Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 204
Filter
1.
J Dev Orig Health Dis ; 4(2): 134-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-25054679

ABSTRACT

Genetic variants in the FTO (fat mass- and obesity-associated) gene have the highest association of all obesity-associated genes. Its placental expression was shown to relate to birth weight, suggesting that it may participate in the control of fetal weight gain. To gain more insight into the implication of FTO in fetal growth, we measured its placental expression in samples including extremes of abnormal fetal growth, such as after intrauterine growth restriction (IUGR) or macrosomia in both rats and humans. In rats, fetal growth was modulated by maternal nutritional modifications. In humans, placental villi were collected from pathological pregnancies (i.e. with IUGR or fetal macrosomia). Placental FTO mRNA expression was reduced by IUGR but was not significantly affected by macrosomia in either rats or humans. Our data suggest that placental FTO may participate in interactions between the in utero environment and the control of fetal growth under IUGR conditions by modulating epigenetic processes.

2.
Theriogenology ; 78(7): 1405-14, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22925651

ABSTRACT

Fetal programming of metabolic diseases is now a well established concept. The scope of the Developmental Origins of Health and Disease has, however, widened and led to the identification of new targets of fetal programming, notably effects on reproductive function. Epidemiologic studies about maternal nutrition and effects on offspring's fertility are rare, but a link between impaired fetal growth, possibly caused by maternal malnutrition, and reproductive function, has been established. The methodologic limitations inherent to human epidemiologic studies can be complemented through the use of animal models, which enable experimental studies on maternal environment and its effect on reproductive functions of the offspring. Altogether, an interaction between inappropriate maternal nutrition (excess or reduced nutritional intake, micronutrient unbalance, or alcohol intake) and reproductive maturation of the offspring has been shown in a majority of experiments as summarized in this review. The exact processes through which maternal nutrition or maternal environment affect reproductive function in the offspring remain unclear but epigenetic modifications are a clear link. Further studies are needed to better understand the mechanisms involved, identify the crucial critical periods, and prevent or treat the adverse effects.


Subject(s)
Maternal Nutritional Physiological Phenomena/physiology , Prenatal Exposure Delayed Effects/physiopathology , Reproduction/physiology , Alcohol Drinking/adverse effects , Animals , Environment , Epigenesis, Genetic/physiology , Female , Fetal Growth Retardation , Humans , Infertility , Male , Malnutrition/complications , Ovary/embryology , Ovary/physiology , Overnutrition/complications , Pregnancy , Pregnancy Complications/physiopathology , Testis/embryology , Testis/physiology
3.
Rev Med Liege ; 67(5-6): 250-7, 2012.
Article in French | MEDLINE | ID: mdl-22891475

ABSTRACT

Epigenetics is the study of heritable changes in gene expression or cellular phenotype caused by mechanisms other than changes in the underlying DNA sequence. Epigenetics is one of the major mechanisms explaining the "Developmental Origin of Health and Diseases" (DOHaD). Besides genetic background inherited from parents, which confers susceptibility to certain pathologies, epigenetic changes constitute the memory of previous events, either positive or negative, along the life cycle, including at the in utero stage. The later exposition to hostile environment may reveal such susceptibility, with the development of various pathologies, among them numerous chronic complex diseases. The demonstration of such a sequence of events has been shown for metabolic diseases as obesity, metabolic syndrome and type 2 diabetes, cardiovascular disease and cancer. In contrast to genetic predisposition, which is irreversible, epigenetic changes are potentially reversible, thus giving targets not only for prevention, but possibly also for the treatment of certain complex diseases.


Subject(s)
Disease/etiology , Epigenesis, Genetic/physiology , Gene-Environment Interaction , Genes/physiology , Animals , Disease/genetics , Environment , Genetic Markers/physiology , Humans , Models, Biological
4.
Endocr Rev ; 32(2): 159-224, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20971919

ABSTRACT

Plasticity in developmental programming has evolved in order to provide the best chances of survival and reproductive success to the organism under changing environments. Environmental conditions that are experienced in early life can profoundly influence human biology and long-term health. Developmental origins of health and disease and life-history transitions are purported to use placental, nutritional, and endocrine cues for setting long-term biological, mental, and behavioral strategies in response to local ecological and/or social conditions. The window of developmental plasticity extends from preconception to early childhood and involves epigenetic responses to environmental changes, which exert their effects during life-history phase transitions. These epigenetic responses influence development, cell- and tissue-specific gene expression, and sexual dimorphism, and, in exceptional cases, could be transmitted transgenerationally. Translational epigenetic research in child health is a reiterative process that ranges from research in the basic sciences, preclinical research, and pediatric clinical research. Identifying the epigenetic consequences of fetal programming creates potential applications in clinical practice: the development of epigenetic biomarkers for early diagnosis of disease, the ability to identify susceptible individuals at risk for adult diseases, and the development of novel preventive and curative measures that are based on diet and/or novel epigenetic drugs.


Subject(s)
Child Development/physiology , Child Welfare , Epigenesis, Genetic/physiology , Adolescent , Aging/physiology , Child , Child, Preschool , Environment , Female , Genomic Imprinting/physiology , Humans , Infant , Infant, Newborn , Male , Sex Differentiation/physiology
5.
Placenta ; 31(9): 785-91, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20615547

ABSTRACT

The brain-derived neurotrophic factor (BDNF) has been shown to exert an important role during implantation, placental development, and fetal growth control in mice. Its expression is closely related to the nutritional status in several tissues such as in the nervous system. In a previous study, we demonstrated that maternal undernutrition (MU), during the perinatal life, modified both the BDNF and its functional receptor, the tyrosine kinase receptor B (TrkB) gene expression in the brain of growth-restricted rat offspring during sensitive developmental windows, suggesting that these early modifications may have long-lasting consequences. In the present study, we measured BDNF/TrkB mRNA and protein levels in rat placentas from mothers submitted to a 50% food restriction during gestation, and in human placentas from pregnancies with fetal growth restriction or fetal macrosomia. In the rat, two subtypes of placental TrkB receptors have been identified: the TrkB-FL and TrkB-T1 receptors. We found that MU induced intrauterine growth restriction (IUGR) of fetuses at term and decreased the placental BDNF mRNA and protein levels. Placentae from undernourished mothers exhibited an increased mRNA expression of TrkB-FL whereas both TrkB-FL and TrkB-T1 receptors proteins levels were not modified. In human IUGR placentas, both BDNF and TrkB receptor mRNA expressions were up-regulated. Finally, although neither BDNF nor TrkB mRNA levels were altered by fetal macrosomia alone, BDNF mRNA levels were decreased when macrosomia was associated with maternal type 1 diabetes. These results show that the placental BDNF/TrkB system is modulated in rats and humans during pregnancies with fetal growth perturbations and is affected by the maternal energetic status. These data suggest that this system may exert an important role for the feto-placental unit development and that it may also be implicated in the etiology of pathologies related to placental and fetal growth disturbances.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Fetal Growth Retardation/metabolism , Receptor, trkB/genetics , Animals , Female , Fetal Macrosomia/metabolism , Humans , Malnutrition/physiopathology , Maternal Nutritional Physiological Phenomena/physiology , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Wistar , Signal Transduction
6.
Proc Nutr Soc ; 69(2): 221-31, 2010 May.
Article in English | MEDLINE | ID: mdl-20202279

ABSTRACT

The phenotype of an individual is the result of complex interactions between genome, epigenome and current, past and ancestral environment leading to a lifelong remodelling of the epigenomes. The genetic information expression contained in the genome is controlled by labile chromatin-associated epigenetic marks. Epigenetic misprogramming during development is widely thought to have a persistent effect on the health of the offspring and may even be transmitted to the next generation. The epigenome serves as an interface between the environment and the genome. Dietary factors, including folate involved in C1 metabolism, and other social and lifestyle exposures have a profound effect on many aspects of health including ageing and do so, at least partly, through interactions with the genome, which result in altered gene expression with consequences for cell function and health throughout the life course. Depending on the nature and intensity of the environmental insult, the critical spatiotemporal windows and developmental or lifelong processes involved, epigenetic alterations can lead to permanent changes in tissue and organ structure and function or to phenotypic changes that can (or cannot) be reversed using appropriate epigenetic tools. Moreover, the flexibility of epigenetic marks may make it possible for environmental, nutritional and hormonal factors or endocrine disruptors to alter, during a particular spatiotemporal window in a sex-specific manner, the sex-specific methylation or demethylation of specific CpG and/or histone modifications underlying sex-specific expression of a substantial proportion of genes. Moreover, genetic factors, the environment and stochastic events change the epigenetic landscape during the lifetime of an individual. Epigenetic alterations leading to gene expression dysregulation accumulate during ageing and are important in tumorigenesis and age-related diseases. Several encouraging trials suggest that prevention and therapy of age- and lifestyle-related diseases by individualised tailoring to optimal epigenetic diets or drugs are conceivable. However, these interventions will require intense efforts to unravel the complexity of these epigenetic, genetic and environment interactions and to evaluate their potential reversibility with minimal side effects.


Subject(s)
Diet , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Metabolic Diseases/genetics , Nutrigenomics , Animals , Female , Humans , Male , Mice , Pregnancy , Rats
7.
Obes Rev ; 8(6): 487-502, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17949354

ABSTRACT

Now that analysis of the organization of the human genome sequence is reaching completion, studies of the finely tuned chromatin epigenetic networks, DNA methylation and histone modifications, are required to determine how the same DNA sequence generates different cells, lineages and organs, i.e. the phenotype. Maternal nutrition, behaviour and metabolic disturbances as well as other environmental factors have been shown to have major effects on these epigenetic processes, potentially affecting the predisposition of offspring to obesity and related adult disorders. The March 2006 Stock Conference considered the latest evidence from studies in the field of obesity and other related areas that elucidate mechanisms by which the environment can modify gene expression and the resulting individual phenotype. Presentations included evaluation of the molecular basis of epigenetic memory and the nature of relevant sequence targets, windows of susceptibility, and maternal dietary and behavioural factors that determine epigenetic changes. Imprinted genes, age and tissue-related exposures, transgenerational and potential interventions were also discussed. In summary, it is clear that epigenetic alterations can no longer be ignored in evaluations of the causes of obesity and its associated disorders. There is a need for systematic large-scale epigenetic studies of obesity, employing appropriate strategies and techniques and appropriately chosen environmental factors in critical spatio-temporal windows.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Epigenesis, Genetic , Maternal Nutritional Physiological Phenomena/physiology , Metabolic Syndrome/genetics , Obesity/genetics , Diabetes Mellitus, Type 2/epidemiology , Environment , Female , Fetal Development , Genetic Predisposition to Disease , Genomics , Genotype , Humans , Male , Metabolic Syndrome/epidemiology , Obesity/epidemiology , Pregnancy
8.
Rheumatology (Oxford) ; 46(4): 622-5, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17166870

ABSTRACT

UNLABELLED: Systemic sclerosis (SSc) is a connective tissue disorder characterized by early generalized microangiopathy with disturbed angiogenesis. Endoglin gene (ENG) encodes a transmembrane glycoprotein which acts as an accessory receptor for the transforming growth factor-beta (TGF-beta) superfamily, and is crucial for maintaining vascular integrity. A 6-base insertion in intron 7 (6bINS) of ENG has been reported to be associated with microvascular disturbance. OBJECTIVES: Our objective was to investigate the relationship between 6bINS and the vascular complication pulmonary arterial hypertension (PAH) in SSc in a French Caucasian population. METHODS: Two hundred eighty SSc cases containing 29/280 having PAH diagnosed by catheterism were compared with 140 patients with osteoarthritis. Genotyping was performed by polymerase-chain-reaction-based fluorescence and direct sequencing of genomic DNA. RESULTS: The polymorphism was in Hardy-Weinberg equilibrium. We observed a significant lower frequency of 6bINS allele in SSc patients with associated PAH compared with controls [10.3 vs 23.9%, P = 0.01; odds ratio (OR) 0.37, 95% confidence interval (CI) 0.15-0.89], and a trend in comparison with SSc patients without PAH (10.3 vs 20.3%, P = 0.05; OR: 0.45, 95% CI: 0.19-1.08). Genotypes carrying allele 6bINS were also less frequent in SSc patients with PAH than in controls (20.7 vs 42.9%, P = 0.02). CONCLUSIONS: Thus the frequency of 6bINS differs between SSc patients with or without PAH, suggesting the implication of ENG in this devastating vascular complication of SSc.


Subject(s)
Antigens, CD/genetics , Hypertension, Pulmonary/genetics , Polymorphism, Genetic , Receptors, Cell Surface/genetics , Scleroderma, Systemic/genetics , Adult , Aged , Aged, 80 and over , Base Sequence , Endoglin , Female , Gene Frequency , Genotype , Humans , Hypertension, Pulmonary/etiology , Male , Middle Aged , Molecular Sequence Data , Scleroderma, Systemic/complications
9.
Cytogenet Genome Res ; 113(1-4): 253-61, 2006.
Article in English | MEDLINE | ID: mdl-16575188

ABSTRACT

Genomic imprinting is an epigenetic phenomenon unique to mammals that causes some genes to be expressed according to their parental origin. It results in developmental asymmetry in the function of the parental genomes. We describe here a method for the profiling of imprinted genes based on the development of a mouse imprinting microchip containing oligonucleotides corresponding to 493 genes, including most of the known imprinted genes (IG = 63), genes involved in epigenetic processes (EPI = 15), in metabolism (= 147), in obesity (= 10) and in neurotransmission (= 256) and housekeeping reference genes (= 2). This custom oligonucleotide microarray has been constructed to make data analysis and handling more manageable than pangenomic microarrays. As a proof of concept we present the differential expression of these 493 genes in different tissues (liver, placenta, embryo) of C57BL6/J mice fed different diets. Appropriate experimental strategies and statistical tools were defined at each step of the data analysis process with regard to the different sources of constraints. Data were confirmed by expression analyses based on quantitative real-time PCR. These oligochips should make it possible to increase our understanding of the involvement of imprinted genes in the timing of expression programs, tissue by tissue, stage by stage, in response to nutrients, lifestyles and other as yet unknown critical environmental factors in a variety of physiopathological situations, and in animals of different strains, ages and sexes. The use of oligonucleotides makes it possible to expand this microchip to include the increasing number of imprinted genes discovered.


Subject(s)
Gene Expression Profiling , Genomic Imprinting , Oligonucleotide Array Sequence Analysis , Animals , DNA, Complementary/genetics , Energy Intake , Mice , Mice, Inbred C57BL , Models, Animal , Models, Genetic , Nucleic Acid Hybridization , Polymerase Chain Reaction/methods
10.
J Med Genet ; 43(3): 248-54, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16033916

ABSTRACT

BACKGROUND: Congenital hyperinsulinism and Beckwith-Wiedemann syndrome both lead to beta islet hyperplasia and neonatal hypoglycaemia. They may be related to complex genetic/epigenetic abnormalities of the imprinted 11p15 region. The possibility of common pathophysiological determinants has not been thoroughly investigated. OBJECTIVE: To report abnormalities of the ploidy in two unrelated patients with congenital hyperinsulinism. METHODS: Two patients with severe congenital hyperinsulinism, one overlapping with Beckwith-Wiedemann syndrome, had pancreatic histology, ex vivo potassium channel electrophysiological studies, and mutation detection of the encoding genes. The parental genetic contribution was explored using genome-wide polymorphism, fluorescent in situ hybridisation (FISH), and blood group typing studies. RESULTS: Histological findings diverged from those described in focal congenital hyperinsulinism or Beckwith-Wiedemann syndrome. No potassium channel dysfunction and no mutation of its encoding genes (SUR1, KIR6.2) were detected. In patient 1 with congenital hyperinsulinism and Beckwith-Wiedemann syndrome, paternal isodisomy for the whole haploid set was homogeneous in the pancreatic lesion, and mosaic in the leucocytes and skin fibroblasts (hemihypertrophic segment). Blood group typing confirmed the presence of two erythroid populations (bi-parental v paternal only contribution). Patient 2 had two pancreatic lesions, both revealing triploidy with paternal heterodisomy. Karyotype and FISH analyses done on the fibroblasts and leucocytes of both patients were unremarkable (diploidy). CONCLUSIONS: Diploid (biparental/paternal-only) mosaicism and diploid/triploid mosaicism were present in two distinct patients with congenital hyperinsulinism. These chromosomal abnormalities led to paternal disomy for the whole haploid set in pancreatic lesions (with isodisomy or heterodisomy), thereby extending the range and complexity of the mechanisms underlying congenital hyperinsulinism, associated or not with Beckwith-Wiedemann syndrome.


Subject(s)
Congenital Abnormalities/genetics , Hyperinsulinism/congenital , Hyperinsulinism/genetics , Mosaicism , Ploidies , Chromosome Aberrations , Female , Humans , Infant, Newborn , Male
11.
Ann Endocrinol (Paris) ; 66(2 Pt 3): 2S19-28, 2005 Apr.
Article in French | MEDLINE | ID: mdl-15959393

ABSTRACT

Epigenetic changes associated with DNA methylation and histone modifications leading to chromatin remodeling and regulation of gene expression underlie the developmental programming of obesity, type 2 diabetes, cardiovascular diseases and metabolic syndrome. This review focuses on converging data supporting the hypothesis that, in addition to "thrifty genotype" inheritance, individuals with obesity, type 2 diabetes, and metabolic syndrome (MetS) with an increased risk of cardiovascular diseases have suffered improper "epigenetic programming" during their fetal/postnatal development due to maternal inadequate nutrition and metabolic disturbances and also during their lifetime, that could even be transmitted to the next generation(s). We highlight the susceptibility of epigenetic mechanisms controlling gene expression to environmental influences due to their inherent malleability, emphasizing the participation of transposable elements and the potential role of imprinted genes during critical time windows in epigenetic programming, from the very beginning of development, throughout life. Increasing our understanding on epigenetic patterns significance and their role in development, evolution and adaptation and on small molecules (nutrients, drugs) that reverse epigenetic (in)activation should provide us with the means to "unlock" silenced (enhanced) genes, and to "convert" the obsolete human thrifty genotype into a "squandering" phenotype.


Subject(s)
Diet/adverse effects , Genomics , Nutritional Physiological Phenomena , Adult , Child , Child, Preschool , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Female , Fetal Development , Genotype , Humans , Infant , Infant, Newborn , Metabolic Syndrome/etiology , Metabolic Syndrome/genetics , Obesity/etiology , Obesity/genetics , Pregnancy
13.
J Med Genet ; 40(11): 797-801, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14627666

ABSTRACT

CONTEXT: Beckwith-Wiedemann syndrome (BWS) arises by several genetic and epigenetic mechanisms affecting the balance of imprinted gene expression in chromosome 11p15.5. The most frequent alteration associated with BWS is the absence of methylation at the maternal allele of KvDMR1, an intronic CpG island within the KCNQ1 gene. Targeted deletion of KvDMR1 suggests that this locus is an imprinting control region (ICR) that regulates multiple genes in 11p15.5. Cell culture based enhancer blocking assays indicate that KvDMR1 may function as a methylation modulated chromatin insulator and/or silencer. OBJECTIVE: To determine the potential consequence of loss of methylation (LOM) at KvDMR1 in the development of BWS. METHODS: The steady state levels of CDKN1C gene expression in fibroblast cells from normal individuals, and from persons with BWS who have LOM at KvDMR1, was determined by both real time quantitative polymerase chain reaction (qPCR) and ribonuclease protection assay (RPA). Methylation of the CDKN1C promoter region was assessed by Southern hybridisation using a methylation sensitive restriction endonuclease. RESULTS: Both qPCR and RPA clearly demonstrated a marked decrease (86-93%) in the expression level of the CDKN1C gene in cells derived from patients with BWS, who had LOM at KvDMR1. Southern analysis indicated that downregulation of CDKN1C in these patients was not associated with hypermethylation at the presumptive CDKN1C promoter. CONCLUSIONS: An epimutation at KvDMR1, the absence of maternal methylation, causes the aberrant silencing of CDKN1C, some 180 kb away on the maternal chromosome. Similar to mutations at this locus, this silencing may give rise to BWS.


Subject(s)
Beckwith-Wiedemann Syndrome/genetics , DNA Methylation , Enzyme Inhibitors/metabolism , Gene Silencing/physiology , Genomic Imprinting/genetics , Membrane Proteins , Nuclear Proteins/genetics , Beckwith-Wiedemann Syndrome/enzymology , Cell Line , Cyclin-Dependent Kinase Inhibitor p57 , Fibroblasts/chemistry , Gene Expression Regulation/genetics , Humans , Potassium Channels, Voltage-Gated , RNA, Long Noncoding , RNA, Untranslated/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods
15.
Hum Mol Genet ; 10(23): 2717-26, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11726559

ABSTRACT

The autosomal dominant mutation causing myotonic dystrophy (DM1) is a CTG repeat expansion in the 3'-UTR of the DM protein kinase (DMPK) gene. This multisystemic disorder includes myotonia, progressive weakness and wasting of skeletal muscle and extramuscular symptoms such as cataracts, testicular atrophy, endocrine and cognitive dysfunction. The mechanisms underlying its pathogenesis are complex. Recent reports have revealed that DMPK gene haploinsufficiency may account for cardiac conduction defects whereas cataracts may be due to haploinsufficiency of the neighboring gene, the DM-associated homeobox protein (DMAHP or SIX5) gene. Furthermore, mice expressing the CUG expansion in an unrelated mRNA develop myotonia and myopathy, consistent with an RNA gain of function. We demonstrated that transgenic mice carrying the CTG expansion in its human DM1 context (>45 kb) and producing abnormal DMPK mRNA with at least 300 CUG repeats, displayed clinical, histological, molecular and electrophysiological abnormalities in skeletal muscle consistent with those observed in DM1 patients. Like DM1 patients, these transgenic mice show abnormal tau expression in the brain. These results provide further evidence for the RNA trans-dominant effect of the CUG expansion, not only in muscle, but also in brain.


Subject(s)
Brain/abnormalities , Muscle, Skeletal/abnormalities , Protein Serine-Threonine Kinases/genetics , Trinucleotide Repeat Expansion/genetics , Animals , Brain/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Electromyography , Electrophoresis, Polyacrylamide Gel , Female , Gene Expression , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/cytology , Myotonia/genetics , Myotonia/physiopathology , Myotonic Dystrophy/genetics , Myotonic Dystrophy/pathology , Myotonin-Protein Kinase , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trinucleotide Repeats/genetics , tau Proteins/metabolism
16.
Ann Med Interne (Paris) ; 152(5): 337-51, 2001 Sep.
Article in French | MEDLINE | ID: mdl-11593147

ABSTRACT

About 5% of colon cancer cases correspond to classic hereditary monogenic mendelian transmission involving at least 8 major genes of predisposition to this tumor. Genes with more moderate effects, in association with other genes can contribute to the occurrence of sporadic polygenic forms. These genes confer susceptibility to environmental factors and can play the role of aggravating or protective modifier genes in the different hereditary forms. Foods can interact with these genes and modulate their expression. Moreover sequence variations (polymorphisms) in these genes may also be responsible for slower or more rapid metabolism of nutrients leading to toxic or carcinogenic compounds. If some foods, or "pharmafoods" can have beneficial effects in some individuals with a particular subtype of the disease, others can be inefficient or even detrimental in patients with the same disease but with a different genetic origin or if the genetic background is different. Moreover tumorigenic processes are diverse. Tumor progression depends on genetic and environmental factors different from tumor initiation and on the site of the tumor along the colon tract. Interactions with the gut flora, the lymphoid system and specific features of growth of the colon mucosa are also important parameters. Today with a formidable genetic knowledge arising from the genome project, new epidemiological data integrating the genetic data for multiple markers and a better knowledge of the tumorigenic processes involved, a new discipline is emerging. "Nutrigenetics" which is the study of hereditary basis of individual variations in response to foods opens for the oncoming decade the era of a personalised predictive medecine based on a nutrition adapted to the genetic make up of each of us.


Subject(s)
Cocarcinogenesis , Colonic Neoplasms , Diet/adverse effects , Genetic Predisposition to Disease/genetics , Molecular Biology , Nutritional Physiological Phenomena , Colonic Neoplasms/epidemiology , Colonic Neoplasms/etiology , Colonic Neoplasms/genetics , Colonic Neoplasms/prevention & control , Environmental Exposure/adverse effects , Gene Frequency/genetics , Genetic Markers/genetics , Genetic Testing/methods , Humans , Intestinal Mucosa/microbiology , Molecular Biology/methods , Polymorphism, Genetic/genetics , Predictive Value of Tests , Risk Factors , Xenobiotics/adverse effects , Xenobiotics/metabolism
17.
Hum Mol Genet ; 10(19): 2079-87, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11590125

ABSTRACT

In this study we have developed an in vitro cell culture system which displays the majority of the defects previously described for congenital myotonic dystrophy (CDM) muscle in vivo. Human satellite cells were isolated from the quadriceps muscles of three CDM fetuses with different clinical severity. By Southern blot analysis all three cultures were found to have approximately 2300 CTG repeats. This CTG expansion was found to progressively increase in size during the proliferative life span, confirming an instability of this triplet in skeletal muscle cells. The CDM myoblasts and myotubes also showed abnormal retention of mutant RNA in nuclear foci, as well as modifications in their myogenic program. The proliferative capacity of the CDM myoblasts was reduced and a delay in fusion, differentiation and maturation was observed in the CDM cultures compared with unaffected myoblast cultures. The clinical severity and delayed maturation observed in the CDM fetuses were closely reflected by the phenotypic modifications observed in vitro. Since the culture conditions were the same, this suggests that the defects we have described are intrinsic to the program expressed by the myoblasts in the absence of any trophic factors. Altogether, our results demonstrate that satellite cells are defective in CDM and are probably implicated in the delay in maturation and muscle atrophy that has been described previously in CDM fetuses.


Subject(s)
Muscle, Skeletal/pathology , Myotonic Dystrophy/pathology , Biopsy , Cell Differentiation , Cell Division , Cells, Cultured , Humans , Immunoenzyme Techniques , In Situ Hybridization , In Vitro Techniques , Infant, Newborn , Muscle, Skeletal/metabolism , Myotonic Dystrophy/metabolism , Myotonin-Protein Kinase , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA/metabolism , Trinucleotide Repeat Expansion
18.
Pharmacogenetics ; 11(6): 521-35, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11505222

ABSTRACT

The von Hippel-Lindau (VHL) tumour suppressor gene is commonly mutated in renal cell carcinoma of clear cell type (CCRCC). We investigated the possible relationship between VHL mutations in sporadic CCRCC and polymorphism of genes encoding enzymes involved in carcinogen metabolism: two cytochrome P450 monooxygenases (CYP1A1 and CYP2D6), one NAD[P]H:quinone oxidoreductase (NQO1), three glutathione S-transferases (GSTM1, GSTT1 and GSTP1) and two arylamine N-acetyltransferases (NAT1 and NAT2). We analysed DNA from tumour and nontumoural kidney tissue from 195 CCRCC patients. Single VHL mutations were identified in 88 patients and double mutations were present in two patients. Nine of 18 transversions were GC to TA, four were AT to TA, four were GC to CG and one was AT to CG. Ten of 19 transitions were GC to AT and nine were AT to GC. We also identified 53 frameshifts and two GC to AT at CpG. An excess of transversions was observed in a subset of patients with active GSTT1 [GSTT1 (+) genotype] and probably defective NAT1 (NAT1 S/R variant genotype). All 18 transversions were in GSTT1 (+) patients, whereas only 76% of transitions (P = 0.05) and 81% of the other mutations (P = 0.06) occurred in this genotype. We found that 28% of the transversions were in the NAT1 S/R genotype versus 12% of the transitions (P = 0.40) and 4% of the other mutations (P = 0.01). This suggests that pharmacogenetic polymorphisms may be associated with the type of acquired VHL mutation, which may modulate CCRCC development.


Subject(s)
Acetyltransferases/genetics , Arylamine N-Acetyltransferase , Carcinoma, Renal Cell/genetics , Genes, Tumor Suppressor , Glutathione Transferase/genetics , Ligases , Mutation , Proteins/genetics , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , Adult , Aged , Chromosome Aberrations , Female , Frameshift Mutation , Gene Frequency , Genotype , Humans , Isoenzymes , Loss of Heterozygosity , Male , Middle Aged , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Polymorphism, Single-Stranded Conformational , Von Hippel-Lindau Tumor Suppressor Protein , Xenobiotics/metabolism
19.
Am J Pathol ; 158(6): 2177-84, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11395395

ABSTRACT

Congenital hyperinsulinism (CHI), previously named persistent hyperinsulinemic hypoglycemia of infancy, is characterized by profound hypoglycemia because of excessive insulin secretion. CHI presents as two different morphological forms: a diffuse form with functional abnormality of islets throughout the pancreas and a focal form with focal islet cell adenomatous hyperplasia, which can be cured by partial pancreatectomy. Recently, we have shown that focal adenomatous hyperplasia involves the specific loss of the maternal 11p15 region and a constitutional mutation of a paternally inherited allele of the gene encoding the regulating subunit of the K(+)(ATP) channel, the sulfonylurea receptor (ABCC8 or SUR1). In the present study on a large series of 31 patients, describing both morphological features and molecular data, we report that 61% of cases (19 out of 31) carried a paternally inherited mutation not only in the ABCC8 gene as previously described but also in the second gene encoding the K(+)(ATP) channel, the inward rectifying potassium channel (KCNJ11 or KIR6.2), in 15 cases and 4 cases, respectively. Moreover our results are consistent with the presence of a duplicated paternal 11p15 allele probably because of mitotic recombination or reduplication of the paternal chromosome after somatic loss of the maternal chromosome. In agreement with the loss of the maternal chromosome, the level of expression of a maternally expressed tumor suppressor gene, H19, was greatly reduced compared to the level of expression of the paternally expressed growth promoter gene, IGF2. The expression of IGF2 was on average only moderately increased. Thus, focal forms of CHI can be considered to be a recessive somatic disease, associating an imbalance in the expression of imprinted genes in the 11p15.5 region to a somatic reduction to homozygosity of an ABCC8- or KCNJ11-recessive mutation. The former is responsible for the abnormal growth rate, as in embryonic tumors, whereas the latter leads to unregulated secretion of insulin.


Subject(s)
ATP-Binding Cassette Transporters , Adenoma, Islet Cell/genetics , Chromosomes, Human, Pair 11 , Genomic Imprinting , Hyperinsulinism/genetics , Pancreatic Neoplasms/genetics , Potassium Channels, Inwardly Rectifying , Potassium Channels/genetics , Receptors, Drug/genetics , Adenoma, Islet Cell/congenital , Adenoma, Islet Cell/metabolism , Gene Dosage , Humans , Hyperinsulinism/congenital , Hyperplasia , Insulin-Like Growth Factor II/biosynthesis , Insulin-Like Growth Factor II/genetics , Loss of Heterozygosity , Mutation , Pancreatic Neoplasms/congenital , Pancreatic Neoplasms/metabolism , RNA, Long Noncoding , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , RNA, Untranslated/biosynthesis , RNA, Untranslated/genetics , Sulfonylurea Receptors
SELECTION OF CITATIONS
SEARCH DETAIL
...