151
|
Gibson JD, Arechavaleta-Velasco ME, Tsuruda JM, Hunt GJ. Biased Allele Expression and Aggression in Hybrid Honeybees may be Influenced by Inappropriate Nuclear-Cytoplasmic Signaling. Front Genet 2015; 6:343. [PMID: 26648977 PMCID: PMC4664729 DOI: 10.3389/fgene.2015.00343] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 11/20/2015] [Indexed: 11/15/2022] Open
Abstract
Hybrid effects are often exhibited asymmetrically between reciprocal families. One way this could happen is if silencing of one parent’s allele occurs in one lineage but not the other, which could affect the phenotypes of the hybrids asymmetrically by silencing that allele in only one of the hybrid families. We have previously tested for allele-specific expression biases in hybrids of European and Africanized honeybees and we found that there was an asymmetric overabundance of genes showing a maternal bias in the family with a European mother. Here, we further analyze allelic bias in these hybrids to ascertain whether they may underlie previously described asymmetries in metabolism and aggression in similar hybrid families and we speculate on what mechanisms may produce this biased allele usage. We find that there are over 500 genes that have some form of biased allele usage and over 200 of these are biased toward the maternal allele but only in the family with European maternity, mirroring the pattern observed for aggression and metabolic rate. This asymmetrically biased set is enriched for genes in loci associated with aggressive behavior and also for mitochondrial-localizing proteins. It contains many genes that play important roles in metabolic regulation. Moreover we find genes relating to the piwi-interacting RNA (piRNA) pathway, which is involved in chromatin modifications and epigenetic regulation and may help explain the mechanism underlying this asymmetric allele use. Based on these findings and previous work investigating aggression and metabolism in bees, we propose a novel hypothesis; that the asymmetric pattern of biased allele usage in these hybrids is a result of inappropriate use of piRNA-mediated nuclear-cytoplasmic signaling that is normally used to modulate aggression in honeybees. This is the first report of widespread asymmetric effects on allelic expression in hybrids and may represent a novel mechanism for gene regulation.
Collapse
Affiliation(s)
- Joshua D Gibson
- Department of Entomology, Purdue University, West Lafayette IN, USA
| | - Miguel E Arechavaleta-Velasco
- CENID-Fisiología y Mejoramiento Animal, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias México, Mexico
| | | | - Greg J Hunt
- Department of Entomology, Purdue University, West Lafayette IN, USA
| |
Collapse
|
152
|
Tunster SJ, Creeth HDJ, John RM. The imprinted Phlda2 gene modulates a major endocrine compartment of the placenta to regulate placental demands for maternal resources. Dev Biol 2015; 409:251-260. [PMID: 26476147 PMCID: PMC4684229 DOI: 10.1016/j.ydbio.2015.10.015] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/02/2015] [Accepted: 10/11/2015] [Indexed: 02/06/2023]
Abstract
Imprinted genes, which are expressed from a single parental allele in response to epigenetic marks first established in the germline, function in a myriad of processes to regulate mammalian development. Recent work suggests that imprinted genes may regulate the signalling function of the placenta by modulating the size of the endocrine compartment. Here we provide in vivo evidence that this hypothesis is well founded. Elevated expression of the imprinted Pleckstrin homology-like domain, family a, member 2 (Phlda2) gene drives a reduction of the spongiotrophoblast endocrine compartment, diminished placental glycogen and asymmetric foetal growth restriction. Using both loss-of-function and gain-in-expression mouse models, here we further show that Phlda2 exclusively modulates the spongiotrophoblast compartment of the placenta without significantly altering the composition of the trophoblast giant cell endocrine lineages that share a common progenitor with this lineage. Additionally, we show that Phlda2 loss-of-function placentae contain nearly three times more placental glycogen than non-transgenic placentae. Remarkably, relative to a fully wild type scenario, wild type placentae also accumulate excessive glycogen. While loss-of-function of Phlda2 increased both placental weight and placental glycogen, the weight of both mutant and non-transgenic fetuses was lower than that found in a fully wild type scenario indicating that excessive glycogen accumulation comes at the cost of foetal growth. This work firstly highlights a novel signalling function for the spongiotrophoblast in stimulating the global accumulation of placental glycogen. Furthermore, this work suggests that Phlda2 manipulates the placenta's demands for maternal resources, a process that must be tightly regulated by epigenetic marks to ensure optimal foetal growth. Phlda2 specifically restrains development of the spongiotrophoblast. The spongiotrophoblast is a major endocrine compartment of the placenta. The spongiotrophoblast locally and globally boosts glycogen accumulation. Excessive glycogen accumulation is associated with foetal growth restriction. Phlda2 regulates placental demands from maternal resources.
Collapse
Affiliation(s)
- S J Tunster
- Cardiff School of Biosciences, Cardiff University, Cardiff, Wales CF10 3AX, UK
| | - H D J Creeth
- Cardiff School of Biosciences, Cardiff University, Cardiff, Wales CF10 3AX, UK
| | - R M John
- Cardiff School of Biosciences, Cardiff University, Cardiff, Wales CF10 3AX, UK.
| |
Collapse
|
153
|
Grandparental morbidity and mortality patterns are associated with infant birth weight in the Lifeways cross-generation cohort study 2001-2010. J Dev Orig Health Dis 2015; 3:458-68. [PMID: 25084299 DOI: 10.1017/s2040174412000451] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The association of infants' birth weight with maternal cardiovascular morbidity (CVD) and mortality substantiates the foetal origins hypothesis. Few studies to date have investigated grandparent-infant risk association. We prospectively examined this relationship in the Lifeways three-generation familial cohort, contrasting lineage and gender differences to understand mechanisms of intergenerational risk transmission. In 2001, a cohort of 1082 families was established at antenatal stage. A total of 539 families (n = 539 infants) had both a participating grandparent (n = 1054) and information on infants' gestational age. At baseline, grandparents provided their diagnosed CVD status and 79% also underwent a cardiovascular risk factors assessment. In 2005, general practitioners provided an update for 61% grandparents. In 2010, a search of civil register confirmed 77 grandparental deaths in 539 families. Grandchildren's birth weight and grandparental cardiovascular risk factors associations were examined with linear regressions. Grandparental CVD associations were analysed using ANCOVA. Cox proportional hazard ratios (HR) were calculated for all-cause mortality associations. Models were adjusted for infants', mothers' and grandparents' demographic, anthropometric and socio-behavioural characteristics, as appropriate. The paternal grandfathers' (PGF) systolic blood pressure (mmHg) [β (95% CI) = 6.6 (0.8 - 12.5); P = 0.03] and paternal grandmothers' serum triglycerides (mmol/l) [β (95% CI) = 78.8 (7.0 - 150.7); P = 0.03] were linearly predictive of infants' birth weight, which was not observed for maternal grandparents. Mean birth weight for infants of maternal grandmothers with diabetes {-272.7 [(-499.7) - (-45.6)] g; P = 0.02} or stroke {-292.1 [(-544.5) - (-39.6)] g; P = 0.02} was lower than those without diabetes or stroke, a pattern not observed for paternal grandparents. Whereas PGFs' mortality was significantly associated with infants' high birth weight (≥4000 g) [HR (95% CI) = 4.9 (1.2 - 19.9); P = 0.03], maternal grandparents' mortality showed a converse pattern with infants' low birth weight (<2500 g) [HR (95% CI) = 1.7 (0.4 - 8.2); P = 0.7], although not statistically significant. These findings suggest that intergenerational transmission of risk differs in maternal and paternal lines.
Collapse
|
154
|
|
155
|
Nuñez CL, Grote MN, Wechsler M, Allen-Blevins CR, Hinde K. Offspring of primiparous mothers do not experience greater mortality or poorer growth: Revisiting the conventional wisdom with archival records of Rhesus Macaques. Am J Primatol 2015; 77:963-973. [PMID: 26031808 PMCID: PMC4666832 DOI: 10.1002/ajp.22426] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 04/02/2015] [Accepted: 04/25/2015] [Indexed: 11/07/2022]
Abstract
Female mammals often begin to reproduce before achieving somatic maturity and therefore face tradeoffs between allocating energy to reproduction or their own continued development. Constraints on primiparous females are associated with greater reproductive failure, and first-born infants often have slower growth and greater mortality and morbidity than infants born to multiparous females. Effects of early life investment may persist even after weaning when juveniles are no longer dependent on maternal care and mother's milk. We investigated the long-term consequences of birth order in a large sample of rhesus macaques, Macaca mulatta, assigned to the outdoor breeding colony at the California National Primate Research Center (n = 2,724). A joint model for growth and mortality over the first three years of life allowed us to explicitly connect growth rates to survival. As expected, males are born heavier and grow faster relative to females. However, contrary to expectations, later-born males face substantially lower survival probability during their first three years, whereas first-born males survive at greater rates similar to both first-born and later-born females. Primiparous mothers are less likely to conceive during the subsequent breeding season, suggesting that their reproductive costs are greater than those of multiparous mothers. We speculate that compensatory tactics, both behavioral and physiological, of first-born offspring and their mothers, as well as the novel ecology of the captive environment, underlie these findings. The results presented here provide new insights into how maternal and infant life history tradeoffs may influence developmental trajectories even after the period of maternal dependence. Am. J. Primatol. 77:963-973, 2015. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Chase L. Nuñez
- University Program in Ecology, Duke University
- Nicholas School of the Environment, Duke University
| | - Mark N. Grote
- Department of Anthropology, University of California-Davis
| | | | | | - Katie Hinde
- Brain, Mind, & Behavior Unit, California National Primate Research Center
- Department of Human Evolutionary Biology, Harvard University
| |
Collapse
|
156
|
Furness AI, Morrison KR, Orr TJ, Arendt JD, Reznick DN. Reproductive mode and the shifting arenas of evolutionary conflict. Ann N Y Acad Sci 2015; 1360:75-100. [PMID: 26284738 DOI: 10.1111/nyas.12835] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/21/2015] [Accepted: 06/05/2015] [Indexed: 12/11/2022]
Abstract
In sexually reproducing organisms, the genetic interests of individuals are not perfectly aligned. Conflicts among family members are prevalent since interactions involve the transfer of limited resources between interdependent players. Intrafamilial conflict has traditionally been considered along three major axes: between the sexes, between parents and offspring, and between siblings. In these interactions, conflict is expected over traits in which the resulting phenotypic value is determined by multiple family members who have only partially overlapping fitness optima. We focus on four major categories of animal reproductive mode (broadcast spawning, egg laying, live bearing, and live bearing with matrotrophy) and identify the shared phenotypes or traits over which conflict is expected, and then review the empirical literature for evidence of their occurrence. Major transitions among reproductive mode, such as a shift from external to internal fertilization, an increase in egg-retention time, modifications of embryos and mothers for nutrient transfer, the evolution of postnatal parental care, and increased interaction with the kin network, mark key shifts that both change and expand the arenas in which conflict is played out.
Collapse
Affiliation(s)
- Andrew I Furness
- Department of Biology, University of California, Riverside, California.,Department of Ecology and Evolutionary Biology, University of California, Irvine, California
| | - Keenan R Morrison
- Department of Biology, University of California, Riverside, California
| | - Teri J Orr
- Department of Biology, University of California, Riverside, California.,Department of Biology, University of Massachusetts, Amherst, Massachusetts
| | - Jeff D Arendt
- Department of Biology, University of California, Riverside, California
| | - David N Reznick
- Department of Biology, University of California, Riverside, California
| |
Collapse
|
157
|
Akamatsu S, Wyatt AW, Lin D, Lysakowski S, Zhang F, Kim S, Tse C, Wang K, Mo F, Haegert A, Brahmbhatt S, Bell R, Adomat H, Kawai Y, Xue H, Dong X, Fazli L, Tsai H, Lotan TL, Kossai M, Mosquera JM, Rubin MA, Beltran H, Zoubeidi A, Wang Y, Gleave ME, Collins CC. The Placental Gene PEG10 Promotes Progression of Neuroendocrine Prostate Cancer. Cell Rep 2015; 12:922-36. [PMID: 26235627 DOI: 10.1016/j.celrep.2015.07.012] [Citation(s) in RCA: 210] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 06/15/2015] [Accepted: 07/07/2015] [Indexed: 01/09/2023] Open
Abstract
More potent targeting of the androgen receptor (AR) in advanced prostate cancer is driving an increased incidence of neuroendocrine prostate cancer (NEPC), an aggressive and treatment-resistant AR-negative variant. Its molecular pathogenesis remains poorly understood but appears to require TP53 and RB1 aberration. We modeled the development of NEPC from conventional prostatic adenocarcinoma using a patient-derived xenograft and found that the placental gene PEG10 is de-repressed during the adaptive response to AR interference and subsequently highly upregulated in clinical NEPC. We found that the AR and the E2F/RB pathway dynamically regulate distinct post-transcriptional and post-translational isoforms of PEG10 at distinct stages of NEPC development. In vitro, PEG10 promoted cell-cycle progression from G0/G1 in the context of TP53 loss and regulated Snail expression via TGF-β signaling to promote invasion. Taken together, these findings show the mechanistic relevance of RB1 and TP53 loss in NEPC and suggest PEG10 as a NEPC-specific target.
Collapse
Affiliation(s)
- Shusuke Akamatsu
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Alexander W Wyatt
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Dong Lin
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Summer Lysakowski
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Fan Zhang
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Soojin Kim
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Charan Tse
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Kendric Wang
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Fan Mo
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Anne Haegert
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Sonal Brahmbhatt
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Robert Bell
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Hans Adomat
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Yoshihisa Kawai
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Hui Xue
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Xin Dong
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Harrison Tsai
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Myriam Kossai
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, USA; Institute for Precision Medicine, New York Presbyterian Hospital-Weill Cornell Medical College, New York, NY 10065, USA
| | - Juan Miguel Mosquera
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, USA; Institute for Precision Medicine, New York Presbyterian Hospital-Weill Cornell Medical College, New York, NY 10065, USA
| | - Mark A Rubin
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, USA; Institute for Precision Medicine, New York Presbyterian Hospital-Weill Cornell Medical College, New York, NY 10065, USA
| | - Himisha Beltran
- Institute for Precision Medicine, New York Presbyterian Hospital-Weill Cornell Medical College, New York, NY 10065, USA; Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Amina Zoubeidi
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Martin E Gleave
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada.
| | - Colin C Collins
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada; Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada.
| |
Collapse
|
158
|
Hoffmann A, Daniel G, Schmidt-Edelkraut U, Spengler D. Roles of imprinted genes in neural stem cells. Epigenomics 2015; 6:515-32. [PMID: 25431944 DOI: 10.2217/epi.14.42] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Imprinted genes and neural stem cells (NSC) play an important role in the developing and mature brain. A central theme of imprinted gene function in NSCs is cell survival and G1 arrest to control cell division, cell-cycle exit, migration and differentiation. Moreover, genomic imprinting can be epigenetically switched off at some genes to ensure stem cell quiescence and differentiation. At the genome scale, imprinted genes are organized in dynamic networks formed by interchromosomal interactions and transcriptional coregulation of imprinted and nonimprinted genes. Such multilayered networks may synchronize NSC activity with the demand from the niche resembling their roles in adjusting fetal size.
Collapse
Affiliation(s)
- Anke Hoffmann
- Max Planck Institute of Psychiatry, Translational Research, Kraepelinstrasse 2-10, 80804 Munich, Germany
| | | | | | | |
Collapse
|
159
|
Kappil MA, Green BB, Armstrong DA, Sharp AJ, Lambertini L, Marsit CJ, Chen J. Placental expression profile of imprinted genes impacts birth weight. Epigenetics 2015; 10:842-9. [PMID: 26186239 DOI: 10.1080/15592294.2015.1073881] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The importance of imprinted genes in regulating feto-placental development has been long established. However, a comprehensive assessment of the role of placental imprinted gene expression on fetal growth has yet to be conducted. In this study, we examined the association between the placental expression of 108 established and putative imprinted genes and birth weight in 677 term pregnancies, oversampled for small for gestational age (SGA) and large for gestational age (LGA) infants. Using adjusted multinomial regression analyses, a 2-fold increase in the expression of 9 imprinted genes was positively associated with LGA status: BLCAP [odds ratio (OR) = 3.78, 95% confidence interval (CI): 1.83, 7.82], DLK1 [OR = 1.63, 95% CI: 1.27, 2.09], H19 [OR = 2.79, 95% CI: 1.77, 4.42], IGF2 [OR = 1.43, 95% CI:1.31, 2.40], MEG3 [OR = 1.42, 95% CI: 1.19, 1.71], MEST [OR = 4.78, 95% CI: 2.64, 8.65], NNAT [OR = 1.40, 95% CI: 1.05, 1.86], NDN [OR = 2.52, 95% CI: 1.72, 3.68], and PLAGL1 [OR = 1.85, 95% CI: 1.40, 2.44]. For SGA status, a 2-fold increase in MEST expression was associated with decreased risk [OR = 0.31, 95% CI: 0.17, 0.58], while a 2-fold increase in NNAT expression was associated with increased risk [OR = 1.52, 95% CI: 1.1, 2.1]. Following a factor analysis, all genes significantly associated with SGA or LGA status loaded onto 2 of the 8 gene-sets underlying the variability in the dataset. Our comprehensive placental profiling of imprinted genes in a large birth cohort supports the importance of these genes for fetal growth. Given that abnormal birth weight is implicated in numerous diseases and developmental abnormalities, the expression pattern of placental imprinted genes has the potential to be developed as a novel biomarker for postnatal health outcomes.
Collapse
Affiliation(s)
- Maya A Kappil
- a Department of Preventive Medicine ; Icahn School of Medicine at Mount Sinai ; New York , NY USA
| | | | | | | | | | | | | |
Collapse
|
160
|
Chiaratti MR, Malik S, Diot A, Rapa E, Macleod L, Morten K, Vatish M, Boyd R, Poulton J. Is Placental Mitochondrial Function a Regulator that Matches Fetal and Placental Growth to Maternal Nutrient Intake in the Mouse? PLoS One 2015; 10:e0130631. [PMID: 26132581 PMCID: PMC4488591 DOI: 10.1371/journal.pone.0130631] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 05/21/2015] [Indexed: 12/18/2022] Open
Abstract
Background Effective fetal growth requires adequate maternal nutrition coupled to active transport of nutrients across the placenta, which, in turn requires ATP. Epidemiological and experimental evidence has shown that impaired maternal nutrition in utero results in an adverse postnatal phenotype for the offspring. Placental mitochondrial function might link maternal food intake to fetal growth since impaired placental ATP production, in response to poor maternal nutrition, could be a pathway linking maternal food intake to reduced fetal growth. Method We assessed the effects of maternal diet on placental water content, ATP levels and mitochondrial DNA (mtDNA) content in mice at embryonic (E) day 18 (E18). Females maintained on either low- (LPD) or normal- (NPD) protein diets were mated with NPD males. Results To investigate the possibility of an underlying mitochondrial stress response, we studied cultured human trophoblast cells (BeWos). High throughput imaging showed that amino acid starvation induces changes in mitochondrial morphology that suggest stress-induced mitochondrial hyperfusion. This is a defensive response, believed to increase mitochondrial efficiency, that could underlie the increase in ATP observed in placenta. Conclusions These findings reinforce the pathophysiological links between maternal diet and conceptus mitochondria, potentially contributing to metabolic programming. The quiet embryo hypothesis proposes that pre-implantation embryo survival is best served by a relatively low level of metabolism. This may extend to post-implantation trophoblast responses to nutrition.
Collapse
Affiliation(s)
- Marcos R. Chiaratti
- Nuffield Department of Obstetrics and Gynaecology, The Women’s Centre, University of Oxford, Oxford, OX3 9DU, United Kingdom
- Departamento de Genética e Evolução, Universidade Federal de São Carlos Rod. Washington Luís, km 235, Jardim Guanabara, São Carlos, SP, CEP 13.565–905, Brazil
| | - Sajida Malik
- Nuffield Department of Obstetrics and Gynaecology, The Women’s Centre, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Alan Diot
- Nuffield Department of Obstetrics and Gynaecology, The Women’s Centre, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Elizabeth Rapa
- Nuffield Department of Obstetrics and Gynaecology, The Women’s Centre, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Lorna Macleod
- Nuffield Department of Obstetrics and Gynaecology, The Women’s Centre, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Karl Morten
- Nuffield Department of Obstetrics and Gynaecology, The Women’s Centre, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Manu Vatish
- Nuffield Department of Obstetrics and Gynaecology, The Women’s Centre, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Richard Boyd
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, OX1 3QX, United Kingdom
| | - Joanna Poulton
- Nuffield Department of Obstetrics and Gynaecology, The Women’s Centre, University of Oxford, Oxford, OX3 9DU, United Kingdom
- * E-mail:
| |
Collapse
|
161
|
Jeong Y, Son JW, Kim BN, Yoo HJ. Evolutionary Perspective on Autism. Soa Chongsonyon Chongsin Uihak 2015. [DOI: 10.5765/jkacap.2015.26.2.67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
162
|
Metsalu T, Viltrop T, Tiirats A, Rajashekar B, Reimann E, Kõks S, Rull K, Milani L, Acharya G, Basnet P, Vilo J, Mägi R, Metspalu A, Peters M, Haller-Kikkatalo K, Salumets A. Using RNA sequencing for identifying gene imprinting and random monoallelic expression in human placenta. Epigenetics 2015; 9:1397-409. [PMID: 25437054 DOI: 10.4161/15592294.2014.970052] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Given the possible critical importance of placental gene imprinting and random monoallelic expression on fetal and infant health, most of those genes must be identified, in order to understand the risks that the baby might meet during pregnancy and after birth. Therefore, the aim of the current study was to introduce a workflow and tools for analyzing imprinted and random monoallelic gene expression in human placenta, by applying whole-transcriptome (WT) RNA sequencing of placental tissue and genotyping of coding DNA variants in family trios. Ten family trios, each with a healthy spontaneous single-term pregnancy, were recruited. Total RNA was extracted for WT analysis, providing the full sequence information for the placental transcriptome. Parental and child blood DNA genotypes were analyzed by exome SNP genotyping microarrays, mapping the inheritance and estimating the abundance of parental expressed alleles. Imprinted genes showed consistent expression from either parental allele, as demonstrated by the SNP content of sequenced transcripts, while monoallelically expressed genes had random activity of parental alleles. We revealed 4 novel possible imprinted genes (LGALS8, LGALS14, PAPPA2 and SPTLC3) and confirmed the imprinting of 4 genes (AIM1, PEG10, RHOBTB3 and ZFAT-AS1) in human placenta. The major finding was the identification of 4 genes (ABP1, BCLAF1, IFI30 and ZFAT) with random allelic bias, expressing one of the parental alleles preferentially. The main functions of the imprinted and monoallelically expressed genes included: i) mediating cellular apoptosis and tissue development; ii) regulating inflammation and immune system; iii) facilitating metabolic processes; and iv) regulating cell cycle.
Collapse
Key Words
- ASE, allele-specific expression
- FDR, false discovery rate
- GEO, Gene Expression Omnibus
- IUGR, intrauterine growth restriction
- MAF, minor allele frequency
- MHC, major histocompatibility complex
- NK cells, natural killer cells
- RNA sequencing
- RNA-Seq, RNA-sequencing
- RPKM, reads per kilobase per million
- UCSC, University of California Santa Cruz
- WT, whole-transcriptome
- allele-specific expression
- imprinting
- placenta
- random monoallelic expression
- short read mapping
Collapse
Affiliation(s)
- Tauno Metsalu
- a Institute of Computer Science ; University of Tartu ; Tartu , Estonia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Byars SG, Stearns SC, Boomsma JJ. Opposite risk patterns for autism and schizophrenia are associated with normal variation in birth size: phenotypic support for hypothesized diametric gene-dosage effects. Proc Biol Sci 2015; 281:20140604. [PMID: 25232142 DOI: 10.1098/rspb.2014.0604] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Opposite phenotypic and behavioural traits associated with copy number variation and disruptions to imprinted genes with parent-of-origin effects have led to the hypothesis that autism and schizophrenia share molecular risk factors and pathogenic mechanisms, but a direct phenotypic comparison of how their risks covary has not been attempted. Here, we use health registry data collected on Denmark's roughly 5 million residents between 1978 and 2009 to detect opposing risks of autism and schizophrenia depending on normal variation (mean ± 1 s.d.) in adjusted birth size, which we use as a proxy for diametric gene-dosage variation in utero. Above-average-sized babies (weight, 3691-4090 g; length, 52.8-54.3 cm) had significantly higher risk for autism spectrum (AS) and significantly lower risk for schizophrenia spectrum (SS) disorders. By contrast, below-average-sized babies (2891-3290 g; 49.7-51.2 cm) had significantly lower risk for AS and significantly higher risk for SS disorders. This is the first study directly comparing autism and schizophrenia risks in the same population, and provides the first large-scale empirical support for the hypothesis that diametric gene-dosage effects contribute to these disorders. Only the kinship theory of genomic imprinting predicts the opposing risk patterns that we discovered, suggesting that molecular research on mental disease risk would benefit from considering evolutionary theory.
Collapse
Affiliation(s)
- Sean G Byars
- Centre for Social Evolution, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Stephen C Stearns
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| | - Jacobus J Boomsma
- Centre for Social Evolution, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
164
|
Hoffmann A, Zimmermann CA, Spengler D. Molecular epigenetic switches in neurodevelopment in health and disease. Front Behav Neurosci 2015; 9:120. [PMID: 26029068 PMCID: PMC4429584 DOI: 10.3389/fnbeh.2015.00120] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/26/2015] [Indexed: 12/16/2022] Open
Abstract
Epigenetic mechanisms encode information above and beyond DNA sequence and play a critical role in brain development and the long-lived effects of environmental cues on the pre- and postnatal brain. Switch-like, rather than graded changes, illustrate par excellence how epigenetic events perpetuate altered activity states in the absence of the initial cue. They occur from early neural development to maturation and can give rise to distinct diseases upon deregulation. Many neurodevelopmental genes harbor bivalently marked chromatin domains, states of balanced inhibition, which guide dynamic “ON or OFF” decisions once the balance is tilted in response to developmental or environmental cues. Examples discussed in this review include neuronal differentiation of embryonic stem cells (ESC) into progenitors and beyond, activation of Kiss1 at puberty onset, and early experience-dependent programming of Avp, a major stress gene. At the genome-scale, genomic imprinting can be epigenetically switched on or off at select genes in a tightly controlled temporospatial manner and provides a versatile mechanism for dosage regulation of genes with important roles in stem cell quiescence or differentiation. Moreover, retrotransposition in neural progenitors provides an intriguing example of an epigenetic-like switch, which is stimulated by bivalently marked neurodevelopmental genes and possibly results in increased genomic flexibility regarding unprecedented challenge. Overall, we propose that molecular epigenetic switches illuminate the catalyzing function of epigenetic mechanisms in guiding dynamic changes in gene expression underpinning robust transitions in cellular and organismal phenotypes as well as in the mediation between dynamically changing environments and the static genetic blueprint.
Collapse
Affiliation(s)
- Anke Hoffmann
- Translational Research, Max Planck Society, Max Planck Institute of Psychiatry Munich, Bavaria, Germany
| | - Christoph A Zimmermann
- Translational Research, Max Planck Society, Max Planck Institute of Psychiatry Munich, Bavaria, Germany
| | - Dietmar Spengler
- Translational Research, Max Planck Society, Max Planck Institute of Psychiatry Munich, Bavaria, Germany
| |
Collapse
|
165
|
Lin X, Zhou S, Wen L, Davie A, Yao X, Liu W, Zhang Y. Potential role of maternal lineage in the thoroughbred breeding strategy. Reprod Fertil Dev 2015; 28:RD15063. [PMID: 25940872 DOI: 10.1071/rd15063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 04/01/2015] [Indexed: 02/28/2024] Open
Abstract
Many studies have focused on identifying the genes or single nucleotide polymorphisms associated with the athletic ability of thoroughbreds, but few have considered differences in maternal and paternal heritability of athletic ability. Herein, we report on our association study of career race performances of 675 Australian thoroughbreds with their pedigrees. Racing performance data (prize money per start) were collected from the Bloodhound database. The performance of all horses was categorised as either poor or elite athletic achievement. Then, 675 foals were divided by their parents' performance (elite or poor) into four groups: (1) elite dams and elite sires; (2) elite dams and poor sires; (3) poor dams and elite sires; and (4) poor dams and poor sires. The performance of foals was then compared between the four groups. The results show that the heritability of race performance between dams and foals (r = 0.141, P < 0.001) is much higher than that between sires and foals (r = 0.035, P = 0.366), and that this difference is statistically significant (P < 0.05). We also examined the effect of the child-bearing age of dams and sires on the ratio of elite foals. We found a strong correlation between the number of elite foals and dams' child-bearing age (r = -0.105, P < 0.001), with the ratio of elite offspring reaching a high level between a child-bearing age of 8 and 11 years (χ2 = 14.31, d.f. = 1, P < 0.001). These findings suggest that the maternal line may play an important role in the selective breeding of athletic performance in thoroughbreds.
Collapse
|
166
|
Kottler ML. [Paternal GNAS mutations: Which phenotypes? What genetic counseling?]. ANNALES D'ENDOCRINOLOGIE 2015; 76:105-9. [PMID: 25952723 DOI: 10.1016/j.ando.2015.03.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/17/2015] [Indexed: 11/20/2022]
Abstract
Parental imprinting and the type of the genetic alteration play a determinant role in the phenotype expression of GNAS locus associated to pseudohypoparathyroidism (PHP). GNAS locus gives rise to several different messenger RNA transcripts that are derived from the paternal allele, the maternal allele, or both and can be either coding or non-coding. As a consequence, GNAS mutations lead to a wide spectrum of phenotypes. An alteration in the coding sequence of the gene leads to a haplo-insufficiency and a dysmorphic phenotype (Albright's syndrome or AHO). AHO is a clinical syndrome defined by specific physical features including short stature, obesity, round-shaped face, subcutaneous ossifications, brachymetarcapy (mainly of the 4th and 5th ray). If the alteration is on the maternal allele, there is a hormonal resistance to the PTH at the kidney level and to the TSH at the thyroid level. The phenotype is known as pseudohypoparathyroidism type 1a (PHP1a). If the alteration is on the paternal allele, there are few clinical signs with no hormonal resistance and the phenotype is known as pseudopseudo hypoparathyroidism (pseudo-PPHP). Heterozygous GNAS mutations on the paternal GNAS allele were associated with intra uterin growth retardation (IUGR). Moreover, birth weights were lower with paternal GNAS mutations affecting exon 2-13 than with exon 1/intron 1 mutations suggesting a role for loss of function XLαs. Progressive osseous heteroplasia (POH) is a rare disease of ectopic bone formation, characterized by cutaneous and subcutaneous ossifications progressing towards deep connective and muscular tissues. POH is caused by a heterozygous GNAS inactivating mutation and has been associated with paternal inheritance. However, genotype/phenotype correlations suggest that there is no direct correlation between the ossifying process and parental origin, as there is high variability in heterotopic ossification. Clinical heterogeneity makes genetic counseling a very delicate matter, specifically where paternal inheritance is concerned as it can lead either to a mild expression of pseudo-PHP or to a severe one of POH.
Collapse
Affiliation(s)
- Marie-Laure Kottler
- Department of Genetics, Reference centre for rare disease of calcium and phosphorus metabolism, Caen University Hospital, 14033 Caen, France.
| |
Collapse
|
167
|
Babak T, DeVeale B, Tsang EK, Zhou Y, Li X, Smith KS, Kukurba KR, Zhang R, Li JB, van der Kooy D, Montgomery SB, Fraser HB. Genetic conflict reflected in tissue-specific maps of genomic imprinting in human and mouse. Nat Genet 2015; 47:544-9. [PMID: 25848752 PMCID: PMC4414907 DOI: 10.1038/ng.3274] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 03/13/2015] [Indexed: 12/12/2022]
Abstract
Genomic imprinting is an epigenetic process that restricts gene expression to either the maternally or paternally inherited allele. Many theories have been proposed to explain its evolutionary origin, but understanding has been limited by a paucity of data mapping the breadth and dynamics of imprinting within any organism. We generated an atlas of imprinting spanning 33 mouse and 45 human developmental stages and tissues. Nearly all imprinted genes were imprinted in early development and either retained their parent-of-origin expression in adults or lost it completely. Consistent with an evolutionary signature of parental conflict, imprinted genes were enriched for coexpressed pairs of maternally and paternally expressed genes, showed accelerated expression divergence between human and mouse, and were more highly expressed than their non-imprinted orthologs in other species. Our approach demonstrates a general framework for the discovery of imprinting in any species and sheds light on the causes and consequences of genomic imprinting in mammals.
Collapse
Affiliation(s)
- Tomas Babak
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| | - Brian DeVeale
- UCSF School of Medicine, UCSF, San Francisco, CA, 94143, USA
| | - Emily K. Tsang
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Yiqi Zhou
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| | - Xin Li
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Kevin S. Smith
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Kim R. Kukurba
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
| | - Rui Zhang
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
| | - Jin Billy Li
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
| | - Derek van der Kooy
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Stephen B. Montgomery
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
| | - Hunter B. Fraser
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
168
|
Binder NK, Beard SA, Kaitu'u-Lino TJ, Tong S, Hannan NJ, Gardner DK. Paternal obesity in a rodent model affects placental gene expression in a sex-specific manner. Reproduction 2015; 149:435-44. [DOI: 10.1530/rep-14-0676] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fetal growth restriction (FGR) is a major obstetric complication stemming from poor placental development. We have previously demonstrated that paternal obesity in mice is associated with impaired embryo development and significantly reduced fetal and placental weights. We hypothesised that the FGR observed in our rodent model of paternal diet-induced obesity is associated with alterations in metabolic, cell signalling and stress pathways. Male C57BL/6 mice were fed either a normal or high-fat diet for 10 weeks before sperm collection for IVF and subsequent embryo transfer. On embryonic day 14, placentas were collected and RNA extracted from both male and female placentas to assess mRNA expression of 24 target genes using custom RT-qPCR arrays. Peroxisome proliferator-activated receptor alpha (Ppara) and caspase-12 (Casp12) expression were significantly altered in male placentas from obese fathers compared with normal (P<0.05), but not female placentas. PPARA and CASP12 proteins were localised within the placenta to trophoblast giant cells by immunohistochemistry, and relative protein abundance was determined by western blot analysis. DNA was also extracted from the same placentas to determine methylation status. Global DNA methylation was significantly increased in female placentas from obese fathers compared with normal (P<0.05), but not male placentas. In this study, we demonstrate for the first time that paternal obesity is associated with changes in gene expression and methylation status of extraembryonic tissue in a sex-specific manner. These findings reinforce the negative consequences of paternal obesity before conception, and emphasise the need for more lifestyle advice for prospective fathers.
Collapse
|
169
|
O'Doherty AM, MacHugh DE, Spillane C, Magee DA. Genomic imprinting effects on complex traits in domesticated animal species. Front Genet 2015; 6:156. [PMID: 25964798 PMCID: PMC4408863 DOI: 10.3389/fgene.2015.00156] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 04/06/2015] [Indexed: 11/13/2022] Open
Abstract
Monoallelically expressed genes that exert their phenotypic effect in a parent-of-origin specific manner are considered to be subject to genomic imprinting, the most well understood form of epigenetic regulation of gene expression in mammals. The observed differences in allele specific gene expression for imprinted genes are not attributable to differences in DNA sequence information, but to specific chemical modifications of DNA and chromatin proteins. Since the discovery of genomic imprinting some three decades ago, over 100 imprinted mammalian genes have been identified and considerable advances have been made in uncovering the molecular mechanisms regulating imprinted gene expression. While most genomic imprinting studies have focused on mouse models and human biomedical disorders, recent work has highlighted the contributions of imprinted genes to complex trait variation in domestic livestock species. Consequently, greater understanding of genomic imprinting and its effect on agriculturally important traits is predicted to have major implications for the future of animal breeding and husbandry. In this review, we discuss genomic imprinting in mammals with particular emphasis on domestic livestock species and consider how this information can be used in animal breeding research and genetic improvement programs.
Collapse
Affiliation(s)
- Alan M O'Doherty
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield Ireland
| | - David E MacHugh
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield Ireland ; Animal Genomics Laboratory, UCD School of Agriculture and Food Science, University College Dublin, Belfield Ireland
| | - Charles Spillane
- Genetics and Biotechnology Laboratory, Plant and AgriBiosciences Research Centre, School of Natural Sciences, National University of Ireland Galway, Galway Ireland
| | - David A Magee
- Animal Genomics Laboratory, UCD School of Agriculture and Food Science, University College Dublin, Belfield Ireland ; Department of Animal Science, University of Connecticut, Storrs, CT USA
| |
Collapse
|
170
|
Sharma R, Agarwal A, Rohra VK, Assidi M, Abu-Elmagd M, Turki RF. Effects of increased paternal age on sperm quality, reproductive outcome and associated epigenetic risks to offspring. Reprod Biol Endocrinol 2015; 13:35. [PMID: 25928123 PMCID: PMC4455614 DOI: 10.1186/s12958-015-0028-x] [Citation(s) in RCA: 246] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 04/09/2015] [Indexed: 12/14/2022] Open
Abstract
Over the last decade, there has been a significant increase in average paternal age when the first child is conceived, either due to increased life expectancy, widespread use of contraception, late marriages and other factors. While the effect of maternal ageing on fertilization and reproduction is well known and several studies have shown that women over 35 years have a higher risk of infertility, pregnancy complications, spontaneous abortion, congenital anomalies, and perinatal complications. The effect of paternal age on semen quality and reproductive function is controversial for several reasons. First, there is no universal definition for advanced paternal ageing. Secondly, the literature is full of studies with conflicting results, especially for the most common parameters tested. Advancing paternal age also has been associated with increased risk of genetic disease. Our exhaustive literature review has demonstrated negative effects on sperm quality and testicular functions with increasing paternal age. Epigenetics changes, DNA mutations along with chromosomal aneuploidies have been associated with increasing paternal age. In addition to increased risk of male infertility, paternal age has also been demonstrated to impact reproductive and fertility outcomes including a decrease in IVF/ICSI success rate and increasing rate of preterm birth. Increasing paternal age has shown to increase the incidence of different types of disorders like autism, schizophrenia, bipolar disorders, and childhood leukemia in the progeny. It is thereby essential to educate the infertile couples on the disturbing links between increased paternal age and rising disorders in their offspring, to better counsel them during their reproductive years.
Collapse
Affiliation(s)
- Rakesh Sharma
- Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA.
| | - Ashok Agarwal
- Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA.
| | - Vikram K Rohra
- Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA.
| | - Mourad Assidi
- Center of Excellence in Genomic Medicine Research, King AbdulAziz University, Jeddah, Saudi Arabia.
- KACST Technology Innovation Center in Personalized Medicine at King AbdulAziz University, Jeddah, Saudi Arabia.
| | - Muhammad Abu-Elmagd
- Center of Excellence in Genomic Medicine Research, King AbdulAziz University, Jeddah, Saudi Arabia.
- KACST Technology Innovation Center in Personalized Medicine at King AbdulAziz University, Jeddah, Saudi Arabia.
| | - Rola F Turki
- KACST Technology Innovation Center in Personalized Medicine at King AbdulAziz University, Jeddah, Saudi Arabia.
- Obstetrics and Gynecology Department, King Abdulaziz University Hospital, Jeddah, Saudi Arabia.
| |
Collapse
|
171
|
Spies N, Smith CL, Rodriguez JM, Baker JC, Batzoglou S, Sidow A. Constraint and divergence of global gene expression in the mammalian embryo. eLife 2015; 4:e05538. [PMID: 25871848 PMCID: PMC4417935 DOI: 10.7554/elife.05538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 04/13/2015] [Indexed: 11/18/2022] Open
Abstract
The effects of genetic variation on gene regulation in the developing mammalian embryo remain largely unexplored. To globally quantify these effects, we crossed two divergent mouse strains and asked how genotype of the mother or of the embryo drives gene expression phenotype genomewide. Embryonic expression of 331 genes depends on the genotype of the mother. Embryonic genotype controls allele-specific expression of 1594 genes and a highly overlapping set of cis-expression quantitative trait loci (eQTL). A marked paucity of trans-eQTL suggests that the widespread expression differences do not propagate through the embryonic gene regulatory network. The cis-eQTL genes exhibit lower-than-average evolutionary conservation and are depleted for developmental regulators, consistent with purifying selection acting on expression phenotype of pattern formation genes. The widespread effect of maternal and embryonic genotype in conjunction with the purifying selection we uncovered suggests that embryogenesis is an important and understudied reservoir of phenotypic variation.
Collapse
Affiliation(s)
- Noah Spies
- Department of Pathology, Stanford University School of Medicine, Stanford, United States
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Cheryl L Smith
- Department of Pathology, Stanford University School of Medicine, Stanford, United States
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Jesse M Rodriguez
- Department of Computer Science, Stanford University, Stanford, United States
- Biomedical Informatics Program, Stanford University School of Medicine, Stanford, United States
| | - Julie C Baker
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Serafim Batzoglou
- Department of Computer Science, Stanford University, Stanford, United States
| | - Arend Sidow
- Department of Pathology, Stanford University School of Medicine, Stanford, United States
- Department of Genetics, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
172
|
Zerres K, Eggermann T. [Genetics and epigenetics. Explanatory approaches for (gender-specific) mechanisms of disease development]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2015; 57:1047-53. [PMID: 25070408 DOI: 10.1007/s00103-014-2013-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Whereas the central role of DNA as the carrier of genetic information has long been well known, the impact of epigenetic mechanisms as mediators between genes and environment is now becoming increasingly clear. Epigenetics helps explain the partially reversible interplay between gene function and environment and even permits observation of the transgenerational transmission of epigenetic modifications. Of special interest are gender-specific mechanisms of gene regulation which, among others, offer an explanation for gender differences in human diseases. Since the study of epigenetic mechanisms and their impact on the etiology of common diseases is in its infancy, it is too early to draw general conclusions from the current state of knowledge. Moreover, completely new strategies are needed to research these effects. In addition to molecular findings, definitions of specific phenotypes are required, including biographic data of affected individuals and their ancestors. Epigenetics needs to be viewed in the context of the theory of evolution, classical genetics, and environmental research. Its aim is not to substitute the knowledge in these disciplines, but rather to provide a key to link their findings, thereby opening up new possibilities in terms of interpretation and understanding of gender differences in medicine. If these epigenetic mechanisms are better understood, particularly in terms of specific diseases, it is conceivable that these disorders could be influenced and treated in a more targeted manner in the future.
Collapse
Affiliation(s)
- K Zerres
- Institut für Humangenetik, Universitätsklinikum der RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Deutschland,
| | | |
Collapse
|
173
|
Bréhin AC, Colson C, Maupetit-Méhouas S, Grybek V, Richard N, Linglart A, Kottler ML, Jüppner H. Loss of methylation at GNAS exon A/B is associated with increased intrauterine growth. J Clin Endocrinol Metab 2015; 100:E623-31. [PMID: 25603460 PMCID: PMC4399294 DOI: 10.1210/jc.2014-4047] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 01/14/2015] [Indexed: 01/26/2023]
Abstract
CONTEXT GNAS is one of few genetic loci that undergo allelic-specific methylation resulting in the parent-specific expression of at least four different transcripts. Due to monoallelic expression, heterozygous GNAS mutations affecting either paternally or maternally derived transcripts cause different forms of pseudohypoparathyroidism (PHP), including autosomal-dominant PHP type Ib (AD-PHP1B) associated with loss of methylation (LOM) at exon A/B alone or sporadic PHP1B (sporPHP1B) associated with broad GNAS methylation changes. Similar to effects other imprinted genes have on early development, we recently observed severe intrauterine growth retardation in newborns, later diagnosed with pseudopseudohypoparathyroidism (PPHP) because of paternal GNAS loss-of-function mutations. OBJECTIVES This study aimed to determine whether GNAS methylation abnormalities affect intrauterine growth. PATIENTS AND METHODS Birth parameters were collected of patients who later developed sporPHP1B or AD-PHP1B, and of their healthy siblings. Comparisons were made to newborns affected by PPHP or PHP1A. RESULTS As newborns, AD-PHP1B patients were bigger than their healthy siblings and well above the reference average; increased sizes were particularly evident if the mothers were unaffected carriers of STX16 deletions. SporPHP1B newborns were slightly above average for weight and length, but their overgrowth was less pronounced than that of AD-PHP1B newborns from unaffected mothers. CONCLUSION LOM at GNAS exon A/B due to maternal STX16 deletions and the resulting biallelic A/B expression are associated with enhanced fetal growth. These findings are distinctly different from those of PPHP patients with paternal GNAS exons 2-13 mutations, whose birth parameters are almost 4.5 z-scores below those of AD-PHP1B patients born to healthy mothers.
Collapse
Affiliation(s)
- Anne-Claire Bréhin
- Department of Genetics (A.-C.B., C.C., N.R., M.-L.K.), Centre Hospitalier Universitaire de Caen, Reference Centre for Rare Disorders of Calcium and Phosphorus Metabolism, F-14000 Caen, France; Paediatric Endocrinology and Diabetology (S.M.-M., V.G., A.L.), Reference Centre for Rare Disorders of the Mineral Metabolism, AP-HP Hôpital Bicêtre, le Kremlin-Bicêtre 94270, France; Faculté de Médecine, Université Paris Sud, le Kremlin-Bicêtre 94270, France; and Pediatric Nephrology Unit and Endocrine Unit (H.J.), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114
| | | | | | | | | | | | | | | |
Collapse
|
174
|
|
175
|
Van Soom A, Peelman L, Holt WV, Fazeli A. An introduction to epigenetics as the link between genotype and environment: a personal view. Reprod Domest Anim 2015; 49 Suppl 3:2-10. [PMID: 25220743 DOI: 10.1111/rda.12341] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2014] [Indexed: 12/18/2022]
Abstract
Lamarck was one of the first scientists who attempted to explain evolution, and he is especially well known for formulating the concept that acquired characteristics can be transmitted to future generations and may therefore steer evolution. Although Lamarckism fell out of favour soon after the publication of Darwin's work on natural selection and evolution, the concept of transmission of acquired characteristics has recently gained renewed attention and has led to some rethinking of the standard evolutionary model. Epigenetics, or the study of heritable (mitotically and/or meiotically) changes in gene activity that are not brought about by changes in the DNA sequence, can explain some types of ill health in offspring, which have been exposed to stressors during early development, when DNA is most susceptible to such epigenetic influences. In this review, we explain briefly the history of epigenetics and we propose some examples of epigenetic and transgenerational effects demonstrated in humans and animals. Growing evidence is available that the health and phenotype of a given individual is already shaped shortly before and after the time of conception. Some evidence suggests that epigenetic markings, which have been established around conception, can also be transmitted to future generations. This knowledge can possibly be used to revolutionize animal breeding and to increase human and animal health worldwide.
Collapse
Affiliation(s)
- A Van Soom
- Faculty of Veterinary Medicine, Department of Reproduction, Obstetrics and Herd Health, Ghent University, Merelbeke, Belgium
| | | | | | | |
Collapse
|
176
|
Daniel G, Schmidt-Edelkraut U, Spengler D, Hoffmann A. Imprinted Zac1 in neural stem cells. World J Stem Cells 2015; 7:300-314. [PMID: 25815116 PMCID: PMC4369488 DOI: 10.4252/wjsc.v7.i2.300] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/24/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
Neural stem cells (NSCs) and imprinted genes play an important role in brain development. On historical grounds, these two determinants have been largely studied independently of each other. Recent evidence suggests, however, that NSCs can reset select genomic imprints to prevent precocious depletion of the stem cell reservoir. Moreover, imprinted genes like the transcriptional regulator Zac1 can fine tune neuronal vs astroglial differentiation of NSCs. Zac1 binds in a sequence-specific manner to pro-neuronal and imprinted genes to confer transcriptional regulation and furthermore coregulates members of the p53-family in NSCs. At the genome scale, Zac1 is a central hub of an imprinted gene network comprising genes with an important role for NSC quiescence, proliferation and differentiation. Overall, transcriptional, epigenomic, and genomic mechanisms seem to coordinate the functional relationships of NSCs and imprinted genes from development to maturation, and possibly aging.
Collapse
|
177
|
Abstract
A gene mediating interactions between mouse mothers and their pups has recently been claimed to support coadaptation rather than the kinship theory of genomic imprinting. This Formal Comment argues that this claim is unfounded.
Collapse
|
178
|
Prasad RB, Groop L. Genetics of type 2 diabetes-pitfalls and possibilities. Genes (Basel) 2015; 6:87-123. [PMID: 25774817 PMCID: PMC4377835 DOI: 10.3390/genes6010087] [Citation(s) in RCA: 290] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 01/28/2015] [Accepted: 02/27/2015] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes (T2D) is a complex disease that is caused by a complex interplay between genetic, epigenetic and environmental factors. While the major environmental factors, diet and activity level, are well known, identification of the genetic factors has been a challenge. However, recent years have seen an explosion of genetic variants in risk and protection of T2D due to the technical development that has allowed genome-wide association studies and next-generation sequencing. Today, more than 120 variants have been convincingly replicated for association with T2D and many more with diabetes-related traits. Still, these variants only explain a small proportion of the total heritability of T2D. In this review, we address the possibilities to elucidate the genetic landscape of T2D as well as discuss pitfalls with current strategies to identify the elusive unknown heritability including the possibility that our definition of diabetes and its subgroups is imprecise and thereby makes the identification of genetic causes difficult.
Collapse
Affiliation(s)
- Rashmi B Prasad
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Lund University, CRC, Skåne University Hospital SUS, SE-205 02 Malmö, Sweden.
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Lund University, CRC, Skåne University Hospital SUS, SE-205 02 Malmö, Sweden.
- Finnish Institute of Molecular Medicine (FIMM), Helsinki University, Helsinki 00014, Finland.
| |
Collapse
|
179
|
Gundling WE, Wildman DE. A review of inter- and intraspecific variation in the eutherian placenta. Philos Trans R Soc Lond B Biol Sci 2015; 370:20140072. [PMID: 25602076 PMCID: PMC4305173 DOI: 10.1098/rstb.2014.0072] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The placenta is one of the most morphologically variable mammalian organs. Four major characteristics are typically discussed when comparing the placentas of different eutherian species: placental shape, maternal-fetal interdigitation, intimacy of the maternal-fetal interface and the pattern of maternal-fetal blood flow. Here, we describe the evolution of three of these features as well as other key aspects of eutherian placentation. In addition to interspecific anatomical variation, there is also variation in placental anatomy and function within a single species. Much of this intraspecific variation occurs in response to different environmental conditions such as altitude and poor maternal nutrition. Examinations of variation in the placenta from both intra- and interspecies perspectives elucidate different aspects of placental function and dysfunction at the maternal-fetal interface. Comparisons within species identify candidate mechanisms that are activated in response to environmental stressors ultimately contributing to the aetiology of obstetric syndromes such as pre-eclampsia. Comparisons above the species level identify the evolutionary lineages on which the potential for the development of obstetric syndromes emerged.
Collapse
Affiliation(s)
- William E Gundling
- Institute for Genomic Biology, University of Illinois, Urbana, IL, USA Department of Molecular and Integrative Physiology, University of Illinois, Urbana, IL, USA Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Derek E Wildman
- Institute for Genomic Biology, University of Illinois, Urbana, IL, USA Department of Molecular and Integrative Physiology, University of Illinois, Urbana, IL, USA Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
180
|
Al Adhami H, Evano B, Le Digarcher A, Gueydan C, Dubois E, Parrinello H, Dantec C, Bouschet T, Varrault A, Journot L. A systems-level approach to parental genomic imprinting: the imprinted gene network includes extracellular matrix genes and regulates cell cycle exit and differentiation. Genome Res 2015; 25:353-67. [PMID: 25614607 PMCID: PMC4352888 DOI: 10.1101/gr.175919.114] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 12/08/2014] [Indexed: 11/24/2022]
Abstract
Genomic imprinting is an epigenetic mechanism that restrains the expression of ∼ 100 eutherian genes in a parent-of-origin-specific manner. The reason for this selective targeting of genes with seemingly disparate molecular functions is unclear. In the present work, we show that imprinted genes are coexpressed in a network that is regulated at the transition from proliferation to quiescence and differentiation during fibroblast cell cycle withdrawal, adipogenesis in vitro, and muscle regeneration in vivo. Imprinted gene regulation is not linked to alteration of DNA methylation or to perturbation of monoallelic, parent-of-origin-dependent expression. Overexpression and knockdown of imprinted gene expression alters the sensitivity of preadipocytes to contact inhibition and adipogenic differentiation. In silico and in cellulo experiments showed that the imprinted gene network includes biallelically expressed, nonimprinted genes. These control the extracellular matrix composition, cell adhesion, cell junction, and extracellular matrix-activated and growth factor-activated signaling. These observations show that imprinted genes share a common biological process that may account for their seemingly diverse roles in embryonic development, obesity, diabetes, muscle physiology, and neoplasm.
Collapse
Affiliation(s)
- Hala Al Adhami
- Institut de Genomique Fonctionnelle, Montpellier 34094, France; CNRS, UMR 5203, Montpellier 34094, France; INSERM, U661, Montpellier 34094, France; Faculté des Sciences, Université de Montpellier, Montpellier 34095, France
| | - Brendan Evano
- Institut de Genomique Fonctionnelle, Montpellier 34094, France; CNRS, UMR 5203, Montpellier 34094, France; INSERM, U661, Montpellier 34094, France; Faculté des Sciences, Université de Montpellier, Montpellier 34095, France
| | - Anne Le Digarcher
- Institut de Genomique Fonctionnelle, Montpellier 34094, France; CNRS, UMR 5203, Montpellier 34094, France; INSERM, U661, Montpellier 34094, France; Faculté des Sciences, Université de Montpellier, Montpellier 34095, France
| | - Charlotte Gueydan
- Institut de Genomique Fonctionnelle, Montpellier 34094, France; CNRS, UMR 5203, Montpellier 34094, France; INSERM, U661, Montpellier 34094, France; Faculté des Sciences, Université de Montpellier, Montpellier 34095, France
| | | | | | | | - Tristan Bouschet
- Institut de Genomique Fonctionnelle, Montpellier 34094, France; CNRS, UMR 5203, Montpellier 34094, France; INSERM, U661, Montpellier 34094, France; Faculté des Sciences, Université de Montpellier, Montpellier 34095, France
| | - Annie Varrault
- Institut de Genomique Fonctionnelle, Montpellier 34094, France; CNRS, UMR 5203, Montpellier 34094, France; INSERM, U661, Montpellier 34094, France; Faculté des Sciences, Université de Montpellier, Montpellier 34095, France
| | - Laurent Journot
- Institut de Genomique Fonctionnelle, Montpellier 34094, France; CNRS, UMR 5203, Montpellier 34094, France; INSERM, U661, Montpellier 34094, France; Faculté des Sciences, Université de Montpellier, Montpellier 34095, France; MGX-Montpellier GenomiX, Montpellier 34094, France
| |
Collapse
|
181
|
King K, Murphy S, Hoyo C. Epigenetic regulation of Newborns' imprinted genes related to gestational growth: patterning by parental race/ethnicity and maternal socioeconomic status. J Epidemiol Community Health 2015; 69:639-47. [PMID: 25678712 DOI: 10.1136/jech-2014-204781] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 01/22/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Children born to parents with lower income and education are at risk for obesity and later-life risk of common chronic diseases, and epigenetics has been hypothesised to link these associations. However, epigenetic targets are unknown. We focus on a cluster of well-characterised genomically imprinted genes because their monoallelic expression is regulated by DNA methylation at differentially methylated regions (DMRs), are critical in fetal growth, and DNA methylation patterns at birth have been associated with increased risk of birth weight extremes and overweight status or obesity in early childhood. METHODS We measured DNA methylation at DMRs regulating genomically imprinted domains (IGF2/H19, DLK1/MEG3, NNAT and PLAGL1) using umbilical cord blood leucocytes from 619 infants recruited in Durham, North Carolina in 2010-2011. We examined differences in DNA methylation levels by race/ethnicity of both parents, and the role that maternal socioeconomic status (SES) may play in the association between race/ethnic epigenetic differences. RESULTS Unadjusted race/ethnic differences only were evident for DMRs regulating MEG3 and IGF2; race/ethnic differences persisted in IGF2/H19 and NNAT after accounting for income and education. CONCLUSIONS Results suggest that parental factors may not only influence DNA methylation, but also do so in ways that vary by DMR. Findings support the hypothesis that epigenetics may link the observed lower SES during the prenatal period and poor outcomes such as low birth weight; lower birth weight has previously been associated with adult-onset chronic diseases and conditions that include cardiovascular diseases, diabetes, obesity and some cancers.
Collapse
Affiliation(s)
- Katherine King
- Environmental Public Health Division, U.S. Environmental Protection Agency, Chapel Hill, North Carolina, USA Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Susan Murphy
- Department of Obstetrics and Gynecology, Duke University, Durham, North Carolina, USA
| | - Cathrine Hoyo
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
182
|
Liu N, Archer E, Srinivasasainagendra V, Allison DB. A statistical framework for testing the causal effects of fetal drive. Front Genet 2015; 5:464. [PMID: 25628644 PMCID: PMC4292723 DOI: 10.3389/fgene.2014.00464] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 12/17/2014] [Indexed: 12/26/2022] Open
Abstract
Maternal genetic and phenotypic characteristics (e.g., metabolic and behavioral) affect both the intrauterine milieu and lifelong health trajectories of their fetuses. Yet at the same time, fetal genotype may affect processes that alter pre and postnatal maternal physiology, and the subsequent health of both fetus and mother. We refer to these latter effects as 'fetal drive.' If fetal genotype is driving physiologic, metabolic, and behavioral phenotypic changes in the mother, there is a possibility of differential effects with different fetal genomes inducing different long-term effects on both maternal and fetal health, mediated through intrauterine environment. This proposed mechanistic path remains largely unexamined and untested. In this study, we offer a statistical method to rigorously test this hypothesis and make causal inferences in humans by relying on the (conditional) randomization inherent in the process of meiosis. For illustration, we apply this method to a dataset from the Framingham Heart Study.
Collapse
Affiliation(s)
- Nianjun Liu
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA
| | - Edward Archer
- Office of Energetics, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA
| | - Vinodh Srinivasasainagendra
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA
| | - David B Allison
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA ; Office of Energetics, School of Public Health, University of Alabama at Birmingham Birmingham, AL, USA
| |
Collapse
|
183
|
Genome-wide methylation analysis in Silver-Russell syndrome patients. Hum Genet 2015; 134:317-332. [PMID: 25563730 DOI: 10.1007/s00439-014-1526-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/29/2014] [Indexed: 01/09/2023]
Abstract
Silver-Russell syndrome (SRS) is a clinically heterogeneous disorder characterised by severe in utero growth restriction and poor postnatal growth, body asymmetry, irregular craniofacial features and several additional minor malformations. The aetiology of SRS is complex and current evidence strongly implicates imprinted genes. Approximately, half of all patients exhibit DNA hypomethylation at the H19/IGF2 imprinted domain, and around 10% have maternal uniparental disomy of chromosome 7. We measured DNA methylation in 18 SRS patients at >485,000 CpG sites using DNA methylation microarrays. Using a novel bioinformatics methodology specifically designed to identify subsets of patients with a shared epimutation, we analysed methylation changes genome-wide as well as at known imprinted regions to identify SRS-associated epimutations. Our analysis identifies epimutations at the previously characterised domains of H19/IGF2 and at imprinted regions on chromosome 7, providing proof of principle that our methodology can detect DNA methylation changes at imprinted loci. In addition, we discovered two novel epimutations associated with SRS and located at imprinted loci previously linked to relevant mouse and human phenotypes. We identify RB1 as an additional imprinted locus associated with SRS, with a region near the RB1 differentially methylated region hypermethylated in 13/18 (~70%) patients. We also report 6/18 (~33%) patients were hypermethylated at a CpG island near the ANKRD11 gene. We do not observe consistent co-occurrence of epimutations at multiple imprinted loci in single SRS individuals. SRS is clinically heterogeneous and the absence of multiple imprinted loci epimutations reflects the heterogeneity at the molecular level. Further stratification of SRS patients by molecular phenotypes might aid the identification of disease causes.
Collapse
|
184
|
Madon-Simon M, Cowley M, Garfield AS, Moorwood K, Bauer SR, Ward A. Antagonistic roles in fetal development and adult physiology for the oppositely imprinted Grb10 and Dlk1 genes. BMC Biol 2014; 12:771. [PMID: 25551289 PMCID: PMC4280702 DOI: 10.1186/s12915-014-0099-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/07/2014] [Indexed: 12/14/2022] Open
Abstract
Background Despite being a fundamental biological problem the control of body size and proportions during development remains poorly understood, although it is accepted that the insulin-like growth factor (IGF) pathway has a central role in growth regulation, probably in all animals. The involvement of imprinted genes has also attracted much attention, not least because two of the earliest discovered were shown to be oppositely imprinted and antagonistic in their regulation of growth. The Igf2 gene encodes a paternally expressed ligand that promotes growth, while maternally expressed Igf2r encodes a cell surface receptor that restricts growth by sequestering Igf2 and targeting it for lysosomal degradation. There are now over 150 imprinted genes known in mammals, but no other clear examples of antagonistic gene pairs have been identified. The delta-like 1 gene (Dlk1) encodes a putative ligand that promotes fetal growth and in adults restricts adipose deposition. Conversely, Grb10 encodes an intracellular signalling adaptor protein that, when expressed from the maternal allele, acts to restrict fetal growth and is permissive for adipose deposition in adulthood. Results Here, using knockout mice, we present genetic and physiological evidence that these two factors exert their opposite effects on growth and physiology through a common signalling pathway. The major effects are on body size (particularly growth during early life), lean:adipose proportions, glucose regulated metabolism and lipid storage in the liver. A biochemical pathway linking the two cell signalling factors remains to be defined. Conclusions We propose that Dlk1 and Grb10 define a mammalian growth axis that is separate from the IGF pathway, yet also features an antagonistic imprinted gene pair. Electronic supplementary material The online version of this article (doi:10.1186/s12915-014-0099-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | - Andrew Ward
- Department of Biology & Biochemistry and Centre for Regenerative Medicine, University of Bath, Building 4 South, Claverton Down, Bath BA2 7AY, UK.
| |
Collapse
|
185
|
Pomeroy E, Wells JCK, Cole TJ, O'Callaghan M, Stock JT. Relationships of maternal and paternal anthropometry with neonatal body size, proportions and adiposity in an Australian cohort. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2014; 156:625-36. [PMID: 25502164 PMCID: PMC4404025 DOI: 10.1002/ajpa.22680] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/29/2014] [Accepted: 11/24/2014] [Indexed: 02/04/2023]
Abstract
The patterns of association between maternal or paternal and neonatal phenotype may offer insight into how neonatal characteristics are shaped by evolutionary processes, such as conflicting parental interests in fetal investment and obstetric constraints. Paternal interests are theoretically served by maximizing fetal growth, and maternal interests by managing investment in current and future offspring, but whether paternal and maternal influences act on different components of overall size is unknown. We tested whether parents' prepregnancy height and body mass index (BMI) were related to neonatal anthropometry (birthweight, head circumference, absolute and proportional limb segment and trunk lengths, subcutaneous fat) among 1,041 Australian neonates using stepwise linear regression. Maternal and paternal height and maternal BMI were associated with birthweight. Paternal height related to offspring forearm and lower leg lengths, maternal height and BMI to neonatal head circumference, and maternal BMI to offspring adiposity. Principal components analysis identified three components of variability reflecting neonatal “head and trunk skeletal size,” “adiposity,” and “limb lengths.” Regression analyses of the component scores supported the associations of head and trunk size or adiposity with maternal anthropometry, and limb lengths with paternal anthropometry. Our results suggest that while neonatal fatness reflects environmental conditions (maternal physiology), head circumference and limb and trunk lengths show differing associations with parental anthropometry. These patterns may reflect genetics, parental imprinting and environmental influences in a manner consistent with parental conflicts of interest. Paternal height may relate to neonatal limb length as a means of increasing fetal growth without exacerbating the risk of obstetric complications. Am J Phys Anthropol 156:625–636, 2015.
Collapse
Affiliation(s)
- Emma Pomeroy
- Newnham College, University of Cambridge, Cambridge, UK; Division of Biological Anthropology, Department of Archaeology and Anthropology, University of Cambridge, Cambridge, UK
| | | | | | | | | |
Collapse
|
186
|
Shi ZH, Lin YT, Hou YM. Mother-derived trans-generational immune priming in the red palm weevil, Rhynchophorus ferrugineus Olivier (Coleoptera, Dryophthoridae). BULLETIN OF ENTOMOLOGICAL RESEARCH 2014; 104:742-750. [PMID: 25208627 DOI: 10.1017/s0007485314000583] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Rhynchophorus ferrugineus (Coleoptera, Curculionidae) is the most destructive pest of palm trees worldwide containing it invasive areas, such as the southern part of China. It is always emphasized to develop integrated pest management based on biological agents, but their success is not very exciting. Presently, the immune defenses of this pest against biological agents attract scarce attention. It is still unclear whether immune priming also generally occurs in insect pests and in response to different pathogens. Our results indicated that previous challenge of bacteria pathogen enhanced the magnitude of phenoloxidase activity and antibacterial activity in R. ferrugineus larvae against the secondary infection. Furthermore, trans-generational immune priming was also determined in this pest, and only challenged R. ferrugineus mothers transferred the immune protection to their offspring which suggested males and females of this pest might have evolved different strategies on the investment of delivering immune protection to their offspring. Importantly, our data provide the evidence to suggest that different kinds of biological control agents might be used alternatively or in combination to fight against R. ferrugineus because of the existence of immune priming with low species-specific level. On the other hand, for this invasive pest, the immune priming may also facilitate its adaptation and dispersal in the new regions.
Collapse
Affiliation(s)
- Z H Shi
- Key Laboratory of Integrated Pest Management of Fujian-Taiwan Crops,Ministry of Agriculture,China
| | - Y T Lin
- Key Laboratory of Integrated Pest Management of Fujian-Taiwan Crops,Ministry of Agriculture,China
| | - Y M Hou
- Key Laboratory of Integrated Pest Management of Fujian-Taiwan Crops,Ministry of Agriculture,China
| |
Collapse
|
187
|
Xiang R, Lee AMC, Eindorf T, Javadmanesh A, Ghanipoor-Samami M, Gugger M, Fitzsimmons CJ, Kruk ZA, Pitchford WS, Leviton AJ, Thomsen DA, Beckman I, Anderson GI, Burns BM, Rutley DL, Xian CJ, Hiendleder S. Widespread differential maternal and paternal genome effects on fetal bone phenotype at mid-gestation. J Bone Miner Res 2014; 29:2392-404. [PMID: 24753181 DOI: 10.1002/jbmr.2263] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 03/09/2014] [Accepted: 04/03/2014] [Indexed: 11/06/2022]
Abstract
Parent-of-origin-dependent (epi)genetic factors are important determinants of prenatal development that program adult phenotype. However, data on magnitude and specificity of maternal and paternal genome effects on fetal bone are lacking. We used an outbred bovine model to dissect and quantify effects of parental genomes, fetal sex, and nongenetic maternal effects on the fetal skeleton and analyzed phenotypic and molecular relationships between fetal muscle and bone. Analysis of 51 bone morphometric and weight parameters from 72 fetuses recovered at day 153 gestation (54% term) identified six principal components (PC1-6) that explained 80% of the variation in skeletal parameters. Parental genomes accounted for most of the variation in bone wet weight (PC1, 72.1%), limb ossification (PC2, 99.8%), flat bone size (PC4, 99.7%), and axial skeletal growth (PC5, 96.9%). Limb length showed lesser effects of parental genomes (PC3, 40.8%) and a significant nongenetic maternal effect (gestational weight gain, 29%). Fetal sex affected bone wet weight (PC1, p < 0.0001) and limb length (PC3, p < 0.05). Partitioning of variation explained by parental genomes revealed strong maternal genome effects on bone wet weight (74.1%, p < 0.0001) and axial skeletal growth (93.5%, p < 0.001), whereas paternal genome controlled limb ossification (95.1%, p < 0.0001). Histomorphometric data revealed strong maternal genome effects on growth plate height (98.6%, p < 0.0001) and trabecular thickness (85.5%, p < 0.0001) in distal femur. Parental genome effects on fetal bone were mirrored by maternal genome effects on fetal serum 25-hydroxyvitamin D (96.9%, p < 0.001) and paternal genome effects on alkaline phosphatase (90.0%, p < 0.001) and their correlations with maternally controlled bone wet weight and paternally controlled limb ossification, respectively. Bone wet weight and flat bone size correlated positively with muscle weight (r = 0.84 and 0.77, p < 0.0001) and negatively with muscle H19 expression (r = -0.34 and -0.31, p < 0.01). Because imprinted maternally expressed H19 regulates growth factors by miRNA interference, this suggests muscle-bone interaction via epigenetic factors.
Collapse
Affiliation(s)
- Ruidong Xiang
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia; JS Davies Epigenetics and Genetics Group, School of Animal and Veterinary Sciences, Roseworthy Campus, The University of Adelaide, Adelaide, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Golding J, Northstone K, Gregory S, Miller LL, Pembrey M. The anthropometry of children and adolescents may be influenced by the prenatal smoking habits of their grandmothers: a longitudinal cohort study. Am J Hum Biol 2014; 26:731-9. [PMID: 25130101 PMCID: PMC4238812 DOI: 10.1002/ajhb.22594] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/11/2014] [Accepted: 07/06/2014] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVES Previously, in the Avon Longitudinal Study of Parents and Children (ALSPAC), we have shown different sex-specific birth anthropometric measurements contingent upon whether or not prenatal smoking was undertaken by paternal grandmother (PGM±), maternal grandmother (MGM±), and the study mother (M±). The findings raised the question as to whether there were long-term associations on the growth of the study children over time. METHODS Measures of weight, height, body mass index, waist circumference, lean mass, and fat mass of children in the ALSPAC study from 7 to 17 years of age were used. We compared growth in four categories at each age: PGM+M- with PGM-M-; MGM+M- with MGM-M-; PGM+M+ with PGM-M+; MGM+M+ with MGM-M+; and adjusted for housing tenure, maternal education, parity, and paternal smoking at the start of the study pregnancy. RESULTS We found that if the PGM had, but the study mother had not, smoked in pregnancy, the girls were taller and both genders had greater bone and lean mass. However, if the MGM had smoked prenatally but the mother had not (MGM+M-), the boys became heavier than expected with increasing age-an association that was particularly due to lean rather than fat mass, reflected in increased strength and fitness. When both the maternal grandmother and the mother had smoked (MGM+M+) girls had reduced height, weight, and fat/lean/bone mass when compared with girls born to smoking mothers whose own mothers had not smoked (MGM-M+). CONCLUSIONS This study indicates that smoking in humans can have sex-specific transgenerational effects.
Collapse
Affiliation(s)
- Jean Golding
- Centre for Child and Adolescent Health, School of Social and Community Medicine, University of BristolBristol, BS8 2BN, United Kingdom
| | - Kate Northstone
- Centre for Child and Adolescent Health, School of Social and Community Medicine, University of BristolBristol, BS8 2BN, United Kingdom
| | - Steven Gregory
- Centre for Child and Adolescent Health, School of Social and Community Medicine, University of BristolBristol, BS8 2BN, United Kingdom
| | - Laura L Miller
- Centre for Child and Adolescent Health, School of Social and Community Medicine, University of BristolBristol, BS8 2BN, United Kingdom
| | - Marcus Pembrey
- Centre for Child and Adolescent Health, School of Social and Community Medicine, University of BristolBristol, BS8 2BN, United Kingdom
| |
Collapse
|
189
|
Shen SQ, Turro E, Corbo JC. Hybrid mice reveal parent-of-origin and Cis- and trans-regulatory effects in the retina. PLoS One 2014; 9:e109382. [PMID: 25340786 PMCID: PMC4207689 DOI: 10.1371/journal.pone.0109382] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 09/02/2014] [Indexed: 11/30/2022] Open
Abstract
A fundamental challenge in genomics is to map DNA sequence variants onto changes in gene expression. Gene expression is regulated by cis-regulatory elements (CREs, i.e., enhancers, promoters, and silencers) and the trans factors (e.g., transcription factors) that act upon them. A powerful approach to dissecting cis and trans effects is to compare F1 hybrids with F0 homozygotes. Using this approach and taking advantage of the high frequency of polymorphisms in wild-derived inbred Cast/EiJ mice relative to the reference strain C57BL/6J, we conducted allele-specific mRNA-seq analysis in the adult mouse retina, a disease-relevant neural tissue. We found that cis effects account for the bulk of gene regulatory divergence in the retina. Many CREs contained functional (i.e., activating or silencing) cis-regulatory variants mapping onto altered expression of genes, including genes associated with retinal disease. By comparing our retinal data with previously published liver data, we found that most of the cis effects identified were tissue-specific. Lastly, by comparing reciprocal F1 hybrids, we identified evidence of imprinting in the retina for the first time. Our study provides a framework and resource for mapping cis-regulatory variants onto changes in gene expression, and underscores the importance of studying cis-regulatory variants in the context of retinal disease.
Collapse
Affiliation(s)
- Susan Q. Shen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ernest Turro
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, National Health Service Blood and Transplant, Cambridge, United Kingdom
| | - Joseph C. Corbo
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
190
|
Tilghman SM. Twists and turns: a scientific journey. Annu Rev Cell Dev Biol 2014; 30:1-21. [PMID: 25288111 DOI: 10.1146/annurev-cellbio-100913-013512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In this perspective I look back on the twists and turns that influenced the direction of my scientific career over the past 40 years. From my early ambition to be a chemist to my training in Philadelphia and Bethesda as a molecular biologist, I benefited enormously from generous and valuable mentoring. In my independent career in Philadelphia and Princeton, I was motivated by a keen interest in the changes in gene expression that direct the development of the mammalian embryo and inspired by the creativity and energy of my students, fellows, and research staff. After twelve years as President of Princeton University, I have happily returned to the faculty of the Department of Molecular Biology.
Collapse
Affiliation(s)
- Shirley M Tilghman
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544;
| |
Collapse
|
191
|
Guo C, Chen X, Song H, Maynard MA, Zhou Y, Lobanov AV, Gladyshev VN, Ganis JJ, Wiley D, Jugo RH, Lee NY, Castroneves LA, Zon LI, Scanlan TS, Feldman HA, Huang SA. Intrinsic expression of a multiexon type 3 deiodinase gene controls zebrafish embryo size. Endocrinology 2014; 155:4069-80. [PMID: 25004091 PMCID: PMC4164935 DOI: 10.1210/en.2013-2029] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thyroid hormone is a master regulator of differentiation and growth, and its action is terminated by the enzymatic removal of an inner-ring iodine catalyzed by the selenoenzyme type 3 deiodinase (dio3). Our studies of the zebrafish reveal that the dio3 gene is duplicated in this species and that embryonic deiodination is an important determinant of embryo size. Although both dio3 paralogs encode enzymatically active proteins with high affinity for thyroid hormones, their anatomic patterns of expression are markedly divergent and only embryos with knockdown of dio3b, a biallelically expressed selenoenzyme expressed in the developing central nervous system, manifest severe thyroid hormone-dependent growth restriction at 72 hours post fertilization. This indicates that the embryonic deficiency of dio3, once considered only a placental enzyme, causes microsomia independently of placental physiology and raises the intriguing possibility that fetal abnormalities in human deiodination may present as intrauterine growth retardation. By mapping the gene structures and enzymatic properties of all four zebrafish deiodinases, we also identify dio3b as the first multiexon dio3 gene, containing a large intron separating its open reading frame from its selenocysteine insertion sequence (SECIS) element.
Collapse
Affiliation(s)
- Cuicui Guo
- State Key Laboratory of Medical Genomics (C.G., X.C., H.S.), Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China; Thyroid Program of the Division of Endocrinology (C.G., X.C., H.S., M.A.M., R.H.J., N.Y.L., L.A.C., S.A.H.) and Clinical Research Center (H.A.F.), Boston Children's Hospital; Stem Cell Program and Division of Hematology/Oncology (Y.Z., J.J.G., D.W., L.I.Z.), Boston Children's Hospital, Harvard Stem Cell Institute, Harvard Medical School, and Howard Hughes Medical Institute; Department of Medicine (A.V.L., V.N.G., S.A.H.), Brigham and Women's Hospital; Dana Farber Cancer Institute (V.N.G., L.I.Z., S.A.H.), Boston, Massachusetts 02115; and Departments of Physiology and Pharmacology (T.S.S.), Oregon Health and Science University, Portland, Oregon 97239
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
192
|
He H, Kim J. Regulation and function of the peg3 imprinted domain. Genomics Inform 2014; 12:105-13. [PMID: 25317109 PMCID: PMC4196374 DOI: 10.5808/gi.2014.12.3.105] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 08/11/2014] [Accepted: 08/14/2014] [Indexed: 01/12/2023] Open
Abstract
A subset of mammalian genes differ functionally between two alleles due to genomic imprinting, and seven such genes (Peg3, Usp29, APeg3, Zfp264, Zim1, Zim2, Zim3) are localized within the 500-kb genomic interval of the human and mouse genomes, constituting the Peg3 imprinted domain. This Peg3 domain shares several features with the other imprinted domains, including an evolutionarily conserved domain structure, along with transcriptional co-regulation through shared cis regulatory elements, as well as functional roles in controlling fetal growth rates and maternal-caring behaviors. The Peg3 domain also displays some unique features, including YY1-mediated regulation of transcription and imprinting; conversion and adaptation of several protein-coding members as ncRNA genes during evolution; and its close connection to human cancers through the potential tumor suppressor functions of Peg3 and Usp29. In this review, we summarize and discuss these
features of the Peg3 domain.
Collapse
Affiliation(s)
- Hongzhi He
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Joomyeong Kim
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
193
|
Eggermann T, Binder G, Brioude F, Maher ER, Lapunzina P, Cubellis MV, Bergadá I, Prawitt D, Begemann M. CDKN1C mutations: two sides of the same coin. Trends Mol Med 2014; 20:614-22. [PMID: 25262539 DOI: 10.1016/j.molmed.2014.09.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 08/13/2014] [Accepted: 09/02/2014] [Indexed: 01/03/2023]
Abstract
Cyclin-dependent kinase (CDK)-inhibitor 1C (CDKN1C) negatively regulates cellular proliferation and it has been shown that loss-of-function mutations in the imprinted CDKN1C gene (11p15.5) are associated with the overgrowth disorder Beckwith-Wiedemann syndrome (BWS). With recent reports of gain-of-function mutations of the PCNA domain of CDKN1C in growth-retarded patients with IMAGe syndrome or Silver-Russell syndrome (SRS), its key role for growth has been confirmed. Thereby, the last gap in the spectrum of molecular alterations in 11p15.5 in growth-retardation and overgrowth syndromes could be closed. Recent functional studies explain the strict association of CDKN1C mutations with clinically opposite phenotypes and thereby contribute to our understanding of the function and regulation of the gene in particular and epigenetic regulation in general.
Collapse
Affiliation(s)
- Thomas Eggermann
- Institute of Human Genetics, University Hospital, Technical University Aachen, Aachen, Germany.
| | - Gerhard Binder
- University Children's Hospital, Paediatric Endocrinology, University of Tübingen, Tübingen, Germany
| | - Frédéric Brioude
- AP-HP, Hôpital Armand Trousseau, Explorations Fonctionnelles Endocriniennes, Paris, France
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge, Cambridge, UK; NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Pablo Lapunzina
- INGEMM, Instituto de Genética Médica y Molecular, Hospital Universitario La Paz, IdiPAZ, CIBERER-ISCIII, Madrid, Spain
| | | | - Ignacio Bergadá
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Dirk Prawitt
- Molekulare Pädiatrie, Zentrum für Kinder- und Jugendmedizin, Universitätsmedizin Mainz, Mainz, Germany
| | - Matthias Begemann
- Institute of Human Genetics, University Hospital, Technical University Aachen, Aachen, Germany
| |
Collapse
|
194
|
El Hajj N, Schneider E, Lehnen H, Haaf T. Epigenetics and life-long consequences of an adverse nutritional and diabetic intrauterine environment. Reproduction 2014; 148:R111-20. [PMID: 25187623 PMCID: PMC4241689 DOI: 10.1530/rep-14-0334] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The phenomenon that adverse environmental exposures in early life are associated with increased susceptibilities for many adult, particularly metabolic diseases, is now referred to as ‘developmental origins of health and disease (DOHAD)’ or ‘Barker’ hypothesis. Fetal overnutrition and undernutrition have similar long-lasting effects on the setting of the neuroendocrine control systems, energy homeostasis, and metabolism, leading to life-long increased morbidity. There are sensitive time windows during early development, where environmental cues can program persistent epigenetic modifications which are generally assumed to mediate these gene–environment interactions. Most of our current knowledge on fetal programing comes from animal models and epidemiological studies in humans, in particular the Dutch famine birth cohort. In industrialized countries, there is more concern about adverse long-term consequences of fetal overnutrition, i.e. by exposure to gestational diabetes mellitus and/or maternal obesity which affect 10–20% of pregnancies. Epigenetic changes due to maternal diabetes/obesity may predispose the offspring to develop metabolic disease later in life and, thus, transmit the adverse environmental exposure to the next generation. This vicious cycle could contribute significantly to the worldwide metabolic disease epidemics. In this review article, we focus on the epigenetics of an adverse intrauterine environment, in particular gestational diabetes, and its implications for the prevention of complex disease.
Collapse
Affiliation(s)
- Nady El Hajj
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| | - Eberhard Schneider
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| | - Harald Lehnen
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| | - Thomas Haaf
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| |
Collapse
|
195
|
Prontera P, Donti E. Hypothesis: gonadal temperature influences sex-specific imprinting. Front Genet 2014; 5:294. [PMID: 25202325 PMCID: PMC4142806 DOI: 10.3389/fgene.2014.00294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 08/07/2014] [Indexed: 11/29/2022] Open
Abstract
Various explanations have been advanced for the evolution of genomic imprinting, the most popular of these being the parental conflict hypothesis. However, while this theory may explain why there has been selection for imprinting certain genes, it does not explain how the maternal and paternal genomes can be distinguished from each other. Here, we hypothesize that the temperature at which male and female gonads are physiologically exposed could be, at least for some loci, the primary factor leading to the different imprinting between the sexes.
Collapse
Affiliation(s)
- Paolo Prontera
- Medical Genetics Unit, Department of Surgical and Biomedical Sciences, University of Perugia Perugia, Italy
| | - Emilio Donti
- Medical Genetics Unit, Department of Surgical and Biomedical Sciences, University of Perugia Perugia, Italy
| |
Collapse
|
196
|
Labialle S, Marty V, Bortolin-Cavaillé ML, Hoareau-Osman M, Pradère JP, Valet P, Martin PGP, Cavaillé J. The miR-379/miR-410 cluster at the imprinted Dlk1-Dio3 domain controls neonatal metabolic adaptation. EMBO J 2014; 33:2216-30. [PMID: 25124681 DOI: 10.15252/embj.201387038] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In mammals, birth entails complex metabolic adjustments essential for neonatal survival. Using a mouse knockout model, we identify crucial biological roles for the miR-379/miR-410 cluster within the imprinted Dlk1-Dio3 region during this metabolic transition. The miR-379/miR-410 locus, also named C14MC in humans, is the largest known placental mammal-specific miRNA cluster, whose 39 miRNA genes are expressed only from the maternal allele. We found that heterozygote pups with a maternal--but not paternal--deletion of the miRNA cluster display partially penetrant neonatal lethality with defects in the maintenance of energy homeostasis. This maladaptive metabolic response is caused, at least in part, by profound changes in the activation of the neonatal hepatic gene expression program, pointing to as yet unidentified regulatory pathways that govern this crucial metabolic transition in the newborn's liver. Not only does our study highlight the physiological importance of miRNA genes that recently evolved in placental mammal lineages but it also unveils additional layers of RNA-mediated gene regulation at the Dlk1-Dio3 domain that impose parent-of-origin effects on metabolic control at birth and have likely contributed to mammal evolution.
Collapse
Affiliation(s)
- Stéphane Labialle
- Laboratoire de Biologie Moléculaire Eucaryote, UPS Université de Toulouse, Toulouse, France CNRS LBME, UMR5099, Toulouse, France
| | - Virginie Marty
- Laboratoire de Biologie Moléculaire Eucaryote, UPS Université de Toulouse, Toulouse, France CNRS LBME, UMR5099, Toulouse, France
| | - Marie-Line Bortolin-Cavaillé
- Laboratoire de Biologie Moléculaire Eucaryote, UPS Université de Toulouse, Toulouse, France CNRS LBME, UMR5099, Toulouse, France
| | - Magali Hoareau-Osman
- Laboratoire de Biologie Moléculaire Eucaryote, UPS Université de Toulouse, Toulouse, France CNRS LBME, UMR5099, Toulouse, France
| | - Jean-Philippe Pradère
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université de Toulouse Université Paul Sabatier, Toulouse, France
| | - Philippe Valet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Toulouse, France Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université de Toulouse Université Paul Sabatier, Toulouse, France
| | - Pascal G P Martin
- Laboratoire de Biologie Moléculaire Eucaryote, UPS Université de Toulouse, Toulouse, France CNRS LBME, UMR5099, Toulouse, France INRA UMR1331 TOXALIM (Research Centre in Food Toxicology), Toulouse, France Université de Toulouse INP UPS TOXALIM, Toulouse, France
| | - Jérôme Cavaillé
- Laboratoire de Biologie Moléculaire Eucaryote, UPS Université de Toulouse, Toulouse, France CNRS LBME, UMR5099, Toulouse, France
| |
Collapse
|
197
|
Wells JCK. Adaptive variability in the duration of critical windows of plasticity: Implications for the programming of obesity. EVOLUTION MEDICINE AND PUBLIC HEALTH 2014; 2014:109-21. [PMID: 25095791 PMCID: PMC4148720 DOI: 10.1093/emph/eou019] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Developmental plasticity underlies widespread associations between early-life exposures and many components of adult phenotype, including the risk of chronic diseases. Humans take almost two decades to reach reproductive maturity, and yet the ‘critical windows’ of physiological sensitivity that confer developmental plasticity tend to close during fetal life or infancy. While several explanations for lengthy human maturation have been offered, the brevity of physiological plasticity has received less attention. I argue that offspring plasticity is only viable within the niche of maternal care, and that as this protection is withdrawn, the offspring is obliged to canalize many developmental traits in order to minimize environmental disruptions. The schedule of maternal care may therefore shape the duration of critical windows, and since the duration of this care is subject to parent–offspring conflict, the resolution of this conflict may shape the duration of critical windows. This perspective may help understand (i) why windows close at different times for different traits, and (ii) why the duration of critical windows may vary across human populations. The issue is explored in relation to population differences in the association between infant weight gain and later body composition. The occupation of more stable environments by western populations may have favoured earlier closure of the critical window during which growth in lean mass is sensitive to nutritional intake. This may paradoxically have elevated the risk of obesity following rapid infant weight gain in such populations.
Collapse
Affiliation(s)
- Jonathan C K Wells
- Childhood Nutrition Research Centre, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
198
|
Jensen A, Tunster S, John R. The significance of elevated placental PHLDA2 in human growth restricted pregnancies. Placenta 2014; 35:528-32. [DOI: 10.1016/j.placenta.2014.04.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 04/23/2014] [Accepted: 04/28/2014] [Indexed: 01/03/2023]
|
199
|
Wolf JB, Brandvain Y. Gene interactions in the evolution of genomic imprinting. Heredity (Edinb) 2014; 113:129-37. [PMID: 24619179 PMCID: PMC4105456 DOI: 10.1038/hdy.2014.7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 12/02/2013] [Accepted: 12/05/2013] [Indexed: 12/24/2022] Open
Abstract
Numerous evolutionary theories have been developed to explain the epigenetic phenomenon of genomic imprinting. Here, we explore a subset of theories wherein non-additive genetic interactions can favour imprinting. In the simplest genic interaction--the case of underdominance--imprinting can be favoured to hide effectively low-fitness heterozygous genotypes; however, as there is no asymmetry between maternally and paternally inherited alleles in this model, other means of enforcing monoallelic expression may be more plausible evolutionary outcomes than genomic imprinting. By contrast, more successful interaction models of imprinting rely on an asymmetry between the maternally and paternally inherited alleles at a locus that favours the silencing of one allele as a means of coordinating the expression of high-fitness allelic combinations. For example, with interactions between autosomal loci, imprinting functionally preserves high-fitness genotypes that were favoured by selection in the previous generation. In this scenario, once a focal locus becomes imprinted, selection at interacting loci favours a matching imprint. Uniparental transmission generates similar asymmetries for sex chromosomes and cytoplasmic factors interacting with autosomal loci, with selection favouring the expression of either maternal or paternally derived autosomal alleles depending on the pattern of transmission of the uniparentally inherited factor. In a final class of models, asymmetries arise when genes expressed in offspring interact with genes expressed in one of its parents. Under such a scenario, a locus evolves to have imprinted expression in offspring to coordinate the interaction with its parent's genome. We illustrate these models and explore key links and differences using a unified framework.
Collapse
Affiliation(s)
- J B Wolf
- Department of Biology and Biochemistry, University of Bath, Bath, Claverton Down, UK
| | - Y Brandvain
- Department of Plant Biology, University of Minnesota, St Paul, MN, USA
| |
Collapse
|
200
|
Iglesias-Platas I, Martin-Trujillo A, Petazzi P, Guillaumet-Adkins A, Esteller M, Monk D. Altered expression of the imprinted transcription factor PLAGL1 deregulates a network of genes in the human IUGR placenta. Hum Mol Genet 2014; 23:6275-85. [PMID: 24993786 DOI: 10.1093/hmg/ddu347] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Genomic imprinting is the epigenetic process that results in monoallelic expression of genes depending on parental origin. These genes are known to be critical for placental development and fetal growth in mammals. Aberrant epigenetic profiles at imprinted loci, such as DNA methylation defects, are surprisingly rare in pregnancies with compromised fetal growth, while variations in transcriptional output from the expressed alleles of imprinted genes are more commonly reported in pregnancies complicated with intrauterine growth restriction (IUGR). To determine if PLAGL1 and HYMAI, two imprinted transcripts deregulated in Transient Neonatal Diabetes Mellitus, are involved in non-syndromic IUGR we compared the expression and DNA methylation levels in a large cohort of placental biopsies from IUGR and uneventful pregnancies. This revealed that despite appropriate maternal methylation at the shared PLAGL1/HYMAI promoter, there was a loss of correlation between PLAGL1 and HYMAI expression in IUGR. This incongruity was due to higher HYMAI expression in IUGR gestations, coupled with PLAGL1 down-regulation in placentas from IUGR girls, but not boys. The PLAGL1 protein is a zinc-finger transcription factor that has been shown to be a master coordinator of a genetic growth network in mice. We observe PLAGL1 binding to the H19/IGF2 shared enhancers in placentae, with significant correlations between PLAGL1 levels with H19 and IGF2 expression levels. In addition, PLAGL1 binding and expression also correlate with expression levels of metabolic regulator genes SLC2A4, TCF4 and PPARγ1. Our results strongly suggest that fetal growth can be influenced by altered expression of the PLAGL1 gene network in human placenta.
Collapse
Affiliation(s)
- Isabel Iglesias-Platas
- Servicio de Neonatología, Hospital Sant Joan de Déu, Fundació Sant Joan de Déu, Barcelona 08950, Spain,
| | | | - Paolo Petazzi
- Cancer Epigenetics Group, Cancer Epigenetic and Biology Program, Institut D'Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona 08907, Spain
| | - Amy Guillaumet-Adkins
- Servicio de Neonatología, Hospital Sant Joan de Déu, Fundació Sant Joan de Déu, Barcelona 08950, Spain, Imprinting and Cancer Group
| | - Manel Esteller
- Cancer Epigenetics Group, Cancer Epigenetic and Biology Program, Institut D'Investigació Biomedica de Bellvitge, Hospital Duran i Reynals, Barcelona 08907, Spain, Department of Physiological Sciences II, School of Medicine, University of Barcelona, Barcelona 08097, Spain and Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia 08010, Spain
| | | |
Collapse
|