1
|
Di Pietrantonio N, Sánchez-Ceinos J, Shumliakivska M, Rakow A, Mandatori D, Di Tomo P, Formoso G, Bonfini T, Baldassarre MPA, Sennström M, Almahmeed W, Pandolfi A, Cosentino F. The inflammatory and oxidative phenotype of gestational diabetes is epigenetically transmitted to the offspring: role of methyltransferase MLL1-induced H3K4me3. Eur Heart J 2024; 45:5171-5185. [PMID: 39471481 DOI: 10.1093/eurheartj/ehae688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/16/2024] [Accepted: 09/25/2024] [Indexed: 11/01/2024] Open
Abstract
BACKGROUND AND AIMS Hyperglycaemia during gestational diabetes (GD) predisposes women and their offspring to later cardiometabolic disease. The hyperglycaemia-mediated epigenetic changes remain to be elucidated. Methyltransferase MLL1-induced trimethylation of histone 3 at lysine 4 (H3K4me3) activates inflammatory and oxidative phenotype. This epigenetic mark in GD women and its transmission to the offspring were investigated. METHODS Peripheral blood mononuclear cells (PBMC) were collected from GD and control (C) women and also from adolescents born to women of both groups. Endothelial human umbilical vein endothelial cells (HUVEC) and cord blood mononuclear cells (CBMC) were from umbilical cords. The NF-κBp65 and NOX4 expressions were investigated by reverse transcription quantitative polymerase chain reaction and immunofluorescence (IF). MLL1 and H3K4me3 were investigated by immunoblotting and IF. H3K4me3 on NF-κBp65 and NOX4 promoters was studied by chromatin immunoprecipitation. Superoxide anion generation was measured by electron spin resonance spectroscopy. Plasma cytokines were measured by enzyme-linked immunosorbent assay. To investigate the role of MLL1, HUVEC were exposed to inhibitor MM102 or siRNA transfection. RESULTS PBMC, CBMC, and HUVEC showed an increase of NF-κBp65, IL-6, ICAM-1, MCP-1, and VCAM-1 mRNAs. These findings were associated with H3K4me3 enrichment in the promoter of NF-κBp65. Elevated H3K4me3 and cytokine levels were observed in GD adolescents. MLL1 drives H3K4me3 not only on NF-kB p65, but also on NOX4 promoter. Inhibition of MLL1 blunted NF-κBp65 and NOX4 by modulating inflammatory and oxidative phenotype. CONCLUSIONS Such proof-of-concept study shows persistence of MLL1-dependent H3K4me3 in offspring born to GD women, suggesting an epigenetic-driven transmission of maternal phenotype. These findings may pave the way for pharmacological reprogramming of adverse histone modifications to mitigate abnormal phenotypes underlying early ASCVD.
Collapse
Affiliation(s)
- Nadia Di Pietrantonio
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm 171 76, Sweden
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti 66100, Italy
| | - Julia Sánchez-Ceinos
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm 171 76, Sweden
| | - Mariana Shumliakivska
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm 171 76, Sweden
| | - Alexander Rakow
- Department of Neonatology, Karolinska University Hospital, Stockholm, Sweden
| | - Domitilla Mandatori
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti 66100, Italy
| | - Pamela Di Tomo
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti 66100, Italy
| | - Gloria Formoso
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Tiziana Bonfini
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Maria Pompea Antonia Baldassarre
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Maria Sennström
- Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Assunta Pandolfi
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, University G. D'Annunzio of Chieti-Pescara, Chieti 66100, Italy
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm 171 76, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, Stockholm 171 76, Sweden
| |
Collapse
|
2
|
Xiao Y, Shi Y, Ni Y, Ni M, Yang Y, Zhang X. Gestational diabetes-combined excess weight gain exacerbates gut microbiota dysbiosis in newborns, associated with reduced abundance of Clostridium, Coriobacteriaceae, and Collinsella. Front Cell Infect Microbiol 2024; 14:1496447. [PMID: 39726807 PMCID: PMC11670820 DOI: 10.3389/fcimb.2024.1496447] [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: 09/14/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Background Existing literature indicates that Gestational diabetes mellitus (GDM) and maternal obesity disrupt the normal colonization of the neonatal gut microbiota alone. Still, the combined impact of GDM and excessive gestational weight gain (EGWG) on this process remains under explored. The association between gestational weight gain before/after GDM diagnosis and neonatal gut microbiota characteristics is also unclear.The purpose of this study is to conduct investigation and analysis on the above-mentioned issues, providing a basis for optimizing clinical management plans. Methods This study involved 98 mother-infant pairs categorized into GDM and non-GDM groups. The GDM group was further subdivided based on gestational weight gain (GWG) into normal (GDM+NGWG) and excessive (GDM+EGWG) weight gain groups. Neonatal stool samples were collected within 24 hours post-delivery for gut microbiota profiling through 16S rRNA gene sequencing. Statistical analyses explored correlations between total GWG/BMI gain and those before/after GDM diagnosis (t-GWG/GBG; b-GWG/GBG; a-GWG/GBG) with key bacterial taxa. Results Notable genus-level changes included enrichment of Escherichia and Klebsiella, and depletion of Bacteroides, Bifidobacterium, Coprococcus, Ruminococcus among GDM-Total and GDM+EGWG groups compared to non-GDM. Further,LEfSe analysis identified 30 differential bacteria taxa between GDM-Total and healthy control groups, which increased to 38 between GDM+EGWG and non-GDM groups, highlighting more pronounced microbial shifts associated with EGWG. Clostridium was negatively correlated with t-GWG and newborn birth weight; The Coriobacteriaceae showed a negative correlation with t-GWG, t-GBG, and a-GBG. Additionally,Collinsella exhibited negative correlations with t-GBG and a-GBG. Conclusion This study has identified that the presence of EGWG in GDM mothers further exacerbated neonatal gut microbial perturbations. Total GWG/GBG and those after the diagnosis of GDM were negatively correlated with the abundance of neonatal gut Clostridium, Coriobacteriaceae, and Collinsella. These findings provide new insights for precise prevention and management of GDM.
Collapse
Affiliation(s)
- Yunshan Xiao
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Clinical Research Center for Perinatal Medicine, Xiamen Obstetric Quality Management Center, Xiamen, China
| | - Yuan Shi
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Clinical Research Center for Perinatal Medicine, Xiamen Obstetric Quality Management Center, Xiamen, China
| | - Yan Ni
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Meilan Ni
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yuxin Yang
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xueqin Zhang
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Clinical Research Center for Perinatal Medicine, Xiamen Obstetric Quality Management Center, Xiamen, China
| |
Collapse
|
3
|
Cho S, Rifas-Shiman SL, Harnois-Leblanc S, Aris IM, Oken E, Hivert MF. Associations of Abnormal Maternal Glucose Regulation in Pregnancy with Offspring Adiposity, Insulin Resistance, and Adipokine Markers During Childhood and Adolescence. J Pediatr 2024; 272:114100. [PMID: 38759779 PMCID: PMC11347092 DOI: 10.1016/j.jpeds.2024.114100] [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: 01/08/2024] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 05/19/2024]
Abstract
OBJECTIVE To examine the associations of abnormal maternal glucose regulation in pregnancy with offspring adiposity, insulin resistance, adipokine, and inflammatory markers during childhood and adolescence. STUDY DESIGN Project Viva is a prospective prebirth cohort (n = 2128 live births) initiated from 1999 through 2002 in Eastern Massachusetts, US. During the second trimester of pregnancy, clinicians used 2-step oral glucose challenge testing to screen for gestational diabetes mellitus. In the offspring, we measured anthropometry, insulin resistance, adipokines, lipids, and inflammatory markers in mid-childhood (n = 1107), early adolescence (n = 1027), and mid-adolescence (n = 693). We used multivariable linear regression models and generalized estimating equations adjusted for child age and sex, and for maternal age, race/ethnicity, education, parity, and smoking during pregnancy; we further adjusted for prepregnancy body mass index (BMI). RESULTS In mid-adolescence (17.1 [0.8] years of age), offspring of mothers with gestational diabetes mellitus (n = 27) had a higher BMI z-score (β; 95% Cl; 0.41 SD; 0.00, 0.82), sum of skinfolds (8.15 mm; 2.48, 13.82), homeostatic model assessment for insulin resistance (0.81 units; 0.13, 1.50), leptin z-score (0.40 SD; 0.01, 0.78), and leptin/adiponectin ratio z-score (0.51 SD; CI 0.09, 0.93) compared with offspring of mothers with normoglycemia (multivariable-adjusted models). The associations with BMI, homeostatic model assessment for insulin resistance, and adiponectin seemed stronger in mid-adolescence compared with earlier time points. The associations were attenuated toward the null after adjustment for maternal prepregnancy BMI. CONCLUSION Exposure to gestational diabetes mellitus is associated with higher adiposity, insulin resistance, and altered adipokines in mid-adolescence. Our findings suggest that the peripubertal period could be a key time for the emergence of prenatally programmed metabolic abnormalities.
Collapse
Affiliation(s)
- Sarah Cho
- The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Sheryl L Rifas-Shiman
- Division of Chronic Disease Research Across the Life Course, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA
| | - Soren Harnois-Leblanc
- Division of Chronic Disease Research Across the Life Course, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA
| | - Izzuddin M Aris
- Division of Chronic Disease Research Across the Life Course, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA
| | - Emily Oken
- Division of Chronic Disease Research Across the Life Course, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Life Course, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA; Diabetes Clinical Center, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
4
|
Kostenko A, Zuffa S, Zhi H, Mildau K, Raffatellu M, Dorrestein PC, Aron AT. Dietary iron intake has long-term effects on the fecal metabolome and microbiome. Metallomics 2024; 16:mfae033. [PMID: 38992131 PMCID: PMC11272056 DOI: 10.1093/mtomcs/mfae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/10/2024] [Indexed: 07/13/2024]
Abstract
Iron is essential for life, but its imbalances can lead to severe health implications. Iron deficiency is the most common nutrient disorder worldwide, and iron dysregulation in early life has been found to cause long-lasting behavioral, cognitive, and neural effects. However, little is known about the effects of dietary iron on gut microbiome function and metabolism. In this study, we sought to investigate the impact of dietary iron on the fecal metabolome and microbiome by using mice fed with three diets with different iron content: an iron deficient, an iron sufficient (standard), and an iron overload diet for 7 weeks. Additionally, we sought to understand whether any observed changes would persist past the 7-week period of diet intervention. To assess this, all feeding groups were switched to a standard diet, and this feeding continued for an additional 7 weeks. Analysis of the fecal metabolome revealed that iron overload and deficiency significantly alter levels of peptides, nucleic acids, and lipids, including di- and tri-peptides containing branched-chain amino acids, inosine and guanosine, and several microbial conjugated bile acids. The observed changes in the fecal metabolome persist long after the switch back to a standard diet, with the cecal gut microbiota composition and function of each group distinct after the 7-week standard diet wash-out. Our results highlight the enduring metabolic consequences of nutritional imbalances, mediated by both the host and gut microbiome, which persist after returning to the original standard diets.
Collapse
Affiliation(s)
- Anastasiia Kostenko
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO, USA
| | - Simone Zuffa
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Hui Zhi
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Kevin Mildau
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
- Bioinformatics Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Manuela Raffatellu
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Chiba University, UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), La Jolla, CA, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Allegra T Aron
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
5
|
Martins MG, Woodside B, Kiss ACI. Effects of maternal mild hyperglycemia associated with snack intake on offspring metabolism and behavior across the lifespan. Physiol Behav 2024; 276:114483. [PMID: 38331375 DOI: 10.1016/j.physbeh.2024.114483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/22/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
The increasing prevalence of diabetes is of particular concern in women of childbearing age because of the short and long-term consequences of maternal diabetes for the health of the offspring, such as a greater risk of developing metabolic impairments and cognitive deficits. In addition, maternal diet during pregnancy and lactation might contribute to preventing or ameliorating adverse offspring outcomes. Recently, we described that access to snacks exacerbates glucose intolerance in mildly hyperglycemic pregnant dams. Therefore, we hypothesized that these offspring would show greater impairment in metabolic and behavioral outcomes across the lifespan. Neonatal STZ treatment was employed to induce maternal mild hyperglycemia in females. After mating, normo- and hyperglycemic dams were given access either to standard chow or standard show plus snacks. Male and female offspring were evaluated on postnatal days (PND) 30, 90, and 360. Offspring behavior was assessed in the marble burying task, the open-field test, the elevated-plus maze, and sucrose preference. Glucose tolerance and morphometric analyses were also carried out. Maternal hyperglycemia increased body weight and fat deposition only on PND 30, while retroperitoneal fat deposition was reduced in the offspring of snack-fed dams. However, maternal snack intake reduced offspring body weight and length on PND 90. Fasting glucose was increased in females born to hyperglycemic, snack-fed dams on PND 90. Glucose clearance was altered by both maternal conditions in male offspring on PND 30, however, this sex difference was reversed on PND 90, with maternal hyperglycemia impairing glucose clearance only in females. In addition, maternal hyperglycemia reduced anxiety-like behavior in female offspring on PND 30, especially in the offspring of snack-fed dams, while maternal snack intake reduced sucrose preference in both males and females in adulthood. These results suggest that the effects of maternal hyperglycemia during pregnancy and lactation on offspring outcomes were not exacerbated by snack intake. Although additive effects of the two maternal conditions were hypothesized, the absence of such effects could be related to the mild maternal hyperglycemia induced by STZ treatment even when combined with snack intake. While maternal hyperglycemia alone impaired some offspring outcomes, its association with snack intake did not aggravate those impairments but rather resulted in outcomes more similar to those of offspring born to normoglycemic dams. Finally, females were found to be more susceptible to both the effects of maternal hyperglycemia and snack intake on metabolism and behavior.
Collapse
Affiliation(s)
- Marina Galleazzo Martins
- Department of Physiology, Institute of Biosciences of the University of São Paulo (IB/USP), Rua do Matão, trav. 14, 321, Cidade Universitária, São Paulo, 05508-090, Brazil; São Paulo State University (Unesp), Institute of Biosciences, Department of Structural and Functional Biology, Rua Prof. Dr. Antonio Celso Wagner Zanin, s/n, Botucatu, São Paulo, 18618-689, Brazil.
| | - Barbara Woodside
- Center for Studies in Behavioral Neurobiology, Psychology Department, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec H4B 1R6, Canada
| | - Ana Carolina Inhasz Kiss
- Department of Physiology, Institute of Biosciences of the University of São Paulo (IB/USP), Rua do Matão, trav. 14, 321, Cidade Universitária, São Paulo, 05508-090, Brazil; São Paulo State University (Unesp), Institute of Biosciences, Department of Structural and Functional Biology, Rua Prof. Dr. Antonio Celso Wagner Zanin, s/n, Botucatu, São Paulo, 18618-689, Brazil
| |
Collapse
|
6
|
Ren S, Wu D, Li P. Evaluation of insulin secretion and insulin sensitivity in pregnant women: Application value of simple indices. Clin Chim Acta 2024; 554:117753. [PMID: 38185282 DOI: 10.1016/j.cca.2023.117753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 12/29/2023] [Accepted: 12/29/2023] [Indexed: 01/09/2024]
Abstract
The prevalence of gestational diabetes mellitus (GDM) is increasing annually, which poses substantial harm to the health of both mothers and children. Therefore, selection of clinically applicable and easily detectable indicators in the assessment of maternal insulin secretory function and insulin sensitivity in pregnant women undoubtedly holds great importance in evaluating the risk of GDM, guiding the choice of GDM therapy modalities, and improving the ability to provide early warning of adverse pregnancy outcomes. Compared with the classic clamp technique, many simple indices are more suited for use among pregnant women due to the low frequency of blood sampling and simple administration involved. While indices derived from fasting blood glucose and fasting insulin levels are most readily available, they are unable to provide information on the ability of insulin to manage the glucose load during pregnancy. Although the indices derived from the insulin and glucose values at each time point of the oral glucose tolerance test can provide a more comprehensive picture of the insulin sensitivity and insulin secretory function of the body, their application is constrained by the complexity of the procedure and associated high costs. Concomitantly, the findings from different studies are influenced by a variety of confounding factors, such as the gestational age during testing, race, and detection method. Furthermore, insulin secretory function and insulin sensitivity in pregnant women differ from those in non-pregnant women in that they change significantly with prolonged pregnancy; hence, there is an urgent need to develop a pregnancy-specific reference range. This article reviews the progress in the application of simple indices to help clinicians better understand their potential application in detecting GDM.
Collapse
Affiliation(s)
- Shuying Ren
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, People's Republic of China
| | - Dan Wu
- Department of Endocrinology, 242 Hospital Affilliated to Shenyang Medical College, Shenyang, Liaoning Province, People's Republic of China
| | - Ping Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, People's Republic of China.
| |
Collapse
|
7
|
Govindarajah V, Sakabe M, Good S, Solomon M, Arasu A, Chen N, Zhang X, Grimes HL, Kendler A, Xin M, Reynaud D. Gestational diabetes in mice induces hematopoietic memory that affects the long-term health of the offspring. J Clin Invest 2024; 134:e169730. [PMID: 37988162 PMCID: PMC10786695 DOI: 10.1172/jci169730] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 11/16/2023] [Indexed: 11/23/2023] Open
Abstract
Gestational diabetes is a common medical complication of pregnancy that is associated with adverse perinatal outcomes and an increased risk of metabolic diseases and atherosclerosis in adult offspring. The mechanisms responsible for this delayed pathological transmission remain unknown. In mouse models, we found that the development of atherosclerosis in adult offspring born to diabetic pregnancy can be in part linked to hematopoietic alterations. Although they do not show any gross metabolic disruptions, the adult offspring maintain hematopoietic features associated with diabetes, indicating the acquisition of a lasting diabetic hematopoietic memory. We show that the induction of this hematopoietic memory during gestation relies on the activity of the advanced glycation end product receptor (AGER) and the nucleotide binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, which lead to increased placental inflammation. In adult offspring, we find that this memory is associated with DNA methyltransferase 1 (DNMT1) upregulation and epigenetic changes in hematopoietic progenitors. Together, our results demonstrate that the hematopoietic system can acquire a lasting memory of gestational diabetes and that this memory constitutes a pathway connecting gestational health to adult pathologies.
Collapse
Affiliation(s)
| | | | - Samantha Good
- Division of Experimental Hematology and Cancer Biology and
| | | | - Ashok Arasu
- Division of Experimental Hematology and Cancer Biology and
| | - Nong Chen
- Division of Experimental Hematology and Cancer Biology and
| | - Xuan Zhang
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
| | - H. Leighton Grimes
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
- Department of Pediatrics and
| | - Ady Kendler
- Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Mei Xin
- Division of Experimental Hematology and Cancer Biology and
- Department of Pediatrics and
| | - Damien Reynaud
- Division of Experimental Hematology and Cancer Biology and
- Department of Pediatrics and
| |
Collapse
|
8
|
Allen LA, Taylor PN, Gillespie KM, Oram RA, Dayan CM. Maternal type 1 diabetes and relative protection against offspring transmission. Lancet Diabetes Endocrinol 2023; 11:755-767. [PMID: 37666263 DOI: 10.1016/s2213-8587(23)00190-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 09/06/2023]
Abstract
Type 1 diabetes is around twice as common in the offspring of men with type 1 diabetes than in the offspring of women with type 1 diabetes, but the reasons for this difference are unclear. This Review summarises the evidence on the rate of transmission of type 1 diabetes to the offspring of affected fathers compared with affected mothers. The findings of nine major studies are presented, describing the magnitude of the effect observed and the relative strengths and weaknesses of these studies. This Review also explores possible underlying mechanisms for this effect, such as genetic mechanisms (eg, the selective loss of fetuses with high-risk genes in mothers with type 1 diabetes, preferential transmission of susceptibility genes from fathers, and parent-of-origin effects influencing gene expression), environmental exposures (eg, exposure to maternal hyperglycaemia, exogenous insulin exposure, and transplacental antibody transfer), and maternal microchimerism. Understanding why type 1 diabetes is more common in the offspring of men versus women with type 1 diabetes will help in the identification of individuals at high risk of the disease and can pave the way in the development of interventions that mimic the protective elements of maternal type 1 diabetes to reduce the risk of disease in individuals at high risk.
Collapse
Affiliation(s)
- Lowri A Allen
- Diabetes Research Group, Cardiff University, University Hospital of Wales, Cardiff, UK.
| | - Peter N Taylor
- Diabetes Research Group, Cardiff University, University Hospital of Wales, Cardiff, UK
| | - Kathleen M Gillespie
- Diabetes and Metabolism, Bristol Medical School, University of Bristol, Southmead Hospital, Bristol, UK
| | - Richard A Oram
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Exeter, UK
| | - Colin M Dayan
- Diabetes Research Group, Cardiff University, University Hospital of Wales, Cardiff, UK
| |
Collapse
|
9
|
Luo J, Geng X, Zhou J, Liang S, Zheng W, Li G. Characteristics of the oral glucose tolerance test in women with different pre-pregnancy body mass index and the effect of gestational diabetes mellitus on twin pregnancy outcomes. Clinics (Sao Paulo) 2023; 78:100272. [PMID: 37604047 PMCID: PMC10445444 DOI: 10.1016/j.clinsp.2023.100272] [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: 05/09/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/23/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the mid-pregnancy blood glucose levels of women with singleton or twin pregnancies. METHOD The relationship between blood glucose levels and Gestational Diabetes Mellitus (GDM) was studied in women with different pre-pregnancy Body Mass Index (BMI), and the effect of GDM on twin pregnancy outcomes was analyzed. Women with twin (n = 1,985) and singleton (n = 1,985) pregnancies were categorized into underweight (BMI < 18.5 kg/m2, n = 597), normal weight (BMI: 18.5-23.9 kg/m2, n = 2,575), and overweight/obese (BMI ≥ 24 kg/m2, n = 798) groups. RESULTS The incidence of GDM was 21.01% in women with twin pregnancies. Among the women with GDM in twin pregnancies, 38.37% had at least two abnormal blood glucose levels. The incidence of these parameters increased with preconception BMI, and the incidence of twin pregnancies was higher than that of singleton pregnancies (p < 0.001). In the normal weight and overweight/obese group, the oral glucose tolerance test glucose level and incidence of GDM were higher in women with twin than singleton pregnancies (p < 0.05). For twin pregnancies, the prevalence of selective fetal growth restriction was higher and anemia was lower in the GDM group than in the non-GDM group (all p < 0.05). CONCLUSION Therefore, a greater emphasis should be placed on BMI before conception, and well-controlled GDM does not increase adverse pregnancy outcomes for twin pregnancies.
Collapse
Affiliation(s)
- Jinying Luo
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, PR China; Beijing Maternal and Child Health Care Hospital, PR China; Department of Obstetrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, PR China
| | - Xiaoyan Geng
- Department of Obstetrics and Gynecology, Xicheng Women and Children's Health Hospital, PR China
| | - Jinfu Zhou
- Department of Obstetrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, PR China
| | - Shengnan Liang
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, PR China; Beijing Maternal and Child Health Care Hospital, PR China
| | - Wei Zheng
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, PR China; Beijing Maternal and Child Health Care Hospital, PR China
| | - Guanghui Li
- Division of Endocrinology and Metabolism, Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, PR China; Beijing Maternal and Child Health Care Hospital, PR China.
| |
Collapse
|
10
|
Ji J, Wu P, Li G, He Z, Wang S, Yu W, Li C, Mi Y, Luo X. The associations of ferritin, serum lipid and plasma glucose levels across pregnancy in women with gestational diabetes mellitus and newborn birth weight. BMC Pregnancy Childbirth 2023; 23:478. [PMID: 37386448 DOI: 10.1186/s12884-023-05806-z] [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: 07/01/2022] [Accepted: 06/21/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Women with gestational diabetes mellitus (GDM) are at greater risk of abnormal birth weight. Since the level of biochemical indicators could often affect the intrauterine growth and development of the fetus, it is of great practical significance to understand the changes of biochemical levels across pregnancy in women with GDM and to find out the indicators that play an important role in predicting birth weight. METHODS The data source of this study was from the Xi'an Longitudinal Mother-Child Cohort study (XAMC), in which women with GDM with normal and high pre-pregnancy body mass index (BMI) and their newborns between January 1st and March 31st in 2018 were included. The data of ferritin, serum lipid profile and fasting plasma glucose (FPG) of mothers in the three trimesters of pregnancy, as well as birth weight of newborns were all collected from medical records. Multiple linear regression and multivariate logistic regression analyses were used to explore the association of the biochemical indexes and birth weight. A P value < 0.05 was considered statistically significant. RESULTS A total of 782 mother-infant pairs were finally included and divided into normal weight group (NG) (n = 530, 67.8%) and overweight/obesity group (OG) (n = 252, 32.2%) according to maternal pre-pregnancy BMI. The level of ferritin in both NG and OG decreased during pregnancy (P for trend < 0.001 for all), whereas the levels of total cholesterol (TC), high density cholesterol (HDL-C), low density cholesterol (LDL-C) and triglycerides (TG) all showed an upward trend (P for trend < 0.05 for all). The levels of FPG in the two groups remained in a relatively stable during the whole pregnancy even though it was higher in OG during the 2nd and 3rd trimesters, whilst HbAlc levels in NG women increased (P for trend = 0.043) during pregnancy. Meanwhile, the risk of macrosomia and large-for-gestational-age (LGA) increased with the increase of FPG level (P for trend < 0.05). Multivariate logistic regression analyses results showed that only FPG level in the 3rd trimester was correlated with birth weight, with birth weight increased by 44.9 g for each SD increase in FPG level. CONCLUSION Maternal FPG in the 3rd trimester is an independent predictor of newborn birth weight, and a higher level of that is associated with an increased risk of macrosomia and LGA.
Collapse
Affiliation(s)
- Jing Ji
- Department of Nutrition and Food Safety, School of Public Health, Xi'an Jiaotong University, Xi'an, 710061, China
- Department of Obstetrics and Gynecology, Northwest Women's and Children's Hospital, Xi'an, 710061, China
| | - Pei Wu
- Department of Nutrition and Food Safety, School of Public Health, Xi'an Jiaotong University, Xi'an, 710061, China
- Key Laboratory of Population Health Across Life Cycle of Ministry of Education, Hefei, China
| | - Guohua Li
- Department of Nutrition and Food Safety, School of Public Health, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhangya He
- Department of Nutrition and Food Safety, School of Public Health, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shanshan Wang
- Department of Nutrition and Food Safety, School of Public Health, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wenlu Yu
- Department of Nutrition and Food Safety, School of Public Health, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Chao Li
- Department of Epidemiology and Health Statistics, School of Public Health, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yang Mi
- Department of Obstetrics and Gynecology, Northwest Women's and Children's Hospital, Xi'an, 710061, China
| | - Xiaoqin Luo
- Department of Nutrition and Food Safety, School of Public Health, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
11
|
Martins MG, Silver Z, Ayoub K, Hyland L, Woodside B, Kiss ACI, Abizaid A. Maternal glucose intolerance during pregnancy affects offspring POMC expression and results in adult metabolic alterations in a sex-dependent manner. Front Endocrinol (Lausanne) 2023; 14:1189207. [PMID: 37396180 PMCID: PMC10311085 DOI: 10.3389/fendo.2023.1189207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction Gestational diabetes (GDM) is associated with negative outcomes in mothers and their offspring, including greater risks of macrosomia at birth and the development of metabolic disorders. While these outcomes are well-established, the mechanisms by which this increased metabolic vulnerability is conferred on the offspring are comparatively lacking. One proposed mechanism is that maternal glycemic dysregulation alters the development of the hypothalamic regions related to metabolism and energy balance. Methods To investigate this possibility, in this study, we first examined the effects of STZ-induced maternal glucose intolerance on the offspring on pregnancy day (PD) 19, and, in a second experiment, in early adulthood (postnatal day (PND) 60). Whether effects would be influenced by sex, or exposure of offspring to a high-fat diet was also investigated. The impact of maternal STZ treatment on POMC neuron number in the ARC of offspring at both time points was also examined. Results As expected, STZ administration on PD 7 decreased maternal glucose tolerance, and increased risk for macrosomia, and loss of pups at birth. Offspring of STZ-treated mothers were also more vulnerable to developing metabolic impairments in adulthood. These were accompanied by sex-specific effects of maternal STZ treatment in the offspring, including fewer POMC neurons in the ARC of female but not male infants in late pregnancy and a higher number of POMC neurons in the ARC of both male and female adult offspring of STZ-treated dams, which was exacerbated in females exposed to a high-fat diet after weaning. Discussion This work suggests that maternal hyperglycemia induced by STZ treatment, in combination with early-life exposure to an obesogenic diet, leads to adult metabolic alterations that correlate with the increased hypothalamic expression of POMC, showing that maternal glycemic dysregulation can impact the development of hypothalamic circuits regulating energy state with a stronger impact on female offspring.
Collapse
Affiliation(s)
- Marina Galleazzo Martins
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
- Department of Physiology, Institute of Biosciences of the University of São Paulo (IB/USP), São Paulo, São Paulo, Brazil
| | - Zachary Silver
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Kiara Ayoub
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Lindsay Hyland
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Barbara Woodside
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Ana Carolina Inhasz Kiss
- Department of Physiology, Institute of Biosciences of the University of São Paulo (IB/USP), São Paulo, São Paulo, Brazil
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| | - Alfonso Abizaid
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
12
|
Francis EC, Kechris K, Jansson T, Dabelea D, Perng W. Novel Metabolic Subtypes in Pregnant Women and Risk of Early Childhood Obesity in Offspring. JAMA Netw Open 2023; 6:e237030. [PMID: 37014638 PMCID: PMC10074224 DOI: 10.1001/jamanetworkopen.2023.7030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/21/2023] [Indexed: 04/05/2023] Open
Abstract
Importance The in utero metabolic milieu is associated with offspring adiposity. Standard definitions of maternal obesity (according to prepregnancy body mass index [BMI]) and gestational diabetes (GDM) may not be adequate to capture subtle yet important differences in the intrauterine environment that could be involved in programming. Objectives To identify maternal metabolic subgroups during pregnancy and to examine associations of subgroup classification with adiposity traits in their children. Design, Setting, and Participants This cohort study included mother-offspring pairs in the Healthy Start prebirth cohort (enrollment: 2010-2014) recruited from University of Colorado Hospital obstetrics clinics in Aurora, Colorado. Follow-up of women and children is ongoing. Data were analyzed from March to December 2022. Exposures Metabolic subtypes of pregnant women ascertained by applying k-means clustering on 7 biomarkers and 2 biomarker indices measured at approximately 17 gestational weeks: glucose, insulin, Homeostatic Model Assessment for Insulin Resistance, total cholesterol, high-density lipoprotein cholesterol (HDL-C), triglycerides, free fatty acids (FFA), HDL-C:triglycerides ratio, and tumor necrosis factor α. Main Outcomes and Measures Offspring birthweight z score and neonatal fat mass percentage (FM%). In childhood at approximately 5 years of age, offspring BMI percentile, FM%, BMI in the 95th percentile or higher, and FM% in the 95th percentile or higher. Results A total of 1325 pregnant women (mean [SD] age, 27.8 [6.2 years]; 322 [24.3%] Hispanic, 207 non-Hispanic Black [15.6%], and 713 [53.8%] non-Hispanic White), and 727 offspring with anthropometric data measured in childhood (mean [SD] age 4.81 [0.72] years, 48% female) were included. We identified the following 5 maternal metabolic subgroups: reference (438 participants), high HDL-C (355 participants), dyslipidemic-high triglycerides (182 participants), dyslipidemic-high FFA (234 participants), and insulin resistant (IR)-hyperglycemic (116 participants). Compared with the reference subgroup, women in the IR-hyperglycemic and dyslipidemic-high FFA subgroups had offspring with 4.27% (95% CI, 1.94-6.59) and 1.96% (95% CI, 0.45-3.47) greater FM% during childhood, respectively. There was a higher risk of high FM% among offspring of the IR-hyperglycemic (relative risk, 8.7; 95% CI, 2.7-27.8) and dyslipidemic-high FFA (relative risk, 3.4; 95% CI, 1.0-11.3) subgroups; this risk was of greater magnitude compared with prepregnancy obesity alone, GDM alone, or both conditions. Conclusions and Relevance In this cohort study, an unsupervised clustering approach revealed distinct metabolic subgroups of pregnant women. These subgroups exhibited differences in risk of offspring adiposity in early childhood. Such approaches have the potential to refine understanding of the in utero metabolic milieu, with utility for capturing variation in sociocultural, anthropometric, and biochemical risk factors for offspring adiposity.
Collapse
Affiliation(s)
- Ellen C. Francis
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, Colorado
| | - Katerina Kechris
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, Colorado
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora
| | - Dana Dabelea
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, Colorado
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora
| | - Wei Perng
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, Colorado
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora
| |
Collapse
|
13
|
Luo SS, Zhu H, Huang HF, Ding GL. Sex differences in glycolipidic disorders after exposure to maternal hyperglycemia during early development. J Endocrinol Invest 2023:10.1007/s40618-023-02069-5. [PMID: 36976483 DOI: 10.1007/s40618-023-02069-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 03/10/2023] [Indexed: 03/29/2023]
Abstract
PURPOSE The aim of this review was to summarize sex differences in glycolipid metabolic phenotypes of human and animal models after exposure to maternal hyperglycemia and overview the underlying mechanisms, providing a new perspective on the maternal hyperglycemia-triggered risk of glycolipidic disorders in offspring. METHODS A comprehensive literature search within PubMed was performed. Selected publications related to studies on offspring exposed to maternal hyperglycemia investigating the sex differences of glycolipid metabolism were reviewed. RESULTS Maternal hyperglycemia increases the risk of glycolipid metabolic disorders in offspring, such as obesity, glucose intolerance and diabetes. Whether with or without intervention, metabolic phenotypes have been shown to exhibit sex differences between male and female offspring in response to maternal hyperglycemia, which may be related to gonadal hormones, organic intrinsic differences, placenta, and epigenetic modifications. CONCLUSION Sex may play a role in the different incidences and pathogenesis of abnormal glycolipid metabolism. More studies investigating both sexes are needed to understand how and why environmental conditions in early life affect long-term health between male and female individuals.
Collapse
Affiliation(s)
- S-S Luo
- School of Medicine, The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - H Zhu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - H-F Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - G-L Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| |
Collapse
|
14
|
Kurniansyah N, Wallace DA, Zhang Y, Yu B, Cade B, Wang H, Ochs-Balcom HM, Reiner AP, Ramos AR, Smith JD, Cai J, Daviglus M, Zee PC, Kaplan R, Kooperberg C, Rich SS, Rotter JI, Gharib SA, Redline S, Sofer T. An integrated multi-omics analysis of sleep-disordered breathing traits implicates P2XR4 purinergic signaling. Commun Biol 2023; 6:125. [PMID: 36721044 PMCID: PMC9889381 DOI: 10.1038/s42003-023-04520-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/23/2023] [Indexed: 02/01/2023] Open
Abstract
Sleep Disordered Breathing (SDB) is a common disease associated with increased risk for cardiometabolic, cardiovascular, and cognitive diseases. How SDB affects the molecular environment is still poorly understood. We study the association of three SDB measures with gene expression measured using RNA-seq in multiple blood tissues from the Multi-Ethnic Study of Atherosclerosis. We develop genetic instrumental variables for the associated transcripts as polygenic risk scores (tPRS), then generalize and validate the tPRS in the Women's Health Initiative. We measure the associations of the validated tPRS with SDB and serum metabolites in Hispanic Community Health Study/Study of Latinos. Here we find differential gene expression by blood cell type in relation to SDB traits and link P2XR4 expression to average oxyhemoglobin saturation during sleep and butyrylcarnitine (C4) levels. These findings can be used to develop interventions to alleviate the effect of SDB on the human molecular environment.
Collapse
Affiliation(s)
- Nuzulul Kurniansyah
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Danielle A Wallace
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Ying Zhang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Brian Cade
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Heming Wang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Heather M Ochs-Balcom
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Alexander P Reiner
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Alberto R Ramos
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joshua D Smith
- Northwest Genomic Center, University of Washington, Seattle, WA, USA
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina, at Chapel Hill, NC, USA
| | - Martha Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Phyllis C Zee
- Division of Sleep Medicine, Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Robert Kaplan
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Epidemiology & Population Health, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Sina A Gharib
- Computational Medicine Core, Center for Lung Biology, UW Medicine Sleep Center, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA.
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA.
- Departments of Medicine and of Biostatistics, Harvard University, Boston, MA, USA.
| |
Collapse
|
15
|
Menendez A, Wanczyk H, Walker J, Zhou B, Santos M, Finck C. Obesity and Adipose Tissue Dysfunction: From Pediatrics to Adults. Genes (Basel) 2022; 13:genes13101866. [PMID: 36292751 PMCID: PMC9601855 DOI: 10.3390/genes13101866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/27/2022] [Accepted: 10/14/2022] [Indexed: 11/04/2022] Open
Abstract
Obesity is a growing health problem that affects both children and adults. The increasing prevalence of childhood obesity is associated with comorbidities such as cardiovascular disease, type 2 diabetes and metabolic syndrome due to chronic low-grade inflammation present at early stages of the disease. In pediatric patients suffering from obesity, the role of epigenetics, the gut microbiome and intrauterine environment have emerged as causative factors Interestingly, pediatric obesity is strongly associated with low birth weight. Accelerated weight gain oftentimes occurs in these individuals during the post-natal period, which can lead to increased risk of adiposity and metabolic disease. The pathophysiology of obesity is complex and involves biological and physiological factors compounded by societal factors such as family and community. On a cellular level, adipocytes contained within adipose tissue become dysregulated and further contribute to development of comorbidities similar to those present in adults with obesity. This review provides an overview of the current understanding of adipose tissue immune, inflammatory and metabolic adaptation of the adipose tissue in obesity. Early cellular changes as well as the role of immune cells and inflammation on the progression of disease in pivotal pediatric clinical trials, adult studies and mouse models are emphasized. Understanding the initial molecular and cellular changes that occur during obesity can facilitate new and improved treatments aimed at early intervention and subsequent prevention of adulthood comorbidities.
Collapse
Affiliation(s)
- Ana Menendez
- Connecticut Children’s Medical Center, Department of Pediatric Endocrinology, Hartford, CT 06106, USA
| | - Heather Wanczyk
- University of Connecticut Health Center, Department of Pediatrics, Farmington, CT 06030, USA
| | - Joanne Walker
- University of Connecticut Health Center, Department of Pediatrics, Farmington, CT 06030, USA
| | - Beiyan Zhou
- University of Connecticut Health Center, Department of Immunology, Farmington, CT 06030, USA
| | - Melissa Santos
- Connecticut Children’s Medical Center, Department of Pediatric Psychology and Director of the Obesity Center, Hartford, CT 06106, USA
| | - Christine Finck
- Connecticut Children’s Medical Center, Department of Surgery and Pediatric Bariatric Surgery, Hartford, CT 06106, USA
- Correspondence: ; Tel.: +860-545-9520
| |
Collapse
|
16
|
Cohen CC, Dabelea D, Michelotti G, Tang L, Shankar K, Goran MI, Perng W. Metabolome Alterations Linking Sugar-Sweetened Beverage Intake with Dyslipidemia in Youth: The Exploring Perinatal Outcomes among CHildren (EPOCH) Study. Metabolites 2022; 12:metabo12060559. [PMID: 35736491 PMCID: PMC9228193 DOI: 10.3390/metabo12060559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/17/2022] Open
Abstract
The objective of this study was to assess intermediary metabolic alterations that link sugar-sweetened beverage (SSB) intake to cardiometabolic (CM) risk factors in youth. A total of 597 participants from the multi-ethnic, longitudinal Exploring Perinatal Outcomes among CHildren (EPOCH) Study were followed in childhood (median 10 yrs) and adolescence (median 16 yrs). We used a multi-step approach: first, mixed models were used to examine the associations of SSB intake in childhood with CM measures across childhood and adolescence, which revealed a positive association between SSB intake and fasting triglycerides (β (95% CI) for the highest vs. lowest SSB quartile: 8.1 (−0.9,17.0); p-trend = 0.057). Second, least absolute shrinkage and selection operator (LASSO) regression was used to select 180 metabolite features (out of 767 features assessed by untargeted metabolomics) that were associated with SSB intake in childhood. Finally, 13 of these SSB-associated metabolites (from step two) were also prospectively associated with triglycerides across follow-up (from step one) in the same direction as with SSB intake (Bonferroni-adj. p < 0.0003). All annotated compounds were lipids, particularly dicarboxylated fatty acids, mono- and diacylglycerols, and phospholipids. In this diverse cohort, we identified a panel of lipid metabolites that may serve as intermediary biomarkers, linking SSB intake to dyslipidemia risk in youth.
Collapse
Affiliation(s)
- Catherine C. Cohen
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (D.D.); (K.S.)
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA;
- Correspondence:
| | - Dana Dabelea
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (D.D.); (K.S.)
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA;
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | | | - Lu Tang
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Kartik Shankar
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (D.D.); (K.S.)
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Michael I. Goran
- Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90007, USA;
| | - Wei Perng
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA;
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| |
Collapse
|
17
|
The newborn metabolome: associations with gestational diabetes, sex, gestation, birth mode, and birth weight. Pediatr Res 2022; 91:1864-1873. [PMID: 34526650 DOI: 10.1038/s41390-021-01672-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 07/07/2021] [Accepted: 07/14/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Pathways towards many adult-onset conditions begin early in life, even in utero. Maternal health in pregnancy influences this process, but little is known how it affects neonatal metabolism. We investigated associations between pregnancy and birth factors and cord blood metabolomic profile in a large, population-derived cohort. METHODS Metabolites were measured using nuclear magnetic resonance in maternal (28 weeks gestation) and cord serum from 912 mother-child pairs in the Barwon Infant Study pre-birth cohort. Associations between maternal (metabolites, age, BMI, smoking), pregnancy (pre-eclampsia, gestational diabetes (GDM)), and birth characteristics (delivery mode, gestational age, weight, infant sex) with 72 cord blood metabolites were examined by linear regression. RESULTS Delivery mode, sex, gestational age, and birth weight were associated with specific metabolite levels in cord blood, including amino acids, fatty acids, and cholesterols. GDM was associated with higher cord blood levels of acetoacetate and 3-hydroxybutyrate. CONCLUSIONS Neonatal factors, particularly delivery mode, were associated with many cord blood metabolite differences, including those implicated in later risk of cardiometabolic disease. Associations between GDM and higher offspring ketone levels at birth are consistent with maternal ketosis in diabetic pregnancies. Further work is needed to determine whether these neonatal metabolome differences associate with later health outcomes. IMPACT Variations in blood metabolomic profile have been linked to health status in adults and children, but corresponding data in neonates are scarce. We report evidence that pregnancy complications, mode of delivery, and offspring characteristics, including sex, are independently associated with a range of circulating metabolites at birth, including ketone bodies, amino acids, cholesterols, and inflammatory markers. Independent of birth weight, exposure to gestational diabetes is associated with higher cord blood ketone bodies and citrate. These findings suggest that pregnancy complications, mode of delivery, gestational age, and measures of growth influence metabolic pathways prior to birth, potentially impacting later health and development.
Collapse
|
18
|
Perng W, Hivert MF, Michelotti G, Oken E, Dabelea D. Metabolomic Predictors of Dysglycemia in Two U.S. Youth Cohorts. Metabolites 2022; 12:404. [PMID: 35629908 PMCID: PMC9147862 DOI: 10.3390/metabo12050404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 01/27/2023] Open
Abstract
Here, we seek to identify metabolite predictors of dysglycemia in youth. In the discovery analysis among 391 youth in the Exploring Perinatal Outcomes among CHildren (EPOCH) cohort, we used reduced rank regression (RRR) to identify sex-specific metabolite predictors of impaired fasting glucose (IFG) and elevated fasting glucose (EFG: Q4 vs. Q1 fasting glucose) 6 years later and compared the predictive capacity of four models: Model 1: ethnicity, parental diabetes, in utero exposure to diabetes, and body mass index (BMI); Model 2: Model 1 covariates + baseline waist circumference, insulin, lipids, and Tanner stage; Model 3: Model 2 + baseline fasting glucose; Model 4: Model 3 + baseline metabolite concentrations. RRR identified 19 metabolite predictors of fasting glucose in boys and 14 metabolite predictors in girls. Most compounds were on lipid, amino acid, and carbohydrate metabolism pathways. In boys, no improvement in aurea under the receiver operating characteristics curve AUC occurred until the inclusion of metabolites in Model 4, which increased the AUC for prediction of IFG (7.1%) from 0.81 to 0.97 (p = 0.002). In girls, %IFG was too low for regression analysis (3.1%), but we found similar results for EFG. We replicated the results among 265 youth in the Project Viva cohort, focusing on EFG due to low %IFG, suggesting that the metabolite profiles identified herein have the potential to improve the prediction of glycemia in youth.
Collapse
Affiliation(s)
- Wei Perng
- Lifcourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse (CoRAL), Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02215, USA; (M.-F.H.); (E.O.)
- Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse (CoRAL), Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02215, USA; (M.-F.H.); (E.O.)
- Department of Nutrition, T. H. Chan Harvard School of Public Health, Boston, MA 02115, USA
| | - Dana Dabelea
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
19
|
Francis EC, Kechris K, Cohen CC, Michelotti G, Dabelea D, Perng W. Metabolomic Profiles in Childhood and Adolescence Are Associated with Fetal Overnutrition. Metabolites 2022; 12:265. [PMID: 35323708 PMCID: PMC8952572 DOI: 10.3390/metabo12030265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Fetal overnutrition predisposes offspring to increased metabolic risk. The current study used metabolomics to assess sustained differences in serum metabolites across childhood and adolescence among youth exposed to three typologies of fetal overnutrition: maternal obesity only, gestational diabetes mellitus (GDM) only, and obesity + GDM. We included youth exposed in utero to obesity only (BMI ≥ 30; n = 66), GDM only (n = 56), obesity + GDM (n = 25), or unexposed (n = 297), with untargeted metabolomics measured at ages 10 and 16 years. We used linear mixed models to identify metabolites across both time-points associated with exposure to any overnutrition, using a false-discovery-rate correction (FDR) <0.20. These metabolites were included in a principal component analysis (PCA) to generate profiles and assess metabolite profile differences with respect to overnutrition typology (adjusted for prenatal smoking, offspring age, sex, and race/ethnicity). Fetal overnutrition was associated with 52 metabolites. PCA yielded four factors accounting for 17−27% of the variance, depending on age of measurement. We observed differences in three factor patterns with respect to overnutrition typology: sphingomyelin-mannose (8−13% variance), skeletal muscle metabolism (6−10% variance), and 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid (CMPF; 3−4% variance). The sphingomyelin-mannose factor score was higher among offspring exposed to obesity vs. GDM. Exposure to obesity + GDM (vs. GDM or obesity only) was associated with higher skeletal muscle metabolism and CMPF scores. Fetal overnutrition is associated with metabolic changes in the offspring, but differences between typologies of overnutrition account for a small amount of variation in the metabolome, suggesting there is likely greater pathophysiological overlap than difference.
Collapse
Affiliation(s)
- Ellen C. Francis
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (C.C.C.); (D.D.); (W.P.)
| | - Katerina Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Catherine C. Cohen
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (C.C.C.); (D.D.); (W.P.)
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | | | - Dana Dabelea
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (C.C.C.); (D.D.); (W.P.)
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Wei Perng
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA; (C.C.C.); (D.D.); (W.P.)
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
20
|
Handakas E, Lau CH, Alfano R, Chatzi VL, Plusquin M, Vineis P, Robinson O. A systematic review of metabolomic studies of childhood obesity: State of the evidence for metabolic determinants and consequences. Obes Rev 2022; 23 Suppl 1:e13384. [PMID: 34797026 DOI: 10.1111/obr.13384] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022]
Abstract
Childhood obesity has become a global epidemic and carries significant long-term consequences to physical and mental health. Metabolomics, the global profiling of small molecules or metabolites, may reveal the mechanisms of development of childhood obesity and clarify links between obesity and metabolic disease. A systematic review of metabolomic studies of childhood obesity was conducted, following Preferred Reporting Items for Systematic Reviews (PRISMA) guidelines, searching across Scopus, Ovid, Web of Science and PubMed databases for articles published from January 1, 2005 to July 8, 2020, retrieving 1271 different records and retaining 41 articles for qualitative synthesis. Study quality was assessed using a modified Newcastle-Ottawa Scale. Thirty-three studies were conducted on blood, six on urine, three on umbilical cord blood, and one on saliva. Thirty studies were primarily cross-sectional, five studies were primarily longitudinal, and seven studies examined effects of weight-loss following a life-style intervention. A consistent metabolic profile of childhood obesity was observed including amino acids (particularly branched chain and aromatic), carnitines, lipids, and steroids. Although the use of metabolomics in childhood obesity research is still developing, the identified metabolites have provided additional insight into the pathogenesis of many obesity-related diseases. Further longitudinal research is needed into the role of metabolic profiles and child obesity risk.
Collapse
Affiliation(s)
- Evangelos Handakas
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Chung Ho Lau
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Rossella Alfano
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.,Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Vaia Lida Chatzi
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Michelle Plusquin
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.,Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Paolo Vineis
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Oliver Robinson
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| |
Collapse
|
21
|
Shashikadze B, Flenkenthaler F, Stöckl JB, Valla L, Renner S, Kemter E, Wolf E, Fröhlich T. Developmental Effects of (Pre-)Gestational Diabetes on Offspring: Systematic Screening Using Omics Approaches. Genes (Basel) 2021; 12:1991. [PMID: 34946940 PMCID: PMC8701487 DOI: 10.3390/genes12121991] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 12/27/2022] Open
Abstract
Worldwide, gestational diabetes affects 2-25% of pregnancies. Due to related disturbances of the maternal metabolism during the periconceptional period and pregnancy, children bear an increased risk for future diseases. It is well known that an aberrant intrauterine environment caused by elevated maternal glucose levels is related to elevated risks for increased birth weights and metabolic disorders in later life, such as obesity or type 2 diabetes. The complexity of disturbances induced by maternal diabetes, with multiple underlying mechanisms, makes early diagnosis or prevention a challenging task. Omics technologies allowing holistic quantification of several classes of molecules from biological fluids, cells, or tissues are powerful tools to systematically investigate the effects of maternal diabetes on the offspring in an unbiased manner. Differentially abundant molecules or distinct molecular profiles may serve as diagnostic biomarkers, which may also support the development of preventive and therapeutic strategies. In this review, we summarize key findings from state-of-the-art Omics studies addressing the impact of maternal diabetes on offspring health.
Collapse
Affiliation(s)
- Bachuki Shashikadze
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377 Munich, Germany; (B.S.); (F.F.); (J.B.S.)
| | - Florian Flenkenthaler
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377 Munich, Germany; (B.S.); (F.F.); (J.B.S.)
| | - Jan B. Stöckl
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377 Munich, Germany; (B.S.); (F.F.); (J.B.S.)
| | - Libera Valla
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; (L.V.); (S.R.); (E.K.)
| | - Simone Renner
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; (L.V.); (S.R.); (E.K.)
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; (L.V.); (S.R.); (E.K.)
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Eckhard Wolf
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377 Munich, Germany; (B.S.); (F.F.); (J.B.S.)
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; (L.V.); (S.R.); (E.K.)
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377 Munich, Germany; (B.S.); (F.F.); (J.B.S.)
| |
Collapse
|
22
|
De Spiegeleer M, De Paepe E, Van Meulebroek L, Gies I, De Schepper J, Vanhaecke L. Paediatric obesity: a systematic review and pathway mapping of metabolic alterations underlying early disease processes. Mol Med 2021; 27:145. [PMID: 34742239 PMCID: PMC8571978 DOI: 10.1186/s10020-021-00394-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The alarming trend of paediatric obesity deserves our greatest awareness to hinder the early onset of metabolic complications impacting growth and functionality. Presently, insight into molecular mechanisms of childhood obesity and associated metabolic comorbidities is limited. This systematic review aimed at scrutinising what has been reported on putative metabolites distinctive for metabolic abnormalities manifesting at young age by searching three literature databases (Web of Science, Pubmed and EMBASE) during the last 6 years (January 2015-January 2021). Global metabolomic profiling of paediatric obesity was performed (multiple biological matrices: blood, urine, saliva and adipose tissue) to enable overarching pathway analysis and network mapping. Among 2792 screened Q1 articles, 40 met the eligibility criteria and were included to build a database on metabolite markers involved in the spectrum of childhood obesity. Differential alterations in multiple pathways linked to lipid, carbohydrate and amino acid metabolisms were observed. High levels of lactate, pyruvate, alanine and acetate marked a pronounced shift towards hypoxic conditions in children with obesity, and, together with distinct alterations in lipid metabolism, pointed towards dysbiosis and immunometabolism occurring early in life. Additionally, aberrant levels of several amino acids, most notably belonging to tryptophan metabolism including the kynurenine pathway and its relation to histidine, phenylalanine and purine metabolism were displayed. Moreover, branched-chain amino acids were linked to lipid, carbohydrate, amino acid and microbial metabolism, inferring a key role in obesity-associated insulin resistance. CONCLUSIONS This systematic review revealed that the main metabolites at the crossroad of dysregulated metabolic pathways underlying childhood obesity could be tracked down to one central disturbance, i.e. impending insulin resistance for which reference values and standardised measures still are lacking. In essence, glycolytic metabolism was evinced as driving energy source, coupled to impaired Krebs cycle flux and ß-oxidation. Applying metabolomics enabled to retrieve distinct metabolite alterations in childhood obesity(-related insulin resistance) and associated pathways at early age and thus could provide a timely indication of risk by elucidating early-stage biomarkers as hallmarks of future metabolically unhealthy phenotypes.
Collapse
Affiliation(s)
- Margot De Spiegeleer
- Laboratory of Chemical Analysis, Department of Translational Physiology, Infectiology and Public Health, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Ellen De Paepe
- Laboratory of Chemical Analysis, Department of Translational Physiology, Infectiology and Public Health, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Lieven Van Meulebroek
- Laboratory of Chemical Analysis, Department of Translational Physiology, Infectiology and Public Health, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium
| | - Inge Gies
- KidZ Health Castle, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090, Brussel, Belgium
| | - Jean De Schepper
- KidZ Health Castle, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090, Brussel, Belgium.,Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Lynn Vanhaecke
- Laboratory of Chemical Analysis, Department of Translational Physiology, Infectiology and Public Health, Ghent University, Salisburylaan 133, 9820, Merelbeke, Belgium. .,Institute for Global Food Security, School of Biological Sciences, Queen's University, University Road, Belfast, BT7 1NN, UK.
| |
Collapse
|
23
|
Juvinao-Quintero DL, Cardenas A, Perron P, Bouchard L, Lutz SM, Hivert MF. Associations between an integrated component of maternal glycemic regulation in pregnancy and cord blood DNA methylation. Epigenomics 2021; 13:1459-1472. [PMID: 34596421 DOI: 10.2217/epi-2021-0220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: Previous studies suggest that fetal programming to hyperglycemia in pregnancy is due to modulation of DNA methylation (DNAm), but they have been limited in their maternal glycemic characterization. Methods: In the Gen3G study, we used a principal component analysis to integrate multiple glucose and insulin values measured during the second trimester oral glucose tolerance test. We investigated associations between principal components and cord blood DNAm levels in an epigenome-wide analysis among 430 mother-child pairs. Results: The first principal component was robustly associated with lower DNAm at cg26974062 (TXNIP; p = 9.9 × 10-9) in cord blood. TXNIP is a well-known DNAm marker for type 2 diabetes in adults. Conclusion: We hypothesize that abnormal glucose metabolism in pregnancy may program dysregulation of TXNIP across the life course.
Collapse
Affiliation(s)
- Diana L Juvinao-Quintero
- Division of Chronic Disease Research Across the Life Course, Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Andres Cardenas
- Division of Environmental Health Sciences, School of Public Health & Center for Computational Biology, University of California, Berkeley, CA 94720-7360, USA
| | - Patrice Perron
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, J1H 5N4, Canada.,Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, J1H 5N4, Canada
| | - Luigi Bouchard
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, J1H 5N4, Canada.,Department of Medical Biology, Centre Intégré Universitaire en Santé et Services Sociaux Saguenay-Lac-Saint-Jean, Hôpital Universitaire de Chicoutimi, Saguenay, QC, G7H 5H6, Canada.,Department of Biochemistry & Functional Genomics, Université de Sherbrooke, Sherbrooke, QC, J1K 2R1, Canada
| | - Sharon M Lutz
- Division of Chronic Disease Research Across the Life Course, Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02215, USA.,Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA 02215, USA
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Life Course, Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, J1H 5N4, Canada.,Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
24
|
Zhu B, Han Y, Deng F, Huang K, Yan S, Hao J, Zhu P, Tao F. The role of triiodothyronine (T3) and T3/free thyroxine (fT4) in glucose metabolism during pregnancy: the Ma'anshan birth cohort study. Endocr Connect 2021; 10:685-693. [PMID: 34077392 PMCID: PMC8240711 DOI: 10.1530/ec-21-0088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 06/02/2021] [Indexed: 01/21/2023]
Abstract
OBJECTIVES Compared with other thyroid markers, fewer studies have explored the associations between triiodothyronine (T3), T3/free thyroxine (fT4) and glucose abnormality during pregnancy. Thus, we aimed to: (i) examine the associations of T3 and T3/fT4 with glucose metabolism indicators and (ii) evaluate, in the first trimester, the performance of the two markers as predictors of gestational diabetes mellitus (GDM) risk. METHODS Longitudinal data from 2723 individuals, consisting of three repeated measurements of T3 and fT4, from the Man'anshan birth cohort study (MABC), China, were analyzed using a time-specific generalized estimating equation (GEE). The receiver operating characteristic curve (ROC) - area under the curve (AUC) and Hosmer-Lemeshow goodness of fit test was used to assess the discrimination and calibration of prediction models. RESULTS T3 and T3/fT4 presented stable associations with the level of fasting glucose, glucose at 1h/2 h during pregnancy. T3 and T3/fT4 in both the first and second trimesters were positively associated with the risk of GDM, with the larger magnitude of association observed in the second trimester (odds ratio (OR) = 2.50, 95% CI = 1.95, 3.21 for T3; OR = 1.09, 95% CI = 1.07, 1.12 for T3/fT4). T3 ((AUC) = 0.726, 95% CI = 0.698, 0.754) and T3/fT4 (AUC = 0.724, 95% CI = 0.696, 0.753) in the first trimester could improve the performance of the prediction model; however, the overall performance is not good. CONCLUSION Significant and stable associations of T3, T3/fT4 and glucose metabolism indicators were documented. Both T3 and T3/fT4 improve the performance of the GDM predictive model.
Collapse
Affiliation(s)
- Beibei Zhu
- School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China
| | - Yan Han
- School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China
| | - Fen Deng
- School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China
| | - Kun Huang
- School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China
| | - Shuangqin Yan
- Ma’anshan Maternal and Child Health Care Center, Ma’anshan, Anhui, China
| | - Jiahu Hao
- School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China
| | - Peng Zhu
- School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China
| | - Fangbiao Tao
- School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, Anhui, China
- Correspondence should be addressed to F Tao:
| |
Collapse
|
25
|
Wang M, Hu RY, Gong WW, Pan J, Fei FR, Wang H, Zhou XY, Zhong JM, Yu M. Trends in prevalence of gestational diabetes mellitus in Zhejiang Province, China, 2016-2018. Nutr Metab (Lond) 2021; 18:12. [PMID: 33468171 PMCID: PMC7814615 DOI: 10.1186/s12986-020-00539-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/30/2020] [Indexed: 11/10/2022] Open
Abstract
Background Limited population-based studies have investigated the secular trend of prevalence of gestational diabetes mellitus (GDM) in mainland China. Therefore, this study aimed to estimate the prevalence of GDM and time trends in Chinese female population. Methods Based on Diabetes Surveillance System of Zhejiang Province, 97,063 diagnosed GDM cases aged 20–50 years were identified from January 1, 2016 to December 31, 2018. Annual prevalence, prevalence rate ratios (PRRs) and average annual percentage change with their 95% confidence intervals (CIs) were reported. Results The age-standardized overall prevalence of GDM was reported to be 7.30% (95% CI 7.27–7.33%);
9.13% (95% CI 9.07–9.19%) in urban areas and 6.24% (95% CI 6.21–6.27%) in rural areas. Compared with 20–24 years age group, women in advanced age groups (25–50 years) were at higher risk for GDM (PRRs ranged from 1.37 to 8.95 and the 95% CIs did not include the null). Compared with rural areas, the risk for GDM was higher in urban areas (PRR: 1.69, 95% CI 1.67–1.72). The standardized annual prevalence increased from 6.02% in 2016 to 7.94% in 2018, with an average annual increase of 5.48%, and grew more rapidly in rural than urban areas (11.28% vs. 0.00%). Conclusions This study suggested a significant increase in the prevalence of GDM among Chinese female population in Zhejiang province during 2016–2018, especially in women characterized by advanced age and rural areas.
Collapse
Affiliation(s)
- Meng Wang
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Binsheng Road, Hangzhou, 310051, China
| | - Ru-Ying Hu
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Binsheng Road, Hangzhou, 310051, China
| | - Wei-Wei Gong
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Binsheng Road, Hangzhou, 310051, China
| | - Jin Pan
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Binsheng Road, Hangzhou, 310051, China
| | - Fang-Rong Fei
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Binsheng Road, Hangzhou, 310051, China
| | - Hao Wang
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Binsheng Road, Hangzhou, 310051, China
| | - Xiao-Yan Zhou
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Binsheng Road, Hangzhou, 310051, China
| | - Jie-Ming Zhong
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Binsheng Road, Hangzhou, 310051, China.
| | - Min Yu
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Binsheng Road, Hangzhou, 310051, China.
| |
Collapse
|
26
|
Ott R, Pawlow X, Weiß A, Hofelich A, Herbst M, Hummel N, Prehn C, Adamski J, Römisch-Margl W, Kastenmüller G, Ziegler AG, Hummel S. Intergenerational Metabolomic Analysis of Mothers with a History of Gestational Diabetes Mellitus and Their Offspring. Int J Mol Sci 2020; 21:E9647. [PMID: 33348910 PMCID: PMC7766614 DOI: 10.3390/ijms21249647] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/05/2022] Open
Abstract
Shared metabolomic patterns at delivery have been suggested to underlie the mother-to-child transmission of adverse metabolic health. This study aimed to investigate whether mothers with gestational diabetes mellitus (GDM) and their offspring show similar metabolomic patterns several years postpartum. Targeted metabolomics (including 137 metabolites) was performed in plasma samples obtained during an oral glucose tolerance test from 48 mothers with GDM and their offspring at a cross-sectional study visit 8 years after delivery. Partial Pearson's correlations between the area under the curve (AUC) of maternal and offspring metabolites were calculated, yielding so-called Gaussian graphical models. Spearman's correlations were applied to investigate correlations of body mass index (BMI), Matsuda insulin sensitivity index (ISI-M), dietary intake, and physical activity between generations, and correlations of metabolite AUCs with lifestyle variables. This study revealed that BMI, ISI-M, and the AUC of six metabolites (carnitine, taurine, proline, SM(-OH) C14:1, creatinine, and PC ae C34:3) were significantly correlated between mothers and offspring several years postpartum. Intergenerational metabolite correlations were independent of shared BMI, ISI-M, age, sex, and all other metabolites. Furthermore, creatinine was correlated with physical activity in mothers. This study suggests that there is long-term metabolic programming in the offspring of mothers with GDM and informs us about targets that could be addressed by future intervention studies.
Collapse
Affiliation(s)
- Raffael Ott
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, 85764 Neuherberg, Germany; (R.O.); (X.P.); (A.W.); (A.H.); (M.H.); (N.H.); (A.-G.Z.)
- Forschergruppe Diabetes e.V., 85764 Neuherberg, Germany
| | - Xenia Pawlow
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, 85764 Neuherberg, Germany; (R.O.); (X.P.); (A.W.); (A.H.); (M.H.); (N.H.); (A.-G.Z.)
- Forschergruppe Diabetes e.V., 85764 Neuherberg, Germany
| | - Andreas Weiß
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, 85764 Neuherberg, Germany; (R.O.); (X.P.); (A.W.); (A.H.); (M.H.); (N.H.); (A.-G.Z.)
- Forschergruppe Diabetes e.V., 85764 Neuherberg, Germany
| | - Anna Hofelich
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, 85764 Neuherberg, Germany; (R.O.); (X.P.); (A.W.); (A.H.); (M.H.); (N.H.); (A.-G.Z.)
- Forschergruppe Diabetes e.V., 85764 Neuherberg, Germany
| | - Melanie Herbst
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, 85764 Neuherberg, Germany; (R.O.); (X.P.); (A.W.); (A.H.); (M.H.); (N.H.); (A.-G.Z.)
- Forschergruppe Diabetes e.V., 85764 Neuherberg, Germany
| | - Nadine Hummel
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, 85764 Neuherberg, Germany; (R.O.); (X.P.); (A.W.); (A.H.); (M.H.); (N.H.); (A.-G.Z.)
| | - Cornelia Prehn
- Research Unit Molecular Endocrinology and Metabolism, Genome Analysis Center, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (C.P.); (J.A.)
| | - Jerzy Adamski
- Research Unit Molecular Endocrinology and Metabolism, Genome Analysis Center, Helmholtz Zentrum München, 85764 Neuherberg, Germany; (C.P.); (J.A.)
- Chair for Experimental Genetics, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764 Neuherberg, Germany; (W.R.-M.); (G.K.)
| | - Werner Römisch-Margl
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764 Neuherberg, Germany; (W.R.-M.); (G.K.)
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Gabi Kastenmüller
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764 Neuherberg, Germany; (W.R.-M.); (G.K.)
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Anette-G. Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, 85764 Neuherberg, Germany; (R.O.); (X.P.); (A.W.); (A.H.); (M.H.); (N.H.); (A.-G.Z.)
- Forschergruppe Diabetes e.V., 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764 Neuherberg, Germany; (W.R.-M.); (G.K.)
| | - Sandra Hummel
- Institute of Diabetes Research, Helmholtz Zentrum München, and Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, 85764 Neuherberg, Germany; (R.O.); (X.P.); (A.W.); (A.H.); (M.H.); (N.H.); (A.-G.Z.)
- Forschergruppe Diabetes e.V., 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764 Neuherberg, Germany; (W.R.-M.); (G.K.)
| |
Collapse
|
27
|
Hromadnikova I, Kotlabova K, Krofta L, Sirc J. Association Analysis in Children Born from Normal and Complicated Pregnancies-Cardiovascular Disease Associated microRNAs and the Incidence of Prehypertension/Hypertension, Overweight/Obesity, Valve Problems and Heart Defects. Int J Mol Sci 2020; 21:ijms21218413. [PMID: 33182505 PMCID: PMC7672623 DOI: 10.3390/ijms21218413] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 12/14/2022] Open
Abstract
The goal was to assess how a history of any kind of pregnancy-related complication altered expression profile of microRNAs played a role in the pathogenesis of diabetes, cardiovascular and cerebrovascular diseases in the peripheral blood leukocytes of children at the age of 3–11 years. The prior exposure to gestational hypertension, preeclampsia, fetal growth restriction, gestational diabetes mellitus, preterm prelabor rupture of membranes or spontaneous preterm birth causes that a significant proportion of children (57.42% to 90.0% specifically) had a substantially altered microRNA expression profile, which might be the origin of a lifelong cardiovascular risk. A total of 23 out of 29 tested microRNAs were upregulated in children born from such complicated gestation. The occurrence of overweight, obesity, valve problems and heart defects even intensified upregulation of microRNAs already present in children exposed to such pregnancy complications. The occurrence of overweight/obesity (miR-92a-3p, and miR-210-3p) and valve problems or heart defects (miR-342-3p) induced microRNA upregulation in children affected with pregnancy complications. Overall, 42.86% overweight/obese children and 27.36% children with valve problems or heart defects had even higher microRNA levels than children with normal clinical findings after complicated pregnancies. In addition, the microRNA expression profile was also able to differentiate between children descending from normal gestation in relation to the occurrence of overweight and obesity. Screening on the base of the combination of 19 microRNAs identified 70.0% overweight/obese children at 90.0% specificity. In general, children after complicated pregnancies, just as children after normal pregnancies, with abnormal findings are at a higher risk of the onset of cardiovascular complications, and their dispensarization, with the aim to implement primary prevention strategies, would be beneficial.
Collapse
Affiliation(s)
- Ilona Hromadnikova
- Department of Molecular Biology and Cell Pathology, Third Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic;
- Correspondence: ; Tel.: +420-296511336
| | - Katerina Kotlabova
- Department of Molecular Biology and Cell Pathology, Third Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic;
| | - Ladislav Krofta
- Institute for the Care of the Mother and Child, Third Faculty of Medicine, Charles University, 147 00 Prague, Czech Republic; (L.K.); (J.S.)
| | - Jan Sirc
- Institute for the Care of the Mother and Child, Third Faculty of Medicine, Charles University, 147 00 Prague, Czech Republic; (L.K.); (J.S.)
| |
Collapse
|
28
|
Perng W, Francis EC, Smith HA, Carey J, Wang D, Kechris KM, Dabelea D. Sex-Specific Metabolite Biomarkers of NAFLD in Youth: A Prospective Study in the EPOCH Cohort. J Clin Endocrinol Metab 2020; 105:dgaa467. [PMID: 32687159 PMCID: PMC7418446 DOI: 10.1210/clinem/dgaa467] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/15/2020] [Indexed: 12/16/2022]
Abstract
CONTEXT Nonalcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver disease in developed nations. There are currently no accurate biomarkers of NAFLD risk in youth. OBJECTIVE Identify sex-specific metabolomics biomarkers of NAFLD in a healthy cohort of youth. DESIGN/SETTING This prospective study included 395 participants of the EPOCH cohort in Colorado, who were recruited 2006-2009 ("T1 visit") and followed for 5 years ("T2 visit"). We entered 767 metabolites measured at T1 into a reduced rank regression model to identify the strongest determinants of hepatic fat fraction (HFF) at T2, separately for boys and girls. We compared the capacity of metabolites versus conventional risk factors (overweight/obesity, insulin, alanine transaminase, aspartate transaminase) to predict NAFLD (HFF ≥5%) and high HFF (fourth vs first quartile) using area under the receiver operating characteristic curve (AUC). RESULTS Prevalence of NAFLD was 7.9% (8.5% of boys, 7.1% of girls). Mean ± SD HFF was 2.5 ± 3.1%. We identified 13 metabolites in girls and 10 metabolites in boys. Metabolites were in lipid, amino acid, and carbohydrate metabolism pathways. At T1, the metabolites outperformed conventional risk factors in prediction of high HFF but not NAFLD. At T2, the metabolites were superior to conventional risk factors as predictors of high HFF (AUC for metabolites vs conventional risk factors for boys: 0.9565 vs 0.8851, P = 0.02; for girls: 0.9450 vs 0.8469, P = 0.02) with similar trends for NAFLD, although the differences were not significant. CONCLUSIONS The metabolite profiles identified herein are superior predictors of high HFF when assessed 5 years prior and concurrently in a general-risk setting.
Collapse
Affiliation(s)
- Wei Perng
- Lifcourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora Colorado
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Colorado
| | - Ellen C Francis
- Lifcourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora Colorado
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Colorado
| | - Harry A Smith
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - John Carey
- Lifcourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora Colorado
| | - Dongqing Wang
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Katerina M Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Dana Dabelea
- Lifcourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora Colorado
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Colorado
- Department of Pediatrics, University of Colorado School of Medicine, Aurora Colorado
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW This review examines the impact of early life exposures on glucose metabolism in the offspring and explores potential metabolic mechanisms leading to type 2 diabetes in childhood. RECENT FINDINGS One in five adolescents is diagnosed with prediabetes. Recent studies have elucidated the impact of early exposures such as maternal diabetes, but also hyperglycemia below the threshold of gestational diabetes, obesity, hyperlipidemia, and paternal obesity on the future metabolic health of the offspring. Mechanisms affecting the developmental programing of offspring toward type 2 diabetes include epigenetic modifications, alterations in stem cell differentiation, metabolome and microbiome variation, immune dysregulation, and neonatal nutrition. The risk of type 2 diabetes in offspring is increased not only by diabetes exposure in utero but also by exposure to a heterogeneous milieu of factors that accompany maternal obesity that provoke a vicious cycle of metabolic disease. The key period for intervention to prevent type 2 diabetes is within the first 1000 days of life.
Collapse
Affiliation(s)
- Ankur Rughani
- Division of Pediatric Diabetes/Endocrinology, Harold Hamm Diabetes Center, Children's Hospital, The University of Oklahoma Health Sciences Center, 1200 Children's Ave Suite 4D, Oklahoma City, OK, 73104, USA
| | - Jacob E Friedman
- Division of Pediatric Diabetes/Endocrinology, Harold Hamm Diabetes Center, Children's Hospital, The University of Oklahoma Health Sciences Center, 1200 Children's Ave Suite 4D, Oklahoma City, OK, 73104, USA
| | - Jeanie B Tryggestad
- Division of Pediatric Diabetes/Endocrinology, Harold Hamm Diabetes Center, Children's Hospital, The University of Oklahoma Health Sciences Center, 1200 Children's Ave Suite 4D, Oklahoma City, OK, 73104, USA.
| |
Collapse
|