Epigenetics of the non-coding RNA nc886 across blood, adipose tissue and skeletal muscle in offspring exposed to diabetes in pregnancy

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Epigenetics of the non-coding RNA nc886 across blood, adipose tissue and skeletal muscle in offspring exposed to diabetes in pregnancy. / Hjort, Line; Bredgaard, Sandra Stokholm; Manitta, Eleonora; Marques, Irene; Sørensen, Anja Elaine; Martino, David; Grunnet, Louise Groth; Kelstrup, Louise; Houshmand-Oeregaard, Azadeh; Clausen, Tine Dalsgaard; Mathiesen, Elisabeth Reinhardt; Olsen, Sjurdur Frodi; Saffery, Richard; Barrès, Romain; Damm, Peter; Vaag, Allan Arthur; Dalgaard, Louise Torp.

In: Clinical Epigenetics, Vol. 16, No. 1, 61, 2024.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Hjort, L, Bredgaard, SS, Manitta, E, Marques, I, Sørensen, AE, Martino, D, Grunnet, LG, Kelstrup, L, Houshmand-Oeregaard, A, Clausen, TD, Mathiesen, ER, Olsen, SF, Saffery, R, Barrès, R, Damm, P, Vaag, AA & Dalgaard, LT 2024, 'Epigenetics of the non-coding RNA nc886 across blood, adipose tissue and skeletal muscle in offspring exposed to diabetes in pregnancy', Clinical Epigenetics, vol. 16, no. 1, 61. https://doi.org/10.1186/s13148-024-01673-3

APA

Hjort, L., Bredgaard, S. S., Manitta, E., Marques, I., Sørensen, A. E., Martino, D., Grunnet, L. G., Kelstrup, L., Houshmand-Oeregaard, A., Clausen, T. D., Mathiesen, E. R., Olsen, S. F., Saffery, R., Barrès, R., Damm, P., Vaag, A. A., & Dalgaard, L. T. (2024). Epigenetics of the non-coding RNA nc886 across blood, adipose tissue and skeletal muscle in offspring exposed to diabetes in pregnancy. Clinical Epigenetics, 16(1), [61]. https://doi.org/10.1186/s13148-024-01673-3

Vancouver

Hjort L, Bredgaard SS, Manitta E, Marques I, Sørensen AE, Martino D et al. Epigenetics of the non-coding RNA nc886 across blood, adipose tissue and skeletal muscle in offspring exposed to diabetes in pregnancy. Clinical Epigenetics. 2024;16(1). 61. https://doi.org/10.1186/s13148-024-01673-3

Author

Hjort, Line ; Bredgaard, Sandra Stokholm ; Manitta, Eleonora ; Marques, Irene ; Sørensen, Anja Elaine ; Martino, David ; Grunnet, Louise Groth ; Kelstrup, Louise ; Houshmand-Oeregaard, Azadeh ; Clausen, Tine Dalsgaard ; Mathiesen, Elisabeth Reinhardt ; Olsen, Sjurdur Frodi ; Saffery, Richard ; Barrès, Romain ; Damm, Peter ; Vaag, Allan Arthur ; Dalgaard, Louise Torp. / Epigenetics of the non-coding RNA nc886 across blood, adipose tissue and skeletal muscle in offspring exposed to diabetes in pregnancy. In: Clinical Epigenetics. 2024 ; Vol. 16, No. 1.

Bibtex

@article{98c14b4aa4c04f3f91a8c08d6529effa,
title = "Epigenetics of the non-coding RNA nc886 across blood, adipose tissue and skeletal muscle in offspring exposed to diabetes in pregnancy",
abstract = "Background: Diabetes in pregnancy is associated with increased risk of long-term metabolic disease in the offspring, potentially mediated by in utero epigenetic variation. Previously, we identified multiple differentially methylated single CpG sites in offspring of women with gestational diabetes mellitus (GDM), but whether stretches of differentially methylated regions (DMRs) can also be identified in adolescent GDM offspring is unknown. Here, we investigate which DNA regions in adolescent offspring are differentially methylated in blood by exposure to diabetes in pregnancy. The secondary aim was to characterize the RNA expression of the identified DMR, which contained the nc886 non-coding RNA. Methods: To identify DMRs, we employed the bump hunter method in samples from young (9–16 yr, n = 92) offspring of women with GDM (O-GDM) and control offspring (n = 94). Validation by pyrosequencing was performed in an adult offspring cohort (age 28–33 years) consisting of O-GDM (n = 82), offspring exposed to maternal type 1 diabetes (O-T1D, n = 67) and control offspring (O-BP, n = 57). RNA-expression was measured using RT-qPCR in subcutaneous adipose tissue and skeletal muscle. Results: One significant DMR represented by 10 CpGs with a bimodal methylation pattern was identified, located in the nc886/VTRNA2-1 non-coding RNA gene. Low methylation status across all CpGs of the nc886 in the young offspring was associated with maternal GDM. While low methylation degree in adult offspring in blood, adipose tissue, and skeletal muscle was not associated with maternal GDM, adipose tissue nc886 expression was increased in O-GDM compared to O-BP, but not in O-T1D. In addition, adipose tissue nc886 expression levels were positively associated with maternal pre-pregnancy BMI (p = 0.006), but not with the offspring{\textquoteright}s own adiposity. Conclusions: Our results highlight that nc886 is a metastable epiallele, whose methylation in young offspring is negatively correlated with maternal obesity and GDM status. The physiological effect of nc886 may be more important in adipose tissue than in skeletal muscle. Further research should aim to investigate how nc886 regulation in adipose tissue by exposure to GDM may contribute to development of metabolic disease.",
keywords = "Adipose, Developmental programming, DNA methylation, Epigenetics, Gene expression, Gestational diabetes, Intrauterine hyperglycemia, Muscle, nc886, ncRNA, Type 1 diabetes, VTRNA2-1",
author = "Line Hjort and Bredgaard, {Sandra Stokholm} and Eleonora Manitta and Irene Marques and S{\o}rensen, {Anja Elaine} and David Martino and Grunnet, {Louise Groth} and Louise Kelstrup and Azadeh Houshmand-Oeregaard and Clausen, {Tine Dalsgaard} and Mathiesen, {Elisabeth Reinhardt} and Olsen, {Sjurdur Frodi} and Richard Saffery and Romain Barr{\`e}s and Peter Damm and Vaag, {Allan Arthur} and Dalgaard, {Louise Torp}",
note = "Publisher Copyright: {\textcopyright} The Author(s) 2024.",
year = "2024",
doi = "10.1186/s13148-024-01673-3",
language = "English",
volume = "16",
journal = "Clinical Epigenetics (Print)",
issn = "1868-7075",
publisher = "BioMed Central Ltd.",
number = "1",

}

RIS

TY - JOUR

T1 - Epigenetics of the non-coding RNA nc886 across blood, adipose tissue and skeletal muscle in offspring exposed to diabetes in pregnancy

AU - Hjort, Line

AU - Bredgaard, Sandra Stokholm

AU - Manitta, Eleonora

AU - Marques, Irene

AU - Sørensen, Anja Elaine

AU - Martino, David

AU - Grunnet, Louise Groth

AU - Kelstrup, Louise

AU - Houshmand-Oeregaard, Azadeh

AU - Clausen, Tine Dalsgaard

AU - Mathiesen, Elisabeth Reinhardt

AU - Olsen, Sjurdur Frodi

AU - Saffery, Richard

AU - Barrès, Romain

AU - Damm, Peter

AU - Vaag, Allan Arthur

AU - Dalgaard, Louise Torp

N1 - Publisher Copyright: © The Author(s) 2024.

PY - 2024

Y1 - 2024

N2 - Background: Diabetes in pregnancy is associated with increased risk of long-term metabolic disease in the offspring, potentially mediated by in utero epigenetic variation. Previously, we identified multiple differentially methylated single CpG sites in offspring of women with gestational diabetes mellitus (GDM), but whether stretches of differentially methylated regions (DMRs) can also be identified in adolescent GDM offspring is unknown. Here, we investigate which DNA regions in adolescent offspring are differentially methylated in blood by exposure to diabetes in pregnancy. The secondary aim was to characterize the RNA expression of the identified DMR, which contained the nc886 non-coding RNA. Methods: To identify DMRs, we employed the bump hunter method in samples from young (9–16 yr, n = 92) offspring of women with GDM (O-GDM) and control offspring (n = 94). Validation by pyrosequencing was performed in an adult offspring cohort (age 28–33 years) consisting of O-GDM (n = 82), offspring exposed to maternal type 1 diabetes (O-T1D, n = 67) and control offspring (O-BP, n = 57). RNA-expression was measured using RT-qPCR in subcutaneous adipose tissue and skeletal muscle. Results: One significant DMR represented by 10 CpGs with a bimodal methylation pattern was identified, located in the nc886/VTRNA2-1 non-coding RNA gene. Low methylation status across all CpGs of the nc886 in the young offspring was associated with maternal GDM. While low methylation degree in adult offspring in blood, adipose tissue, and skeletal muscle was not associated with maternal GDM, adipose tissue nc886 expression was increased in O-GDM compared to O-BP, but not in O-T1D. In addition, adipose tissue nc886 expression levels were positively associated with maternal pre-pregnancy BMI (p = 0.006), but not with the offspring’s own adiposity. Conclusions: Our results highlight that nc886 is a metastable epiallele, whose methylation in young offspring is negatively correlated with maternal obesity and GDM status. The physiological effect of nc886 may be more important in adipose tissue than in skeletal muscle. Further research should aim to investigate how nc886 regulation in adipose tissue by exposure to GDM may contribute to development of metabolic disease.

AB - Background: Diabetes in pregnancy is associated with increased risk of long-term metabolic disease in the offspring, potentially mediated by in utero epigenetic variation. Previously, we identified multiple differentially methylated single CpG sites in offspring of women with gestational diabetes mellitus (GDM), but whether stretches of differentially methylated regions (DMRs) can also be identified in adolescent GDM offspring is unknown. Here, we investigate which DNA regions in adolescent offspring are differentially methylated in blood by exposure to diabetes in pregnancy. The secondary aim was to characterize the RNA expression of the identified DMR, which contained the nc886 non-coding RNA. Methods: To identify DMRs, we employed the bump hunter method in samples from young (9–16 yr, n = 92) offspring of women with GDM (O-GDM) and control offspring (n = 94). Validation by pyrosequencing was performed in an adult offspring cohort (age 28–33 years) consisting of O-GDM (n = 82), offspring exposed to maternal type 1 diabetes (O-T1D, n = 67) and control offspring (O-BP, n = 57). RNA-expression was measured using RT-qPCR in subcutaneous adipose tissue and skeletal muscle. Results: One significant DMR represented by 10 CpGs with a bimodal methylation pattern was identified, located in the nc886/VTRNA2-1 non-coding RNA gene. Low methylation status across all CpGs of the nc886 in the young offspring was associated with maternal GDM. While low methylation degree in adult offspring in blood, adipose tissue, and skeletal muscle was not associated with maternal GDM, adipose tissue nc886 expression was increased in O-GDM compared to O-BP, but not in O-T1D. In addition, adipose tissue nc886 expression levels were positively associated with maternal pre-pregnancy BMI (p = 0.006), but not with the offspring’s own adiposity. Conclusions: Our results highlight that nc886 is a metastable epiallele, whose methylation in young offspring is negatively correlated with maternal obesity and GDM status. The physiological effect of nc886 may be more important in adipose tissue than in skeletal muscle. Further research should aim to investigate how nc886 regulation in adipose tissue by exposure to GDM may contribute to development of metabolic disease.

KW - Adipose

KW - Developmental programming

KW - DNA methylation

KW - Epigenetics

KW - Gene expression

KW - Gestational diabetes

KW - Intrauterine hyperglycemia

KW - Muscle

KW - nc886

KW - ncRNA

KW - Type 1 diabetes

KW - VTRNA2-1

U2 - 10.1186/s13148-024-01673-3

DO - 10.1186/s13148-024-01673-3

M3 - Journal article

C2 - 38715048

AN - SCOPUS:85192240327

VL - 16

JO - Clinical Epigenetics (Print)

JF - Clinical Epigenetics (Print)

SN - 1868-7075

IS - 1

M1 - 61

ER -

ID: 391622334