Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res Ther ; 14(1): 50, 2023 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-36959625

RESUMO

BACKGROUND: Three-dimensional (3D) human brain spheroids are instrumental to study central nervous system (CNS) development and (dys)function. Yet, in current brain spheroid models the limited variety of cell types hampers an integrated exploration of CNS (disease) mechanisms. METHODS: Here we report a 5-month culture protocol that reproducibly generates H9 embryonic stem cell-derived human cortical spheroids (hCSs) with a large cell-type variety. RESULTS: We established the presence of not only neuroectoderm-derived neural progenitor populations, mature excitatory and inhibitory neurons, astrocytes and oligodendrocyte (precursor) cells, but also mesoderm-derived microglia and endothelial cell populations in the hCSs via RNA-sequencing, qPCR, immunocytochemistry and transmission electron microscopy. Transcriptomic analysis revealed resemblance between the 5-months-old hCSs and dorsal frontal rather than inferior regions of human fetal brains of 19-26 weeks of gestational age. Pro-inflammatory stimulation of the generated hCSs induced a neuroinflammatory response, offering a proof-of-principle of the applicability of the spheroids. CONCLUSIONS: Our protocol provides a 3D human brain cell model containing a wide variety of innately developing neuroectoderm- as well as mesoderm-derived cell types, furnishing a versatile platform for comprehensive examination of intercellular CNS communication and neurological disease mechanisms.


Assuntos
Encéfalo , Neurônios , Humanos , Lactente , Encéfalo/metabolismo , Neurônios/metabolismo , Células Cultivadas , Esferoides Celulares , Astrócitos
2.
Biomedicines ; 10(5)2022 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-35625761

RESUMO

Crucial in the pathogenesis of neurodegenerative diseases is the process of neuroinflammation that is often linked to the pro-inflammatory cytokines Tumor necrosis factor alpha (TNFα) and Interleukin-1beta (IL-1ß). Human cortical spheroids (hCSs) constitute a valuable tool to study the molecular mechanisms underlying neurological diseases in a complex three-dimensional context. We recently designed a protocol to generate hCSs comprising all major brain cell types. Here we stimulate these hCSs for three time periods with TNFα and with IL-1ß. Transcriptomic analysis reveals that the main process induced in the TNFα- as well as in the IL-1ß-stimulated hCSs is neuroinflammation. Central in the neuroinflammatory response are endothelial cells, microglia and astrocytes, and dysregulated genes encoding cytokines, chemokines and their receptors, and downstream NFκB- and STAT-pathway components. Furthermore, we observe sets of neuroinflammation-related genes that are specifically modulated in the TNFα-stimulated and in the IL-1ß-stimulated hCSs. Together, our results help to molecularly understand human neuroinflammation and thus a key mechanism of neurodegeneration.

3.
J Neuromuscul Dis ; 8(5): 801-814, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34024774

RESUMO

BACKGROUND: FSHD is caused by specific genetic mutations resulting in activation of the Double Homeobox 4 gene (DUX4). DUX4 targets hundreds of downstream genes eventually leading to muscle atrophy, oxidative stress, abnormal myogenesis, and muscle inflammation. We hypothesized that DUX4-induced aberrant expression of genes triggers a sustained autoimmune response against skeletal muscle cells. OBJECTIVE: This study aimed at the identification of autoantibodies directed against muscle antigens in FSHD. Moreover, a possible relationship between serum antibody reactivity and DUX4 expression was also investigated. METHODS: FSHD sera (N = 138, 48±16 years, 48% male) and healthy control sera (N = 20, 47±14 years, 50% male) were analyzed by immunoblotting for antibodies against several skeletal muscle protein extracts: healthy muscle, FSHD muscle, healthy and FSHD myotubes, and inducible DUX4 expressing myoblasts. In addition, DUX4 expressing myoblasts were analyzed by immunofluorescence with FSHD and healthy control sera. RESULTS: The results showed that the reactivity of FSHD sera did not significantly differ from that of healthy controls, with all the tested muscle antigen extracts. Besides, the immunofluorescent staining of DUX4-expressing myoblasts was not different when incubated with either FSHD or healthy control sera. CONCLUSION: Since the methodology used did not lead to the identification of disease-specific autoantibodies in the FSHD cohort, we suggest that autoantibody-mediated pathology may not be an important disease mechanism in FSHD. Nevertheless, it is crucial to further unravel if and which role the immune system plays in FSHD pathogenesis. Other innate as well as adaptive immune players could be involved in the complex DUX4 cascade of events and could become appealing druggable targets.


Assuntos
Autoanticorpos/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapuloumeral/sangue , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Atrofia Muscular/metabolismo , Mioblastos/metabolismo
4.
J Med Genet ; 55(7): 469-478, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29563141

RESUMO

BACKGROUND: 18p deletion syndrome is a rare disorder caused by partial or full monosomy of the short arm of chromosome 18. Clinical symptoms caused by 18p hemizygosity include cognitive impairment, mild facial dysmorphism, strabismus and ptosis. Among other genes, structural maintenance of chromosomes flexible hinge domain containing 1 (SMCHD1) is hemizygous in most patients with 18p deletions. Digenic inheritance of a SMCHD1 mutation and a moderately sized D4Z4 repeat on a facioscapulohumeral muscular dystrophy (FSHD) permissive genetic background of chromosome 4 can cause FSHD type 2 (FSHD2). OBJECTIVES: Since 12% of Caucasian individuals harbour moderately sized D4Z4 repeats on an FSHD permissive background, we tested if people with 18p deletions are at risk of developing FSHD. METHODS: To test our hypothesis we studied different cellular systems originating from individuals with 18p deletions not presenting FSHD2 phenotype for transcriptional and epigenetic characteristics of FSHD at D4Z4. Furthermore, individuals with an idiopathic muscle phenotype and an 18p deletion were subjected to neurological examination. RESULTS: Primary fibroblasts hemizygous for SMCHD1 have a D4Z4 chromatin structure comparable with FSHD2 concomitant with DUX4 expression after transdifferentiation into myocytes. Neurological examination of 18p deletion individuals from two independent families with a moderately sized D4Z4 repeat identified muscle features compatible with FSHD. CONCLUSIONS: 18p deletions leading to haploinsufficiency of SMCHD1, together with a moderately sized FSHD permissive D4Z4 allele, can associate with symptoms and molecular features of FSHD. We propose that patients with 18p deletion should be characterised for their D4Z4 repeat size and haplotype and monitored for clinical features of FSHD.


Assuntos
Proteínas Cromossômicas não Histona/genética , Transtornos Cromossômicos/genética , Epigênese Genética , Distrofia Muscular Facioescapuloumeral/genética , Adolescente , Adulto , Cromatina/genética , Deleção Cromossômica , Transtornos Cromossômicos/diagnóstico , Transtornos Cromossômicos/fisiopatologia , Cromossomos Humanos Par 18/genética , Metilação de DNA/genética , Feminino , Haploinsuficiência/genética , Humanos , Masculino , Pessoa de Meia-Idade , Monossomia/genética , Monossomia/patologia , Distrofia Muscular Facioescapuloumeral/epidemiologia , Distrofia Muscular Facioescapuloumeral/fisiopatologia , Mutação , Fatores de Risco , Adulto Jovem
5.
Eur J Hum Genet ; 26(1): 94-106, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29162933

RESUMO

Facioscapulohumeral muscular dystrophy is caused by incomplete repression of the transcription factor DUX4 in skeletal muscle as a consequence of D4Z4 macrosatellite repeat contraction in chromosome 4q35 (FSHD1) or variants in genes encoding D4Z4 chromatin repressors (FSHD2). A clinical hallmark of FSHD is variability in onset and progression suggesting the presence of disease modifiers. A well-known cis modifier is the polymorphic DUX4 polyadenylation signal (PAS) that defines FSHD permissive alleles: D4Z4 chromatin relaxation on non-permissive alleles which lack the DUX4-PAS cannot cause disease in the absence of stable DUX4 mRNA. We have explored the nature and relevance of a common variant of the major FSHD haplotype 4A161, which is defined by 1.6 kb size difference of the most distal D4Z4 repeat unit. While the short variant (4A161S) has been extensively studied, we demonstrate that the long variant (4A161L) is relatively common in the European population, is capable of expressing DUX4, but that DUX4 mRNA processing differs from 4A161S. While we do not find evidence for a difference in disease severity between FSHD carriers of an 4A161S or 4A161L allele, our study does uncover biallelic DUX4 expression in FSHD2 patients. Compared to control individuals, we observed an increased frequency of FSHD2 patients homozygous for disease permissive alleles, and who are thus capable of biallelic DUX4 expression, while SMCHD1 variant carriers with only one permissive allele were significantly more often asymptomatic. This suggests that biallelic DUX4 expression lowers the threshold for disease presentation and is a modifier for disease severity in FSHD2.


Assuntos
Genes Modificadores , Proteínas de Homeodomínio/genética , Distrofia Muscular Facioescapuloumeral/genética , Penetrância , Células Cultivadas , Feminino , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Facioescapuloumeral/patologia , Mutação de Sentido Incorreto
6.
Am J Hum Genet ; 98(5): 1020-1029, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-27153398

RESUMO

Facioscapulohumeral dystrophy (FSHD) is associated with somatic chromatin relaxation of the D4Z4 repeat array and derepression of the D4Z4-encoded DUX4 retrogene coding for a germline transcription factor. Somatic DUX4 derepression is caused either by a 1-10 unit repeat-array contraction (FSHD1) or by mutations in SMCHD1, which encodes a chromatin repressor that binds to D4Z4 (FSHD2). Here, we show that heterozygous mutations in DNA methyltransferase 3B (DNMT3B) are a likely cause of D4Z4 derepression associated with low levels of DUX4 expression from the D4Z4 repeat and increased penetrance of FSHD. Recessive mutations in DNMT3B were previously shown to cause immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome. This study suggests that transcription of DUX4 in somatic cells is modified by variations in its epigenetic state and provides a basis for understanding the reduced penetrance of FSHD within families.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Repressão Epigenética/genética , Distrofia Muscular Facioescapuloumeral/genética , Mutação/genética , Penetrância , Sequências de Repetição em Tandem/genética , Adolescente , Adulto , Idoso , Sequência de Aminoácidos , Criança , Pré-Escolar , Cromatina/genética , DNA (Citosina-5-)-Metiltransferases/química , Metilação de DNA , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Linhagem , Conformação Proteica , Homologia de Sequência de Aminoácidos , DNA Metiltransferase 3B
8.
Epigenetics ; 10(12): 1133-42, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26575099

RESUMO

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.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Epigênese Genética , Proteínas de Homeodomínio/metabolismo , Desenvolvimento Muscular/genética , Distrofia Muscular Facioescapuloumeral/genética , Diferenciação Celular/genética , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Metilação de DNA , Regulação da Expressão Gênica , Código das Histonas , Proteínas de Homeodomínio/genética , Humanos , Músculo Esquelético/metabolismo
9.
Proc Natl Acad Sci U S A ; 110(51): 20783-8, 2013 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-24297891

RESUMO

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.


Assuntos
Autoanticorpos/imunologia , Imunoglobulina G/imunologia , Proteínas Relacionadas a Receptor de LDL/imunologia , Miastenia Gravis/imunologia , Receptores Proteína Tirosina Quinases/imunologia , Receptores Colinérgicos/imunologia , Receptores de LDL/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Agrina/imunologia , Animais , Autoanticorpos/farmacologia , Linhagem Celular , Criança , Pré-Escolar , Epitopos/imunologia , Feminino , Humanos , Imunização Passiva , Imunoglobulina G/farmacologia , Proteínas Relacionadas a Receptor de LDL/antagonistas & inibidores , Masculino , Camundongos , Pessoa de Meia-Idade , Miastenia Gravis/induzido quimicamente , Miastenia Gravis/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/imunologia , Multimerização Proteica/efeitos dos fármacos , Multimerização Proteica/imunologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores de LDL/antagonistas & inibidores
10.
Am J Hum Genet ; 93(4): 744-51, 2013 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-24075187

RESUMO

Facioscapulohumeral muscular dystrophy type 1 (FSHD1) is caused by contraction of the D4Z4 repeat array on chromosome 4 to a size of 1-10 units. The residual number of D4Z4 units inversely correlates with clinical severity, but significant clinical variability exists. Each unit contains a copy of the DUX4 retrogene. Repeat contractions are associated with changes in D4Z4 chromatin structure that increase the likelihood of DUX4 expression in skeletal muscle, but only when the repeat resides in a genetic background that contains a DUX4 polyadenylation signal. Mutations in the structural maintenance of chromosomes flexible hinge domain containing 1 (SMCHD1) gene, encoding a chromatin modifier of D4Z4, also result in the increased likelihood of DUX4 expression in individuals with a rare form of FSHD (FSHD2). Because SMCHD1 directly binds to D4Z4 and suppresses somatic expression of DUX4, we hypothesized that SMCHD1 may act as a genetic modifier in FSHD1. We describe three unrelated individuals with FSHD1 presenting an unusual high clinical severity based on their upper-sized FSHD1 repeat array of nine units. Each of these individuals also carries a mutation in the SMCHD1 gene. Familial carriers of the FSHD1 allele without the SMCHD1 mutation were only mildly affected, suggesting a modifier effect of the SMCHD1 mutation. Knocking down SMCHD1 in FSHD1 myotubes increased DUX4 expression, lending molecular support to a modifier role for SMCHD1 in FSHD1. We conclude that FSHD1 and FSHD2 share a common pathophysiological pathway in which the FSHD2 gene can act as modifier for disease severity in families affected by FSHD1.


Assuntos
Proteínas Cromossômicas não Histona/genética , Distrofia Muscular Facioescapuloumeral/genética , Adolescente , Adulto , Idoso , Alelos , Sequência de Aminoácidos , Sequência de Bases , Criança , Feminino , Proteínas de Homeodomínio/genética , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Fibras Musculares Esqueléticas/metabolismo , Mutação , Linhagem , Adulto Jovem
11.
Nat Genet ; 44(12): 1370-4, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23143600

RESUMO

Facioscapulohumeral dystrophy (FSHD) is characterized by chromatin relaxation of the D4Z4 macrosatellite array on chromosome 4 and expression of the D4Z4-encoded DUX4 gene in skeletal muscle. The more common form, autosomal dominant FSHD1, is caused by contraction of the D4Z4 array, whereas the genetic determinants and inheritance of D4Z4 array contraction-independent FSHD2 are unclear. Here, we show that mutations in SMCHD1 (encoding structural maintenance of chromosomes flexible hinge domain containing 1) on chromosome 18 reduce SMCHD1 protein levels and segregate with genome-wide D4Z4 CpG hypomethylation in human kindreds. FSHD2 occurs in individuals who inherited both the SMCHD1 mutation and a normal-sized D4Z4 array on a chromosome 4 haplotype permissive for DUX4 expression. Reducing SMCHD1 levels in skeletal muscle results in D4Z4 contraction-independent DUX4 expression. Our study identifies SMCHD1 as an epigenetic modifier of the D4Z4 metastable epiallele and as a causal genetic determinant of FSHD2 and possibly other human diseases subject to epigenetic regulation.


Assuntos
Proteínas Cromossômicas não Histona/genética , Hereditariedade/genética , Proteínas de Homeodomínio/genética , Distrofia Muscular Facioescapuloumeral/genética , Mutação , Adulto , Idoso , Cromossomos Humanos Par 18/genética , Ilhas de CpG/genética , Metilação de DNA/genética , Epigênese Genética , Feminino , Haplótipos , Humanos , Masculino , Pessoa de Meia-Idade
12.
Brain ; 135(Pt 4): 1081-101, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22396395

RESUMO

Myasthenia gravis is a paralytic disorder with autoantibodies against acetylcholine receptors at the neuromuscular junction. A proportion of patients instead has antibodies against muscle-specific kinase, a protein essential for acetylcholine receptor clustering. These are generally of the immunoglobulin-G4 subclass and correlate with disease severity, suggesting specific myasthenogenic activity. However, immunoglobulin-G4 subclass antibodies are generally considered to be 'benign' and direct proof for their pathogenicity in muscle-specific kinase myasthenia gravis (or other immunoglobulin-G4-associated disorders) is lacking. Furthermore, the exact electrophysiological synaptic defects caused at neuromuscular junctions by human anti-muscle-specific kinase autoantibodies are hitherto unknown. We show that purified immunoglobulin-G4, but not immunoglobulin-G1-3, from patients with muscle-specific kinase myasthenia gravis binds to mouse neuromuscular junctions in vitro, and that injection into immunodeficient mice causes paralysis. Injected immunoglobulin-G4 caused reduced density and fragmented area of neuromuscular junction acetylcholine receptors. Detailed electrophysiological synaptic analyses revealed severe reduction of postsynaptic acetylcholine sensitivity, and exaggerated depression of presynaptic acetylcholine release during high-rate activity, together causing the (fatigable) muscle weakness. Intriguingly, compensatory transmitter release upregulation, which is the normal homeostatic response in acetylcholine receptor myasthenia gravis, was absent. This conveys extra vulnerability to neurotransmission at muscle-specific kinase myasthenia gravis neuromuscular junctions. Thus, we demonstrate that patient anti-muscle-specific kinase immunoglobulin-G4 is myasthenogenic, independent of additional immune system components, and have elucidated the underlying electrophysiological neuromuscular junction abnormalities.


Assuntos
Imunoglobulina G/efeitos adversos , Imunoglobulina G/sangue , Miastenia Gravis/sangue , Doenças da Junção Neuromuscular/complicações , Receptores Proteína Tirosina Quinases/imunologia , Receptores Colinérgicos/imunologia , Potenciais de Ação/efeitos dos fármacos , Adulto , Animais , Autoanticorpos/sangue , Modelos Animais de Doenças , Eletromiografia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Placa Motora/efeitos dos fármacos , Placa Motora/fisiopatologia , Contração Muscular/efeitos dos fármacos , Força Muscular/efeitos dos fármacos , Força Muscular/fisiologia , Miastenia Gravis/complicações , Miastenia Gravis/imunologia , Miastenia Gravis/terapia , Condução Nervosa/efeitos dos fármacos , Condução Nervosa/fisiologia , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/patologia , Junção Neuromuscular/fisiopatologia , Junção Neuromuscular/ultraestrutura , Doenças da Junção Neuromuscular/patologia , Plasmaferese/métodos , Adulto Jovem
13.
Am J Pathol ; 179(4): 1988-2000, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21854744

RESUMO

Oculopharyngeal muscular dystrophy (OPMD) is an autosomal dominant disease caused by an alanine tract expansion mutation in poly(A) binding protein nuclear 1 (expPABPN1). To model OPMD in a myogenic and physiological context, we generated mouse myoblast cell clones stably expressing either human wild type (WT) or expPABPN1 at low levels. Transgene expression is induced on myotube differentiation and results in formation of insoluble nuclear PABPN1 aggregates that are similar to those observed in patients with OPMD. Quantitative analysis of PABPN1 in myotube cultures revealed that expPABPN1 accumulation and aggregation is greater than that of the WT protein. We found that aggregation of expPABPN1 is more affected than WT PABPN1 by inhibition of proteasome activity. Consistent with this, in myotube cultures expressing expPABPN1, deregulation of the proteasome was identified as the most significantly perturbed pathway. Differences in the accumulation of soluble WT and expPABPN1 were consistent with differences in ubiquitination and rate of protein turnover. This study demonstrates, for the first time to our knowledge, that, in myotubes, the ratio of soluble/insoluble expPABPN1 is significantly lower compared with that of the WT protein. We suggest that this difference can contribute to muscle weakness in OPMD.


Assuntos
Fibras Musculares Esqueléticas/patologia , Distrofia Muscular Oculofaríngea/metabolismo , Distrofia Muscular Oculofaríngea/patologia , Proteínas Mutantes/metabolismo , Proteína II de Ligação a Poli(A)/metabolismo , Animais , Sequência de Bases , Células Cultivadas , Desmina/genética , Modelos Animais de Doenças , Humanos , Corpos de Inclusão Intranuclear/metabolismo , Camundongos , Dados de Sequência Molecular , Fibras Musculares Esqueléticas/metabolismo , Músculos/patologia , Distrofia Muscular Oculofaríngea/genética , Proteínas Mutantes/química , Proteína II de Ligação a Poli(A)/química , Proteína II de Ligação a Poli(A)/genética , Poliubiquitina/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Estrutura Quaternária de Proteína , Transdução de Sinais , Solubilidade , Transcriptoma , Transfecção , Expansão das Repetições de Trinucleotídeos/genética , Ubiquitinação
14.
Am J Hum Genet ; 88(6): 796-804, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21596365

RESUMO

Autosomal-recessive immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome is mainly characterized by recurrent, often fatal, respiratory and gastrointestinal infections. About 50% of patients carry mutations in the DNA methyltransferase 3B gene (DNMT3B) (ICF1). The remaining patients carry unknown genetic defects (ICF2) but share with ICF1 patients the same immunological and epigenetic features, including hypomethylation of juxtacentromeric repeat sequences. We performed homozygosity mapping in five unrelated ICF2 patients with consanguineous parents and then performed whole-exome sequencing in one of these patients and Sanger sequencing in all to identify mutations in the zinc-finger- and BTB (bric-a-bric, tramtrack, broad complex)-domain-containing 24 (ZBTB24) gene in four consanguineously descended ICF2 patients. Additionally, we found ZBTB24 mutations in an affected sibling pair and in one patient for whom it was not known whether his parents were consanguineous. ZBTB24 belongs to a large family of transcriptional repressors that include members, such as BCL6 and PATZ1, with prominent regulatory roles in hematopoietic development and malignancy. These data thus indicate that ZBTB24 is involved in DNA methylation of juxtacentromeric DNA and in B cell development and/or B and T cell interactions. Because ZBTB24 is a putative DNA-binding protein highly expressed in the lymphoid lineage, we predict that by studying the molecular function of ZBTB24, we will improve our understanding of the molecular pathophysiology of ICF syndrome and of lymphocyte biology in general.


Assuntos
Centrômero/genética , Metilação de DNA/genética , Proteínas Repressoras/genética , Dedos de Zinco , Adolescente , Adulto , Criança , Pré-Escolar , Análise Mutacional de DNA , Epigenômica , Face/anormalidades , Feminino , Humanos , Síndromes de Imunodeficiência/genética , Masculino , Mutação , Linhagem , Doenças da Imunodeficiência Primária
15.
Science ; 329(5999): 1650-3, 2010 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-20724583

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is a common form of muscular dystrophy in adults that is foremost characterized by progressive wasting of muscles in the upper body. FSHD is associated with contraction of D4Z4 macrosatellite repeats on chromosome 4q35, but this contraction is pathogenic only in certain "permissive" chromosomal backgrounds. Here, we show that FSHD patients carry specific single-nucleotide polymorphisms in the chromosomal region distal to the last D4Z4 repeat. This FSHD-predisposing configuration creates a canonical polyadenylation signal for transcripts derived from DUX4, a double homeobox gene of unknown function that straddles the last repeat unit and the adjacent sequence. Transfection studies revealed that DUX4 transcripts are efficiently polyadenylated and are more stable when expressed from permissive chromosomes. These findings suggest that FSHD arises through a toxic gain of function attributable to the stabilized distal DUX4 transcript.


Assuntos
Cromossomos Humanos Par 4/genética , Proteínas de Homeodomínio/genética , Distrofia Muscular Facioescapuloumeral/genética , Sequências Repetitivas de Ácido Nucleico , Adolescente , Adulto , Idoso , Sequência de Bases , Pré-Escolar , Cromossomos Humanos Par 10/genética , Feminino , Predisposição Genética para Doença , Haplótipos , Proteínas de Homeodomínio/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Genéticos , Dados de Sequência Molecular , Poliadenilação , Polimorfismo de Nucleotídeo Único , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica , Transfecção , Adulto Jovem
16.
Chromosoma ; 116(1): 53-64, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17103222

RESUMO

FRG1 is considered a candidate gene for facioscapulohumeral muscular dystrophy (FSHD) based on its location at chromosome 4qter and its upregulation in FSHD muscle. The FRG1 protein (FRG1P) localizes to nucleoli, Cajal bodies (and speckles), and has been suggested to be a component of the human spliceosome but its exact function is unknown. Recently, transgenic mice overexpressing high levels of FRG1P in skeletal muscle were described to present with muscular dystrophy. Moreover, upregulation of FRG1P was demonstrated to correlate with missplicing of specific pre-mRNAs. In this study, we have combined colocalization studies with yeast two-hybrid screens to identify proteins that associate with FRG1P. We demonstrate that artificially induced nucleolar aggregates of VSV-FRG1P specifically sequester proteins involved in pre-mRNA processing. In addition, we have identified SMN, PABPN1, and FAM71B, a novel speckle and Cajal body protein, as binding partners of FRG1P. All these proteins are, or seem to be, involved in RNA biogenesis. Our data confirm the presence of FRG1P in protein complexes containing human spliceosomes and support a potential role of FRG1P in either splicing or another step in nuclear RNA biogenesis. Intriguingly, among FRG1P-associated proteins are SMN and PABPN1, both being involved in neuromuscular disorders, possibly through RNA biogenesis-related processes.


Assuntos
Proteínas Nucleares/metabolismo , Proteínas/metabolismo , Precursores de RNA/metabolismo , Processamento Pós-Transcricional do RNA , Processamento Alternativo , Animais , Linhagem Celular , Nucléolo Celular/metabolismo , Humanos , Imunoprecipitação , Proteínas dos Microfilamentos , Distrofia Muscular Facioescapuloumeral/genética , Proteínas Nucleares/genética , Proteínas/genética , Precursores de RNA/genética , Proteínas de Ligação a RNA , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Troponina T/genética , Troponina T/metabolismo , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...