Scopus, PubMed, MEDLINE, Embase, Cochrane Central Register of Controlled Trials, and Google Scholar, from January 1, 2010, to March 20, 2024. Results: A total of 19 articles, with 2517 participants (1128 with GDM and 1389 without GDM), were included in the qualitative synthesis. Due to high heterogeneity among the articles, a meta-analysis was not conducted. The studies assessed 35 different lymphocyte subsets or proportions. The most commonly assessed subsets were CD3+ T cell (five articles, mostly no difference between GDM and non-GDM), CD4+ T cell (five articles with contradictory results), CD8+ T cell (four articles with contradictory results), B cell and NK cell (three articles, mostly no difference between GDM and non-GDM), and Tregs (three articles with contradictory results). Additionally, 32 cytokines or proportions were assessed in the studies. The most commonly assessed cytokines were IL-6 (eight articles, higher or similar levels in GDM compared to non-GDM), TNF-α (seven articles, mostly higher or similar levels in GDM compared to non-GDM), IL-10 (six articles, mostly no difference between GDM and non-GDM), IL-2 (three articles, mostly no difference between GDM and non-GDM), and IFN-γ (three articles with contradictory results). Conclusion: According to the results, there were no significant changes in CD3+ T cells, B cells, NK cells, IL-10, and IL-2 in GDM. However, the levels of IL-6 and TNF-α were higher or similar in GDM compared to non-GDM. The changes of other lymphocyte subsets and cytokines in GDM remained unclear.
Collapse
Affiliation(s)
- Wang Yu
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Huang Miao
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Yunhui Gong
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Lilliecreutz C, Söderström E, Ersson M, Bendtsen M, Brown V, Kaegi-Braun N, Linder R, Maddison R, Chisalita SI, Löf M. SPARK: an mHealth intervention for self-management and treatment of gestational diabetes mellitus in Sweden - protocol for a randomised controlled trial. BMJ Open 2025; 15:e089355. [PMID: 40032379 DOI: 10.1136/bmjopen-2024-089355] [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] [Indexed: 03/05/2025] Open
Abstract
INTRODUCTION Gestational diabetes mellitus (GDM) is increasingly becoming a serious public health challenge. Innovative, effective and scalable lifestyle interventions to support women with GDM to manage their condition and prevent adverse obstetric and neonatal outcomes as well as later morbidity are required. This study aims to evaluate whether a novel, multilingual and scalable mobile health (mHealth) intervention (SPARK; SmartPhone App for gestational diabetes patients suppoRting Key lifestyle behaviours and glucose control) can improve self-management and treatment of GDM and prevent adverse maternal and offspring outcomes. METHODS/ANALYSES SPARK is a multicentre two-arm randomised controlled trial recruiting women diagnosed with GDM in south-eastern Sweden. A total of 412 women will be randomised to either standard care (control) or the SPARK intervention. The SPARK online platform (accessed through a mobile app) provides a behaviour change programme for healthy eating, physical activity and glycaemic control. To increase reach, SPARK is available in Swedish, English, Arabic and Somali. SPARK also comes with a clinician portal where healthcare professionals monitor and intervene when glycaemic control is unsatisfactory (above certain cut-offs). Primary outcomes are glycaemic control that is, time in range and HbA1c, while diet, physical activity (ActiGraph), gestational weight gain, metabolic and inflammatory biomarkers in weeks 37-38, adherence to protocol for daily glucose sampling, as well as adverse obstetric and neonatal outcomes are secondary outcomes. Secondary outcomes also include cardiometabolic risk evaluation, physical activity and healthy eating behaviours 1 year postpartum. A health economic evaluation of SPARK vs standard care will also be conducted. ETHICS AND DISSEMINATION This study has been approved by the Swedish Ethical Review Authority (2021-06627-01; 2022-03842-02; 2023-05911-02). Results will be disseminated through scientific papers in peer-reviewed journals, posts in traditional and social media, and presentations at scientific and healthcare professionals' conferences. TRIAL REGISTRATION NUMBER This trial was registered at the ClinicalTrials.gov register platform (ID NCT05348863) 27 April 2022.
Collapse
Affiliation(s)
- Caroline Lilliecreutz
- Department of Biomedical and Clinical Sciences, Linköping University, Linkoping, Sweden
| | - Emmie Söderström
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Matilda Ersson
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Bendtsen
- Department of Health, Medicine and Caring Sciences, Linköping University, Linkoping, Sweden
| | | | - Nina Kaegi-Braun
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Rebecka Linder
- Endocrinology Clinic, Linköping University Hospital, Linkoping, Sweden
| | | | | | - Marie Löf
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
- Deakin University, Melbourne, Victoria, Australia
| |
Collapse
|
11
|
Muntean M, Mărginean C, Bernad ES, Bănescu C, Nyulas V, Muntean IE, Săsăran V. Adiponectin C1Q and Collagen Domain Containing rs266729, Cyclin-Dependent Kinase Inhibitor 2A and 2B rs10811661, and Signal Sequence Receptor Subunit 1 rs9505118 Polymorphisms and Their Association with Gestational Diabetes Mellitus: A Case-Control Study in a Romanian Population. Int J Mol Sci 2025; 26:1654. [PMID: 40004118 PMCID: PMC11855124 DOI: 10.3390/ijms26041654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Gestational diabetes mellitus (GDM) and type 2 diabetes mellitus (T2DM) are public health concerns worldwide. These two diseases share the same pathophysiological and genetic similarities. This study aimed to investigate the T2DM known single nucleotide polymorphisms (SNPs) of the adiponectin C1Q and collagen domain containing (ADIPOQ), cyclin-dependent kinase inhibitor 2A and 2B (CDKN2A/2B), and signal sequence receptor subunit 1 (SSR1) genes in a cohort of Romanian GDM pregnant women and perinatal outcomes. DNA was isolated from the peripheral blood of 213 pregnant women with (n = 71) or without (n = 142) GDM. Afterward, ADIPOQ (rs266729), CDKN2A/2B (rs10811661), and SSR1 (rs9505118) gene polymorphisms were genotyped using TaqMan Real-Time PCR analysis. Women with GDM had a higher pre-pregnancy body mass index (BMI) (p < 0.0001), higher BMI (p < 0.0001), higher insulin resistance homeostatic model assessment (IR-HOMA) (p = 0.0002), higher insulin levels (p = 0.003), and lower adiponectin levels (p = 0.004) at birth compared to pregnant women with normoglycemia. GDM pregnant women had gestational hypertension (GH) more frequently during pregnancy (p < 0.0001), perineal lacerations more frequently during vaginal birth (p = 0.03), and more macrosomic newborns (p < 0.0001) than pregnant women from the control group. We did not find an association under any model (allelic, genotypic, dominant, or recessive) of ADIPOQ rs266729, CDKN2A/2B rs10811661, and SSR1 rs9505118 polymorphisms and GDM. In correlation analysis, we found a weak positive correlation (r = 0.24) between the dominant model GG + CG vs. CC of rs266729 and labor induction failure. In the dominant model TT vs. CC + CT of rs10811661, we found a weak negative correlation between this model and perineal lacerations. Our results suggest that the ADIPOQ rs266729, the CDKN2A/2B rs10811661, and the SSR1 rs9505118 gene polymorphisms are not associated with GDM in a cohort of Romanian pregnant women.
Collapse
Affiliation(s)
- Mihai Muntean
- Department of Obstetrics and Gynecology 2, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (M.M.); (V.S.)
| | - Claudiu Mărginean
- Department of Obstetrics and Gynecology 2, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (M.M.); (V.S.)
| | - Elena Silvia Bernad
- Department of Obstetrics and Gynecology, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Clinic of Obstetrics and Gynecology, “Pius Brinzeu” County Clinical Emergency Hospital, 300723 Timisoara, Romania
- Center for Laparoscopy, Laparoscopic Surgery and In Vitro Fertilization, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Claudia Bănescu
- Genetics Laboratory, Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Victoria Nyulas
- Departament of Informatics and Medical Biostatistics, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | | | - Vladut Săsăran
- Department of Obstetrics and Gynecology 2, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (M.M.); (V.S.)
| |
Collapse
|
12
|
Habtewold TD, Wijesiriwardhana P, Biedrzycki RJ, Zhang C, Grantz KL, Grewal J, Tekola-Ayele F. Longitudinal maternal glycemia during pregnancy and placental epigenetic age acceleration. Clin Epigenetics 2025; 17:19. [PMID: 39915864 PMCID: PMC11803985 DOI: 10.1186/s13148-025-01825-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/27/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Dysregulation of maternal glucose homeostasis has been related to an increased risk of morbidity and mortality in mothers and fetuses, yet the mechanism remains unclear. This study investigated the association between maternal glycemic levels and placental epigenetic age acceleration (PAA) in a multiethnic cohort. METHODS In a sample of 301 pregnant women (102 Hispanic, 77 White, 72 Black, and 50 Asian/Pacific Islander), the association of glycemic markers cumulative exposure with PAA was tested using linear regression adjusting for fetal sex, maternal age, educational status, and health insurance status. Models were applied in the full cohort and stratified by race/ethnicity. Further, sensitivity analyses were performed after excluding women with GDM or preeclampsia. RESULTS Among Black women, high glucose, HbA1c, and insulin cumulative exposure levels were associated with lower PAA compared to low cumulative exposure levels (β = - 0.75 weeks, 95% CI = - 1.41 to - 0.08); β = - 0.86, 95% CI = - 1.51 to - 0.21; and β = - 0.76, 95% CI = - 1.49 to - 0.03, respectively). Among Asian/Pacific Islander women, medium insulin cumulative exposure level was associated with lower PAA (β = - 0.94 weeks, 95% CI = - 1.74 to - 0.14). No significant association was observed among White and Hispanic women as well as in the full cohort. CONCLUSIONS Elevated glucose, HbA1c, and insulin cumulative levels throughout pregnancy were associated with lower PAA in Black and Asian/Pacific Islander women. Placental epigenetic aging may be altered by maternal elevated glycemia and may in part underlie early programming of health outcomes in pregnancy and childhood health outcomes.
Collapse
Grants
- HHSN275200800013C NICHD NIH HHS
- HHSN275200800002I NICHD NIH HHS
- HHSN27500006 Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH); American Recovery and Reinvestment Act
- HHSN275200800003IC Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH); American Recovery and Reinvestment Act
- HHSN275200800014C NICHD NIH HHS
- HHSN275200800012C NICHD NIH HHS
- HHSN275200800028C NICHD NIH HHS
- HHSN275201000009C NICHD NIH HHS
- HHSN27500008 Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH); American Recovery and Reinvestment Act
- NIH Office of the Director, the National Institute on Minority Health and Health Disparities and the National Institute of Diabetes and Digestive and Kidney Diseases
- National Institutes of Health (NIH)
Collapse
Affiliation(s)
- Tesfa Dejenie Habtewold
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, Room 3204, Bethesda, MD, 20892-7004, USA
| | - Prabhavi Wijesiriwardhana
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, Room 3204, Bethesda, MD, 20892-7004, USA
| | - Richard J Biedrzycki
- Glotech, Inc., Contractor for Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Cuilin Zhang
- Global Centre for Asian Women's Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Bia-Echo Asia Centre for Reproductive Longevity and Equality (ACRLE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Katherine L Grantz
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, Room 3204, Bethesda, MD, 20892-7004, USA
| | - Jagteshwar Grewal
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, Room 3204, Bethesda, MD, 20892-7004, USA
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, Room 3204, Bethesda, MD, 20892-7004, USA.
| |
Collapse
|
13
|
Saxena A, Minal, Pahwa P, Maras JS, Siddiqui H, Sevak JK, Mala YM, Tyagi S, Sarin SK, Trehanpati N. Immune-metabolic shifts in acute liver failure caused by HEV infection during pregnancy and their association with obstetric outcomes. Hepatol Commun 2025; 9:e0608. [PMID: 40209115 DOI: 10.1097/hc9.0000000000000608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/08/2024] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Hepatitis-E virus (HEV)-induced liver failure during pregnancy leads to maternal and fetal complications. This study investigates the HEV-associated metabolomic and immunological changes to elucidate the worsening of obstetric outcomes in patients with acute liver failure (ALF) due to HEV. METHODS Pregnant women with (i) acute viral hepatitis, IgM HEV positive (AVH-E, n = 31, Gr.I), (ii) acute liver failure (ALF-E, n = 15, Gr.II), (iii) acute hepatitis but negative for viral infections (non-HEV, n = 30, Gr.III), and healthy (HC, n = 21, Gr.IV) were evaluated at delivery for plasma untargeted metabolomics, cytokine, and immune profiling. RESULTS AVH-E and ALF-E (Gr.I, II) showed elevated TNF-α, IL-1β, IL-9, IL-22, and IL-33 compared to HC. In addition, in ALF-E, IFN-γ and IL-12p70 were decreased, but MIP-1α, fractalkine, SDF-1α, IL-22, and IL-33 were increased compared to AVH-E. Both AVH-E and ALF-E had decreased choline, sn-glycero-3-phosphocholine, O-palmitoyl-r-carnitine, and increased taurocholic acid. However, patients with ALF-E had a 2-5-fold decline in these metabolites with raised taurochenodeoxycholic acid. ALF-E showed increased naive T/B cells, decreased CD4, CD8 Tcm, Tem, and plasmablasts, compared to AVH-E contributing to higher failed inductions, preterm births, maternal complications like eclampsia, disseminated intravascular coagulation, preterm premature rupture of membranes, small-for-gestational-age infants, higher rates of intrauterine death, abortion, and mortality. CONCLUSIONS HEV infection reduces choline, phosphocholine, and palmitoyl carnitine, enhancing inflammation in ALF-E, while increasing taurocholic and taurochenodeoxycholic acids impairs the immune response. These factors together likely contribute to severe obstetric complications, including higher failed inductions, intrauterine death, and maternal and fetal mortality in ALF-E.
Collapse
Affiliation(s)
- Anoushka Saxena
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Minal
- Department of Obstetrics and Gynaecology, Maulana Azad Medical College, New Delhi, India
| | - Prabhjyoti Pahwa
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Jaswinder Singh Maras
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Hamda Siddiqui
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Jayesh Kumar Sevak
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Yedla Manikya Mala
- Department of Obstetrics and Gynaecology, Maulana Azad Medical College, New Delhi, India
| | - Shakun Tyagi
- Department of Obstetrics and Gynaecology, Maulana Azad Medical College, New Delhi, India
| | - Shiv K Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Nirupama Trehanpati
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
14
|
Ullah I, Khan SA, Nayab D, Ahmad S, Khan A, Ali N, Ahmad J, Ali Z, Safdar S, Khan MI. Long-Term Cardiovascular and Metabolic Health Outcomes of Gestational Diabetes Mellitus: A Systematic Review. Cureus 2025; 17:e79555. [PMID: 40144448 PMCID: PMC11938991 DOI: 10.7759/cureus.79555] [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] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
The public health concern associated with gestational diabetes mellitus (GDM) generates serious cardiovascular and metabolic impacts that affect both mothers whose condition went undiagnosed and their offspring. The review evaluates contemporary data about how GDM affects long-term health results. We searched published studies from databases such as PubMed, Scopus, and the Cochrane Library. A total of 710 records were identified, with 173 duplicates and 61 irrelevant records removed, leaving 476 for screening. After excluding 282, 194 reports were sought for retrieval, 97 were not retrieved, 97 were assessed for eligibility, 85 were excluded, and 12 studies were included in the review. Women with GDM face higher risks of type 2 diabetes (HR 2.5-7.1), hypertension (HR 3.2), and metabolic syndrome (HR 4.1). Their offspring have increased risks of obesity (OR 2.9), insulin resistance, cardiovascular disease, and metabolic disorders from childhood to adolescence. This review examines early detection biomarkers alongside strategies like lactation, probiotics, and CRP levels to potentially reduce maternal GDM risks. Although research had some methodological limitations regarding diagnostic irregularities and variable follow-up durations the findings demonstrate that early identification of GDM matters alongside customized care plans and long-term monitoring of women's health status. The review offers vital clinical guidelines and research pathways for advancing maternal and child health practices.
Collapse
Affiliation(s)
- Inayat Ullah
- General Medicine, Lady Reading Hospital Peshawar, Peshawar, PAK
| | - Samad Ali Khan
- General Surgery, Peshawar Institute of Medical Sciences, Peshawar, ESP
| | - Dure Nayab
- Internal Medicine, Lady Reading Hospital Peshawar, Peshawar, PAK
| | - Sohail Ahmad
- Internal Medicine, Lady Reading Hospital Peshawar, Peshawar, PAK
| | - Asmad Khan
- Internal Medicine, Hayatabad Medical Complex, Peshawar, PAK
| | - Nughman Ali
- Nephrology Medicine, One Brooklyn Hospital, New York City, USA
| | - Jamil Ahmad
- Internal Medicine, Lady Reading Hospital Peshawar, Peshawar, PAK
| | - Zeeshan Ali
- Medicine and Surgery, Liaquat College of Medicine and Dentistry, Karachi, PAK
| | - Sundas Safdar
- Diagnostic Radiology, Lady Reading Hospital Peshawar, Peshawar, PAK
| | | |
Collapse
|
15
|
Quaresima P, Myers SH, Pintaudi B, D’Anna R, Morelli M, Unfer V. Gestational diabetes mellitus and polycystic ovary syndrome, a position statement from EGOI-PCOS. Front Endocrinol (Lausanne) 2025; 16:1501110. [PMID: 39959624 PMCID: PMC11825335 DOI: 10.3389/fendo.2025.1501110] [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: 09/24/2024] [Accepted: 01/15/2025] [Indexed: 02/18/2025] Open
Abstract
Gestational diabetes mellitus is a worldwide health issue in pregnancy, posing a threat to both mother and child. One of the major risk factors for the development of gestational diabetes mellitus is polycystic ovary syndrome, primarily due to the biochemical hyperandrogenism and metabolic issues, commonly observed in these patients. In recent years, the Expert Group on Inositol in Basic and Clinical Research and on PCOS (EGOI-PCOS) has sought to better understand the pathogenesis behind polycystic ovary syndrome, in order to accurately diagnose and treat patients according to their individual needs. Through the scope of polycystic ovary syndrome, this position paper examines the characteristics of both conditions, and underlying biological mechanisms, before moving on to common treatment strategies to avoid or treat gestational diabetes mellitus in women with polycystic ovary syndrome.
Collapse
Affiliation(s)
- Paola Quaresima
- Department of Obstetrics and Gynecology, Azienda Sanitaria Provinciale di Cosenza, Cosenza, Italy
| | | | - Basilio Pintaudi
- The Experts Group on Inositol in Basic and Clinical Research and on PCOS (EGOI-PCOS), Rome, Italy
- Azienda Socio Sanitaria Territoriale (ASST) Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Rosario D’Anna
- The Experts Group on Inositol in Basic and Clinical Research and on PCOS (EGOI-PCOS), Rome, Italy
- Department of Human Pathology, University of Messina, Messina, Italy
| | - Michele Morelli
- Department of Obstetrics and Gynecology, Annunziata Hospital, Cosenza, Italy
| | - Vittorio Unfer
- The Experts Group on Inositol in Basic and Clinical Research and on PCOS (EGOI-PCOS), Rome, Italy
- UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy
| |
Collapse
|
16
|
Huang Q, Shire D, Hollis F, Abuaish S, Picard M, Monk C, Duman EA, Trumpff C. Associations between prenatal distress, mitochondrial health, and gestational age: findings from two pregnancy studies in the USA and Turkey. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.16.618719. [PMID: 39464008 PMCID: PMC11507865 DOI: 10.1101/2024.10.16.618719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Objective This study examined associations between mitochondrial markers-circulating cell-free mitochondrial DNA (cf-mtDNA) and Growth Differentiation Factor-15 (GDF15)-with maternal distress and pregnancy outcomes. Method Participants were drawn from two pregnancy studies, EPI (N=187, USA) and BABIP (N=198, Turkey). Plasma cf-mtDNA and GDF15 levels were quantified using qPCR and ELISA assays. Results Plasma cf-mtDNA levels did not significantly vary across pregnancy, while plasma GDF15 levels increased from early to late pregnancy and decreased postpartum. Late 2nd trimester plasma GDF15 was negatively correlated with pre-pregnancy BMI (p=0.035) and gestational age (p=0.0048) at birth. Early 2nd trimester maternal distress was associated with lower cf-mtDNA (p<0.05) and a trend for lower GDF15. Higher pre-pregnancy BMI and late-pregnancy maternal distress were linked to smaller postpartum GDF15 declines in EPI (p<0.05). Conclusion This study reveals distinct plasma cf-mtDNA and GDF15 patterns during the perinatal period, linking mitochondrial markers to maternal distress and pregnancy outcomes.
Collapse
Affiliation(s)
- Qiuhan Huang
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - David Shire
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Fiona Hollis
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Sameera Abuaish
- Department of Basic Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Kingdom of Saudi Arabia
| | - Martin Picard
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology, H. Houston Merritt Center, Neuromuscular Medicine Division, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Catherine Monk
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Elif Aysimi Duman
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem University, Istanbul, Turkey
- Institute of Natural and Applied Sciences, Acibadem University, Istanbul, Turkey
| | - Caroline Trumpff
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
17
|
Yuan Z, Su T, Yang L, Xi L, Wang HJ, Ji Y. Maternal Glycemia and Its Pattern Associated with Offspring Neurobehavioral Development: A Chinese Birth Cohort Study. Nutrients 2025; 17:257. [PMID: 39861387 PMCID: PMC11767945 DOI: 10.3390/nu17020257] [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: 11/30/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES This study investigates the impact of maternal glycemic levels during early and late pregnancy on offspring neurodevelopment in China. METHODS Fasting plasma glucose (FPG) and triglyceride (TG) levels were measured in maternal blood during pregnancy, and the TyG index was calculated to assess insulin resistance. Hyperglycemia was defined as FPG > 5.1 mmol/L. Neurodevelopmental outcomes in offspring aged 6-36 months were evaluated using the China Developmental Scale for Children, focusing on developmental delay (DD) and developmental quotient (DQ). Mothers were categorized into four glycemic groups: healthy glycemia group (HGG), early pregnancy hyperglycemia group (EHG), late pregnancy hyperglycemia group (LHG), and full-term hyperglycemia group (FHG). Linear and logistic regression models were applied. RESULTS Among 1888 mother-child pairs, hyperglycemia and FPG were associated with an increased risk of overall DD (aOR = 1.68; 95% CI 1.07-2.64) and lower DQ (aBeta = -1.53; 95% CI -2.70 to -0.36). Elevated FPG was linked to DD in fine motor and social behaviors. Compared to HGG, LHG and FHG significantly increased the risk of overall DD (aOR = 2.18; 95% CI 1.26-3.77; aOR = 2.64; 95% CI 1.38-5.05), whereas EHG did not. Male offspring were particularly vulnerable to early pregnancy hyperglycemia (aBeta = -2.80; 95% CI -4.36 to -1.34; aOR = 2.05; 95% CI 1.10-3.80). CONCLUSIONS Maternal glycemic levels during pregnancy influence offspring neurodevelopment, with persistent hyperglycemia significantly increasing DD risk. Early pregnancy hyperglycemia particularly affects male offspring, underscoring the need for glycemic management during pregnancy.
Collapse
Affiliation(s)
- Zhichao Yuan
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China
| | - Tao Su
- Tongzhou Maternal and Child Health Care Hospital of Beijing, Beijing 101101, China
| | - Li Yang
- Tongzhou Maternal and Child Health Care Hospital of Beijing, Beijing 101101, China
| | - Lei Xi
- Tongzhou Maternal and Child Health Care Hospital of Beijing, Beijing 101101, China
| | - Hai-Jun Wang
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China
| | - Yuelong Ji
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China
| |
Collapse
|
18
|
Açar Y, Köksal E. Anthropometric Measurements and Laboratory Methods for Pregnancy: An Update Review to Evaluation of Body Composition. Curr Nutr Rep 2025; 14:13. [PMID: 39776373 PMCID: PMC11706875 DOI: 10.1007/s13668-024-00597-x] [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] [Accepted: 11/18/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE OF REVIEW The aim of this review is to summarize and put into context the current evidence on anthropometric measurements and laboratory methods used in the evaluation of body composition in pregnancy, in the light of current studies. RECENT FINDINGS Protecting women's health during pregnancy, childbirth and the postnatal period is important for maternal health. Pregnancy is a critical period during which the female body undergoes significant changes to support fetal growth and development. Maternal changes in body composition are associated with fatal development and maternal health during pregnancy. Anthropometry is a simple, reliable, and low-cost method that can be applied globally in primary care for evaluating maternal nutritional status. Maternal anthropometry is important in predicting various complications that may occur during pregnancy, such as intrauterine growth retardation and the risk of low birth weight. In this context, anthropometric measurements such as body weight, skinfold thickness, and middle upper arm circumference, and laboratory methods such as body water, body density, bioelectrical impedance analysis, ultrasound, dual-energy X-ray absorptiometry, and magnetic resonance imaging are frequently used in estimating the body composition of pregnant women. In addition to body weight gain monitoring, it is possible to determine the body composition of a pregnant woman by using different anthropometric measurements and the proposed equations. Accurate evaluation of anthropometric measurements and body composition in pregnant women is important in protecting the health of the mother and baby and in the early diagnosis of complications that may occur during pregnancy.
Collapse
Affiliation(s)
- Yasemin Açar
- Department of Nutrition and Dietetics, Ondokuz Mayıs University, Samsun, Türkiye.
| | - Eda Köksal
- Department of Nutrition and Dietetics, Gazi University, Ankara, Türkiye
| |
Collapse
|
19
|
Khiat N, Girouard J, Kana Tsapi ES, Vaillancourt C, Van Themsche C, Reyes-Moreno C. TGFβ1 Restores Energy Homeostasis of Human Trophoblast Cells Under Hyperglycemia In Vitro by Inducing PPARγ Expression, AMPK Activation, and HIF1α Degradation. Cells 2025; 14:45. [PMID: 39791746 PMCID: PMC11720224 DOI: 10.3390/cells14010045] [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: 11/12/2024] [Revised: 12/23/2024] [Accepted: 01/01/2025] [Indexed: 01/12/2025] Open
Abstract
Elevated glucose levels at the fetal-maternal interface are associated with placental trophoblast dysfunction and increased incidence of pregnancy complications. Trophoblast cells predominantly utilize glucose as an energy source, metabolizing it through glycolysis in the cytoplasm and oxidative respiration in the mitochondria to produce ATP. The TGFβ1/SMAD2 signaling pathway and the transcription factors PPARγ, HIF1α, and AMPK are key regulators of cell metabolism and are known to play critical roles in extravillous trophoblast cell differentiation and function. While HIF1α promotes glycolysis over mitochondrial respiration, PPARγ and AMPK encourage the opposite. However, the interplay between TGFβ1 and these energy-sensing regulators in trophoblast cell glucose metabolism remains unclear. This study aimed to investigate whether and how TGFβ1 regulates energy metabolism in trophoblast cells exposed to normal and high glucose conditions. The trophoblast JEG-3 cells were incubated in normal (5 mM) and high (25 mM) glucose conditions for 24 h in the absence and the presence of TGFβ1. The protein expression levels of phosphor (p)-SMAD2, GLUT1/3, HIF1α, PPARγ, p-AMPK, and specific OXPHOS protein subunits were determined by western blotting, and ATP and lactate production by bioluminescent assay kits. JEG-3 cells exposed to 25 mM glucose decreased ATP production but did not affect lactate production. These changes led to a reduction in the expression levels of GLUT1/3, mitochondrial respiratory chain proteins, and PPARγ, coinciding with an increase in HIF1α expression. Conversely, TGFβ1 treatment at 25 mM glucose reduced HIF1α expression while enhancing the expression levels of GLUT1/3, PPARγ, p-AMPK, and mitochondrial respiratory chain proteins, thereby rejuvenating ATP production. Our findings reveal that high glucose conditions disrupt cellular glucose metabolism in trophoblast cells by perturbing mitochondrial oxidative respiration and decreasing ATP production. Treatment with TGFβ1 appears to counteract this trend, probably by enhancing both glycolytic and mitochondrial metabolism, suggesting a potential regulatory role of TGFβ1 in placental trophoblast cell glucose metabolism.
Collapse
Affiliation(s)
- Nihad Khiat
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boulevard des Forges, Trois-Rivières, QC G8Z 4M3, Canada; (N.K.); (J.G.); (E.S.K.T.); (C.V.T.)
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
| | - Julie Girouard
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boulevard des Forges, Trois-Rivières, QC G8Z 4M3, Canada; (N.K.); (J.G.); (E.S.K.T.); (C.V.T.)
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
| | - Emmanuelle Stella Kana Tsapi
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boulevard des Forges, Trois-Rivières, QC G8Z 4M3, Canada; (N.K.); (J.G.); (E.S.K.T.); (C.V.T.)
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
| | - Cathy Vaillancourt
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
- Institut National de la Recherche Scientifique (INRS)-Centre Armand Frappier Santé Biotechnologie and Research Centre CIUSSS-Nord-de-l’île-de-Montréal, Laval, QC H7V 1B7, Canada
| | - Céline Van Themsche
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boulevard des Forges, Trois-Rivières, QC G8Z 4M3, Canada; (N.K.); (J.G.); (E.S.K.T.); (C.V.T.)
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
| | - Carlos Reyes-Moreno
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boulevard des Forges, Trois-Rivières, QC G8Z 4M3, Canada; (N.K.); (J.G.); (E.S.K.T.); (C.V.T.)
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
| |
Collapse
|
20
|
Lin L, Chen R, Lin H, Yao T, Chen Z, Zheng L. Influence of parity on weight gain during pregnancy in women with Gestational Diabetes: A retrospective cohort study. Pak J Med Sci 2025; 41:29-36. [PMID: 39867753 PMCID: PMC11755282 DOI: 10.12669/pjms.41.1.11412] [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: 11/13/2024] [Revised: 11/22/2024] [Accepted: 12/20/2024] [Indexed: 01/28/2025] Open
Abstract
Objective This retrospective cohort study aimed to investigate the effects of parity on gestational weight gain (GWG) and its association with maternal and neonatal outcomes in women with gestational diabetes mellitus (GDM). Methods This retrospective cohort study data from 2,909 pregnant women with GDM who delivered between 2021 and 2023 at Fujian Maternity and Child Health hospital, were analyzed. Participants were categorized into nulliparous (no previous births), primiparous (one previous birth), and multiparous (two or more previous births) groups. Women aged 18-45 years with singleton pregnancies were included, while those with multiple pregnancies, pre-existing diabetes, hypertension, thyroid disorders, or incomplete BMI data were excluded. Data extracted from hospital records included demographic information, BMI, GWG at various gestational stages, and pregnancy outcomes. Statistical analysis involved logistic regression models, adjusting for confounders such as maternal age, BMI, and gestational age at delivery. P-values < 0.05 were considered significant. Results Primiparous and multiparous women exhibited significantly higher GWG than nulliparous women (p < 0.01). Primiparous women had reduced risks of pregnancy-induced hypertension (adjusted OR, 0.64; 95% CI, 0.45-0.92) and premature rupture of membranes (adjusted OR, 0.58; 95% CI, 0.48-0.70). Both primiparous and multiparous women had lower risks of delivering large-for-gestational-age newborns or those with neonatal jaundice. Conclusion Parity significantly influences GWG and maternal/neonatal outcomes in GDM pregnancies, highlighting the need for parity-specific guidelines.
Collapse
Affiliation(s)
- Lin Lin
- Lin Lin Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics, Gynecology and Pediatrics, Fujian Medical University. P.R. China
| | - Ruiyun Chen
- Ruiyun Chen Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics, Gynecology and Pediatrics, Fujian Medical University. P.R. China
| | - Haifeng Lin
- Haifeng Lin Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics, Gynecology and Pediatrics, Fujian Medical University. P.R. China
| | - Ting Yao
- Ting Yao Fujian Medical University, Fuzhou, Fujian Province 350001, P.R. China
| | - Zihua Chen
- Zihua Chen Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics, Gynecology and Pediatrics, Fujian Medical University. P.R. China
| | - Lianghui Zheng
- Lianghui Zheng Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics, Gynecology and Pediatrics, Fujian Medical University. P.R. China
| |
Collapse
|
21
|
Meda C, Dolce A, Torre SD. Metabolic dysfunction-associated steatotic liver disease across women's reproductive lifespan and issues. Clin Mol Hepatol 2025; 31:327-332. [PMID: 39098816 PMCID: PMC11791579 DOI: 10.3350/cmh.2024.0419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024] Open
Affiliation(s)
- Clara Meda
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Arianna Dolce
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Sara Della Torre
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
22
|
Ding L, Yin X, Wen G, Sun D, Xian D, Zhao Y, Zhang M, Yang W, Chen W. Prediction of preterm birth using machine learning: a comprehensive analysis based on large-scale preschool children survey data in Shenzhen of China. BMC Pregnancy Childbirth 2024; 24:810. [PMID: 39633287 PMCID: PMC11616287 DOI: 10.1186/s12884-024-06980-4] [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: 08/03/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Preterm birth (PTB) is a significant cause of neonatal mortality and long-term health issues. Accurate prediction and timely prevention of PTB are essential for reducing associated child mortality and morbidity. Traditional predictive methods face challenges due to heterogeneous risk factors and their interaction effects. This study aims to develop and evaluate six machine learning (ML) models to predict PTB using large-scale children survey data from Shenzhen, China, and to identify key predictors through Shapley Additive Explanations (SHAP) analysis. METHODS Data from 84,050 mother-child pairs, collected in 2021 and 2022, were processed and divided into training, validation, and test sets. Six ML models were tested: L1-Regularised Logistic Regression, Light Gradient Boosting Machine (LightGBM), Naive Bayes, Random Forests, Support Vector Machine, and Extreme Gradient Boosting (XGBoost). Model performance was evaluated based on discrimination, calibration and clinical utility. SHAP analysis was used to interpret the importance and impact of individual features on PTB prediction. RESULTS The XGBoost model demonstrated the best overall performance, with the area under the receiver operating characteristic curve (AUC) scores of 0.752 and 0.757 in the validation and test sets, respectively, along with favorable calibration and clinical utility. Key predictors identified were multiple pregnancies, threatened abortion, and maternal age of conception. SHAP analysis highlighted the positive impacts of multiple pregnancies and threatened abortion, as well as the negative impact of micronutrient supplementation on PTB. CONCLUSION Our study found that ML models, particularly XGBoost, show promise in accurately predicting PTB and identifying key risk factors. These findings provide the potential of ML for enhancing clinical interventions, personalizing prenatal care, and informing public health initiatives.
Collapse
Affiliation(s)
- Liwen Ding
- Department of Epidemiology and Health Statistics, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xiaona Yin
- Women's and Children's Hospital of Longhua District of Shenzhen, Shenzhen, 518109, China
| | - Guomin Wen
- Women's and Children's Hospital of Longhua District of Shenzhen, Shenzhen, 518109, China
| | - Dengli Sun
- Women's and Children's Hospital of Longhua District of Shenzhen, Shenzhen, 518109, China
| | - Danxia Xian
- Women's and Children's Hospital of Longhua District of Shenzhen, Shenzhen, 518109, China
| | - Yafen Zhao
- Women's and Children's Hospital of Longhua District of Shenzhen, Shenzhen, 518109, China
| | - Maolin Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Weikang Yang
- Women's and Children's Hospital of Longhua District of Shenzhen, Shenzhen, 518109, China.
| | - Weiqing Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China.
- School of Health Management, Xinhua College of Guangzhou, Guangzhou, 510080, China.
| |
Collapse
|
23
|
Farghaly MAA, Abuelazm S, Farrell RM, Elgendy MM, Grove D, Abu-Shaweesh JM, Dweik RA, Aly H. Exhaled Breath Volatile Organic Compounds in Pregnancy: A Pilot Study. Am J Perinatol 2024. [PMID: 39515786 DOI: 10.1055/a-2463-5352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
OBJECTIVE This study aimed to assess the volatile organic compounds (VOC)s in breath samples collected noninvasively from pregnant women during pregnancy and postpartum and compare it with nonpregnant controls. STUDY DESIGN This pilot study included 50 subjects: 10 pregnant patients in their first trimester, 10 in second trimester, 10 in third trimester, 10 in the first postpartum week, and 10 nonpregnant subjects as a control. We collected exhaled breath from subjects who reported to be healthy and free of any respiratory symptoms. Clinical and demographic data were collected. The samples were analyzed for VOCs using a selected-ion flow-tube mass spectrometer. RESULTS The VOCs monitored were 22 compounds selected for their common presence in exhaled breath. During pregnancy and postpartum period, there were differences in five compounds mainly: 2-propanol, acrylonitrile, 1-nonene, 2-nonene, and hydrogen sulfide. Significant differences in VOCs were identified during each trimester compared with controls. CONCLUSION VOCs could be measured safely and noninvasively in pregnant women. VOCs differed significantly among nonpregnant, pregnant women, and postpartum period. The utilization of this novel assay to identify fetal conditions or identifying women at risk of premature delivery, which should be further investigated in future studies. KEY POINTS · This study demonstrated that VOCs could be measured noninvasively in exhaled breath of pregnant women.. · VOCs differed significantly among women who were nonpregnant, pregnant, and in postpartum period.. · These findings might serve as a base to look for potential markers for maternal or fetal conditions..
Collapse
Affiliation(s)
| | | | - Ruth M Farrell
- OB/GYN Health Institute, Cleveland Clinic, Cleveland, Ohio
- Center for Bioethics, Cleveland Clinic, Cleveland, Ohio
| | | | - David Grove
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | | | - Raed A Dweik
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Hany Aly
- Neonatology Department, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
24
|
Chávez-Manzanera EA, Vera-Zertuche JM, Kaufer-Horwitz M, Vázquez-Velázquez V, Flores-Lázaro JR, Mireles-Zavala L, Calzada-León R, Garnica-Cuellar JC, Sánchez-Muñoz V, Ramírez-Butanda E, Hernández-González R, Vargas-Martínez MA, Laviada-Molina H, Violante-Ortíz R, Esquivias-Zavala H, García-García E, Lavalle-González FJ, Mancillas-Adame L, López-Alvarenga JC, Pérez-Hernández JF, Soto-Fuentes EV, Soriano-Cortés RR, Goicoechea-Turcott EW, Magallanes-Díaz G, Herrera-Hernández MF, Barquera-Cervera S, Vargas-Contreras E, Díaz-Wionczek CB, Salmon M, Jesús DRD, Villaseñor-Díaz JP, Peña J, Ramos-Rojas J, Ávila-Oliver C, Rada G, Hussey B, Salas XR. Mexican Clinical Practice Guidelines for Adult Overweight and Obesity Management. Curr Obes Rep 2024; 13:643-666. [PMID: 39356455 PMCID: PMC11522083 DOI: 10.1007/s13679-024-00585-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2024] [Indexed: 10/03/2024]
Abstract
PURPOSE To develop Mexico's first methodologically rigorous clinical practice guideline for the management of adult overweight and obesity. The target audiences are interdisciplinary healthcare professionals across healthcare systems who are the first point of contact for patients with obesity in Mexico, patients, and health system decision makers. RECENT FINDINGS A review of recent international obesity clinical practice guidelines and an expert consensus process identified: i) common recommendations appropriate for implementation in Mexico and ii) knowledge gaps requiring the formulation of new recommendations. In all, 20 new recommendations and 20 good practice statements were developed using the GRADE Evidence-to-Decision Framework and expert consensus. Overweight and obesity negatively impact the health and well-being of individuals and populations in Mexico. This guideline aims to establish a new evidence-based, patient-centered, non-stigmatizing, and practical treatment and management framework, based on the fundamental principles of chronic disease prevention and management.
Collapse
Affiliation(s)
- Emma A Chávez-Manzanera
- Obesity Workgroup, Sociedad Mexicana de Nutrición y Endocrinología, Mexico City, México.
- Obesity and Eating Disorders Clinic, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México.
| | - Juan M Vera-Zertuche
- Obesity Workgroup, Sociedad Mexicana de Nutrición y Endocrinología, Mexico City, México
- ABC Medical Center, Mexico City, México
| | - Martha Kaufer-Horwitz
- Obesity Workgroup, Sociedad Mexicana de Nutrición y Endocrinología, Mexico City, México
- Nutrition Division, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Verónica Vázquez-Velázquez
- Obesity and Eating Disorders Clinic, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
- Obesidades S.C, Mexico City, México
| | - José R Flores-Lázaro
- Sports Medicine Division, Universidad Nacional Autónoma de México, Mexico City, México
| | - Leonor Mireles-Zavala
- Obesity Workgroup, Sociedad Mexicana de Nutrición y Endocrinología, Mexico City, México
| | - Raúl Calzada-León
- Obesity Workgroup, Sociedad Mexicana de Nutrición y Endocrinología, Mexico City, México
- Service of Endocrinology, Instituto Nacional de Pediatría, Mexico City, México
| | - Juan C Garnica-Cuellar
- Obesity Workgroup, Sociedad Mexicana de Nutrición y Endocrinología, Mexico City, México
- Endocrinology Division, Centro Médico Nacional 20 de Noviembre, Institute for Social Security and Services for State, Mexico City, México
| | | | - Eduardo Ramírez-Butanda
- Obesity and Eating Disorders Clinic, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | | | - María A Vargas-Martínez
- Department of Neurology and Psychiatry, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Hugo Laviada-Molina
- Obesity Workgroup, Sociedad Mexicana de Nutrición y Endocrinología, Mexico City, México
- School of Health Sciences, Universidad Marista de Mérida, Mérida, México
| | - Rafael Violante-Ortíz
- Obesity Workgroup, Sociedad Mexicana de Nutrición y Endocrinología, Mexico City, México
- Tampico Faculty of Medicine Alberto Romo Caballero, Universidad Autónoma de Tamaulipas, Tampico, Tamaulipas, México
| | - Héctor Esquivias-Zavala
- Department for Continuing Education, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City, México
| | - Eduardo García-García
- Obesity Workgroup, Sociedad Mexicana de Nutrición y Endocrinología, Mexico City, México
| | - Fernando J Lavalle-González
- Obesity Workgroup, Sociedad Mexicana de Nutrición y Endocrinología, Mexico City, México
- Endocrine Service, Hospital Universitario Dr José E. González, Medicine School, Universidad Autónoma de Nuevo León, Nuevo Leon, México
| | - Leonardo Mancillas-Adame
- Obesity Workgroup, Sociedad Mexicana de Nutrición y Endocrinología, Mexico City, México
- Institute for Obesity Research, Instituto Tecnológico de Monterrey, Nuevo Leon, México
- Internal Medicine Division, Medical School, and University Hospital, Universidad Autónoma de Nuevo Leon, Nuevo Leon, México
| | - Juan C López-Alvarenga
- Obesity Workgroup, Sociedad Mexicana de Nutrición y Endocrinología, Mexico City, México
- Population Health & Biostatistics, School of Medicine, University of Texas Rio Grande Valley, Texas, United States
| | - Juan F Pérez-Hernández
- Obesity and Eating Disorders Clinic, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Erika V Soto-Fuentes
- Obesity and Eating Disorders Clinic, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Reina R Soriano-Cortés
- Obesity and Eating Disorders Clinic, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | | | - Gerardo Magallanes-Díaz
- Department of Secondary Prevention Central level, Servicios de Salud IMSS-BIENESTAR, Mexico City, México
| | | | - Simón Barquera-Cervera
- School of Public Health of Mexico at, Instituto Nacional de Salud Pública, Morelos, México
| | - Edith Vargas-Contreras
- Department of Endocrinology, Hospital General de México "Dr. Eduardo Liceaga, Mexico City, México
| | | | | | | | | | | | | | | | | | - Brad Hussey
- Replica Communications, Hamilton, Ontario, Canada
| | | |
Collapse
|
25
|
Pisek A, McKinney CM, Muktabhant B, Pitiphat W. Maternal Metabolic Status and Orofacial Cleft Risk: A Case-Control Study in Thailand. Int Dent J 2024; 74:1413-1423. [PMID: 38614877 PMCID: PMC11551577 DOI: 10.1016/j.identj.2024.02.005] [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: 12/01/2023] [Revised: 01/31/2024] [Accepted: 02/13/2024] [Indexed: 04/15/2024] Open
Abstract
OBJECTIVES Metabolic syndrome (MetS) has been suggested to play a role in congenital defects. This study investigated the association of MetS and its components with orofacial clefts (OFCs). METHODS We conducted a case-control study in Northeast Thailand. Ninety-four cases with cleft lip, with or without cleft palate, were frequency matched with 94 controls on the infant's age and mother's education. We administered a mother's health questionnaire and collected anthropometric measurements and blood samples. Multiple logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs). Subgroup analyses were performed among infants without a family history of OFCs, mothers who were not currently breastfeeding, and mothers who were >6 months postpartum. RESULTS When compared to mothers of normal weight, the OR associated with OFCs were 2.44 (95% CI, 1.04-5.76, P = .04) in overweight mothers, and 3.30 (95% CI, 1.14-9.57, P = .03) in obese mothers. Low HDL-C raised the risk of OFCs 2.95 times (95% CI, 1.41-6.14, P = .004) compared to normal HDL-C levels. Mothers with 4 or 5 features of MetS were 2.77 times as likely to have the affected child than those who did not (95% CI, 0.43-17.76), but this difference was not statistically significant (P = .28). Subgroup analyses showed similar results, uncovering an additional significant association between underweight mothers and OFCs. CONCLUSIONS The results indicate a robust association between underweight and overweight/obese maternal body mass index and increased OFC risk. Additionally, low HDL-C in mothers is linked to an elevated risk of OFCs. Further research is needed to evaluate if promoting strategies to maintain optimal body weight and enhance HDL-C levels in reproductive-age and pregnant women icould contribute to a reduction of the risk of OFCs in their progeny.
Collapse
Affiliation(s)
- Araya Pisek
- Division of Dental Public Health, Department of Preventive Dentistry, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | - Christy M McKinney
- Division of Craniofacial Medicine, Department of Pediatrics, University of Washington, and Seattle Children's Research Institute, Seattle, Washington, USA
| | - Benja Muktabhant
- Department of Public Health Administration, Health Promotion and Nutrition, Faculty of Public Health, Khon Kaen University, Khon Kaen, Thailand
| | - Waranuch Pitiphat
- Division of Dental Public Health, Department of Preventive Dentistry, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
26
|
Pepin AS, Jazwiec PA, Dumeaux V, Sloboda DM, Kimmins S. Determining the effects of paternal obesity on sperm chromatin at histone H3 lysine 4 tri-methylation in relation to the placental transcriptome and cellular composition. eLife 2024; 13:e83288. [PMID: 39612469 PMCID: PMC11717366 DOI: 10.7554/elife.83288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/28/2024] [Indexed: 12/01/2024] Open
Abstract
Paternal obesity has been implicated in adult-onset metabolic disease in offspring. However, the molecular mechanisms driving these paternal effects and the developmental processes involved remain poorly understood. One underexplored possibility is the role of paternally induced effects on placenta development and function. To address this, we investigated paternal high-fat diet-induced obesity in relation to sperm histone H3 lysine 4 tri-methylation signatures, the placenta transcriptome, and cellular composition. C57BL6/J male mice were fed either a control or high-fat diet for 10 weeks beginning at 6 weeks of age. Males were timed-mated with control-fed C57BL6/J females to generate pregnancies, followed by collection of sperm, and placentas at embryonic day (E)14.5. Chromatin immunoprecipitation targeting histone H3 lysine 4 tri-methylation (H3K4me3) followed by sequencing (ChIP-seq) was performed on sperm to define obesity-associated changes in enrichment. Paternal obesity corresponded with altered sperm H3K4me3 at promoters of genes involved in metabolism and development. Notably, altered sperm H3K4me3 was also localized at placental enhancers. Bulk RNA-sequencing on placentas revealed paternal obesity-associated sex-specific changes in expression of genes involved in hypoxic processes such as angiogenesis, nutrient transport, and imprinted genes, with a subset of de-regulated genes showing changes in H3K4me3 in sperm at corresponding promoters. Paternal obesity was also linked to impaired placenta development; specifically, a deconvolution analysis revealed altered trophoblast cell lineage specification. These findings implicate paternal obesity effects on placenta development and function as one potential developmental route to offspring metabolic disease.
Collapse
Affiliation(s)
- Anne-Sophie Pepin
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill UniversityMontrealCanada
| | - Patrycja A Jazwiec
- Department of Biochemistry and Biomedical Sciences, McMaster UniversityHamiltonCanada
| | - Vanessa Dumeaux
- Departments of Anatomy & Cell Biology and Oncology, Western UniversityLondonCanada
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster UniversityHamiltonCanada
- Farncombe Family Digestive Health Research Institute, McMaster University HamiltonHamiltonCanada
- Departments of Obstetrics and Gynecology, and Pediatrics, McMaster UniversityHamiltonCanada
| | - Sarah Kimmins
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill UniversityMontrealCanada
- Department of Pathology and Molecular Biology, University of Montreal, University of Montreal Hospital Research CenterMontrealCanada
| |
Collapse
|
27
|
Karcz K, Królak-Olejnik B. Impact of Gestational Diabetes Mellitus on Fetal Growth and Nutritional Status in Newborns. Nutrients 2024; 16:4093. [PMID: 39683486 DOI: 10.3390/nu16234093] [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: 11/09/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is one of the most prevalent complications associated with pregnancy, exhibiting a gradual rise in prevalence worldwide. Given the potential for numerous short- and long-term complications for both mother and child, patients diagnosed with GDM require individualised treatment to compensate for metabolic abnormalities and ultimately reduce the risk of the known adverse consequences of impaired glucose tolerance. METHODS AND RESULTS The manuscript presents a summary of the current knowledge on changes in maternal metabolism during physiological pregnancy and pregnancy complicated by gestational diabetes. Furthermore, the article provides a synthesis of the findings from recent research examining the impacts of gestational diabetes and the therapeutic modalities employed on the nutritional status of the fetus and neonate. Additionally, the review elucidates the function of the placenta and placental hormones in fetal development, as well as the impact of hyperglycemia, insulin resistance and adipokines on fetal and neonatal nutritional programming and predisposition to metabolic complications in adulthood. CONCLUSIONS The metabolic environment, resulting from abnormal glucose tolerance during pregnancy, exerts a particularly significant impact on fetal growth and, consequently, on the birth weight and fat mass of the newborn infants. This is a pivotal factor influencing the nutritional and metabolic programming of the developing fetus, predisposing the individual to the development of metabolic complications throughout their lifetime.
Collapse
Affiliation(s)
- Karolina Karcz
- Department of Neonatology, Wroclaw Medical University, 50-367 Wrocław, Poland
| | | |
Collapse
|
28
|
Łuszczki E, Wyszyńska J, Dymek A, Drożdż D, González-Ramos L, Hartgring I, García-Carbonell N, Mazur A, Erdine S, Parnarauskienė J, Alvarez-Pitti J. The Effect of Maternal Diet and Lifestyle on the Risk of Childhood Obesity. Metabolites 2024; 14:655. [PMID: 39728436 DOI: 10.3390/metabo14120655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: Childhood obesity is a global health problem that affects at least 41 million children under the age of five. Increased BMI in children is associated with serious long-term health consequences, such as type 2 diabetes, cardiovascular disease, and psychological problems, including depression and low self-esteem. Although the etiology of obesity is complex, research suggests that the diet and lifestyle of pregnant women play a key role in shaping metabolic and epigenetic changes that can increase the risk of obesity in their children. Excessive gestational weight gain, unhealthy dietary patterns (including the Western diet), and pregnancy complications (such as gestational diabetes) are some of the modifiable factors that contribute to childhood obesity. The purpose of this narrative review is to summarize the most important and recent information on the impact of the diet and lifestyle of pregnant women on the risk of childhood obesity. Methods: This article is a narrative review that aims to summarize the available literature on the impact of pregnant women's diet and lifestyle on the risk of obesity in their offspring, with a focus on metabolic and epigenetic mechanisms. Results/Conclusions: Current evidence suggests that a pregnant woman's lifestyle and diet can significantly contribute to lowering the risk of obesity in their offspring. However, further high-quality research is needed to understand better the metabolic and epigenetic relationships concerning maternal factors that predispose offspring to obesity.
Collapse
Affiliation(s)
- Edyta Łuszczki
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Justyna Wyszyńska
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Agnieszka Dymek
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Dorota Drożdż
- Department of Pediatric Nephrology and Hypertension, Pediatric Institute, Jagiellonian University Medical College, 31-007 Krakow, Poland
| | - Laura González-Ramos
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
| | - Isa Hartgring
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
| | - Nuria García-Carbonell
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
- Pediatric Department, Consorcio Hospital General, University of Valencia, 46014 Valencia, Spain
| | - Artur Mazur
- Institute of Medical Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Serap Erdine
- Cerrahpasa Faculty of Medicine, Department of Cardiology, Istanbul University-Cerrahpasa, 34320 Istanbul, Turkey
| | - Justė Parnarauskienė
- Pediatric Department, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Julio Alvarez-Pitti
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
- Pediatric Department, Consorcio Hospital General, University of Valencia, 46014 Valencia, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
29
|
Sharifat R, Borazjani F, Araban M, Pakpour AH, Angali KA, Aiiashi S. Nutritional education on health beliefs, metabolic profiles, and quality of life among high-risk pregnant women for gestational diabetes mellitus: a randomized controlled trial. Sci Rep 2024; 14:27712. [PMID: 39532953 PMCID: PMC11558005 DOI: 10.1038/s41598-024-78447-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
In recent years, nutrition has become increasingly important in treating and managing gestational diabetes mellitus. The Health Belief Model (HBM) is a conceptual framework in health behavior research used in some medical research. The present study aimed to evaluate the effect of glycemic index training based on the HBM on metabolic indicators and pregnant women's health-related quality of life. In this open-label, parallel-controlled randomized trial, 90 pregnant women from primary health centers in Iran were recruited and randomly assigned to either the intervention group or the control group, using a block randomization method with a block size of six. The intervention group underwent 12 weeks of nutritional education on the glycemic index and load of foodstuff. The control group only received routine health care. Valid and reliable questionnaires included demographic and prenatal information, physical activity, three days of food records, quality of life (SF-12), and HBM constructs. Fasting blood samples were taken at baseline and end of the study, to assess fasting blood glucose (FBS), fasting insulin, hs-CRP, and lipids. Eighty-four pregnant women with an age mean of 30.12 ± 4.35 completed 12 weeks of intervention. At baseline, there were no significant differences between groups in the HBM subscales (P > 0.05). However, within the intervention group, there was an increase in perceived susceptibilities (mean change 1.45 ± 4.21; P = 0.03) and cue to action (mean change: 1.22 ± 3.38; P = 0.02). In the adjusted model, the General Health subscale of Quality of Life showed a significant increase in the comparison between groups (mean change 13.69 ± 29.83 vs. 0.00 ± 29.58; P = 0.04). Additionally, the adjusted model revealed a notable difference between the groups in serum hs-CRP level, (mean change -0.877 ± 3.47 vs. -0.067 ± 3.40; P = 0.01) and triglyceride level (mean change - 55.24 ± 111.21 vs. 40.92 ± 142.01; P = 0.001). However, in the adjusted model, the total cholesterol levels increased between groups (mean change 22.75 ± 66.17 vs. 30.12 ± 61.33; P = 0.01) at the end of the study. There was no significant difference in glycemic indices (P > 0.05). Participation in a nutrition education program might positively impact nutritional beliefs, behaviors, and some biochemical indicators among pregnant women. Future studies with larger sample sizes and longer follow-ups are warranted.Trial registration number: IRCTID: IRCT20190227042858N1. Registration date: 2019-07-18.
Collapse
Affiliation(s)
- Robab Sharifat
- Nutrition and Metabolic Diseases Research Center, Clinical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Borazjani
- Nutrition and Metabolic Diseases Research Center, Clinical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Marzieh Araban
- Menopause Andropause Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Amir H Pakpour
- Department of Nursing, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Kambiz Ahmadi Angali
- Social Determinants of Health Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Statistics and Epidemiology, School of Health Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | |
Collapse
|
30
|
Hua L, Yang Y, Zhang H, Jiang X, Jin C, Feng B, Che L, Xu S, Lin Y, Wu D, Zhuo Y. Adipocyte FGF21 Signaling Defect Aggravated Adipose Tissue Inflammation in Gestational Diabetes Mellitus. Nutrients 2024; 16:3826. [PMID: 39599611 PMCID: PMC11597770 DOI: 10.3390/nu16223826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is associated with increased inflammation in adipose tissues. Fibroblast growth factor 21 (FGF21) is an endocrine hormone which signals to multiple tissues to regulate metabolism. However, its role in GDM remains largely unknown. In this study, we found that impaired FGF21 signaling in GDM correlates with worsened inflammation and insulin resistance in white adipose tissues in mice. Mechanistically, the pregnancy-related upregulation of FGF21 signaling in adipocytes promotes the differentiation of regulatory T cells (Tregs), which are critical for reducing pregnancy-induced adipose tissue inflammation. The anti-inflammatory effects of FGF21 may involve linolenic acid-mediated PGE2 synthesis in adipocytes. These findings underscore FGF21's role in mediating crosstalk between mature adipocytes and immune cells in white adipose tissue and suggest that targeting FGF21 signaling and its downstream metabolites could offer a potential therapeutic approach for GDM in humans.
Collapse
Affiliation(s)
- Lun Hua
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (L.H.); (X.J.); (Y.L.)
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yi Yang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (L.H.); (X.J.); (Y.L.)
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Haoqi Zhang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (L.H.); (X.J.); (Y.L.)
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xuemei Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (L.H.); (X.J.); (Y.L.)
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Chao Jin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (L.H.); (X.J.); (Y.L.)
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (L.H.); (X.J.); (Y.L.)
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lianqiang Che
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (L.H.); (X.J.); (Y.L.)
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shengyu Xu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (L.H.); (X.J.); (Y.L.)
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Lin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (L.H.); (X.J.); (Y.L.)
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - De Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (L.H.); (X.J.); (Y.L.)
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yong Zhuo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (L.H.); (X.J.); (Y.L.)
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
31
|
Poudel SP, Behura SK. Sex-bias metabolism of fetal organs, and their relationship to the regulation of fetal brain-placental axis. Metabolomics 2024; 20:126. [PMID: 39495316 DOI: 10.1007/s11306-024-02189-w] [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: 08/05/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION The placenta plays influential role in the fetal development of mammals. But how the metabolic need of the fetal organs is related to that of the placenta, and whether this relationship is influenced by the sex of the fetus remain poorly understood. OBJECTIVES This study used pigs to investigate metabolomic signatures of male and female fetal organs, and determine the relevance of gene expression of the placenta and brain to the metabolism of peripheral organs. METHODS Untargeted metabolomics analysis was performed with the day-45 placenta, kidney, heart, liver, lung and brain of male and female pig fetuses to model sex differences in the metabolism of the peripheral organs relative to that of the brain and placenta. Transcriptomic analysis was performed to investigate the expression of metabolic genes in the placenta and fetal brain of both sexes. RESULTS The results of this study show that the fetoplacental metabolic regulation was not only influenced by the fetal sex but also dependent on the metabolic requirement of the individual organs of the fetus. Neural network modeling of metabolomics data revealed differential relationship of the metabolic changes of the peripheral organs with the placenta and fetal brain between males and females. RNA sequencing further showed that genes associated with the metabolism of the peripheral organs were differentially expressed in the placenta and fetal brain. CONCLUSION The findings of this study suggest a regulatory role of the fetal brain and placenta axis in the sex-bias metabolism of the peripheral organs.
Collapse
Affiliation(s)
- Shankar P Poudel
- Division of Animal Sciences, University of Missouri, 920 East Campus Drive, Columbia, MO, 65211, USA
| | - Susanta K Behura
- Division of Animal Sciences, University of Missouri, 920 East Campus Drive, Columbia, MO, 65211, USA.
- MU Institute for Data Science and Informatics, University of Missouri, 920 East Campus Drive, Columbia, MO, 65211, USA.
- Interdisciplinary Reproduction and Health Group, University of Missouri, 920 East Campus Drive, Columbia, MO, 65211, USA.
- Interdisciplinary Neuroscience Program, University of Missouri, 920 East Campus Drive, Columbia, MO, 65211, USA.
| |
Collapse
|
32
|
Valencia-Ortega J, Castillo-Santos A, Molerés-Orduña M, Solis-Paredes JM, Saucedo R, Estrada-Gutierrez G, Camacho-Arroyo I. Influence of Maternal Adipokines on Anthropometry, Adiposity, and Neurodevelopmental Outcomes of the Offspring. Int J Mol Sci 2024; 25:11655. [PMID: 39519203 PMCID: PMC11547085 DOI: 10.3390/ijms252111655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/17/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Pregnancy is distinguished by a multitude of intricate interactions between the mother and the new individual, commencing at implantation and persisting until the maturation and integration of the fetal apparatus and systems. The physiological increase in fat mass during pregnancy and the association of maternal obesity with adverse neonatal outcomes have directed attention to the study of maternal adipokines as participants in fetal development. Interestingly, maternal concentrations of certain adipokines such as adiponectin, leptin, tumor necrosis factor-alpha, and interleukin-6 have been found to be associated with offspring anthropometry and adiposity at birth and at three months of age, even with neurodevelopmental alterations later in life. This is partly explained by the functions of these adipokines in the regulation of maternal metabolism and placental nutrient transport. This review compiles, organizes, and analyzes the most relevant studies on the association between maternal adipokines with anthropometry, adiposity, and neurodevelopmental outcomes of the offspring. Furthermore, it proposes the underlying mechanisms involved in this association.
Collapse
Affiliation(s)
- Jorge Valencia-Ortega
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, Mexico;
| | - Andrea Castillo-Santos
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (A.C.-S.); (M.M.-O.); (J.M.S.-P.)
| | - Miranda Molerés-Orduña
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (A.C.-S.); (M.M.-O.); (J.M.S.-P.)
| | - Juan Mario Solis-Paredes
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (A.C.-S.); (M.M.-O.); (J.M.S.-P.)
| | - Renata Saucedo
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Guadalupe Estrada-Gutierrez
- Department of Immunobiochemistry, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico;
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, Mexico;
| |
Collapse
|
33
|
Torres-Torres J, Monroy-Muñoz IE, Perez-Duran J, Solis-Paredes JM, Camacho-Martinez ZA, Baca D, Espino-Y-Sosa S, Martinez-Portilla R, Rojas-Zepeda L, Borboa-Olivares H, Reyes-Muñoz E. Cellular and Molecular Pathophysiology of Gestational Diabetes. Int J Mol Sci 2024; 25:11641. [PMID: 39519193 PMCID: PMC11546748 DOI: 10.3390/ijms252111641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Gestational diabetes (GD) is a metabolic disorder characterized by glucose intolerance during pregnancy, significantly impacting maternal and fetal health. Its global prevalence is approximately 14%, with risk factors including obesity, family history of diabetes, advanced maternal age, and ethnicity, which are linked to cellular and molecular disruptions in glucose regulation and insulin resistance. GD is associated with short- and long-term complications for both the mother and the newborn. For mothers, GD increases the risk of developing type 2 diabetes, cardiovascular diseases, and metabolic syndrome. In the offspring, exposure to GD in utero predisposes them to obesity, glucose intolerance, and metabolic disorders later in life. This review aims to elucidate the complex cellular and molecular mechanisms underlying GD to inform the development of effective therapeutic strategies. A systematic review was conducted using medical subject headings (MeSH) terms related to GD's cellular and molecular pathophysiology. Inclusion criteria encompassed original studies, systematic reviews, and meta-analyses focusing on GD's impact on maternal and fetal health, adhering to PRISMA guidelines. Data extraction captured study characteristics, maternal and fetal outcomes, key findings, and conclusions. GD disrupts insulin signaling pathways, leading to impaired glucose uptake and insulin resistance. Mitochondrial dysfunction reduces ATP production and increases reactive oxygen species, exacerbating oxidative stress. Hormonal influences, chronic inflammation, and dysregulation of the mammalian target of rapamycin (mTOR) pathway further impair insulin signaling. Gut microbiota alterations, gene expression, and epigenetic modifications play significant roles in GD. Ferroptosis and placental dysfunction primarily contribute to intrauterine growth restriction. Conversely, fetal macrosomia arises from maternal hyperglycemia and subsequent fetal hyperinsulinemia, resulting in excessive fetal growth. The chronic inflammatory state and oxidative stress associated with GD exacerbate these complications, creating a hostile intrauterine environment. GD's complex pathophysiology involves multiple disruptions in insulin signaling, mitochondrial function, inflammation, and oxidative stress. Effective management requires early detection, preventive strategies, and international collaboration to standardize care and improve outcomes for mothers and babies.
Collapse
Affiliation(s)
- Johnatan Torres-Torres
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Irma Eloisa Monroy-Muñoz
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Javier Perez-Duran
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Juan Mario Solis-Paredes
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | | | - Deyanira Baca
- Obstetric and Gynecology Department, Hospital General de México Dr. Eduardo Liceaga, Mexico City 06720, Mexico
| | - Salvador Espino-Y-Sosa
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
- Centro de Investigacion en Ciencias de la Salud, Universidad Anahuac Mexico, Campus Norte, Huixquilucan 52786, Mexico
| | - Raigam Martinez-Portilla
- Department of Reproductive and Perinatal Health Research, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Lourdes Rojas-Zepeda
- Maternal-Fetal Department, Instituto Materno Infantil del Estado de Mexico, Toluca 50170, Mexico
| | - Hector Borboa-Olivares
- Community Interventions Research Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Enrique Reyes-Muñoz
- Research Division, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| |
Collapse
|
34
|
Valero P, Silva K, Valenzuela-Hinrichsen A, Vásquez A, Espinoza F, Lira F, Cornejo M, Fuentes G, González D, Moore-Carrasco R, van der Beek EM, Hillebrands JL, van Goor H, Grismaldo A, Sobrevia L. Shortcomings, limitations and gaps in physiological roles of extracellular vesicles in obesity. J Physiol 2024. [PMID: 39470472 DOI: 10.1113/jp286955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024] Open
Abstract
Extracellular vesicles (EVs) play a crucial role in mediating communication between cells across species and kingdoms. The intercellular communication facilitated by EVs through autocrine and paracrine signalling mechanisms is essential for cell survival, maintaining normal metabolic functions and ensuring overall bodily homeostasis and health. Extracellular vesicles are present in various bodily fluids, such as pleural effusions, plasma, breast milk, amniotic fluid, semen and saliva. Additionally, the generation and release of EVs contribute to the removal of cellular waste. Patients with obesity exhibit a higher release and amount of circulating EVs than individuals with normal weight. This increased EV release in obesity might contribute to the inflammatory state characteristic of this metabolic condition, because higher levels of pro-inflammatory molecules are found within their cargo. However, interpreting results related to EV abundance, cargo and biological actions can be complicated by several factors; these include variations in cell sources, a wide age range (from children to the elderly), a mix of females and males, medication use and health status, a range of body weights (from normal weight to morbid obesity) and differences between in vitro assays using cell lines versus primary cultures. This article addresses the shortcomings, limitations and gaps in knowledge, providing a framework for enhancing our understanding of the physiological effects of EVs on obesity.
Collapse
Affiliation(s)
- Paola Valero
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | - Katherin Silva
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Institute of Chemistry, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Andrés Valenzuela-Hinrichsen
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Antonia Vásquez
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernanda Espinoza
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernanda Lira
- Faculty of Medicine, Universidad de Antofagasta, Antofagasta, Chile
| | - Marcelo Cornejo
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Faculty of Health Sciences, Universidad de Talca, Talca, Chile
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
- Biomedical Department, Faculty of Health Sciences, Universidad de Antofagasta, Antofagasta, Chile
| | - Gonzalo Fuentes
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Faculty of Health Sciences, Universidad de Talca, Talca, Chile
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
| | - Daniel González
- Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | | | - Eline M van der Beek
- Department of Pediatrics, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
- Nestlé Institute for Health Sciences, Nestlé Research, Societé des Produits de Nestlé, Lausanne, Switzerland
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
| | - Adriana Grismaldo
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- **Faculty of Excellence program, School of Medicine and Health Sciences, The Institute for Obesity Research (IOR), Eutra, Tecnologico de Monterrey, Monterrey, Nuevo León, Mexico
| | - Luis Sobrevia
- **Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, The Netherlands
- **Faculty of Excellence program, School of Medicine and Health Sciences, The Institute for Obesity Research (IOR), Eutra, Tecnologico de Monterrey, Monterrey, Nuevo León, Mexico
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville, Spain
- Medical School (Faculty of Medicine), Sao Paulo State University (UNESP), Botucatu, Sao Paulo, Brazil
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
35
|
Al Balawi AN, Alblwi NAN, Soliman R, El-Far AH, Hassan MG, El-Sewedy T, Ameen F, Ismail NF, Elmetwalli A. Impact of Vitamin D deficiency on immunological and metabolic responses in women with recurrent pregnancy loss: focus on VDBP/HLA-G1/CTLA-4/ENTPD1/adenosine-fetal-maternal conflict crosstalk. BMC Pregnancy Childbirth 2024; 24:709. [PMID: 39472874 PMCID: PMC11523824 DOI: 10.1186/s12884-024-06914-0] [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: 09/05/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND AND AIM Recurrent pregnancy loss (RPL), also known as recurrent implantation failure (RIF), is a distressing condition affecting women characterized by two or more consecutive miscarriages or the inability to carry a pregnancy beyond 20 weeks. Immunological factors and genetic variations, particularly in Vit D Binding Protein (VDBP), have gained attention as potential contributors to RPL. This study aimed to provide insight into the immunological, genetic, and metabolic networks underlying RPL, placing a particular emphasis on the interactions between VDBP, HLA-G1, CTLA-4, ENTPD1, and adenosine-fetal-maternal conflict crosstalk. METHODS A retrospective study included 198 women with three or more consecutive spontaneous abortions. Exclusion criteria comprised uterine abnormalities, endocrine disorders, parental chromosomal abnormalities, infectious factors, autoimmune diseases, or connective tissue diseases. Immunological interplay was investigated in 162 female participants, divided into two groups based on their Vit D levels: normal Vit D-RPL and low Vit D-RPL. Various laboratory techniques were employed, including LC/MS/MS for Vit D measurement, ELISA for protein detection, flow cytometry for immune function analysis, and molecular docking for protein-ligand interaction assessment. RESULTS General characteristics between groups were significant regarding Vit D and glucose levels. Low Vit D levels were associated with decreased NK cell activity and downregulation of HLA-G1 and HLA-G5 proteins, while CTLA-4 revealed upregulation. VDBP was significantly downregulated in the low Vit D group. Our findings highlight the intricate relationship between Vit D status and adenosine metabolism by the downregulation of SGLT1, and NT5E, key components of adenosine metabolism, suggests that Vit D deficiency may disrupt the regulation of adenosine levels, leading to an impaired reproductive outcome. HNF1β, a negative regulator of VDBP, was upregulated, while HNF1α, a positive regulator, was downregulated in low Vit D women after RPL. Molecular docking analysis revealed crucial residues involved in the interaction between Vit D and HNF1β. CONCLUSION Collectively, these findings underscore the importance of Vit D in modulating immune function and molecular pathways relevant to pregnancy maintenance, highlighting the need for further research to elucidate the mechanisms and potential therapeutic interventions for improving pregnancy outcomes in individuals with Vit D deficiency and RPL.
Collapse
Affiliation(s)
- Aisha Nawaf Al Balawi
- Biology Department, University College of Haql, University of Tabuk, Tabuk, Saudi Arabia.
| | | | - Riham Soliman
- Tropical Medicine Department, Faculty of Medicine, Port Said University, Port Said, Egypt
- Egyptian Liver Research Institute and Hospital (ELRIAH), Sherbin, Mansoura, Egypt
| | - Ali H El-Far
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, China
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt
| | - Mervat G Hassan
- Department of Botany and Microbiology, Faculty of Science, Benha University, Benha, 13511, Egypt
| | - Tarek El-Sewedy
- Department of Applied Medical Chemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Fuad Ameen
- Department of Botany and Microbiology, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Nadia F Ismail
- Health Information Management Program, Biochemistry, Faculty of Health Science Technology, Borg El Arab Technological University, Alexandria, Egypt
| | - Alaa Elmetwalli
- Department of Clinical Trial Research Unit and Drug Discovery, Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt.
- Microbiology Division, Higher Technological Institute of Applied Health Sciences, Egypt Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt.
| |
Collapse
|
36
|
Kyrkou C, Athanasiadis AP, Chourdakis M, Kada S, Biliaderis CG, Menexes G, Michaelidou AM. Are Maternal Dietary Patterns During Pregnancy Associated with the Risk of Gestational Diabetes Mellitus? A Systematic Review of Observational Studies. Nutrients 2024; 16:3632. [PMID: 39519467 PMCID: PMC11547687 DOI: 10.3390/nu16213632] [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: 10/08/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Maternal nutritional status is a "key" contributor to Gestational Diabetes Mellitus (GDM). However, the role of maternal dietary patterns (DPs) during pregnancy remains poorly understood. Thus, we conducted a systematic review to assess associations between "a posteriori-derived" DPs and GDM. METHODS A systematic search was conducted in PubMed, ScienceDirect, Web of Science, and Scopus for cohort, cross-sectional, and case-control studies published until June 2024. A total of twenty-eight studies involving 39,735 pregnant women were included, and their quality was evaluated by the Newcastle-Ottawa Scale. The 91 identified DPs were classified into four categories: "Westernized", "Nutritious", "Plant-based", and "Miscellaneous". RESULTS Our findings do not reveal definitive associations between maternal DPs during pregnancy and GDM risk. Notably, "Westernized" DPs tended to be associated with an increased risk. However, a very small portion of patterns within this category exhibited protective associations. Conversely, "Nutritious" and "Plant-based" appear beneficial for GDM prevention in specific populations. The "Miscellaneous" category presented an almost equal distribution of DPs with both detrimental and protective associations, pinpointing the absence of a clear directional trend regarding GDM risk. CONCLUSIONS The heterogeneity in findings can be attributed to geographic and sociocultural variations and methodological differences across studies. Thus, there is a need for more standardized research methodologies to provide more precise insights that will ultimately help develop effective and tailored dietary guidelines for GDM prevention.
Collapse
Affiliation(s)
- Charikleia Kyrkou
- Department of Food Science and Technology, School of Agriculture, Faculty of Agriculture, Forestry and Natural Environment, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (C.K.); (C.G.B.)
| | - Apostolos P. Athanasiadis
- 3rd Department of Obstetrics and Gynecology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Michael Chourdakis
- Laboratory of Hygiene, Social & Preventive Medicine and Medical Statistics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.C.); (S.K.)
| | - Stefania Kada
- Laboratory of Hygiene, Social & Preventive Medicine and Medical Statistics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.C.); (S.K.)
| | - Costas G. Biliaderis
- Department of Food Science and Technology, School of Agriculture, Faculty of Agriculture, Forestry and Natural Environment, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (C.K.); (C.G.B.)
| | - Georgios Menexes
- Department of Field Crops and Ecology, School of Agriculture, Faculty of Agriculture, Forestry and Natural Environment, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Alexandra-Maria Michaelidou
- Department of Food Science and Technology, School of Agriculture, Faculty of Agriculture, Forestry and Natural Environment, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (C.K.); (C.G.B.)
| |
Collapse
|
37
|
Franzago M, Borrelli P, Di Nicola M, Cavallo P, D’Adamo E, Di Tizio L, Gazzolo D, Stuppia L, Vitacolonna E. From Mother to Child: Epigenetic Signatures of Hyperglycemia and Obesity during Pregnancy. Nutrients 2024; 16:3502. [PMID: 39458497 PMCID: PMC11510513 DOI: 10.3390/nu16203502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND In utero exposure to maternal hyperglycemia and obesity can trigger detrimental effects in the newborn through epigenetic programming. We aimed to assess the DNA methylation levels in the promoters of MC4R and LPL genes from maternal blood, placenta, and buccal swab samples collected in children born to mothers with and without obesity and Gestational Diabetes Mellitus (GDM). METHODS A total of 101 Caucasian mother-infant pairs were included in this study. Sociodemographic characteristics, clinical parameters, physical activity, and adherence to the Mediterranean diet were evaluated in the third trimester of pregnancy. Clinical parameters of the newborns were recorded at birth. RESULTS A negative relationship between MC4R DNA methylation on the fetal side of the GDM placenta and birth weight (r = -0.630, p = 0.011) of newborns was found. MC4R DNA methylation level was lower in newborns of GDM women (CpG1: 2.8% ± 3.0%, CpG2: 3.8% ± 3.3%) as compared to those of mothers without GDM (CpG1: 6.9% ± 6.2%, CpG2: 6.8% ± 5.6%; p < 0.001 and p = 0.0033, respectively), and it was negatively correlated with weight (r = -0.229; p = 0.035), head circumference (r = -0.236; p = 0.030), and length (r = -0.240; p = 0.027) at birth. LPL DNA methylation was higher on the fetal side of the placenta in obese patients as compared to normal-weight patients (66.0% ± 14.4% vs. 55.7% ± 15.2%, p = 0.037), and it was associated with maternal total cholesterol (r = 0.770, p = 0.015) and LDL-c (r = 0.783, p = 0.012). CONCLUSIONS These results support the role of maternal MC4R and LPL methylation in fetal programming and in the future metabolic health of children.
Collapse
Affiliation(s)
- Marica Franzago
- Department of Medicine and Aging, School of Medicine, and Health Sciences, “G. D’Annunzio” University, Via dei Vestini, Chieti-Pescara, 66100 Chieti, Italy; (M.F.); (D.G.)
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy;
| | - Paola Borrelli
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy; (P.B.); (M.D.N.)
| | - Marta Di Nicola
- Laboratory of Biostatistics, Department of Medical, Oral and Biotechnological Sciences, “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy; (P.B.); (M.D.N.)
| | - Pierluigi Cavallo
- Department of Medicine and Aging, School of Medicine, and Health Sciences, “G. D’Annunzio” University, Via dei Vestini, Chieti-Pescara, 66100 Chieti, Italy; (M.F.); (D.G.)
| | - Ebe D’Adamo
- Neonatal Intensive Care Unit, “G. D’Annunzio” University, 66100 Chieti, Italy;
| | - Luciano Di Tizio
- Department of Obstetrics and Gynaecology, SS. Annunziata Hospital, “G. D’Annunzio” University, 66100 Chieti, Italy;
| | - Diego Gazzolo
- Department of Medicine and Aging, School of Medicine, and Health Sciences, “G. D’Annunzio” University, Via dei Vestini, Chieti-Pescara, 66100 Chieti, Italy; (M.F.); (D.G.)
- Neonatal Intensive Care Unit, “G. D’Annunzio” University, 66100 Chieti, Italy;
| | - Liborio Stuppia
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy;
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy
| | - Ester Vitacolonna
- Department of Medicine and Aging, School of Medicine, and Health Sciences, “G. D’Annunzio” University, Via dei Vestini, Chieti-Pescara, 66100 Chieti, Italy; (M.F.); (D.G.)
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti-Pescara, 66100 Chieti, Italy;
| |
Collapse
|
38
|
Li J, Sun Z, Sun F, Lai Y, Yi X, Wang Z, Yuan J, Hu Y, Pan A, Pan XF, Zheng Y, Chen D. Gut antibiotic resistome during pregnancy associates with the risk of gestational diabetes mellitus: New evidence from a prospective nested case-control study. JOURNAL OF HAZARDOUS MATERIALS 2024; 478:135434. [PMID: 39146585 DOI: 10.1016/j.jhazmat.2024.135434] [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: 12/24/2023] [Revised: 07/24/2024] [Accepted: 08/04/2024] [Indexed: 08/17/2024]
Abstract
Antibiotic resistome has emerged as a global threat to public health. However, gestational antibiotic resistome and potential link with adverse pregnancy outcomes remains poorly understood. Our study reports for the first time an association between gut antibiotic resistome during early pregnancy and the risk of gestational diabetes mellitus (GDM) based on a prospective nested case-control cohort including 120 cases and 120 matched controls. A total of 214 antibiotic resistance gene (ARG) subtypes belonging to 17 ARG types were identified in > 10 % fecal samples collected during each trimester. The data revealed dynamic profiles of gut antibiotic resistome through pregnancy, and significant positive associations between selected features (i.e., ARG abundances and a GDM-ARG score which is a new feature characterizing the association between ARGs and GDM) of gut antibiotic resistome during early pregnancy and GDM risk as well as selected endogenous metabolites. The findings demonstrate ubiquitous presence of ARGs in pregnant women and suggest it could constitute an important risk factor for the development of GDM.
Collapse
Affiliation(s)
- Jing Li
- College of Environment and Climate, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China; School of Public Health, Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Zhonghan Sun
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Fudan University, 200433, Shanghai, China
| | - Fengjiang Sun
- College of Environment and Climate, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China
| | - Yuwei Lai
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xinzhu Yi
- Institute of Ecological Sciences, School of Life Sciences, South China Normal University, Guangzhou 510631, Guangdong, China
| | - Zhang Wang
- Institute of Ecological Sciences, School of Life Sciences, South China Normal University, Guangzhou 510631, Guangdong, China
| | - Jiaying Yuan
- Department of Science and Education, Shuangliu Maternal and Child Health Hospital, Chengdu 610200, Sichuan, China
| | - Yayi Hu
- Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - An Pan
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xiong-Fei Pan
- Shuangliu Institute of Women's and Children's Health, Shuangliu Maternal and Child Health Hospital, Chengdu 610041, Sichuan, China; Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & National Medical Products Administration Key Laboratory for Technical Research on Drug Products In Vitro and In Vivo Correlation, West China Second University Hospital, Sichuan University, Shuangliu Maternal and Child Health Hospital, Chengdu 610041, Sichuan, China.
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Fudan University, 200433, Shanghai, China.
| | - Da Chen
- College of Environment and Climate, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China.
| |
Collapse
|
39
|
Hill KB, Mullen GP, Nagareddy PR, Zimmerman KA, Rudolph MC. Key questions and gaps in understanding adipose tissue macrophages and early-life metabolic programming. Am J Physiol Endocrinol Metab 2024; 327:E478-E497. [PMID: 39171752 PMCID: PMC11482221 DOI: 10.1152/ajpendo.00140.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 08/23/2024]
Abstract
The global obesity epidemic, with its associated comorbidities and increased risk of early mortality, underscores the urgent need for enhancing our understanding of the origins of this complex disease. It is increasingly clear that metabolism is programmed early in life and that metabolic programming can have life-long health consequences. As a critical metabolic organ sensitive to early-life stimuli, proper development of adipose tissue (AT) is crucial for life-long energy homeostasis. Early-life nutrients, especially fatty acids (FAs), significantly influence the programming of AT and shape its function and metabolism. Of growing interest are the dynamic responses during pre- and postnatal development to proinflammatory omega-6 (n6) and anti-inflammatory omega-3 (n3) FA exposures in AT. In the US maternal diet, the ratio of "pro-inflammatory" n6- to "anti-inflammatory" n3-FAs has grown dramatically due to the greater prevalence of n6-FAs. Notably, AT macrophages (ATMs) form a significant population within adipose stromal cells, playing not only an instrumental role in AT formation and maintenance but also acting as key mediators of cell-to-cell lipid and cytokine signaling. Despite rapid advances in ATM and immunometabolism fields, research has focused on responses to obesogenic diets and during adulthood. Consequently, there is a significant gap in identifying the mechanisms contributing metabolic health, especially regarding lipid exposures during the establishment of ATM physiology. Our review highlights the current understanding of ATM diversity, their critical role in AT, their potential role in early-life metabolic programming, and the broader implications for metabolism and health.
Collapse
Affiliation(s)
- Kaitlyn B Hill
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Gregory P Mullen
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Prabhakara R Nagareddy
- Department of Internal Medicine, Cardiovascular Section, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Kurt A Zimmerman
- Department of Internal Medicine, Division of Nephrology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Michael C Rudolph
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
40
|
Rocha M, Daniels K, Chandrasekaran S, Michopoulos V. Trauma and Posttraumatic Stress Disorder as Important Risk Factors for Gestational Metabolic Dysfunction. Am J Perinatol 2024; 41:1895-1907. [PMID: 38307105 PMCID: PMC11436347 DOI: 10.1055/a-2260-5051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2024]
Abstract
Gestational metabolic diseases adversely impact the health of pregnant persons and their offspring. Pregnant persons of color are impacted disproportionately by gestational metabolic disease, highlighting the need to identify additional risk factors contributing to racial-ethnic pregnancy-related health disparities. Trauma exposure and posttraumatic stress disorder (PTSD) are associated with increased risk for cardiometabolic disorders in nonpregnant persons, making them important factors to consider when identifying contributors to gestational metabolic morbidity and mortality health disparities. Here, we review current literature investigating trauma exposure and posttraumatic stress disorder as psychosocial risk factors for gestational metabolic disorders, inclusive of gestational diabetes, low birth weight and fetal growth restriction, gestational hypertension, and preeclampsia. We also discuss the physiological mechanisms by which trauma and PTSD may contribute to gestational metabolic disorders. Ultimately, understanding the biological underpinnings of how trauma and PTSD, which disproportionately impact people of color, influence risk for gestational metabolic dysfunction is critical to developing therapeutic interventions that reduce complications arising from gestational metabolic disease. KEY POINTS: · Gestational metabolic diseases disproportionately impact the health of pregnant persons of color.. · Trauma and PTSD are associated with increased risk for cardiometabolic disorders in nonpregnant per.. · Trauma and PTSD impact physiological cardiometabolic mechanisms implicated in gestational metabolic..
Collapse
Affiliation(s)
- Mariana Rocha
- Graduate Program in Neuroscience, Emory University, Atlanta, Georgia
| | | | - Suchitra Chandrasekaran
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Vasiliki Michopoulos
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
41
|
Kurbatfinski S, Dosani A, Dewey DM, Letourneau N. Proposed Physiological Mechanisms Underlying the Association between Adverse Childhood Experiences and Mental Health Conditions: A Narrative Review. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1112. [PMID: 39334644 PMCID: PMC11430311 DOI: 10.3390/children11091112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024]
Abstract
Adverse childhood experiences (ACEs; e.g., physical abuse) can impact lifelong mental health both directly and intergenerationally, with effects transmitted from the parent to the child. Several physiological mechanisms have been proposed to explain the impacts of ACEs on mental health. The purpose of this narrative review was to synthesize and critique the peer-reviewed literature on physiological mechanisms proposed to underlie the impacts of ACEs on mental health, specifically: (1) hypothalamic-pituitary-adrenal axis functioning, (2) inflammation, (3) genetic inheritance and differential susceptibility, (4) epigenetics, (5) brain structure and function, (6) oxidative stress, and (7) metabolic profiles. We searched Google Scholar using variations of the terms "adverse childhood experiences", "mechanisms", and "mental health" to locate relevant peer-reviewed literature. We also mined citations of the identified literature to find additional important sources. The role of inflammation in the etiology of mental health conditions among those exposed to ACEs appeared promising, followed by hypothalamic-pituitary-adrenal axis functioning, brain structure and function, genetics, epigenetics, metabolism, and lastly, oxidative stress. Replication studies that examine the associations among ACEs, genetic inheritance and differential susceptibility, epigenetics, oxidative stress, and metabolism are required to better define links with mental health.
Collapse
Affiliation(s)
- Stefan Kurbatfinski
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (S.K.); (A.D.); (D.M.D.)
- Owerko Centre, Alberta Children's Hospital Research Institute, Calgary, AB T2N 1N4, Canada
| | - Aliyah Dosani
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (S.K.); (A.D.); (D.M.D.)
- Faculty of Health, Community and Education, School of Nursing and Midwifery, Mount Royal University, Calgary, AB T3E 6K6, Canada
- O’Brien Institute for Public Health, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Deborah M. Dewey
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (S.K.); (A.D.); (D.M.D.)
- Owerko Centre, Alberta Children's Hospital Research Institute, Calgary, AB T2N 1N4, Canada
- Departments of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Nicole Letourneau
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; (S.K.); (A.D.); (D.M.D.)
- Owerko Centre, Alberta Children's Hospital Research Institute, Calgary, AB T2N 1N4, Canada
- Departments of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
- Faculty of Nursing, University of Calgary, Calgary, AB T2N 1N4, Canada
- Departments of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
42
|
He L, Wang X, Chen X. Unveiling the role of microRNAs in metabolic dysregulation of Gestational Diabetes Mellitus. Reprod Biol 2024; 24:100924. [PMID: 39013209 DOI: 10.1016/j.repbio.2024.100924] [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: 05/01/2024] [Revised: 05/26/2024] [Accepted: 06/05/2024] [Indexed: 07/18/2024]
Abstract
Gestational Diabetes Mellitus (GDM) presents a significant health concern globally, necessitating a comprehensive understanding of its metabolic intricacies for effective management. MicroRNAs (miRNAs) have emerged as pivotal regulators in GDM pathogenesis, influencing glucose metabolism, insulin signaling, and lipid homeostasis during pregnancy. Dysregulated miRNA expression, both upregulated and downregulated, contributes to GDM-associated metabolic abnormalities. Ethnic and temporal variations in miRNA expression underscore the multifaceted nature of GDM susceptibility. This review examines the dysregulation of miRNAs in GDM and their regulatory functions in metabolic disorders. We discuss the involvement of specific miRNAs in modulating key pathways implicated in GDM pathogenesis, such as glucose metabolism, insulin signaling, and lipid homeostasis. Furthermore, we explore the potential diagnostic and therapeutic implications of miRNAs in GDM management, highlighting the promise of miRNA-based interventions for mitigating the adverse consequences of GDM on maternal and offspring health.
Collapse
Affiliation(s)
- Ling He
- Department of Obstetrics, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoli Wang
- Department of Obstetrics, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangyi Chen
- Department of Obstetrics, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
43
|
Kirovakov Z, Gyokova E, Hinkova N, Stoilov B. Management of Endocrinopathies During Pregnancy: A Systematic Review. Cureus 2024; 16:e70554. [PMID: 39479091 PMCID: PMC11524603 DOI: 10.7759/cureus.70554] [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] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Uncertainty surrounds the efficacy and security of several medications in treating endocrinopathies, such as gestational diabetes mellitus (GDM) in individuals whose normal glucose levels cannot be maintained by diet and exercise alone. To improve pregnancy results for GDM individuals, the present review is conducted to measure the effectiveness of several antidiabetic medications for glucose management. Up until 2024, we looked through PubMed and Google Scholar. Patients with GDM were enrolled in randomized controlled studies that examined several medications. Using the Cochrane risk of bias method, we obtained the pertinent data and evaluated the bias probability. To determine the odds ratio and the surface of the cumulative ranking function of the maternal and neonatal consequences of various therapies in GDM individuals, we first performed pair-wise meta-assessments and subsequently used a systematic review. Macrosomia, higher gestational ages, infant hypoglycemia, and birth weight are the neonatal outcomes. Glycohemoglobin (HbA1c), and pregnancy-induced hypertension (PIH) are the maternal outcomes. This thorough analysis of 25 trial designs found that metformin had fewer cases of macrosomia, higher gestational ages, infant hypoglycemia, and decreased birth weight when compared to glyburide. Metformin was found to be the fastest way to control blood sugar levels in individuals with GDM, whereas glyburide was found to be the most successful medicine for the same purpose.
Collapse
Affiliation(s)
- Zlatko Kirovakov
- Department of Midwifery Care, Faculty of Health Care, Medical University - Pleven, Pleven, BGR
| | - Elitsa Gyokova
- Department of Obstetrics and Gynecology, Faculty of Medicine, Medical University - Pleven, Pleven, BGR
- Department of Obstetrics and Gynecology, University Hospital Saint Marina - Pleven, Pleven, BGR
| | - Nadezhda Hinkova
- Department of Midwifery Care, Faculty of Health Care, Medical University - Pleven, Pleven, BGR
| | - Boris Stoilov
- Department of Obstetrics and Gynecology, Medical University of Plovdiv, Plovdiv, BGR
| |
Collapse
|
44
|
Sousa D, Magalhães C, Matafome P, Pereira S. Adipose tissue-liver cross-talk: a route to hepatic dysfunction in pregnant women with obesity. Biosci Rep 2024; 44:BSR20231679. [PMID: 39083072 PMCID: PMC11327218 DOI: 10.1042/bsr20231679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/24/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
Obesity during pregnancy has been escalating, becoming a huge problem that poses consequences not only for the health of the offspring but also for the maternal well-being. Women's adipose and hepatic tissue metabolism undergoes significant changes during the gestational period. During pregnancy, obesity is a primary instigator of steatosis, increasing the risk of non-alcholic fatty liver disease (NAFLD), now recognized under the updated nomenclature metabolic dysfunction-associated steatotic liver disease (MASLD). Pregnant women with obesity present higher levels of free fatty acids and glucose, reduction in insulin sensitivity, and adipose tissue endocrine dysregulation. Furthermore, obesity-induced modifications in clock genes and lipid-associated gene expression within adipose tissue disrupt crucial metabolic adaptations, potentially culminating in adipose tissue dysfunction. Thus, the liver experiences increased exposure to free fatty acids through the portal vein. Higher uptake of free fatty acids into the liver disrupts hepatic lipid oxidation while enhances lipogenesis, thereby predisposing to ectopic fat deposition within the liver. This review focuses on the obesity-induced changes during pregnancy in both liver and adipose tissue metabolism, elucidating how the metabolic crosstalk between these two organs can be dysregulated in pregnant women living with obesity.
Collapse
Affiliation(s)
- Diana Sousa
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
- Ph.D. Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Carina C. Magalhães
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Paulo Matafome
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Polytechnic University of Coimbra, Coimbra Health School, Rua 5 de Outubro—S. Martinho do Bispo, 3046-854 Coimbra, Portugal
| | - Susana P. Pereira
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra,3004-504 Coimbra, Portugal
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra; 3004-517 Coimbra, Portugal
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory of for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal
| |
Collapse
|
45
|
Karimi M, Mofidi Nejad M, Tabaeifard R, Omid N, Rezaei Z, Azadbakht L. The association between dietary habits and self-care behavior of pregnant women with pregnancy complications. Sci Rep 2024; 14:19681. [PMID: 39181929 PMCID: PMC11344835 DOI: 10.1038/s41598-024-70162-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
Pregnancy as a sensitive period has a critical effect on the mother and infant's life. It is well understood that dietary habits and mother awareness can improve health status and prevent pregnancy complications such as gestational diabetes, pre-eclampsia, and nausea and vomiting. The current study was conducted to investigate the association between dietary habits and self-care behaviors with pregnancy complications. This cross-sectional study was conducted across 300 pregnant women in their third trimester referred to healthcare centers and Yas hospital in Tehran, Iran. Dietary habits and self-care behaviors were assessed by valid and reliable questionnaires. Dietary intake and physical activity were evaluated using valid and reliable Food Frequency Questionnaires and Pregnancy Physical Activity questionnaires, respectively. Women with self-care behaviors had a lower rate of pre-eclampsia (P = 0.9). Regarding food habits, pregnant women who ate their meals faster had a non-significantly higher rate of gestational diabetes and pre-eclampsia. Pregnant women who consumed red meat on a daily frequency had a significantly higher rate of nausea and vomiting (OR 1.85; 95% CI 1.05-3.28, P = 0.03). Also, there was a significant positive association between the mother's pre-pregnancy BMI and gestational diabetes and pre-eclampsia [(OR 2.03; 95% CI 1.03-3.98, P = 0.03) and (OR 4.23; 95% CI 1.12-16.0, P = 0.03) respectively]. There was no significant association between pregnant women's dietary habits and GDM and pre-eclampsia. However, pregnant women with pre-pregnancy overweight and obesity had increased odds of gestational diabetes and pre-eclampsia significantly.
Collapse
Affiliation(s)
- Mehdi Karimi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mofidi Nejad
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, P.O. Box 14155-6117, Tehran, Iran
| | - Razieh Tabaeifard
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, P.O. Box 14155-6117, Tehran, Iran
| | - Noushin Omid
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, P.O. Box 14155-6117, Tehran, Iran
| | - Zahra Rezaei
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Leila Azadbakht
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, P.O. Box 14155-6117, Tehran, Iran.
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
46
|
Abdalqader MM, Hussein SS, Jadi HF, Nori W. Kisspeptin-10: A Predictor for Fetal Growth Restriction among Preeclamptic Women that Discriminated Early Onset Cases. CLIN EXP OBSTET GYN 2024; 51. [DOI: 10.31083/j.ceog5108177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Background: Preeclampsia (PE) is a major cause of maternal and neonatal morbidity. Fetal growth restriction (FGR) shares many pathophysiological roles with PE. Kisspeptin-10 is a peptide secreted by placental syncytium. It was linked to many adverse pregnancy events. The current study aimed to examine Kisspeptin’s-10 role in predicting FGR in PE pregnancies and to verify whether it can predict its onset as early or late FGR. Methods: An observational case-control study enrolled 120 eligible cases at matched gestational age (28–40 weeks) and body mass index (BMI); they were divided into 2-groups: (60) healthy controls and (60) PE cases. PE cases were subdivided into early onset FGR (28/60), who had a gestational age less than 34 weeks, and late-onset FGR (32/60) with a gestational age equal to 34 weeks. A collection was made of the following data: first: pregnant primary criteria [age, BMI, systolic and diastolic blood pressure (BP), and urine for albumin], second: serum Kisspetein-10 was evaluated via enzyme-linked immunosorbent assay (ELISA), and third: ultrasonic criteria [estimated fetal weight, resistance, and pulsatility index (RI, PI)] were recorded for all. Results: Serum Kisspeptin-10 was significantly higher among the controls (309.56 ± 67.72) followed by late-onset FGR and early onset FGR (235.46 ± 68.97) vs. (212.09 ± 58.44) ng/dL; p = 0.0001 respectively. It was negatively linked to systolic, diastolic BP, and urine for albumin; Pearson correlation coefficient (r) was (–0.29, –0.48, –0.28) respectively; p < 0.0001, 0.0018, 0.028 respectively. Kisspeptin-10 was positively linked to estimated fetal weight (r = 0.27; p = 0.034); it had an odds ratio (OR) of 3.04; 95% confidence interval of (1.37–4.765); p = 0.0001 in discriminating healthy pregnancies from FGR cases. Conclusions: The significant correlation of Kisspeptin-10 with PE parameters and estimated fetal weight with high sensitivity, specificity and reliable area under the curve in predicting early onset FGR cases make it recommended for practice in predicting FGR onset.
Collapse
Affiliation(s)
- Manal Madany Abdalqader
- Department of Obstetrics and Gynecology, College of Medicine, Mustansiriyah University, 10052 Baghdad, Iraq
| | - Shatha Sami Hussein
- Department of Obstetrics and Gynecology, College of Medicine, Mustansiriyah University, 10052 Baghdad, Iraq
| | - Huda Fadhil Jadi
- Department of Obstetrics and Gynecology, Fatima Al Zahraa Maternity Hospital, 10052 Baghdad, Iraq
| | - Wassan Nori
- Department of Obstetrics and Gynecology, College of Medicine, Mustansiriyah University, 10052 Baghdad, Iraq
| |
Collapse
|
47
|
Stankovic M, Zeljkovic A, Vekic J, Antonic T, Ardalic D, Miljkovic-Trailovic M, Munjas J, Saric Matutinovic M, Gojkovic T, Jovicic S, Mikovic Z, Stefanovic A. Differences in HDL Remodeling during Healthy Pregnancy and Pregnancy with Cardiometabolic Complications. Antioxidants (Basel) 2024; 13:948. [PMID: 39199194 PMCID: PMC11352027 DOI: 10.3390/antiox13080948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 09/01/2024] Open
Abstract
This study investigated the longitudinal trajectory of changes in antioxidative and anti-inflammatory high-density lipoprotein (HDL) components during healthy pregnancy and pregnancy with cardiometabolic complications. We recruited and longitudinally followed 84 women with healthy pregnancies and 46 pregnant women who developed cardiometabolic pregnancy complications (gestational diabetes mellitus and hypertensive disorders of pregnancy). Their general lipid profiles, oxidative stress status, inflammatory status, and antioxidative and anti-inflammatory HDL components were analyzed. The results of our study confirmed the expected trajectory for the routine lipid parameters. Our study results indicate more intensive oxidative stress and a higher level of inflammation in the group with complications compared with the control group. Sphingosine-1-phosphate (S1P) was significantly lower in the first trimester in the group with complications compared with the control group (p < 0.05). We did not find significant differences in the apolipoprotein A1 (Apo A1) concentrations in the first trimester between the control group and the group with complications, but in the second and third trimesters, the group with complications had significantly higher concentrations (p < 0.001, p < 0.05, respectively). The S1P, paraoxonase 1 (PON1), and serum amyloid A (SAA) concentrations were significantly lower in the group with complications in the first trimester. During the second trimester, only the SAA concentrations were identified as significantly lower in the group with complications compared with the control group, while in the third trimester, the PON1, apolipoprotein M (Apo M), and SAA concentrations were all significantly lower in the group with complications. Through a multivariate binary logistic regression analysis, the S1P concentration in the first trimester was distinguished as an HDL-associated marker independently associated with cardiometabolic pregnancy complications. In conclusion, our study results showed that HDL remodeling differs between healthy pregnancies and pregnancies with maternal cardiometabolic complications, with changed HDL composition and functionality consequently impacting its biological functionality in the latter case.
Collapse
Affiliation(s)
- Marko Stankovic
- Gynecology and Obstetrics Clinic Narodni Front, 11000 Belgrade, Serbia; (M.S.); (D.A.); (Z.M.)
| | - Aleksandra Zeljkovic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (A.Z.); (J.V.); (T.A.); (M.M.-T.); (J.M.); (M.S.M.); (T.G.); (S.J.)
| | - Jelena Vekic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (A.Z.); (J.V.); (T.A.); (M.M.-T.); (J.M.); (M.S.M.); (T.G.); (S.J.)
| | - Tamara Antonic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (A.Z.); (J.V.); (T.A.); (M.M.-T.); (J.M.); (M.S.M.); (T.G.); (S.J.)
| | - Daniela Ardalic
- Gynecology and Obstetrics Clinic Narodni Front, 11000 Belgrade, Serbia; (M.S.); (D.A.); (Z.M.)
| | - Milica Miljkovic-Trailovic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (A.Z.); (J.V.); (T.A.); (M.M.-T.); (J.M.); (M.S.M.); (T.G.); (S.J.)
| | - Jelena Munjas
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (A.Z.); (J.V.); (T.A.); (M.M.-T.); (J.M.); (M.S.M.); (T.G.); (S.J.)
| | - Marija Saric Matutinovic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (A.Z.); (J.V.); (T.A.); (M.M.-T.); (J.M.); (M.S.M.); (T.G.); (S.J.)
| | - Tamara Gojkovic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (A.Z.); (J.V.); (T.A.); (M.M.-T.); (J.M.); (M.S.M.); (T.G.); (S.J.)
| | - Snezana Jovicic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (A.Z.); (J.V.); (T.A.); (M.M.-T.); (J.M.); (M.S.M.); (T.G.); (S.J.)
| | - Zeljko Mikovic
- Gynecology and Obstetrics Clinic Narodni Front, 11000 Belgrade, Serbia; (M.S.); (D.A.); (Z.M.)
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandra Stefanovic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (A.Z.); (J.V.); (T.A.); (M.M.-T.); (J.M.); (M.S.M.); (T.G.); (S.J.)
| |
Collapse
|
48
|
Al-Saleh I, Elkhatib R, Alghamdi R, Alrushud N, Alnuwaysir H, Alnemer M, Aldhalaan H, Shoukri M. Phthalate exposure during pregnancy and its association with thyroid hormones: A prospective cohort study. Int J Hyg Environ Health 2024; 261:114421. [PMID: 39002474 DOI: 10.1016/j.ijheh.2024.114421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024]
Abstract
Phthalate esters (PAEs) possess endocrine-disrupting properties. Studies in humans have indicated that in utero phthalate exposure affects maternal thyroid hormones, which are essential for fetal growth and development. However, these studies also reported inconsistent results on the relationship between phthalates and thyroid hormones. This prospective cohort study aimed to assess phthalate exposure across the three trimesters of pregnancy and its association with thyroid hormone levels. From 2019 to 2022, we recruited 672 pregnant women, and two urine samples and one blood sample were collected from each participant during the pregnancy. We examined the urine samples from 663, 335, and 294 women in the first, second, and third trimester, respectively, for the following seven phthalate metabolites: monoethyl phthalate (MEP) from diethyl phthalate (DEP); mono-n-butyl phthalate (MnBP) and mono-iso-butyl phthalate (MiBP) from dibutyl phthalate (DBP); monobenzyl phthalate (MBzP) from butyl benzyl phthalate; and three di(2-ethylhexyl) phthalate (DEHP) metabolites, mono-(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP), mono-(2-ethyl-5-oxohexyl) phthalate (MEOHP), and mono-(2-ethyl-5-carboxypentyl) phthalate (MECPP). Additionally, we examined the levels of free thyroxine (FT4), thyroid-stimulating hormone (TSH), and total triiodothyronine (TT3) in the serum samples of the following participants: 596, 627, and 576 in the first trimester; 292, 293, and 282 in the second trimester; and 250, 250, and 248 in the third trimester, respectively. Other than MBzP, which was detected in 25%-33% of the samples, other metabolites were detectable in >86% of urine samples, indicating widespread exposure to DEP, DBP, and DEHP. The detected phthalate exposure levels in our cohort were significantly higher than those reported in other countries. Metabolite levels varied across the trimesters, implying changes in exposure and metabolism throughout pregnancy. The observed variability in urinary concentrations of phthalate metabolites, which ranged from poor to moderate, underscores the importance of taking multiple measurements during pregnancy for precise exposure assessment. Using a linear mixed model, we analyzed the effects of repeated phthalate exposure on thyroid hormone levels while adjusting for potential confounders. We observed significant linear trends in FT4, TSH, and, to a lesser extent, TT3 across quartiles of specific phthalate metabolites. Comparing the highest to the lowest quartiles, we found a significant increase in FT4 levels, ranging from 2 to 3.7%, associated with MEP; MECPP; MEHHP; and the sum of seven metabolites (∑7PAE), three DEHP metabolites (∑3DEHP), two DBP metabolites (∑DBP), and both low molecular weight (∑LMW) and high molecular weight metabolites. Increased TSH levels (5%-16%) were observed for all phthalate metabolites (except MEHHP) and their molar sums, including ∑7PAE. For TT3, a significant increase was observed with MEP (2.2%) and a decrease was observed with ∑DBP (-2.7%). A higher TSH/FT4 ratio was observed with the highest quartiles (third or fourth) of several phthalate metabolites: MEP (8.8%), MiBP (8.7%), MnBP (22.2%), ∑7PAE (15.3%), ∑DBP (16.4%), and ∑LMW (18.6%). These hormonal alterations, most notably in the second and third trimesters, suggest that phthalate exposure may impact fetal growth and development by affecting maternal thyroid function.
Collapse
Affiliation(s)
- Iman Al-Saleh
- Environmental Health Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.
| | - Rola Elkhatib
- Environmental Health Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Reem Alghamdi
- Environmental Health Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Nujud Alrushud
- Environmental Health Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Hissah Alnuwaysir
- Environmental Health Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Maha Alnemer
- Obstetrics and Gynecology Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Hesham Aldhalaan
- Center for Autism Research, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mohamed Shoukri
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
49
|
Samare-Najaf M, Dehghanian A, Asadikaram G, Mohamadi M, Jafarinia M, Savardashtaki A, Afshari A, Vakili S. Designing an Electrochemical Biosensor Based on Voltammetry for Measurement of Human Chorionic Gonadotropin. JOURNAL OF MEDICAL SIGNALS & SENSORS 2024; 14:21. [PMID: 39234593 PMCID: PMC11373787 DOI: 10.4103/jmss.jmss_64_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/19/2024] [Accepted: 04/12/2024] [Indexed: 09/06/2024]
Abstract
Background Human chorionic gonadotropin (hCG) is a polypeptide hormone synthesized during pregnancy and is also upregulated in some pathologic conditions such as certain tumors. Its measurement is essential for diagnosing pregnancy and malignancies. Despite numerous attempts to introduce an accurate method capable of detecting hCG levels, several limitations are found in previous techniques. This study aimed to address the limitations of current hCG assay methods by designing an electrochemical biosensor based on voltammetry for the rapid, selective, inexpensive, and sensitive measurement of hCG levels. Methods A carbon paste electrode was prepared and functionalized by para-aminobenzoic acid. The primary anti-β-hCG monoclonal antibody was immobilized on the electrode surface by activating the carboxyl groups with 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide and N-hydroxysuccinimide solutions. The study also involved optimizing parameters such as the time for primary antibody fixation, the time for hCG attachment, and the pH of the hydrogen peroxide solution to maximize the biosensor response. Different concentrations of hCG hormone were prepared and loaded on the electrode surface, the secondary antibody labeled with HRP enzyme was applied, thionine in phosphate-buffered saline solution was placed on the electrode surface, and the differential pulse electrical signal was recorded. Results The linear range ranged from 5 to 100 mIU/ml, and the limit of detection was calculated as 0.11 mIU. The relative standard deviation was 3% and 2% for five repeated measurements of commercial standard samples with concentrations of 2 and 20 mIU/mL, respectively. The percent recovery was obtained from 98.3% to 101.5%. Conclusion The sensor represents a promising advancement in hCG level measurement, offering a potential solution to overcome the existing limitations in current diagnostic strategies. Simple and inexpensive design, detecting hCG in its important clinical range during early pregnancy, and successful measurement of hCG in real serum samples are the advantages of this sensor.
Collapse
Affiliation(s)
- Mohammad Samare-Najaf
- Blood Transfusion Research Center, Kerman Regional Blood Transfusion Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amirreza Dehghanian
- Trauma Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pathology, Molecular Pathology and Cytogenetics Division, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamreza Asadikaram
- Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Mohamadi
- Pistachio Safety Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Morteza Jafarinia
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Afrooz Afshari
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Vakili
- Infertility Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
50
|
Peng J, Xu T, Tan X, He Y, Zeng Y, Tang J, Sun M. Eating Styles Profiles and Correlates in Chinese Postpartum Women: A Latent Profile Analysis. Nutrients 2024; 16:2299. [PMID: 39064741 PMCID: PMC11280277 DOI: 10.3390/nu16142299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 06/27/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Postpartum women present a high risk of disordered eating behaviors, but the heterogeneity between groups was not identified. This cross-sectional study aimed to identify eating styles profiles in postpartum women and explore the correlates based on demographic characteristics and psychosocial factors. Questionnaires were administered to 507 Chinese postpartum women. Latent profile analysis (LPA) was conducted to identify eating styles profiles. Multinomial logistic regression was used to investigate the correlates of these profiles among postpartum women. The LPA identified three eating styles profiles: postpartum women with low emotional, external, and restrained eating (Profile 1, 6.9%); postpartum women with medium emotional, external, and restrained eating (Profile 2, 66.1%); and postpartum women with high emotional, external, and restrained eating (Profile 3, 27.0%). Compared to Profile 1, higher postpartum depression (PPD) and body mass index (BMI) were more likely to be associated with Profile 2 and Profile 3, whereas higher postpartum weight retention (PPWR) was more likely to be associated with Profile 1. Compared to Profile 2, higher PPD and BMI were more likely associated with Profile 3. Disordered eating behaviors in postpartum women with three eating styles were associated with BMI, PPD, and PPWR. This study can guide healthcare professionals in developing targeted interventions to improve maternal and child health globally.
Collapse
Affiliation(s)
- Jiayuan Peng
- Xiangya School of Nursing, Central South University, No.172 Tongzipo Road, Yuelu District, Changsha 410013, China; (J.P.); (T.X.); (Y.H.); (Y.Z.); (J.T.)
| | - Tian Xu
- Xiangya School of Nursing, Central South University, No.172 Tongzipo Road, Yuelu District, Changsha 410013, China; (J.P.); (T.X.); (Y.H.); (Y.Z.); (J.T.)
| | - Xiangmin Tan
- School of Rural Health, Monash University, 15 Sargeant Street, Warragul, VIC 3820, Australia;
| | - Yuqing He
- Xiangya School of Nursing, Central South University, No.172 Tongzipo Road, Yuelu District, Changsha 410013, China; (J.P.); (T.X.); (Y.H.); (Y.Z.); (J.T.)
| | - Yi Zeng
- Xiangya School of Nursing, Central South University, No.172 Tongzipo Road, Yuelu District, Changsha 410013, China; (J.P.); (T.X.); (Y.H.); (Y.Z.); (J.T.)
| | - Jingfei Tang
- Xiangya School of Nursing, Central South University, No.172 Tongzipo Road, Yuelu District, Changsha 410013, China; (J.P.); (T.X.); (Y.H.); (Y.Z.); (J.T.)
| | - Mei Sun
- Xiangya School of Nursing, Central South University, No.172 Tongzipo Road, Yuelu District, Changsha 410013, China; (J.P.); (T.X.); (Y.H.); (Y.Z.); (J.T.)
- School of Nursing, Xinjiang Medical University, No.168 Youyi South Road, Urumqi 830054, China
| |
Collapse
|