Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(3)2022 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-35163099

RESUMO

De novo somatic mutations are well documented in diseases such as neoplasia but are rarely reported in rare diseases. Hovewer, severe genetic diseases that are not compatible with embryonic development are caused exclusively by deleterious mutations that could only be found as mosaic and not as inherited mutations. We will review here the paradigmatic case of Incontinentia Pigmenti, a rare X-linked dominant disease caused by deficiency of the NEMO (also called IKKgamma) protein, which plays a pivotal role in tissue homeostasis. The loss-of-function mutations of NEMO are embryonically lethal in males while females survive because of unbalanced X-inactivation due to NEMO wild type (WT) expressing cells survival despite of NEMO mutant expressing cells. The few surviving IP males are obligatory mosaic mutants with the typical clinical presentation of IP in female. Indeed, the IP pathogenesis in the female and most likely also in the male somatic mosaics is based on the cellular effects of an impaired NEMO activity, but in the context of the interaction of genetically different cells in the affected tissue, which might underline the inflammatory status.


Assuntos
Quinase I-kappa B/deficiência , Quinase I-kappa B/genética , Incontinência Pigmentar/patologia , Mutação com Perda de Função , Mosaicismo , Humanos , Incontinência Pigmentar/etiologia , Incontinência Pigmentar/metabolismo , Masculino
2.
J Cell Physiol ; 231(1): 152-61, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26060140

RESUMO

NEMO/IKKγ is the regulatory subunit of the IκB Kinase (IKK) complex, required for the activation of the NF-κB pathway, which is involved in a variety of key processes, including immunity, inflammation, differentiation, and cell survival. Termination of NF-κB activity on specific -κB responsive genes, which is crucial for the resolution of inflammatory responses, can be achieved by direct degradation of the chromatin-bound NF-κB subunit RelA/p65, a process mediated by a protein complex that contains Copper Metabolism Murr1 Domain 1 (COMMD1). In this study, we identify COMMD7, another member of the COMMDs protein family, as a novel NEMO-interacting protein. We show that COMMD7 exerts an inhibitory effect on NF-κB activation upon TNFα stimulation. COMMD7 interacts with COMMD1 and together they cooperate to down-regulate NF-κB activity. Accordingly, termination of TNFα-induced NF-κB activity on the -κB responsive gene, Icam1, is defective in cells silenced for COMMD7 expression. Furthermore, this impairment is not greatly increased when we silence the expression of both COMMD7 and COMMD1 indicating that the two proteins participate in the same pathway of termination of TNFα-induced NF-κB activity. Importantly, we have demonstrated that COMMD7's binding to NEMO does not interfere with the binding to the IKKs, and that the disruption of the IKK complex through the use of the NBP competitor impairs the termination of NF-κB activity. We propose that an intact IKK complex is required for the termination of NF-κB-dependent transcription and that COMMD7 acts as a scaffold in the IKK-mediated NF-κB termination.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Diferenciação Celular/fisiologia , Quinase I-kappa B/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Diferenciação Celular/genética , Linhagem Celular , Sobrevivência Celular/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Fosforilação
3.
Am J Hum Genet ; 92(1): 114-25, 2013 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23246292

RESUMO

Intellectual disability (ID) and epilepsy often occur together and have a dramatic impact on the development and quality of life of the affected children. Polyalanine (polyA)-expansion-encoding mutations of aristaless-related homeobox (ARX) cause a spectrum of X-linked ID (XLID) diseases and chronic epilepsy, including infantile spasms. We show that lysine-specific demethylase 5C (KDM5C), a gene known to be mutated in XLID-affected children and involved in chromatin remodeling, is directly regulated by ARX through the binding in a conserved noncoding element. We have studied altered ARX carrying various polyA elongations in individuals with XLID and/or epilepsy. The changes in polyA repeats cause hypomorphic ARX alterations, which exhibit a decreased trans-activity and reduced, but not abolished, binding to the KDM5C regulatory region. The altered functioning of the mutants tested is likely to correlate with the severity of XLID and/or epilepsy. By quantitative RT-PCR, we observed a dramatic Kdm5c mRNA downregulation in murine Arx-knockout embryonic and neural stem cells. Such Kdm5c mRNA diminution led to a severe decrease in the KDM5C content during in vitro neuronal differentiation, which inversely correlated with an increase in H3K4me3 signal. We established that ARX polyA alterations damage the regulation of KDM5C expression, and we propose a potential ARX-dependent path acting via chromatin remodeling.


Assuntos
Epilepsia/genética , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Deficiência Intelectual Ligada ao Cromossomo X/genética , Oxirredutases N-Desmetilantes/genética , Fatores de Transcrição/genética , Animais , Criança , Expansão das Repetições de DNA , Histona Desmetilases , Humanos , Camundongos , Camundongos Knockout , Peptídeos/genética
4.
Clin Immunol ; 161(2): 131-5, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26307434

RESUMO

Hypohidrotic ectodermal dysplasia (HED) consists of disorders resulting from molecular alterations of ectodysplasin-A (EDA) pathway. Hypomorphic mutations in NF-kB essential modulator, downstream EDA, result in HED with immunodeficiency (HED-ID), characterized by susceptibility to encapsulated pyogenic bacteria infections. Increased susceptibility to pneumococcal infections and poor response to polysaccharide antigens are associated with defect in T-independent B-cell immunity. We investigated B-cell differentiation and immunoglobulin secretion induced by the TLR9 ligand CpG in two HED-ID and in a HED patient caused by EDA mutations (XLHED). In HED-ID, only few B cells differentiated into plasma cells upon TLR9 stimulation and memory B cells did not produce IgG and IgA, but small amounts of IgM. Unexpectedly, memory B cells from XLHED patient failed to produce normal IgA or IgG amount upon TLR9 stimulation. Our findings expand the knowledge about the pathogenesis of humoral alterations in HED patients and help explain the susceptibility to pneumococcal infections.


Assuntos
Células Produtoras de Anticorpos/imunologia , Linfócitos B/imunologia , Diferenciação Celular/imunologia , Síndromes de Imunodeficiência/imunologia , NF-kappa B/imunologia , Receptor Toll-Like 9/imunologia , Criança , Pré-Escolar , Suscetibilidade a Doenças/imunologia , Displasia Ectodérmica Anidrótica Tipo 1/imunologia , Humanos , Imunoglobulinas/imunologia , Ligantes , Masculino , Infecções Pneumocócicas/imunologia , Linfócitos T/imunologia
5.
Hum Mutat ; 35(2): 165-77, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24339369

RESUMO

Incontinentia pigmenti (IP) is an X-linked-dominant Mendelian disorder caused by mutation in the IKBKG/NEMO gene, encoding for NEMO/IKKgamma, a regulatory protein of nuclear factor kappaB (NF-kB) signaling. In more than 80% of cases, IP is due to recurrent or nonrecurrent deletions causing loss-of-function (LoF) of NEMO/IKKgamma. We review how the local architecture of the IKBKG/NEMO locus with segmental duplication and a high frequency of repetitive elements favor de novo aberrant recombination through different mechanisms producing genomic microdeletion. We report here a new microindel (c.436_471delinsT, p.Val146X) arising through a DNA-replication-repair fork-stalling-and-template-switching and microhomology-mediated-end-joining mechanism in a sporadic IP case. The LoF mutations of IKBKG/NEMO leading to IP include small insertions/deletions (indel) causing frameshift and premature stop codons, which account for 10% of cases. We here present 21 point mutations previously unreported, which further extend the spectrum of pathologic variants: 14/21 predict LoF because of premature stop codon (6/14) or frameshift (8/14), whereas 7/21 predict a partial loss of NEMO/IKKgamma activity (two splicing and five missense). We review how the analysis of IP-associated IKBKG/NEMO hypomorphic mutants has contributed to the understanding of the pathophysiological mechanism of IP disease and has provided important information on affected NF-kB signaling. We built a locus-specific database listing all IKBKG/NEMO variants, accessible at http://IKBKG.lovd.nl.


Assuntos
Códon sem Sentido , Mutação da Fase de Leitura , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Incontinência Pigmentar/genética , NF-kappa B/metabolismo , Animais , Sequência de Bases , Cromossomos Humanos X , Variação Genética , Genótipo , Humanos , Incontinência Pigmentar/patologia , Mutação de Sentido Incorreto , NF-kappa B/genética , Fenótipo , Mutação Puntual , Deleção de Sequência , Transdução de Sinais
6.
Hum Mol Genet ; 21(6): 1260-71, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22121116

RESUMO

IKBKG/NEMO gene mutations cause an X-linked, dominant neuroectodermal disorder named Incontinentia Pigmenti (IP). Located at Xq28, IKBKG/NEMO has a unique genomic organization, as it is part of a segmental duplication or low copy repeat (LCR1-LCR2, >99% identical) containing the gene and its pseudogene copy (IKBKGP). In the opposite direction and outside LCR1, IKBKG/NEMO partially overlaps G6PD, whose mutations cause a common X-linked human enzymopathy. The two LCRs in the IKBKG/NEMO locus are able to recombine through non-allelic homologous recombination producing either a pathological recurrent exon 4-10 IKBKG/NEMO deletion (IKBKGdel) or benign small copy number variations. We here report that the local high frequency of micro/macro-homologies, tandem repeats and repeat/repetitive sequences make the IKBKG/NEMO locus susceptible to novel pathological IP alterations. Indeed, we describe the first two independent instances of inter-locus gene conversion, occurring between the two LCRs, that copies the IKBKGP pseudogene variants into the functional IKBKG/NEMO, causing the de novo occurrence of p.Glu390ArgfsX61 and the IKBKGdel mutations, respectively. Subsequently, by investigating a group of 20 molecularly unsolved IP subjects using a high-density quantitative polymerase chain reaction assay, we have identified seven unique de novo deletions varying from 4.8 to ∼115 kb in length. Each deletion removes partially or completely both IKBKG/NEMO and the overlapping G6PD, thereby uncovering the first deletions disrupting the G6PD gene which were found in patients with IP. Interestingly, the 4.8 kb deletion removes the conserved bidirectional promoterB, shared by the two overlapping IKBKG/NEMO and G6PD genes, leaving intact the alternative IKBKG/NEMO unidirectional promoterA. This promoter, although active in the keratinocytes of the basal dermal layer, is down-regulated during late differentiation. Genomic analysis at the breakpoint sites indicated that other mutational forces, such as non-homologous end joining, Alu-Alu-mediated recombination and replication-based events, might enhance the vulnerability of the IP locus to produce de novo pathological IP alleles.


Assuntos
Genômica , Glucosefosfato Desidrogenase/genética , Quinase I-kappa B/genética , Incontinência Pigmentar/genética , Incontinência Pigmentar/patologia , Regiões Promotoras Genéticas/genética , Deleção de Sequência/genética , Alelos , Diferenciação Celular , Células Cultivadas , Feminino , Recombinação Homóloga , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Masculino , Repetições de Microssatélites , Pseudogenes/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Sequências Repetitivas de Ácido Nucleico/genética
8.
Hum Mol Genet ; 19(16): 3138-49, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20529958

RESUMO

NF-kappaB Essential MOdulator (NEMO) has been shown to play a critical role in NF-kappaB activation, as the regulatory subunit of IkappaB kinase. Upon cell stimulation, NEMO can be modified through phosphorylation, sumoylation or ubiquitination. In the latter case, not much is known regarding the exact function of this posttranslational modification. One of the E3 ligase responsible for K63-linked NEMO polyubiquitination is TRAF6, which participates in several signaling pathways controlling immunity, osteoclastogenesis, skin development and brain functions. We previously observed a potentially important interaction between NEMO and TRAF6. In this study, we defined in more detail the domains required for this interaction, uncovering a new binding site for TRAF6 located at the amino-terminus of NEMO and recognized by the coiled-coil domain of TRAF6. This site appears to work in concert with the previously identified NEMO ubiquitin-binding domain which binds polyubiquitinated chains, suggesting a dual mode of TRAF6 recognition. We also showed that E57K mutation of NEMO found in a mild form of the genetic disease incontinentia pigmenti, resulted in impaired TRAF6 binding and IL-1beta signaling. In contrast, activation of NF-kappaB by TNF-alpha was not affected. These data demonstrate that NEMO/TRAF6 interaction has physiological relevance and might represent a new target for therapeutic purposes.


Assuntos
Quinase I-kappa B/genética , Incontinência Pigmentar/genética , Mutação , Fator 6 Associado a Receptor de TNF/genética , Substituição de Aminoácidos , Animais , Sítios de Ligação/genética , Western Blotting , Linhagem Celular , Células Cultivadas , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Quinase I-kappa B/química , Quinase I-kappa B/metabolismo , Imunoprecipitação , Incontinência Pigmentar/metabolismo , Incontinência Pigmentar/patologia , Interleucina-1beta/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Knockout , Modelos Moleculares , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Fator 6 Associado a Receptor de TNF/metabolismo
10.
Nat Commun ; 13(1): 5529, 2022 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-36130971

RESUMO

Dysregulated secretion in neutrophil leukocytes associates with human inflammatory disease. The exocytosis response to triggering stimuli is sequential; gelatinase granules modulate the initiation of the innate immune response, followed by the release of pro-inflammatory azurophilic granules, requiring stronger stimulation. Exocytosis requires actin depolymerization which is actively counteracted under non-stimulatory conditions. Here we show that the actin nucleator, WASH, is necessary to maintain azurophilic granules in their refractory state by granule actin entrapment and interference with the Rab27a-JFC1 exocytic machinery. On the contrary, gelatinase granules of WASH-deficient neutrophil leukocytes are characterized by decreased Rac1, shortened granule-associated actin comets and impaired exocytosis. Rac1 activation restores exocytosis of these granules. In vivo, WASH deficiency induces exacerbated azurophilic granule exocytosis, inflammation, and decreased survival. WASH deficiency thus differentially impacts neutrophil granule subtypes, impairing exocytosis of granules that mediate the initiation of the neutrophil innate response while exacerbating pro-inflammatory granule secretion.


Assuntos
Actinas , Neutrófilos , Grânulos Citoplasmáticos , Exocitose , Gelatinases , Humanos , Inflamação , Proteínas dos Microfilamentos
11.
J Biol Chem ; 285(22): 16951-7, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20308062

RESUMO

The Arp2/3 complex is essential for actin filament nucleation in a variety of cellular processes. The activation of the Arp2/3 complex is mediated by nucleation-promoting factors, such as the Wiskott-Aldrich syndrome family proteins, which share a WCA (WH2 domain, central region, acidic region) catalytic module at the C-terminal region, required for Arp2/3 activation, but diverge at the N-terminal region, required for binding to specific activators. Here, we report the characterization of WASH, a new member of the WAS family that has nucleation-promoting factor activity and recently has been demonstrated to play a role in endosomal sorting. We found that overexpression of the WASH-WCA domain induced disruption of the actin cytoskeleton, whereas overexpression of full-length WASH in mammalian cells did not affect stress fiber organization. Furthermore, our analysis has revealed that nerve growth factor treatment of PC12 cells overexpressing full-length WASH leads to disruption of the actin cytoskeleton. We have also found that WASH interacts through its N-terminal region with BLOS2, a centrosomal protein belonging to the BLOC-1 complex that functions as a scaffolding factor in the biogenesis of lysosome-related organelles. In addition to BLOS2, WASH also interacts with centrosomal gamma-tubulin and with pallidin, an additional component of the BLOC-1 complex. Collectively, our data propose that WASH is a bimodular protein in which the C terminus is involved in Arp2/3-mediated actin nucleation, whereas the N-terminal portion is required for its regulation and localization in the cells. Moreover, our data suggest that WASH is also a component of the BLOC-1 complex that is associated with the centrosomes.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Regulação da Expressão Gênica , Lisossomos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas/metabolismo , Tubulina (Proteína)/metabolismo , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Camundongos , Modelos Biológicos , Células PC12 , Ratos
12.
Hum Reprod ; 26(11): 3186-96, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21859812

RESUMO

BACKGROUND: Diminished ovarian reserve (DOR) is a heterogeneous disorder causing infertility, characterized by a decreased number of oocytes, the genetic cause of which is still unknown. METHODS AND RESULTS: We describe a family with a new unbalanced X;18 translocation der(X) associated with either fully attenuated or DOR phenotype in the same family. Cytogenetics and array comparative genomic hybridization (aCGH) studies have revealed the same partial Xq monosomy and partial 18q trisomy in both the 32-year-old female with DOR and the unaffected mother. The genetic analysis has defined a subtelomeric deletion spanning 13.3 Mb from Xq27.3 to -Xqter, which covers the premature ovarian failure locus 1 (POF1); and a duplication spanning 13.4 Mb, from 18q22.1 to 18qter. From a parental-origin study, we have inferred that the rearranged X chromosome is maternally derived. The Xq27 and 18q22 breakpoint regions fall in a region extremely rich in long interspersed nuclear element, a class of retrotransposons able to trigger mispairing and unusual crossovers. X-inactivation studies reveal a skewing of der(X) both in the mother and the proband. Therefore, the phenotypic expression of der(X) is fully attenuated in the fertile mother and partially attenuated in the DOR daughter. CONCLUSIONS: We report on an unbalanced maternally derived translocation (X;18)(q27;q22) with different intra-familial reproductive performances, ranging from fertility to DOR. Skewed X-inactivation seems to restore the unbalanced genetic make-up, fully silencing the 18q22 trisomy and at least in part the Xq27 monosomy. The chromosomal abnormality observed in this family supports the presence of a DOR susceptibility locus in the distal Xq region and targets the POF1 region for further investigation.


Assuntos
Aberrações Cromossômicas , Cromossomos Humanos Par 18/genética , Cromossomos Humanos X/genética , Doenças Ovarianas/genética , Ovário/fisiologia , Insuficiência Ovariana Primária/genética , Adolescente , Adulto , Núcleo Celular/metabolismo , Feminino , Inativação Gênica , Humanos , Hibridização in Situ Fluorescente , Masculino , Repetições de Microssatélites/genética , Pessoa de Meia-Idade , Hibridização de Ácido Nucleico , Oócitos/citologia , Fenótipo , Reação em Cadeia da Polimerase em Tempo Real/métodos , Trissomia/genética
14.
Methods Mol Biol ; 2366: 243-254, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34236642

RESUMO

The engagement of TNF on TNFR can result in cell survival or cell death depending on the different complex formation downstream this interaction. Here we describe reagents and assay procedures that can be used to study caspase-independent cell death (necroptosis) in cultured cells, in response to pharmacological interventions with NF-kappaB and death inhibitors. We provide protocol to detect death-specific proteins using immunoblot and to dissect necrosome complex by sequential co-immunoprecipitation of death-specific components during necroptosis.


Assuntos
Apoptose , Necroptose , Células Cultivadas , Humanos , NF-kappa B/metabolismo , Necrose , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
15.
Cell Death Dis ; 11(3): 210, 2020 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-32231206

RESUMO

Escaping programmed cell death is a hallmark of cancer. NF-κB transcription factors are key regulator of cell survival and aberrant NF-κB signaling has been involved in the pathogenesis of most human malignancies. Although NF-κB is best known for its antiapoptotic role, other processes regulating the life/death balance, such as autophagy and necroptosis, seem to network with NF-κB. This review discusses how the reciprocal regulation of NF-κB, autophagy and programmed cell death affect cancer development and progression.


Assuntos
NF-kappa B/metabolismo , Neoplasias/genética , Autofagia , Humanos , Transdução de Sinais
17.
Hum Mutat ; 30(9): 1284-91, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19603533

RESUMO

The Incontinentia Pigmenti (IP) locus contains the IKBKG/NEMO/IKKgamma gene and its truncated pseudogene copy, IKBKGP/deltaNEMO. The major genetic defect in IP is a heterozygous exon4_10 IKBKG deletion (IKBKGdel) caused by a recombination between two consecutive MER67B repeats. We analyzed 91 IP females carrying the IKBKGdel, 59 of whom carrying de novo mutations (65%). In eight parents, we found two recurrent nonpathological variants of IP locus, which were also present as rare polymorphism in control population: the IKBKGPdel, corresponding to the exon4_10 deletion in the pseudogene, and the MER67Bdup, that replicates the exon4_10 region downstream of the normal IKBKG gene. Using quantitative DNA analysis and microsatellite mapping, we established that both variants might promote the generation of the pathological IKBKGdel. Indeed, in family IP-516, the exon4_10 deletion was repositioned in the same allele from the pseudogene to the gene, whereas in family IP-688, the MER67Bdup generated the pathological IKBKGdel by recombination between two direct nonadjacent MER67Bs. Moreover, we found an instance of somatic recombination in a MER67Bdup variant, creating the IKBKGdel in an IP male. Our data suggest that the IP locus undergoes recombination producing recurrent variants that might be "at risk" of generating de novo IKBKGdel by NAHR during either meiotic or mitotic division.


Assuntos
Cromossomos Humanos X/genética , Éxons/genética , Duplicação Gênica , Quinase I-kappa B/genética , Incontinência Pigmentar/genética , Deleção de Sequência , Sequência de Bases , Família , Feminino , Humanos , Quinase I-kappa B/metabolismo , Incontinência Pigmentar/metabolismo , Masculino , Modelos Genéticos , Dados de Sequência Molecular , Linhagem
18.
J Cell Physiol ; 220(2): 410-7, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19365808

RESUMO

NEMO/IKKgamma is the essential regulatory subunit of the IkB Kinase (IKK) complex, required for the activation of Nuclear Factor kB (NF-kB) in many physiological processes such as inflammation, immunity, apoptosis, or development. NEMO works at a converging point of the NF-kB pathway as it interacts with upstream signaling molecules to orchestrate its activation. Here we report on the identification of a novel NEMO-interacting protein, NESCA, an adapter molecule previously shown to be involved in the NGF-pathway via the TrkA receptor. We demonstrated that NESCA and NEMO interact by their N-terminal region. Beside to NEMO, we revealed that NESCA directly associates to the E3 ubiquitin ligase TRAF6, which in turn catalyzes NESCA polyubiquitination. Finally, we demonstrated that NESCA overexpression strongly inhibits TRAF6-mediated polyubiquitination of NEMO. In summary, our results highlight that NESCA represents a novel missing link in the NEMO-mediated NF-kB activation pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Quinase I-kappa B/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular , Humanos , Quinase I-kappa B/genética , Poliubiquitina/metabolismo , Ligação Proteica , Fator 6 Associado a Receptor de TNF/genética , Técnicas do Sistema de Duplo-Híbrido , Ubiquitinação
19.
Eur J Hum Genet ; 27(10): 1509-1518, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31231133

RESUMO

Incontinentia pigmenti (IP; OMIM#308300) is a rare genetic disease resulting in neuroectodermal defects, which can lead to disability. At present, there is neither definitive cure available nor are there any sufficiently reliable insights to predict the severity of the disease. We launched the Incontinentia Pigmenti Genetic Biobank (IPGB) project ( http://www.igb.cnr.it/ipgb ) in 2015 to establish a large-scale deposit of biological samples, to provide detailed clinical information about children diagnosed with IP and to facilitate research. We have built a cohort comprising samples of 381 clinically confirmed patients with IP and 633 healthy individuals recruited through IP patients' associations. The collection includes 269 trios, 83 duos, and 95 families with at least two affected members and represents an extensive dataset (200 cooperative medical institutes, 139 in Italy and 61 worldwide) that enables a comprehensive phenotyping. Joining the IPGB guarantees all participants access to the results including the genetic testing of IP and the long-term storage of the samples. The IPGB is the largest IP sample collection and one of the largest rare-disease-oriented collections in the world and will be open to requests for access to data by the national and international scientific community.


Assuntos
Bancos de Espécimes Biológicos , Incontinência Pigmentar/genética , Doenças Raras , Bases de Dados Genéticas , Feminino , Geografia Médica , Saúde Global , Humanos , Incontinência Pigmentar/diagnóstico , Incontinência Pigmentar/epidemiologia , Masculino , Linhagem , Sistema de Registros , Pesquisa , Projetos de Pesquisa , Fatores Socioeconômicos , Navegador
20.
Hum Mutat ; 29(5): 595-604, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18350553

RESUMO

Mutations in the inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase gamma (IKBKG), also called nuclear factor-kappaB (NF-kB) essential modulator (NEMO), gene are the most common single cause of incontinentia pigmenti (IP) in females and anhydrotic ectodermal dysplasia with immunodeficiency (EDA-ID) in males. The IKBKG gene, located in the Xq28 chromosomal region, encodes for the regulatory subunit of the inhibitor of kappaB (IkB) kinase (IKK) complex required for the activation of the NF-kB pathway. Therefore, the remarkably heterogeneous and often severe clinical presentation reported in IP is due to the pleiotropic role of this signaling transcription pathway. A recurrent exon 4_10 genomic rearrangement in the IKBKG gene accounts for 60 to 80% of IP-causing mutations. Besides the IKBKG rearrangement found in IP females (which is lethal in males), a total of 69 different small mutations (missense, frameshift, nonsense, and splice-site mutations) have been reported, including 13 novel ones in this work. The updated distribution of all the IP- and EDA-ID-causing mutations along the IKBKG gene highlights a secondary hotspot mutation in exon 10, which contains only 11% of the protein. Furthermore, familial inheritance analysis revealed an unexpectedly high incidence of sporadic cases (>65%). The sum of the observations can aid both in determining the molecular basis of IP and EDA-ID allelic diseases, and in genetic counseling in affected families.


Assuntos
Quinase I-kappa B/genética , Mutação , Sequência de Aminoácidos , Animais , Sequência de Bases , Cromossomos Humanos X , DNA , Displasia Ectodérmica/genética , Feminino , Humanos , Quinase I-kappa B/química , Incontinência Pigmentar/genética , Masculino , Camundongos , Dados de Sequência Molecular , Mosaicismo , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa