Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Exp Med ; 217(11)2020 11 02.
Article in English | MEDLINE | ID: mdl-32865561

ABSTRACT

The autosomal recessive immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome is a genetically heterogeneous disorder. Despite the identification of the underlying gene defects, it is unclear how mutations in any of the four known ICF genes cause a primary immunodeficiency. Here we demonstrate that loss of ZBTB24 in B cells from mice and ICF2 patients affects nonhomologous end-joining (NHEJ) during immunoglobulin class-switch recombination and consequently impairs immunoglobulin production and isotype balance. Mechanistically, we found that ZBTB24 associates with poly(ADP-ribose) polymerase 1 (PARP1) and stimulates its auto-poly(ADP-ribosyl)ation. The zinc-finger in ZBTB24 binds PARP1-associated poly(ADP-ribose) chains and mediates the PARP1-dependent recruitment of ZBTB24 to DNA breaks. Moreover, through its association with poly(ADP-ribose) chains, ZBTB24 protects them from degradation by poly(ADP-ribose) glycohydrolase (PARG). This facilitates the poly(ADP-ribose)-dependent assembly of the LIG4/XRCC4 complex at DNA breaks, thereby promoting error-free NHEJ. Thus, we uncover ZBTB24 as a regulator of PARP1-dependent NHEJ and class-switch recombination, providing a molecular basis for the immunodeficiency in ICF2 syndrome.


Subject(s)
DNA End-Joining Repair/genetics , Face/abnormalities , Immunoglobulin Class Switching/genetics , Mutation , Primary Immunodeficiency Diseases/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Animals , B-Lymphocytes/immunology , DNA Breaks , Face/pathology , HEK293 Cells , Humans , Immunoglobulin Switch Region , Mice , Poly (ADP-Ribose) Polymerase-1/metabolism , Primary Immunodeficiency Diseases/blood , Primary Immunodeficiency Diseases/pathology , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transfection
2.
Am J Med Genet A ; 170(12): 3253-3257, 2016 12.
Article in English | MEDLINE | ID: mdl-27604394

ABSTRACT

ICF syndrome is a primary immunodeficiency disease characterized by hypo- or agammaglobulinemia, centromeric instability mainly on chromosomes 1, 9, and 16 and facial anomalies. ICF syndrome presents with frequent respiratory tract infections in infancy. A 20-month-old female patient was referred to our clinic due to frequent lower respiratory tract infections. ICF syndrome was considered because of comorbidity of hypogammaglobulinemia, facial anomalies, and neuromotor growth retardation. Metaphase chromosome analysis revealed centromeric instability on chromosomes 1, 9, and 16 and through Sanger a previously unreported homozygous missense mutation (c.1805T>C; [p.V602A]) was identified in the DNMT3B, confirming ICF1. The patient was found to have a breakdown in renal function 1 year later; the urinary system was examined and bilateral vesicoureteral reflux was found, warranting the need for dialysis in time. This report expands the mutation spectrum of ICF1 and is the first to describe bilateral vesicoureteral reflux accompanying ICF syndrome. © 2016 Wiley Periodicals, Inc.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Mutation , Renal Insufficiency/diagnosis , Renal Insufficiency/etiology , Vesico-Ureteral Reflux/complications , Biomarkers , Chromosomal Instability , Chromosome Aberrations , DNA Mutational Analysis , Facies , Female , Genetic Association Studies , Humans , Immunoglobulins, Intravenous , Immunologic Deficiency Syndromes/drug therapy , Infant , Pedigree , Phenotype , Physical Examination , Syndrome , Vesico-Ureteral Reflux/diagnosis , DNA Methyltransferase 3B
3.
Hum Mol Genet ; 25(18): 4041-4051, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27466202

ABSTRACT

For genetically heterogeneous diseases a better understanding of how the underlying gene defects are functionally interconnected will be important for dissecting disease etiology. The Immunodeficiency, Centromeric instability, Facial anomalies (ICF) syndrome is a chromatin disorder characterized by mutations in DNMT3B, ZBTB24, CDCA7 or HELLS Here, we generated a Zbtb24 BTB domain deletion mouse and found that loss of functional Zbtb24 leads to early embryonic lethality. Transcriptome analysis identified Cdca7 as the top down-regulated gene in Zbtb24 homozygous mutant mESCs, which can be restored by ectopic ZBTB24 expression. We further demonstrate enrichment of ZBTB24 at the CDCA7 promoter suggesting that ZBTB24 can function as a transcription factor directly controlling Cdca7 expression. Finally, we show that this regulation is conserved between species and that CDCA7 levels are reduced in patients carrying ZBTB24 nonsense mutations. Together, our findings demonstrate convergence of the two ICF genes ZBTB24 and CDCA7 at the level of transcription.


Subject(s)
Face/abnormalities , Immunologic Deficiency Syndromes/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Transcription, Genetic , Animals , Codon, Nonsense/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Helicases/genetics , Face/physiopathology , Female , Gene Expression Regulation, Developmental , Humans , Immunologic Deficiency Syndromes/physiopathology , Male , Mice , Mouse Embryonic Stem Cells/metabolism , Nuclear Proteins/biosynthesis , Primary Immunodeficiency Diseases , Transcriptome/genetics , DNA Methyltransferase 3B
8.
Epigenetics ; 10(12): 1133-42, 2015.
Article in English | MEDLINE | ID: mdl-26575099

ABSTRACT

Facioscapulohumeral muscular dystrophy is caused by incomplete epigenetic repression of the transcription factor DUX4 in skeletal muscle. A copy of DUX4 is located within each unit of the D4Z4 macrosatellite repeat array and its derepression in somatic cells is caused by either repeat array contraction (FSHD1) or by mutations in the chromatin repressor SMCHD1 (FSHD2). While DUX4 expression has thus far only been detected in FSHD muscle and muscle cell cultures, and increases with in vitro myogenic differentiation, the D4Z4 chromatin structure has only been studied in proliferating myoblasts or non-myogenic cells. We here show that SMCHD1 protein levels at D4Z4 decline during muscle cell differentiation and correlate with DUX4 derepression. In FSHD2, but not FSHD1, the loss of SMCHD1 repressor activity is partially compensated by increased Polycomb Repressive Complex 2 (PRC2)-mediated H3K27 trimethylation at D4Z4, a situation that can be mimicked by SMCHD1 knockdown in control myotubes. In contrast, moderate overexpression of SMCHD1 results in DUX4 silencing in FSHD1 and FSHD2 myotubes demonstrating that DUX4 derepression in FSHD is reversible. Together, we show that in FSHD1 and FSHD2 the decline in SMCHD1 protein levels during muscle cell differentiation renders skeletal muscle sensitive to DUX4.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic , Homeodomain Proteins/metabolism , Muscle Development/genetics , Muscular Dystrophy, Facioscapulohumeral/genetics , Cell Differentiation/genetics , Cell Line , Chromosomal Proteins, Non-Histone/genetics , DNA Methylation , Gene Expression Regulation , Histone Code , Homeodomain Proteins/genetics , Humans , Muscle, Skeletal/metabolism
9.
Nat Commun ; 6: 7870, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26216346

ABSTRACT

The life-threatening Immunodeficiency, Centromeric Instability and Facial Anomalies (ICF) syndrome is a genetically heterogeneous autosomal recessive disorder. Twenty percent of patients cannot be explained by mutations in the known ICF genes DNA methyltransferase 3B or zinc-finger and BTB domain containing 24. Here we report mutations in the cell division cycle associated 7 and the helicase, lymphoid-specific genes in 10 unexplained ICF cases. Our data highlight the genetic heterogeneity of ICF syndrome; however, they provide evidence that all genes act in common or converging pathways leading to the ICF phenotype.


Subject(s)
DNA Helicases/genetics , Face/abnormalities , Immunologic Deficiency Syndromes/genetics , Nuclear Proteins/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Male , Mutation , Mutation, Missense , Primary Immunodeficiency Diseases , Young Adult
11.
Nucleic Acids Res ; 43(14): 6919-33, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26101254

ABSTRACT

The faithful repair of DNA double-strand breaks (DSBs) is essential to safeguard genome stability. DSBs elicit a signaling cascade involving the E3 ubiquitin ligases RNF8/RNF168 and the ubiquitin-dependent assembly of the BRCA1-Abraxas-RAP80-MERIT40 complex. The association of BRCA1 with ubiquitin conjugates through RAP80 is known to be inhibitory to DSB repair by homologous recombination (HR). However, the precise regulation of this mechanism remains poorly understood. Through genetic screens we identified USP26 and USP37 as key de-ubiquitylating enzymes (DUBs) that limit the repressive impact of RNF8/RNF168 on HR. Both DUBs are recruited to DSBs where they actively remove RNF168-induced ubiquitin conjugates. Depletion of USP26 or USP37 disrupts the execution of HR and this effect is alleviated by the simultaneous depletion of RAP80. We demonstrate that USP26 and USP37 prevent excessive spreading of RAP80-BRCA1 from DSBs. On the other hand, we also found that USP26 and USP37 promote the efficient association of BRCA1 with PALB2. This suggests that these DUBs limit the ubiquitin-dependent sequestration of BRCA1 via the BRCA1-Abraxas-RAP80-MERIT40 complex, while promoting complex formation and cooperation of BRCA1 with PALB2-BRCA2-RAD51 during HR. These findings reveal a novel ubiquitin-dependent mechanism that regulates distinct BRCA1-containing complexes for efficient repair of DSBs by HR.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Cysteine Endopeptidases/metabolism , Endopeptidases/metabolism , Nuclear Proteins/antagonists & inhibitors , Recombinational DNA Repair , BRCA1 Protein/metabolism , Carrier Proteins/metabolism , Cell Line , DNA Breaks, Double-Stranded , DNA-Binding Proteins , Histone Chaperones , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Rad51 Recombinase/metabolism , Tumor Suppressor p53-Binding Protein 1 , Ubiquitin/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
12.
Nat Commun ; 5: 5592, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25424739

ABSTRACT

Periconceptional diet may persistently influence DNA methylation levels with phenotypic consequences. However, a comprehensive assessment of the characteristics of prenatal malnutrition-associated differentially methylated regions (P-DMRs) is lacking in humans. Here we report on a genome-scale analysis of differential DNA methylation in whole blood after periconceptional exposure to famine during the Dutch Hunger Winter. We show that P-DMRs preferentially occur at regulatory regions, are characterized by intermediate levels of DNA methylation and map to genes enriched for differential expression during early development. Validation and further exploratory analysis of six P-DMRs highlight the critical role of gestational timing. Interestingly, differential methylation of the P-DMRs extends along pathways related to growth and metabolism. P-DMRs located in INSR and CPT1A have enhancer activity in vitro and differential methylation is associated with birth weight and serum LDL cholesterol. Epigenetic modulation of pathways by prenatal malnutrition may promote an adverse metabolic phenotype in later life.


Subject(s)
Antigens, CD/metabolism , DNA Methylation , Fetal Development , Fetal Nutrition Disorders/metabolism , Prenatal Exposure Delayed Effects/metabolism , Receptor, Insulin/metabolism , Starvation , Antigens, CD/genetics , Birth Weight , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Epigenesis, Genetic , Female , Fetal Nutrition Disorders/genetics , Humans , Male , Middle Aged , Molecular Sequence Data , Netherlands , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Receptor, Insulin/genetics
13.
Int J Epidemiol ; 43(3): 878-86, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24425829

ABSTRACT

BACKGROUND: Human leukocyte telomere length (LTL) decreases with age and shorter LTL has previously been associated with increased prospective mortality. However, it is not clear whether LTL merely marks the health status of an individual by its association with parameters of immune function, for example, or whether telomere shortening also contributes causally to lifespan variation in humans. METHODS: We measured LTL in 870 nonagenarian siblings (mean age 93 years), 1580 of their offspring and 725 spouses thereof (mean age 59 years) from the Leiden Longevity Study (LLS). RESULTS: We found that shorter LTL is associated with increased prospective mortality in middle (30-80 years; hazard ratio (HR)=0.75, P=0.001) and highly advanced age (≥90 years; HR=0.92, P=0.028), and show that this association cannot be explained by the association of LTL with the immune-related markers insulin-like growth factor 1 to insulin-like growth factor binding protein 3 molar ratio, C-reactive protein, interleukin 6, cytomegalovirus serostatus or white blood cell counts. We found no difference in LTL between the middle-aged LLS offspring and their spouses (ß=0.006, P=0.932). Neither did we observe an association of LTL-associated genetic variants with mortality in a prospective meta-analysis of multiple cohorts (n=8165). CONCLUSIONS: We confirm LTL to be a marker of prospective mortality in middle and highly advanced age and additionally show that this association could not be explained by the association of LTL with various immune-related markers. Furthermore, the approaches performed here do not further support the hypothesis that LTL variation contributes to the genetic propensity for longevity.


Subject(s)
Aging/immunology , Family , Leukocytes/immunology , Longevity/immunology , Telomere/immunology , Aged , Aged, 80 and over , Biomarkers , Female , Health Status , Humans , Immunoproteins/analysis , Inflammation Mediators/blood , Male , Middle Aged , Prospective Studies
14.
Skelet Muscle ; 4: 19, 2014.
Article in English | MEDLINE | ID: mdl-25789155

ABSTRACT

BACKGROUND: The most common form of facioscapulohumeral muscular dystrophy (FSHD) is caused by a genetic contraction of the polymorphic D4Z4 macrosatellite repeat array in the subtelomeric region of chromosome 4q. In some studies, genes centromeric to the D4Z4 repeat array have been reported to be over-expressed in FSHD, including FRG1 and FRG2, presumably due to decreased long-distance repression by the shorter array through a mechanism similar to position-effect variegation. Differential regulation of FRG1 in FSHD has never been unequivocally proven, however, FRG2 has been reproducibly shown to be induced in primary FSHD-derived muscle cells when differentiated in vitro. The molecular function of FRG2 and a possible contribution to FSHD pathology remain unclear. Recent evidence has identified the mis-expression of DUX4, located within the D4Z4 repeat unit, in skeletal muscle as the cause of FSHD. DUX4 is a double homeobox transcription factor that has been shown to be toxic when expressed in muscle cells. METHODS: We used a combination of expression analysis by qRT/PCR and RNA sequencing to determine the transcriptional activation of FRG2 and DUX4. We examined this in both differentiating control and FSHD derived muscle cell cultures or DUX4 transduced control cell lines. Next, we used ChIP-seq analysis and luciferase reporter assays to determine the potential DUX4 transactivation effect on the FRG2 promoter. RESULTS: We show that DUX4 directly activates the expression of FRG2. Increased expression of FRG2 was observed following expression of DUX4 in myoblasts and fibroblasts derived from control individuals. Moreover, we identified DUX4 binding sites at the FRG2 promoter by chromatin immunoprecipitation followed by deep sequencing and confirmed the direct regulation of DUX4 on the FRG2 promoter by luciferase reporter assays. Activation of luciferase was dependent on both DUX4 expression and the presence of the DUX4 DNA binding motifs in the FRG2 promoter. CONCLUSION: We show that the FSHD-specific upregulation of FRG2 is a direct consequence of the activity of DUX4 protein rather than representing a regional de-repression secondary to fewer D4Z4 repeats.

15.
Proc Natl Acad Sci U S A ; 110(51): 20783-8, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24297891

ABSTRACT

Myasthenia gravis (MG) is a severely debilitating autoimmune disease that is due to a decrease in the efficiency of synaptic transmission at neuromuscular synapses. MG is caused by antibodies against postsynaptic proteins, including (i) acetylcholine receptors, the neurotransmitter receptor, (ii) muscle-specific kinase (MuSK), a receptor tyrosine kinase essential for the formation and maintenance of neuromuscular synapses, and (iii) low-density lipoprotein receptor-related protein 4 (Lrp4), which responds to neural Agrin by binding and stimulating MuSK. Passive transfer studies in mice have shown that IgG4 antibodies from MuSK MG patients cause disease without requiring complement or other immune components, suggesting that these MuSK antibodies cause disease by directly interfering with MuSK function. Here we show that pathogenic IgG4 antibodies to MuSK bind to a structural epitope in the first Ig-like domain of MuSK, prevent binding between MuSK and Lrp4, and inhibit Agrin-stimulated MuSK phosphorylation. In contrast, these IgG4 antibodies have no direct effect on MuSK dimerization or MuSK internalization. These results provide insight into the unique pathogenesis of MuSK MG and provide clues toward development of specific treatment options.


Subject(s)
Autoantibodies/immunology , Immunoglobulin G/immunology , LDL-Receptor Related Proteins/immunology , Myasthenia Gravis/immunology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Cholinergic/immunology , Receptors, LDL/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Agrin/immunology , Animals , Autoantibodies/pharmacology , Cell Line , Child , Child, Preschool , Epitopes/immunology , Female , Humans , Immunization, Passive , Immunoglobulin G/pharmacology , LDL-Receptor Related Proteins/antagonists & inhibitors , Male , Mice , Middle Aged , Myasthenia Gravis/chemically induced , Myasthenia Gravis/pathology , Phosphorylation/drug effects , Phosphorylation/immunology , Protein Multimerization/drug effects , Protein Multimerization/immunology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, LDL/antagonists & inhibitors
16.
Cell Cycle ; 12(18): 3070-82, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23974106

ABSTRACT

The cellular response to ionizing radiation (IR)-induced DNA double-strand breaks (DSBs) in native chromatin requires a tight coordination between the activities of DNA repair machineries and factors that modulate chromatin structure. SMARCA5 is an ATPase of the SNF2 family of chromatin remodeling factors that has recently been implicated in the DSB response. It forms distinct chromatin remodeling complexes with several non-canonical subunits, including the remodeling and spacing factor 1 (RSF1) protein. Despite the fact that RSF1 is often overexpressed in tumors and linked to tumorigenesis and genome instability, its role in the DSB response remains largely unclear. Here we show that RSF1 accumulates at DSB sites and protects human cells against IR-induced DSBs by promoting repair of these lesions through homologous recombination (HR) and non-homologous end-joining (NHEJ). Although SMARCA5 regulates the RNF168-dependent ubiquitin response that targets BRCA1 to DSBs, we found RSF1 to be dispensable for this process. Conversely, we found that RSF1 facilitates the assembly of centromere proteins CENP-S and CENP-X at sites of DNA damage, while SMARCA5 was not required for these events. Mechanistically, we uncovered that CENP-S and CENP-X, upon their incorporation by RSF1, promote assembly of the NHEJ factor XRCC4 at damaged chromatin. In contrast, CENP-S and CENP-X were dispensable for HR, suggesting that RSF1 regulates HR independently of these centromere proteins. Our findings reveal distinct functions of RSF1 in the 2 major pathways of DSB repair and explain how RSF1, through the loading of centromere proteins and XRCC4 at DSBs, promotes repair by non-homologous end-joining.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , BRCA1 Protein/metabolism , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/genetics , DNA Breaks, Double-Stranded/radiation effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , Homologous Recombination , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Radiation, Ionizing , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
17.
Epigenetics ; 8(5): 512-21, 2013 May.
Article in English | MEDLINE | ID: mdl-23644601

ABSTRACT

Subtelomeres are patchworks of evolutionary conserved sequence blocks and harbor the transcriptional start sites for telomere repeat containing RNAs (TERRA). Recent studies suggest that the interplay between telomeres and subtelomeric chromatin is required for maintaining telomere function. To further characterize chromatin remodeling of subtelomeres in relation to telomere shortening and cellular senescence, we systematically quantified histone modifications and DNA methylation at the subtelomeres of chromosomes 7q and 11q in primary human WI-38 fibroblasts. Upon senescence, both subtelomeres were characterized by a decrease in markers of constitutive heterochromatin, suggesting relative chromatin relaxation. However, we did not find increased levels of markers of euchromatin or derepression of the 7q VIPR2 gene. The repressed state of the subtelomeres was maintained upon senescence, which could be attributed to a rise in levels of facultative heterochromatin markers at both subtelomeres. While senescence-induced subtelomeric chromatin remodeling was similar for both chromosomes, chromatin remodeling at TERRA promoters displayed chromosome-specific patterns. At the 7q TERRA promoter, chromatin structure was co-regulated with the more proximal subtelomere. In contrast, the 11q TERRA promoter, which was previously shown to be bound by CCCTC-binding factor CTCF, displayed lower levels of markers of constitutive heterochromatin that did not change upon senescence, whereas levels of markers of facultative heterochromatin decreased upon senescence. In line with the chromatin state data, transcription of 11q TERRA but not 7q TERRA was detected. Our study provides a detailed description of human subtelomeric chromatin dynamics and shows distinct regulation of the TERRA promoters of 7q and 11q upon cellular senescence.


Subject(s)
Cellular Senescence/genetics , Chromatin Assembly and Disassembly/genetics , Chromosomes, Human/genetics , Promoter Regions, Genetic/genetics , RNA/genetics , Telomere/genetics , Biomarkers/metabolism , Euchromatin/metabolism , Fibroblasts/metabolism , Heterochromatin/metabolism , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Phenotype , Repetitive Sequences, Nucleic Acid/genetics , Telomere Shortening , Transcription, Genetic
18.
PLoS Genet ; 9(4): e1003415, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23593020

ABSTRACT

Facioscapulohumeral dystrophy (FSHD) is a progressive muscular dystrophy caused by decreased epigenetic repression of the D4Z4 macrosatellite repeats and ectopic expression of DUX4, a retrogene encoding a germline transcription factor encoded in each repeat. Unaffected individuals generally have more than 10 repeats arrayed in the subtelomeric region of chromosome 4, whereas the most common form of FSHD (FSHD1) is caused by a contraction of the array to fewer than 10 repeats, associated with decreased epigenetic repression and variegated expression of DUX4 in skeletal muscle. We have generated transgenic mice carrying D4Z4 arrays from an FSHD1 allele and from a control allele. These mice recapitulate important epigenetic and DUX4 expression attributes seen in patients and controls, respectively, including high DUX4 expression levels in the germline, (incomplete) epigenetic repression in somatic tissue, and FSHD-specific variegated DUX4 expression in sporadic muscle nuclei associated with D4Z4 chromatin relaxation. In addition we show that DUX4 is able to activate similar functional gene groups in mouse muscle cells as it does in human muscle cells. These transgenic mice therefore represent a valuable animal model for FSHD and will be a useful resource to study the molecular mechanisms underlying FSHD and to test new therapeutic intervention strategies.


Subject(s)
Epigenesis, Genetic/genetics , Homeodomain Proteins , Muscular Dystrophy, Facioscapulohumeral/genetics , Repetitive Sequences, Nucleic Acid/genetics , Animals , Cells, Cultured , Chromatin/genetics , DNA Methylation/genetics , Disease Models, Animal , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Germ Cells/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/metabolism
19.
Eur J Hum Genet ; 21(11): 1219-25, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23486536

ABSTRACT

Immunodeficiency with centromeric instability and facial anomalies (ICF) syndrome is a primary immunodeficiency, predominantly characterized by agammaglobulinemia or hypoimmunoglobulinemia, centromere instability and facial anomalies. Mutations in two genes have been discovered to cause ICF syndrome: DNMT3B and ZBTB24. To characterize the clinical features of this syndrome, as well as genotype-phenotype correlations, we compared clinical and genetic data of 44 ICF patients. Of them, 23 had mutations in DNMT3B (ICF1), 13 patients had mutations in ZBTB24 (ICF2), whereas for 8 patients, the gene defect has not yet been identified (ICFX). While at first sight these patients share the same immunological, morphological and epigenetic hallmarks of the disease, systematic evaluation of all reported informative cases shows that: (1) the humoral immunodeficiency is generally more pronounced in ICF1 patients, (2) B- and T-cell compartments are both involved in ICF1 and ICF2, (3) ICF2 patients have a significantly higher incidence of intellectual disability and (4) congenital malformations can be observed in some ICF1 and ICF2 cases. It is expected that these observations on prevalence and clinical presentation will facilitate mutation-screening strategies and help in diagnostic counseling.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Face/abnormalities , Genetic Heterogeneity , Genetic Predisposition to Disease , Immunologic Deficiency Syndromes/genetics , Mutation/genetics , Repressor Proteins/genetics , Adolescent , Adult , Child , Demography , Female , Hematopoietic Stem Cell Transplantation , Humans , Immunoglobulin G/blood , Immunologic Deficiency Syndromes/blood , Immunologic Deficiency Syndromes/therapy , Lymphocyte Count , Male , Middle Aged , Primary Immunodeficiency Diseases , Young Adult , DNA Methyltransferase 3B
20.
Age (Dordr) ; 35(1): 235-49, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22113349

ABSTRACT

In genome-wide association studies (GWAS) of complex traits, single SNP analysis is still the most applied approach. However, the identified SNPs have small effects and provide limited biological insight. A more appropriate approach to interpret GWAS data of complex traits is to analyze the combined effect of a SNP set grouped per pathway or gene region. We used this approach to study the joint effect on human longevity of genetic variation in two candidate pathways, the insulin/insulin-like growth factor (IGF-1) signaling (IIS) pathway and the telomere maintenance (TM) pathway. For the analyses, we used genotyped GWAS data of 403 unrelated nonagenarians from long-lived sibships collected in the Leiden Longevity Study and 1,670 younger population controls. We analyzed 1,021 SNPs in 68 IIS pathway genes and 88 SNPs in 13 TM pathway genes using four self-contained pathway tests (PLINK set-based test, Global test, GRASS and SNP ratio test). Although we observed small differences between the results of the different pathway tests, they showed consistent significant association of the IIS and TM pathway SNP sets with longevity. Analysis of gene SNP sets from these pathways indicates that the association of the IIS pathway is scattered over several genes (AKT1, AKT3, FOXO4, IGF2, INS, PIK3CA, SGK, SGK2, and YWHAG), while the association of the TM pathway seems to be mainly determined by one gene (POT1). In conclusion, this study shows that genetic variation in genes involved in the IIS and TM pathways is associated with human longevity.


Subject(s)
Insulin-Like Growth Factor I/genetics , Insulin/metabolism , Longevity/genetics , Polymorphism, Single Nucleotide , Adult , Aged , Aged, 80 and over , Female , Genome-Wide Association Study , Genotype , Humans , Insulin/genetics , Male , Middle Aged , Signal Transduction/genetics , Telomere
SELECTION OF CITATIONS
SEARCH DETAIL
...