Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nat Genet ; 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38969833

ABSTRACT

Many variants that we inherit from our parents or acquire de novo or somatically are rare, limiting the precision with which we can associate them with disease. We performed exhaustive saturation genome editing (SGE) of BAP1, the disruption of which is linked to tumorigenesis and altered neurodevelopment. We experimentally characterized 18,108 unique variants, of which 6,196 were found to have abnormal functions, and then used these data to evaluate phenotypic associations in the UK Biobank. We also characterized variants in a large population-ascertained tumor collection, in cancer pedigrees and ClinVar, and explored the behavior of cancer-associated variants compared to that of variants linked to neurodevelopmental phenotypes. Our analyses demonstrated that disruptive germline BAP1 variants were significantly associated with higher circulating levels of the mitogen IGF-1, suggesting a possible pathological mechanism and therapeutic target. Furthermore, we built a variant classifier with >98% sensitivity and specificity and quantify evidence strengths to aid precision variant interpretation.

2.
Nat Commun ; 14(1): 7702, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38057330

ABSTRACT

Loss-of-function of DDX3X is a leading cause of neurodevelopmental disorders (NDD) in females. DDX3X is also a somatically mutated cancer driver gene proposed to have tumour promoting and suppressing effects. We perform saturation genome editing of DDX3X, testing in vitro the functional impact of 12,776 nucleotide variants. We identify 3432 functionally abnormal variants, in three distinct classes. We train a machine learning classifier to identify functionally abnormal variants of NDD-relevance. This classifier has at least 97% sensitivity and 99% specificity to detect variants pathogenic for NDD, substantially out-performing in silico predictors, and resolving up to 93% of variants of uncertain significance. Moreover, functionally-abnormal variants can account for almost all of the excess nonsynonymous DDX3X somatic mutations seen in DDX3X-driven cancers. Systematic maps of variant effects generated in experimentally tractable cell types have the potential to transform clinical interpretation of both germline and somatic disease-associated variation.


Subject(s)
Neoplasms , Neurodevelopmental Disorders , Female , Humans , Gene Editing , Virulence , Neurodevelopmental Disorders/genetics , Neoplasms/genetics , Germ Cells , Germ-Line Mutation , DEAD-box RNA Helicases/genetics
3.
Bioinformatics ; 38(4): 892-899, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34791067

ABSTRACT

MOTIVATION: CRISPR/Cas9-based technology allows for the functional analysis of genetic variants at single nucleotide resolution whilst maintaining genomic context. This approach, known as saturation genome editing (SGE), a form of deep mutational scanning, systematically alters each position in a target region to explore its function. SGE experiments require the design and synthesis of oligonucleotide variant libraries which are introduced into the genome. This technology is applicable to diverse fields such as disease variant identification, drug development, structure-function studies, synthetic biology, evolutionary genetics and host-pathogen interactions. Here, we present the Variant Library Annotation Tool (VaLiAnT) which can be used to generate variant libraries from user-defined genomic coordinates and standard input files. The software can accommodate user-specified species, reference sequences and transcript annotations. RESULTS: Coordinates for a genomic range are provided by the user to retrieve a corresponding oligonucleotide reference sequence. A user-specified range within this sequence is then subject to systematic, nucleotide and/or amino acid saturating mutator functions. VaLiAnT provides a novel way to retrieve, mutate and annotate genomic sequences for oligonucleotide library generation. Specific features for SGE library generation can be employed. In addition, VaLiAnT is configurable, allowing for cDNA and prime editing saturation library generation, with other diverse applications possible. AVAILABILITY AND IMPLEMENTATION: VaLiAnT is a command line tool written in Python. Source code, testing data, example input and output files and executables are available (https://github.com/cancerit/VaLiAnT) in addition to a detailed user manual (https://github.com/cancerit/VaLiAnT/wiki). VaLiAnT is licensed under AGPLv3. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Gene Editing , Oligonucleotides , Genomics , Software , Genome
5.
Proc Natl Acad Sci U S A ; 116(20): 10103-10112, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31010925

ABSTRACT

Physical exercise has positive effects on cognition, but very little is known about the inheritance of these effects to sedentary offspring and the mechanisms involved. Here, we use a patrilineal design in mice to test the transmission of effects from the same father (before or after training) and from different fathers to compare sedentary- and runner-father progenies. Behavioral, stereological, and whole-genome sequence analyses reveal that paternal cognition improvement is inherited by the offspring, along with increased adult neurogenesis, greater mitochondrial citrate synthase activity, and modulation of the adult hippocampal gene expression profile. These results demonstrate the inheritance of exercise-induced cognition enhancement through the germline, pointing to paternal physical activity as a direct factor driving offspring's brain physiology and cognitive behavior.


Subject(s)
Brain/physiology , Cognition/physiology , Fathers/psychology , Paternal Inheritance , Running/physiology , Animals , Female , Gene Expression , Male , Mice , Pregnancy
6.
Science ; 362(6419): 1161-1164, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30409806

ABSTRACT

We estimated the genome-wide contribution of recessive coding variation in 6040 families from the Deciphering Developmental Disorders study. The proportion of cases attributable to recessive coding variants was 3.6% in patients of European ancestry, compared with 50% explained by de novo coding mutations. It was higher (31%) in patients with Pakistani ancestry, owing to elevated autozygosity. Half of this recessive burden is attributable to known genes. We identified two genes not previously associated with recessive developmental disorders, KDM5B and EIF3F, and functionally validated them with mouse and cellular models. Our results suggest that recessive coding variants account for a small fraction of currently undiagnosed nonconsanguineous individuals, and that the role of noncoding variants, incomplete penetrance, and polygenic mechanisms need further exploration.


Subject(s)
Developmental Disabilities/genetics , Genes, Recessive , Genetic Code , Genetic Variation , Penetrance , Animals , Disease Models, Animal , Eukaryotic Initiation Factor-3/genetics , Europe , Genome-Wide Association Study , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Nuclear Proteins/genetics , Pakistan , Phylogeny , Repressor Proteins/genetics
7.
Nature ; 562(7726): 268-271, 2018 10.
Article in English | MEDLINE | ID: mdl-30258228

ABSTRACT

There are thousands of rare human disorders that are caused by single deleterious, protein-coding genetic variants1. However, patients with the same genetic defect can have different clinical presentations2-4, and some individuals who carry known disease-causing variants can appear unaffected5. Here, to understand what explains these differences, we study a cohort of 6,987 children assessed by clinical geneticists to have severe neurodevelopmental disorders such as global developmental delay and autism, often in combination with abnormalities of other organ systems. Although the genetic causes of these neurodevelopmental disorders are expected to be almost entirely monogenic, we show that 7.7% of variance in risk is attributable to inherited common genetic variation. We replicated this genome-wide common variant burden by showing, in an independent sample of 728 trios (comprising a child plus both parents) from the same cohort, that this burden is over-transmitted from parents to children with neurodevelopmental disorders. Our common-variant signal is significantly positively correlated with genetic predisposition to lower educational attainment, decreased intelligence and risk of schizophrenia. We found that common-variant risk was not significantly different between individuals with and without a known protein-coding diagnostic variant, which suggests that common-variant risk affects patients both with and without a monogenic diagnosis. In addition, previously published common-variant scores for autism, height, birth weight and intracranial volume were all correlated with these traits within our cohort, which suggests that phenotypic expression in individuals with monogenic disorders is affected by the same variants as in the general population. Our results demonstrate that common genetic variation affects both overall risk and clinical presentation in neurodevelopmental disorders that are typically considered to be monogenic.


Subject(s)
Genetic Predisposition to Disease , Genetic Variation , Neurodevelopmental Disorders/genetics , Rare Diseases/genetics , Autistic Disorder/genetics , Birth Weight/genetics , Body Height/genetics , Case-Control Studies , Cohort Studies , Developmental Disabilities/genetics , Female , Genome-Wide Association Study , Humans , Intelligence/genetics , Linkage Disequilibrium , Male , Multifactorial Inheritance/genetics , Phenotype , Schizophrenia/genetics
8.
Prog Mol Biol Transl Sci ; 158: 29-48, 2018.
Article in English | MEDLINE | ID: mdl-30072058

ABSTRACT

The astonishing array of cellular phenotypes required to make a complex organism such as a human is generated from an identical genetic sequence in the nucleus of each cell. The central nervous system is a highly ordered, complex system composed of multiple neuronal and glial cell types. Both neurons and glia are derived from neural stem/precursor cells (NPCs) in a temporally and spatially patterned process of cellular division and differentiation, migration, maturation and the establishment of neuronal connectivity. In part, this is testament to the power and flexibility of epigenetic mechanisms to direct transcription, producing a myriad of outcomes. The progressive restriction of developmental potential involves the acquisition of a hierarchy of epigenetic regulation which restricts and directs the developmental program of gene expression. Differentiation into each cell type is associated with the acquisition of a distinct epigenetic profile. The importance of epigenetic mechanisms for neurodevelopment is exemplified by the many neurodevelopmental disorders caused by the genetic disruption of epigenetic readers, writers or modifiers. In this chapter I will introduce the different classes of epigenetic mechanisms, and I will briefly discuss some of the known neurodevelopmental disorders caused by genetic disruption of these pathways to illustrate the importance of epigenetic processes in brain development and function.


Subject(s)
Epigenesis, Genetic , Animals , Cellular Reprogramming/genetics , DNA Methylation/genetics , Genomic Imprinting , Histones/metabolism , Humans , Inheritance Patterns/genetics
9.
Prog Mol Biol Transl Sci ; 158: 299-323, 2018.
Article in English | MEDLINE | ID: mdl-30072059

ABSTRACT

The development of mental disorders constitutes a complex phenomenon driven by unique social, psychological and biological factors such as genetics and epigenetics, throughout an individual's life course. Both environmental and genetic factors have an impact on mental health phenotypes and act simultaneously to induce changes in brain and behavior. Here, we describe and critically evaluate the current literature on gene-environment interactions and epigenetics on mental health by highlighting recent human and animal studies. We furthermore review some of the main ethical and social implications concerning gene-environment interactions and epigenetics and provide explanations and suggestions on how to move from statistical and epigenetic associations to biological and psychological explanations within a multi-disciplinary and integrative approach of understanding mental health.


Subject(s)
Epigenesis, Genetic , Mental Disorders/genetics , Mental Disorders/psychology , Mental Health , Animals , Environmental Exposure , Gene-Environment Interaction , Humans , Interdisciplinary Research
10.
Nat Rev Endocrinol ; 14(6): 345-355, 2018 06.
Article in English | MEDLINE | ID: mdl-29666451

ABSTRACT

Environmental factors, particularly during early life, are important for the later metabolic health of the individual. In our obesogenic environment, it is of major socio-economic importance to investigate the mechanisms that contribute to the risk of metabolic ill health. Increasing evidence from a variety of model organisms suggests that non-genetically determined phenotypes, including metabolic effects such as glucose intolerance and obesity, can be passed between generations, which encourages us to revisit heredity. Inheritance of altered epigenetic information through the germ line has been proposed as one plausible mechanism. Whether the germline epigenome can be altered by environmental conditions such as diet and the extent to which this occurs in humans are the subject of intense current interest and debate, especially given that extensive germline epigenetic reprogramming is known to occur. As epigenetic mechanisms are often highly conserved between organisms, studying epigenetic inheritance in plants and lower metazoans has the potential to inform our investigation in mammals. This Review explores the extent to which epigenetic inheritance contributes to heredity in these different organisms, whether the environment can affect epigenetic inheritance and whether there is any evidence for the inheritance of acquired phenotypes.


Subject(s)
Genetic Diseases, Inborn/physiopathology , Genetic Predisposition to Disease/epidemiology , Heredity/genetics , Phenotype , Animals , Epigenesis, Genetic , Female , Genetic Diseases, Inborn/epidemiology , Germ Cells/metabolism , Humans , Male , Mammals/genetics , Mendelian Randomization Analysis/methods , Mice , Plants/genetics , Species Specificity
11.
FASEB J ; 31(11): 4928-4934, 2017 11.
Article in English | MEDLINE | ID: mdl-28778976

ABSTRACT

The prenatal environment can alter an individual's developmental trajectory with long-lasting effects on health. Animal models demonstrate that the impact of the early life environment extends to subsequent generations, but there is a paucity of data from human populations on intergenerational transmission of environmentally induced phenotypes. Here we investigated the association of parental exposure to energy and nutrient restriction in utero on their children's growth in rural Gambia. In a Gambian cohort with infants born between 1972 and 2011, we used multiple regression to test whether parental season of birth predicted offspring birth weight (n = 2097) or length (n = 1172), height-for-age z score (HAZ), weight-for-height z score (WHZ), and weight-for-age z score (WAZ) at 2 yr of age (n = 923). We found that maternal exposure to seasonal energy restriction in utero was associated with reduced offspring birth length (crude:-4.2 mm, P = 0.005; adjusted: -4.0 mm, P = 0.02). In contrast, paternal birth season predicted offspring HAZ at 24 mo (crude: -0.21, P = 0.005; adjusted: -0.22, P = 0.004) but had no discernible impact at birth. Our results indicate that periods of nutritional restriction in a parent's fetal life can have intergenerational consequences in human populations. Fetal growth appears to be under matriline influence, and postnatal growth appears to be under patriline intergenerational influences.-Eriksen, K. G., Radford, E. J., Silver, M. J., Fulford, A. J. C., Wegmüller, R., Prentice, A. M. Influence of intergenerational in utero parental energy and nutrient restriction on offspring growth in rural Gambia.


Subject(s)
Birth Weight , Caloric Restriction , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects/epidemiology , Prenatal Exposure Delayed Effects/physiopathology , Female , Gambia/epidemiology , Humans , Male , Pregnancy , Prenatal Exposure Delayed Effects/pathology
12.
PLoS Genet ; 12(3): e1005898, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26938916

ABSTRACT

BAHD1 is a vertebrate protein that promotes heterochromatin formation and gene repression in association with several epigenetic regulators. However, its physiological roles remain unknown. Here, we demonstrate that ablation of the Bahd1 gene results in hypocholesterolemia, hypoglycemia and decreased body fat in mice. It also causes placental growth restriction with a drop of trophoblast glycogen cells, a reduction of fetal weight and a high neonatal mortality rate. By intersecting transcriptome data from murine Bahd1 knockout (KO) placentas at stages E16.5 and E18.5 of gestation, Bahd1-KO embryonic fibroblasts, and human cells stably expressing BAHD1, we also show that changes in BAHD1 levels alter expression of steroid/lipid metabolism genes. Biochemical analysis of the BAHD1-associated multiprotein complex identifies MIER proteins as novel partners of BAHD1 and suggests that BAHD1-MIER interaction forms a hub for histone deacetylases and methyltransferases, chromatin readers and transcription factors. We further show that overexpression of BAHD1 leads to an increase of MIER1 enrichment on the inactive X chromosome (Xi). In addition, BAHD1 and MIER1/3 repress expression of the steroid hormone receptor genes ESR1 and PGR, both playing important roles in placental development and energy metabolism. Moreover, modulation of BAHD1 expression in HEK293 cells triggers epigenetic changes at the ESR1 locus. Together, these results identify BAHD1 as a core component of a chromatin-repressive complex regulating placental morphogenesis and body fat storage and suggest that its dysfunction may contribute to several human diseases.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Nuclear Proteins/genetics , Placentation/genetics , Steroids/metabolism , Transcription Factors/genetics , Animals , Chromatin/genetics , Chromosomal Proteins, Non-Histone/biosynthesis , DNA-Binding Proteins , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Mice , Mice, Knockout , Nuclear Proteins/biosynthesis , Placenta/metabolism , Pregnancy , Transcription Factors/biosynthesis , Transcriptome/genetics
13.
Proc Natl Acad Sci U S A ; 111(45): 16088-93, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25349437

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is associated with insulin resistance and obesity, as well as progressive liver dysfunction. Recent animal studies have underscored the importance of hepatic growth hormone (GH) signaling in the development of NAFLD. The imprinted Delta-like homolog 1 (Dlk1)/preadipocyte factor 1 (Pref1) gene encodes a complex protein producing both circulating and membrane-tethered isoforms whose expression dosage is functionally important because even modest elevation during embryogenesis causes lethality. DLK1 is up-regulated during embryogenesis, during suckling, and in the mother during pregnancy. We investigated the normal role for elevated DLK1 dosage by overexpressing Dlk1 from endogenous control elements. This increased DLK1 dosage caused improved glucose tolerance with no primary defect in adipose tissue expansion even under extreme metabolic stress. Rather, Dlk1 overexpression caused reduced fat stores, pituitary insulin-like growth factor 1 (IGF1) resistance, and a defect in feedback regulation of GH. Increased circulatory GH culminated in a switch in whole body fuel metabolism and a reduction in hepatic steatosis. We propose that the function of DLK1 is to shift the metabolic mode of the organism toward peripheral lipid oxidation and away from lipid storage, thus mediating important physiological adaptations associated with early life and with implications for metabolic disease resistance.


Subject(s)
Embryonic Development , Fatty Liver/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism , Animals , Calcium-Binding Proteins , Fatty Liver/genetics , Fatty Liver/pathology , Fatty Liver/prevention & control , Female , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Pregnancy
14.
Science ; 345(6198): 1255903, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25011554

ABSTRACT

Adverse prenatal environments can promote metabolic disease in offspring and subsequent generations. Animal models and epidemiological data implicate epigenetic inheritance, but the mechanisms remain unknown. In an intergenerational developmental programming model affecting F2 mouse metabolism, we demonstrate that the in utero nutritional environment of F1 embryos alters the germline DNA methylome of F1 adult males in a locus-specific manner. Differentially methylated regions are hypomethylated and enriched in nucleosome-retaining regions. A substantial fraction is resistant to early embryo methylation reprogramming, which may have an impact on F2 development. Differential methylation is not maintained in F2 tissues, yet locus-specific expression is perturbed. Thus, in utero nutritional exposures during critical windows of germ cell development can impact the male germline methylome, associated with metabolic disease in offspring.


Subject(s)
DNA Methylation , Fetal Nutrition Disorders/metabolism , Prenatal Exposure Delayed Effects , Spermatozoa/metabolism , Animals , Caloric Restriction , Epigenesis, Genetic , Female , Fetal Nutrition Disorders/genetics , Insulin/metabolism , Insulin Secretion , Male , Metabolic Diseases/metabolism , Mice , Mice, Inbred ICR , Nucleosomes/metabolism , Pregnancy , Spermatozoa/physiology
15.
PLoS Genet ; 8(4): e1002605, 2012.
Article in English | MEDLINE | ID: mdl-22511876

ABSTRACT

Environmental factors during early life are critical for the later metabolic health of the individual and of future progeny. In our obesogenic environment, it is of great socioeconomic importance to investigate the mechanisms that contribute to the risk of metabolic ill health. Imprinted genes, a class of functionally mono-allelic genes critical for early growth and metabolic axis development, have been proposed to be uniquely susceptible to environmental change. Furthermore, it has also been suggested that perturbation of the epigenetic reprogramming of imprinting control regions (ICRs) may play a role in phenotypic heritability following early life insults. Alternatively, the presence of multiple layers of epigenetic regulation may in fact protect imprinted genes from such perturbation. Unbiased investigation of these alternative hypotheses requires assessment of imprinted gene expression in the context of the response of the whole transcriptome to environmental assault. We therefore analyse the role of imprinted genes in multiple tissues in two affected generations of an established murine model of the developmental origins of health and disease using microarrays and quantitative RT-PCR. We demonstrate that, despite the functional mono-allelicism of imprinted genes and their unique mechanisms of epigenetic dosage control, imprinted genes as a class are neither more susceptible nor protected from expression perturbation induced by maternal undernutrition in either the F1 or the F2 generation compared to other genes. Nor do we find any evidence that the epigenetic reprogramming of ICRs in the germline is susceptible to nutritional restriction. However, we propose that those imprinted genes that are affected may play important roles in the foetal response to undernutrition and potentially its long-term sequelae. We suggest that recently described instances of dosage regulation by relaxation of imprinting are rare and likely to be highly regulated.


Subject(s)
Gene Expression Regulation, Developmental , Gene-Environment Interaction , Genomic Imprinting , Malnutrition , Animals , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Humans , Liver/growth & development , Liver/metabolism , Male , Malnutrition/genetics , Malnutrition/metabolism , Mice , Placenta/metabolism , Placentation , Pregnancy
16.
FASEB J ; 26(4): 1431-41, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22186874

ABSTRACT

Ascent to high altitude is associated with a fall in the partial pressure of inspired oxygen (hypobaric hypoxia). For oxidative tissues such as skeletal muscle, resultant cellular hypoxia necessitates acclimatization to optimize energy metabolism and restrict oxidative stress, with changes in gene and protein expression that alter mitochondrial function. It is known that lowlanders returning from high altitude have decreased muscle mitochondrial densities, yet the underlying transcriptional mechanisms and time course are poorly understood. To explore these, we measured gene and protein expression plus ultrastructure in muscle biopsies of lowlanders at sea level and following exposure to hypobaric hypoxia. Subacute exposure (19 d after initiating ascent to Everest base camp, 5300 m) was not associated with mitochondrial loss. After 66 d at altitude and ascent beyond 6400 m, mitochondrial densities fell by 21%, with loss of 73% of subsarcolemmal mitochondria. Correspondingly, levels of the transcriptional coactivator PGC-1α fell by 35%, suggesting down-regulation of mitochondrial biogenesis. Sustained hypoxia also decreased expression of electron transport chain complexes I and IV and UCP3 levels. We suggest that during subacute hypoxia, mitochondria might be protected from oxidative stress. However, following sustained exposure, mitochondrial biogenesis is deactivated and uncoupling down-regulated, perhaps to improve the efficiency of ATP production.


Subject(s)
Acclimatization/physiology , Altitude , Hypoxia/metabolism , Hypoxia/physiopathology , Mitochondria, Muscle/metabolism , Mountaineering/physiology , Muscle, Skeletal/physiology , Adult , Biopsy , Energy Metabolism/physiology , Gene Expression , Humans , Male , Middle Aged , Mitochondria, Muscle/ultrastructure , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/ultrastructure , RNA/metabolism , Transcription, Genetic
17.
Nature ; 475(7356): 381-5, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21776083

ABSTRACT

The gene for the atypical NOTCH ligand delta-like homologue 1 (Dlk1) encodes membrane-bound and secreted isoforms that function in several developmental processes in vitro and in vivo. Dlk1, a member of a cluster of imprinted genes, is expressed from the paternally inherited chromosome. Here we show that mice that are deficient in Dlk1 have defects in postnatal neurogenesis in the subventricular zone: a developmental continuum that results in depletion of mature neurons in the olfactory bulb. We show that DLK1 is secreted by niche astrocytes, whereas its membrane-bound isoform is present in neural stem cells (NSCs) and is required for the inductive effect of secreted DLK1 on self-renewal. Notably, we find that there is a requirement for Dlk1 to be expressed from both maternally and paternally inherited chromosomes. Selective absence of Dlk1 imprinting in both NSCs and niche astrocytes is associated with postnatal acquisition of DNA methylation at the germ-line-derived imprinting control region. The results emphasize molecular relationships between NSCs and the niche astrocyte cells of the microenvironment, identifying a signalling system encoded by a single gene that functions coordinately in both cell types. The modulation of genomic imprinting in a stem-cell environment adds a new level of epigenetic regulation to the establishment and maintenance of the niche, raising wider questions about the adaptability, function and evolution of imprinting in specific developmental contexts.


Subject(s)
Animals, Newborn/metabolism , Astrocytes/metabolism , Genomic Imprinting , Intercellular Signaling Peptides and Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Stem Cell Niche/cytology , Aging/genetics , Animals , Base Sequence , Calcium-Binding Proteins , Cell Membrane/metabolism , Cells, Cultured , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Genotype , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Olfactory Bulb/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Stem Cell Niche/metabolism
18.
FEBS Lett ; 585(13): 2059-66, 2011 Jul 07.
Article in English | MEDLINE | ID: mdl-21672541

ABSTRACT

Developmental plasticity can be defined as the ability of one genotype to produce a range of phenotypes in response to environmental conditions. Such plasticity can be manifest at the level of individual cells, an organ, or a whole organism. Imprinted genes are a group of approximately 100 genes with functionally monoallelic, parental-origin specific expression. As imprinted genes are critical for prenatal growth and metabolic axis development and function, modulation of imprinted gene dosage has been proposed to play a key role in the plastic development of the unborn foetus in response to environmental conditions. Evidence is accumulating that imprinted dosage may also be involved in controlling the plastic potential of individual cells or stem cell populations. Imprinted gene dosage can be modulated through canonical, transcription factor mediated mechanisms, or through the relaxation of imprinting itself, reactivating the normally silent allele.


Subject(s)
Genomic Imprinting/genetics , Animals , DNA Methylation/genetics , DNA Methylation/physiology , Epigenomics , Genomic Imprinting/physiology , Humans , Models, Biological
19.
Dev Biol ; 337(1): 1-8, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19833122

ABSTRACT

The control of foetal growth is poorly understood and yet it is critically important that at birth the body has attained appropriate size and proportions. Growth and survival of the mammalian foetus is dependent upon a functional placenta throughout most of gestation. A few genes are known that influence both foetal and placental growth and might therefore coordinate growth of the conceptus, including the imprinted Igf2 and Grb10 genes. Grb10 encodes a signalling adapter protein, is expressed predominantly from the maternally-inherited allele and acts to restrict foetal and placental growth. Here, we show that following disruption of the maternal allele in mice, the labyrinthine volume was increased in a manner consistent with a cell-autonomous function of Grb10 and the enlarged placenta was more efficient in supporting foetal growth. Thus, Grb10 is the first example of a gene that acts to limit placental size and efficiency. In addition, we found that females inheriting a mutant Grb10 allele from their mother had larger litters and smaller offspring than those inheriting a mutant allele from their father. This grandparental effect suggests Grb10 can influence reproductive strategy through the allocation of maternal resources such that offspring number is offset against size.


Subject(s)
GRB10 Adaptor Protein/physiology , Placenta/physiology , Alleles , Animals , Endothelium/metabolism , Female , GRB10 Adaptor Protein/analysis , GRB10 Adaptor Protein/genetics , Genomic Imprinting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Placenta/pathology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...