Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Biochem Biophys Res Commun ; 645: 147-153, 2023 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-36689811

RESUMO

PIDDosome formation followed by caspase-2 activation is critical for genotoxic stress-induced apoptotic cell death. Failure of proper caspase-2 activation causes a neurodevelopmental disorder and intellectual disability. R815W, R862W, and Q863stop mutations in p53-induced protein with a death domain (PIDD), a component of the PIDDosome, also lead to this disorder. However, the molecular mechanisms underlying this pathogenesis remain elusive. In this study, we analyzed the molecular mechanisms underlying the pathogenesis of the PIDD DD pathogenic variants R815W, R862W, and Q863stop. We determined that these mutations prevented the interaction between PIDD and RIP-associated Ich-1/Ced-3 homologous protein with a death domain (RAIDD), a molecule that mediates PIDDosome formation. The disruption of this interaction affects PIDDosome formation and caspase-2 activation.


Assuntos
Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte , Transtornos do Neurodesenvolvimento , Humanos , Apoptose/genética , Caspase 2/genética , Caspase 2/metabolismo , Proteína Adaptadora de Sinalização CRADD/genética , Proteína Adaptadora de Sinalização CRADD/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Transtornos do Neurodesenvolvimento/genética
2.
Biochem Soc Trans ; 50(2): 813-824, 2022 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-35343572

RESUMO

The death fold domain-containing protein PIDD1 has recently attracted renewed attention as a regulator of the orphan cell death-related protease, Caspase-2. Caspase-2 can activate p53 to promote cell cycle arrest in response to centrosome aberrations, and its activation requires formation of the PIDDosome multi-protein complex containing multimers of PIDD1 and the adapter RAIDD/CRADD at its core. However, PIDD1 appears to be able to engage with multiple client proteins to promote an even broader range of biological responses, such as NF-κB activation, translesion DNA synthesis or cell death. PIDD1 shows features of inteins, a class of self-cleaving proteins, to create different polypeptides from a common precursor protein that allow it to serve these diverse functions. This review summarizes structural information and molecular features as well as recent experimental advances that highlight the potential pathophysiological roles of this unique death fold protein to highlight its drug-target potential.


Assuntos
Proteína Adaptadora de Sinalização CRADD , Caspase 2 , Apoptose/fisiologia , Proteína Adaptadora de Sinalização CRADD/genética , Proteína Adaptadora de Sinalização CRADD/metabolismo , Caspase 2/genética , Caspase 2/metabolismo , Caspases/metabolismo , Pontos de Checagem do Ciclo Celular , Morte Celular , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Humanos , Inflamação
3.
Eur J Med Genet ; 64(4): 104181, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33647455

RESUMO

In a consanguineous Pakistani kinship afflicted with mild to moderate intellectual disability (ID), mild lissencephaly, brain atrophy and skeletal anomalies, we detected homozygous CRADD c.2T > G (p.Met1?) and USP44 c.873_886delinsT (p.Leu291Phefs*8), two good candidates 1.85-Mb apart that segregated with the disorder. Biallelic damaging variants in CRADD cause recessive mental retardation-34 (MRT34; MIM 614499) with mild to moderate ID, "thin" lissencephaly, and variable megalencephaly and seizures. For USP44, only a single ID family has been reported with a homozygous deleterious variant, which is the same as the variant we detected. In affected individuals we present, at ages 29-32 years, clinical findings are similar yet not fully concordant with phenotypes for either gene considering the skeletal findings, and ID is not as severe as would be expected for defects in two genes with additive effect. Some variable CRADD-related features such as language impairment and seizures are not observed in the presented family. The presence of the two variants in the family is a very rare example of familial linked homozygous variants, and whether the damaging USP44 variant contributed to the disease in the family we present is not clear. As for the skeletal findings, facial dysmorphism and digestive problems, we did not find a candidate variant. This study is an example of both clinical variation and difficulty in variant detection and evaluation. Our findings highlight that even an extensive exome sequence analysis can fail to fully uncover the complex molecular basis of a syndrome even if potentially causative variants are identified.


Assuntos
Proteína Adaptadora de Sinalização CRADD/genética , Deficiências do Desenvolvimento/genética , Deficiência Intelectual/genética , Lisencefalia/genética , Anormalidades Musculoesqueléticas/genética , Estrabismo/genética , Ubiquitina Tiolesterase/genética , Adulto , Consanguinidade , Deficiências do Desenvolvimento/patologia , Feminino , Humanos , Deficiência Intelectual/patologia , Lisencefalia/patologia , Masculino , Anormalidades Musculoesqueléticas/patologia , Mutação , Linhagem , Estrabismo/patologia , Síndrome
4.
Transl Psychiatry ; 11(1): 1, 2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33414379

RESUMO

PIDD1 encodes p53-Induced Death Domain protein 1, which acts as a sensor surveilling centrosome numbers and p53 activity in mammalian cells. Early results also suggest a role in DNA damage response where PIDD1 may act as a cell-fate switch, through interaction with RIP1 and NEMO/IKKg, activating NF-κB signaling for survival, or as an apoptosis-inducing protein by activating caspase-2. Biallelic truncating mutations in CRADD-the protein bridging PIDD1 and caspase-2-have been reported in intellectual disability (ID), and in a form of lissencephaly. Here, we identified five families with ID from Iran, Pakistan, and India, with four different biallelic mutations in PIDD1, all disrupting the Death Domain (DD), through which PIDD1 interacts with CRADD or RIP1. Nonsense mutations Gln863* and Arg637* directly disrupt the DD, as does a missense mutation, Arg815Trp. A homozygous splice mutation in the fifth family is predicted to disrupt splicing upstream of the DD, as confirmed using an exon trap. In HEK293 cells, we show that both Gln863* and Arg815Trp mutants fail to co-localize with CRADD, leading to its aggregation and mis-localization, and fail to co-precipitate CRADD. Using genome-edited cell lines, we show that these three PIDD1 mutations all cause loss of PIDDosome function. Pidd1 null mice show decreased anxiety, but no motor abnormalities. Together this indicates that PIDD1 mutations in humans may cause ID (and possibly lissencephaly) either through gain of function or secondarily, due to altered scaffolding properties, while complete loss of PIDD1, as modeled in mice, may be well tolerated or is compensated for.


Assuntos
Proteína Adaptadora de Sinalização CRADD , Deficiência Intelectual , Animais , Proteína Adaptadora de Sinalização CRADD/genética , Proteína Adaptadora de Sinalização CRADD/metabolismo , Caspase 2/genética , Caspase 2/metabolismo , Domínio de Morte , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Células HEK293 , Humanos , Índia , Deficiência Intelectual/genética , Camundongos , Mutação
5.
EMBO J ; 40(4): e104844, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33350486

RESUMO

Centrosome amplification results into genetic instability and predisposes cells to neoplastic transformation. Supernumerary centrosomes trigger p53 stabilization dependent on the PIDDosome (a multiprotein complex composed by PIDD1, RAIDD and Caspase-2), whose activation results in cleavage of p53's key inhibitor, MDM2. Here, we demonstrate that PIDD1 is recruited to mature centrosomes by the centriolar distal appendage protein ANKRD26. PIDDosome-dependent Caspase-2 activation requires not only PIDD1 centrosomal localization, but also its autoproteolysis. Following cytokinesis failure, supernumerary centrosomes form clusters, which appear to be necessary for PIDDosome activation. In addition, in the context of DNA damage, activation of the complex results from a p53-dependent elevation of PIDD1 levels independently of centrosome amplification. We propose that PIDDosome activation can in both cases be promoted by an ANKRD26-dependent local increase in PIDD1 concentration close to the centrosome. Collectively, these findings provide a paradigm for how centrosomes can contribute to cell fate determination by igniting a signalling cascade.


Assuntos
Proteína Adaptadora de Sinalização CRADD/metabolismo , Caspase 2/metabolismo , Centrossomo/metabolismo , Cisteína Endopeptidases/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Células A549 , Proteína Adaptadora de Sinalização CRADD/genética , Caspase 2/genética , Diferenciação Celular , Cisteína Endopeptidases/genética , Dano ao DNA , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Transdução de Sinais , Proteína Supressora de Tumor p53/genética
6.
Eur J Hum Genet ; 27(8): 1235-1243, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30914828

RESUMO

Intellectual disability (ID), megalencephaly, frontal predominant pachygyria, and seizures, previously called "thin" lissencephaly, are reported to be caused by recessive variants in CRADD. Among five families of different ethnicities identified, one homozygous missense variant, c.509G>A p.(Arg170His), was of Finnish ancestry. Here we report on the phenotypic variability associated for this potential CRADD founder variant in 22 Finnish individuals. Exome sequencing was used to identify candidate genes in Finnish patients presenting with ID. Targeted Sanger sequencing and restriction enzyme analysis were applied to screen for the c.509G>A CRADD variant in cohorts from Finland. Detailed phenotyping and genealogical studies were performed. Twenty two patients were identified with the c.509G>A p.(Arg170His) homozygous variant in CRADD. The majority of the ancestors originated from Northeastern Finland indicating a founder effect. The hallmark of the disease is frontotemporal predominant pachygyria with mild cortical thickening. All patients show ID of variable severity. Aggressive behavior was found in nearly half of the patients, EEG abnormalities in five patients and megalencephaly in three patients. This study provides detailed data about the phenotypic spectrum of patients with lissencephaly due to a CRADD variant that affects function. High inter- and intrafamilial phenotypic heterogeneity was identified in patients with pachygyria caused by the homozygous CRADD founder variant. The phenotype variability suggests that additional genetic and/or environmental factors play a role in the clinical presentation. Since frontotemporal pachygyria is the hallmark of the disease, brain imaging studies are essential to support the molecular diagnosis for individuals with ID and a CRADD variant.


Assuntos
Proteína Adaptadora de Sinalização CRADD/genética , Efeito Fundador , Predisposição Genética para Doença/genética , Lisencefalia/genética , Mutação de Sentido Incorreto , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Encéfalo/patologia , Saúde da Família , Feminino , Finlândia , Geografia , Homozigoto , Humanos , Lisencefalia/diagnóstico por imagem , Lisencefalia/patologia , Imageamento por Ressonância Magnética/métodos , Masculino , Linhagem , Fenótipo , Sequenciamento do Exoma
7.
Nat Commun ; 10(1): 410, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30679432

RESUMO

The contribution of de novo variants in severe intellectual disability (ID) has been extensively studied whereas the genetics of mild ID has been less characterized. To elucidate the genetics of milder ID we studied 442 ID patients enriched for mild ID (>50%) from a population isolate of Finland. Using exome sequencing, we show that rare damaging variants in known ID genes are observed significantly more often in severe (27%) than in mild ID (13%) patients. We further observe a significant enrichment of functional variants in genes not yet associated with ID (OR: 2.1). We show that a common variant polygenic risk significantly contributes to ID. The heritability explained by polygenic risk score is the highest for educational attainment (EDU) in mild ID (2.2%) but lower for more severe ID (0.6%). Finally, we identify a Finland enriched homozygote variant in the CRADD ID associated gene.


Assuntos
Variações do Número de Cópias de DNA/genética , Variação Genética/genética , Genoma Humano/genética , Deficiência Intelectual/epidemiologia , Deficiência Intelectual/genética , Proteína Adaptadora de Sinalização CRADD/genética , Disfunção Cognitiva/epidemiologia , Disfunção Cognitiva/genética , Estudos de Coortes , Exoma , Feminino , Finlândia/epidemiologia , Estudos de Associação Genética , Doenças Genéticas Inatas/epidemiologia , Doenças Genéticas Inatas/genética , Predisposição Genética para Doença , Geografia , Homozigoto , Humanos , Deficiência Intelectual/diagnóstico , Masculino , Herança Multifatorial , Mutação , Transtornos do Neurodesenvolvimento/epidemiologia , Transtornos do Neurodesenvolvimento/genética , Patologia Molecular , Prevalência , Sequenciamento do Exoma
8.
PLoS One ; 13(10): e0205042, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30281648

RESUMO

Abnormal regulation of caspase-2-mediated neuronal cell death causes neurodegenerative diseases and defective brain development. PIDDosome is caspase-2 activating complex composed of PIDD, RAIDD, and caspase-2. Recent whole-exome sequencing study showed that the RAIDD mutations in the death domain (DD), including G128R, F164C, R170C, and R170H mutations, cause thin lissencephaly (TLIS) by reducing caspase-2-mediated neuronal apoptosis. Given that the molecular structure of the RAIDD DD:PIDD DD complex is available, in this study, we analyzed the molecular mechanisms underlying TLIS caused by the RAIDD TLIS variants by performing mutagenesis and biochemical assays.


Assuntos
Proteína Adaptadora de Sinalização CRADD/genética , Lisencefalia/etiologia , Lisencefalia/genética , Sequência de Aminoácidos , Animais , Proteína Adaptadora de Sinalização CRADD/química , Proteína Adaptadora de Sinalização CRADD/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação Puntual , Domínios Proteicos
9.
Cell Death Dis ; 9(1): 13, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29317636

RESUMO

Chronic lymphocytic leukaemia (CLL) is the most common B-cell malignancy with a variable clinical outcome. Biomarkers of CLL progression are required for optimising prognosis and therapy. The Inhibitor of Bruton's tyrosine kinase-isoform α (IBTKα) gene encodes a substrate receptor of Cullin 3-dependent E3 ubiquitin ligase, and promotes cell survival in response to the reticulum stress. Searching for novel markers of CLL progression, we analysed the expression of IBTKα in the peripheral blood B-cells of CLL patients, before and after first line therapy causing remission. The expression of IBTKα was significantly increased in disease progression, and decreased in remission after chemotherapy. Consistently with a pro-survival action, RNA interference of IBTKα increased the spontaneous and Fludarabine-induced apoptosis of MEC-1 CLL cells, and impaired the cell cycle of DeFew B-lymphoma cells by promoting the arrest in G0/G1 phase and apoptosis. Consistently, RNA interference of IBTKα up regulated the expression of pro-apoptotic genes, including TNF, CRADD, CASP7, BNIP3 and BIRC3. Our results indicate that IBTKα is a novel marker of CLL progression promoting cell growth and resistance to apoptosis. In this view, IBTKα may represent an attractive cancer drug target for counteracting the therapy-resistance of tumour cells.


Assuntos
Apoptose , Proteínas de Transporte/metabolismo , Leucemia Linfocítica Crônica de Células B/patologia , Proteínas Adaptadoras de Transdução de Sinal , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Linfócitos B/citologia , Linfócitos B/metabolismo , Proteína Adaptadora de Sinalização CRADD/genética , Proteína Adaptadora de Sinalização CRADD/metabolismo , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Caspase 7/genética , Caspase 7/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Linfocítica Crônica de Células B/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Regulação para Cima , Vidarabina/análogos & derivados , Vidarabina/farmacologia , Vidarabina/uso terapêutico
10.
Am J Med Genet A ; 173(9): 2539-2544, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28686357

RESUMO

Lissencephaly is a severe malformation of cortical development, most often attributed to abnormalities in neuronal migration. It is associated with a severe prognosis including developmental delay, intellectual disability, and seizures. Lissencephaly can be reliably diagnosed during late gestation by neurosonography or fetal magnetic resonance imaging (MRI). We report two sibling male fetuses who were diagnosed with delayed cortical sulcation highly suggestive of lissencephaly during late pregnancy. After receiving genetic counseling, the parents elected to terminate the pregnancies based on the neuroradiological findings and the associated severe prognosis. Whole exome sequencing (WES) of an affected fetus, and subsequent Sanger sequencing of the second fetus, revealed a homozygous frameshift variant in CRADD, which encodes an adaptor protein that interacts with PIDD and caspase-2 to initiate apoptosis. Biallelic variants in this gene have been recently reported to cause "thin" lissencephaly and intellectual disability. Interestingly, the allegedly healthy father was also found to be homozygous for the variant, prompting evaluation by brain MRI which revealed hypogyration. This study underscores the phenotypic variability of pathogenic variants in CRADD and the challenges of prenatal genetic counseling.


Assuntos
Apoptose/genética , Proteína Adaptadora de Sinalização CRADD/genética , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Lisencefalia/genética , Proteínas Adaptadoras de Transdução de Sinal , Caspase 2/genética , Exoma/genética , Feminino , Feto/diagnóstico por imagem , Feto/fisiopatologia , Aconselhamento Genético , Homozigoto , Humanos , Lisencefalia/fisiopatologia , Imageamento por Ressonância Magnética , Masculino , Gravidez , Diagnóstico Pré-Natal
11.
Oral Dis ; 23(5): 653-659, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28160766

RESUMO

OBJECTIVE: In this study, we investigated the role of phenytoin (PHT) in death receptor-induced apoptosis of gingival fibroblasts to clarify the mechanism of PHT-induced gingival overgrowth. METHODS: Human gingival fibroblasts were cultured to semiconfluence and treated with PHT (0.025, 0.1, 0.25, and 1.0 µM) for 48 h, and then, the apoptotic cell numbers were relatively determined by absorptiometry. After 24 h of 0.25 µM PHT treatment, caspase activity was measured by absorptiometry, apoptotic and cell cycle phase distribution was analyzed by flow cytometry, expression levels of apoptotic genes were quantified by real-time qPCR, and expression of apoptotic proteins was detected by Western blot analysis. After 48 h of 0.25 µM PHT treatment, appearance of apoptotic cells was detected by TUNEL assay. RESULTS: PHT treatment decreased the proportion of apoptotic cells in gingival fibroblasts compared to a serum-free control culture in response to the protein changes as follows: PHT upregulated c-FLIP and, in turn, downregulated FADD, caspase-8, and caspase-3; PHT upregulated c-IAP2 and downregulated TRAF2; PHT downregulated caspase-9 and caspase-3 via decreased RIPK1 activity and increased Bcl-2 activity. CONCLUSION: PHT-induced gingival overgrowth may result from the above-mentioned mechanisms involving apoptosis inhibition in gingival fibroblasts.


Assuntos
Anticonvulsivantes/farmacologia , Caspases/metabolismo , Fenitoína/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Proteína 3 com Repetições IAP de Baculovírus/metabolismo , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Proteína Adaptadora de Sinalização CRADD/genética , Células Cultivadas , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/metabolismo , Fibroblastos , Expressão Gênica , Gengiva/citologia , Crescimento Excessivo da Gengiva/induzido quimicamente , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fator 2 Associado a Receptor de TNF/genética , Fator 2 Associado a Receptor de TNF/metabolismo
12.
Am J Hum Genet ; 99(5): 1117-1129, 2016 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-27773430

RESUMO

Lissencephaly is a malformation of cortical development typically caused by deficient neuronal migration resulting in cortical thickening and reduced gyration. Here we describe a "thin" lissencephaly (TLIS) variant characterized by megalencephaly, frontal predominant pachygyria, intellectual disability, and seizures. Trio-based whole-exome sequencing and targeted re-sequencing identified recessive mutations of CRADD in six individuals with TLIS from four unrelated families of diverse ethnic backgrounds. CRADD (also known as RAIDD) is a death-domain-containing adaptor protein that oligomerizes with PIDD and caspase-2 to initiate apoptosis. TLIS variants cluster in the CRADD death domain, a platform for interaction with other death-domain-containing proteins including PIDD. Although caspase-2 is expressed in the developing mammalian brain, little is known about its role in cortical development. CRADD/caspase-2 signaling is implicated in neurotrophic factor withdrawal- and amyloid-ß-induced dendritic spine collapse and neuronal apoptosis, suggesting a role in cortical sculpting and plasticity. TLIS-associated CRADD variants do not disrupt interactions with caspase-2 or PIDD in co-immunoprecipitation assays, but still abolish CRADD's ability to activate caspase-2, resulting in reduced neuronal apoptosis in vitro. Homozygous Cradd knockout mice display megalencephaly and seizures without obvious defects in cortical lamination, supporting a role for CRADD/caspase-2 signaling in mammalian brain development. Megalencephaly and lissencephaly associated with defective programmed cell death from loss of CRADD function in humans implicate reduced apoptosis as an important pathophysiological mechanism of cortical malformation. Our data suggest that CRADD/caspase-2 signaling is critical for normal gyration of the developing human neocortex and for normal cognitive ability.


Assuntos
Apoptose , Proteína Adaptadora de Sinalização CRADD/genética , Caspase 2/metabolismo , Cisteína Endopeptidases/metabolismo , Lisencefalia/genética , Megalencefalia/genética , Neurônios/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Caspase 2/genética , Sobrevivência Celular , Clonagem Molecular , Cognição , Cisteína Endopeptidases/genética , Células Dendríticas/metabolismo , Etnicidade/genética , Genes Recessivos , Estudo de Associação Genômica Ampla , Células HEK293 , Humanos , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Células PC12 , Ratos , Transdução de Sinais
13.
Cell Physiol Biochem ; 39(4): 1271-80, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27606466

RESUMO

BACKGROUND/AIMS: Viral infections represent a global health problem with the need for new viral therapies and better understanding of the immune response during infection. The most immediate and potent anti-viral defense mechanism is the production of type I interferon (IFN-I) which are activated rapidly following recognition of viral infection by host pathogen recognition receptors (PRR). The mechanisms of innate cellular signaling downstream of PRR activation remain to be fully understood. In the present study, we demonstrate that CASP2 and RIPK1 domain-containing adaptor with death domain (CRADD/RAIDD) is a critical component in type I IFN production. METHODS: The role of RAIDD during IFN-I production was investigated using western blot, shRNA mediated lentiviral knockdown, immunoprecipitation and IFN-I driven dual luciferase assay. RESULTS: Immunoprecipitation analysis revealed the molecular interaction of RAIDD with interferon regulatory factor 7 (IRF7) and its phosphorylating kinase IKKε. Using an IFN-4α driven dual luciferase analysis in RAIDD deficient cells, type I IFN activation by IKKε and IRF7 was dramatically reduced. Furthermore, deletion of either the caspase recruitment domain (CARD) or death domain (DD) of RAIDD inhibited IKKε and IRF7 mediated interferon-4α activation. CONCLUSION: We have identified that the adaptor molecule RAIDD coordinates IKKε and IRF7 interaction to ensure efficient expression of type I interferon.


Assuntos
Proteína Adaptadora de Sinalização CRADD/genética , Quinase I-kappa B/genética , Fator Regulador 7 de Interferon/genética , Receptor 3 Toll-Like/genética , Animais , Proteína Adaptadora de Sinalização CRADD/imunologia , Domínio de Ativação e Recrutamento de Caspases , Regulação da Expressão Gênica , Genes Reporter , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Células HEK293 , Humanos , Quinase I-kappa B/imunologia , Fator Regulador 7 de Interferon/imunologia , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/genética , Interferon beta/imunologia , Lentivirus/genética , Lentivirus/metabolismo , Luciferases/genética , Luciferases/metabolismo , Camundongos , Plasmídeos/química , Plasmídeos/metabolismo , Poli I-C/farmacologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Transdução de Sinais , Receptor 3 Toll-Like/imunologia
14.
Cell Death Differ ; 22(11): 1803-11, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25857265

RESUMO

The receptor-interacting protein-associated ICH-1/CED-3 homologous protein with a death domain (RAIDD/CRADD) functions as a dual adaptor and is a constituent of different multi-protein complexes implicated in the regulation of inflammation and cell death. Within the PIDDosome complex, RAIDD connects the cell death-related protease, Caspase-2, with the p53-induced protein with a death domain 1 (PIDD1). As such, RAIDD has been implicated in DNA-damage-induced apoptosis as well as in tumorigenesis. As loss of Caspase-2 leads to an acceleration of tumor onset in the Eµ-Myc mouse lymphoma model, whereas loss of Pidd1 actually delays onset of this disease, we set out to interrogate the role of Raidd in cancer in more detail. Our data obtained analyzing Eµ-Myc/Raidd(-/-) mice indicate that Raidd is unable to protect from c-Myc-driven lymphomagenesis. Similarly, we failed to observe a modulatory effect of Raidd deficiency on DNA-damage-driven cancer. The role of Caspase-2 as a tumor suppressor and that of Pidd1 as a tumor promoter can therefore be uncoupled from their ability to interact with the Raidd scaffold, pointing toward the existence of alternative signaling modules engaging these two proteins in this context.


Assuntos
Proteína Adaptadora de Sinalização CRADD/metabolismo , Caspase 2/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Proteína Adaptadora de Sinalização CRADD/genética , Caspase 2/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Dano ao DNA/genética , Dano ao DNA/efeitos da radiação , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Fibrossarcoma/genética , Fibrossarcoma/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-myc/genética
15.
J Mol Biol ; 427(4): 737-752, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25528640

RESUMO

Homotypic death domain (DD)-DD interactions are important in the assembly of oligomeric signaling complexes such as the PIDDosome that acts as a platform for activation of caspase-2-dependent apoptotic signaling. The structure of the PIDDosome core complex exhibits an asymmetric three-layered arrangement containing five PIDD-DDs in one layer, five RAIDD-DDs in a second layer and an additional two RAIDD-DDs. We addressed complex formation between PIDD-DD and RAIDD-DD in solution using heteronuclear nuclear magnetic resonance (NMR) spectroscopy, nanoflow electrospray ionization mass spectrometry and size-exclusion chromatography with multi-angle light scattering. The DDs assemble into complexes displaying molecular masses in the range 130-158kDa and RAIDD-DD:PIDD-DD stoichiometries of 5:5, 6:5 and 7:5. These data suggest that the crystal structure is representative of only the heaviest species in solution and that two RAIDD-DDs are loosely attached to the 5:5 core. Two-dimensional (1)H,(15)N-NMR experiments exhibited signal loss upon complexation consistent with the formation of high-molecular-weight species. (13)C-Methyl-transverse relaxation optimized spectroscopy measurements of the PIDDosome core exhibit signs of differential line broadening, cross-peak splitting and chemical shift heterogeneity that reflect the presence of non-equivalent sites at interfaces within an asymmetric complex. Experiments using a mutant RAIDD-DD that forms a monodisperse 5:5 complex with PIDD-DD show that the spectroscopic signature derives from the quasi- but non-exact equivalent environments of each DD. Since this characteristic was previously demonstrated for the complex between the DDs of CD95 and FADD, the NMR data for this system are consistent with the formation of a structure homologous to the PIDDosome core.


Assuntos
Proteína Adaptadora de Sinalização CRADD/metabolismo , Proteína Adaptadora de Sinalização CRADD/ultraestrutura , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/ultraestrutura , Sequência de Aminoácidos , Proteína Adaptadora de Sinalização CRADD/genética , Cristalografia por Raios X , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/química , Humanos , Modelos Moleculares , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/ultraestrutura , Ressonância Magnética Nuclear Biomolecular , Estrutura Terciária de Proteína , Espectrometria de Massas por Ionização por Electrospray
16.
J Biol Chem ; 289(32): 21973-83, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-24958727

RESUMO

A hallmark of inflammation, increased vascular permeability, is induced in endothelial cells by multiple agonists through stimulus-coupled assembly of the CARMA3 signalosome, which contains the adaptor protein BCL10. Previously, we reported that BCL10 in immune cells is targeted by the "death" adaptor CRADD/RAIDD (CRADD), which negatively regulates nuclear factor κB (NFκB)-dependent cytokine and chemokine expression in T cells (Lin, Q., Liu, Y., Moore, D. J., Elizer, S. K., Veach, R. A., Hawiger, J., and Ruley, H. E. (2012) J. Immunol. 188, 2493-2497). This novel anti-inflammatory CRADD-BCL10 axis prompted us to analyze CRADD expression and its potential anti-inflammatory action in non-immune cells. We focused our study on microvascular endothelial cells because they play a key role in inflammation. We found that CRADD-deficient murine endothelial cells display heightened BCL10-mediated expression of the pleotropic proinflammatory cytokine IL-6 and chemokine monocyte chemoattractant protein-1 (MCP-1/CCL2) in response to LPS and thrombin. Moreover, these agonists also induce significantly increased permeability in cradd(-/-), as compared with cradd(+/+), primary murine endothelial cells. CRADD-deficient cells displayed more F-actin polymerization with concomitant disruption of adherens junctions. In turn, increasing intracellular CRADD by delivery of a novel recombinant cell-penetrating CRADD protein (CP-CRADD) restored endothelial barrier function and suppressed the induction of IL-6 and MCP-1 evoked by LPS and thrombin. Likewise, CP-CRADD enhanced barrier function in CRADD-sufficient endothelial cells. These results indicate that depletion of endogenous CRADD compromises endothelial barrier function in response to inflammatory signals. Thus, we define a novel function for CRADD in endothelial cells as an inducible suppressor of BCL10, a key mediator of responses to proinflammatory agonists.


Assuntos
Proteína Adaptadora de Sinalização CRADD/metabolismo , Células Endoteliais/metabolismo , Inflamação/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteína 10 de Linfoma CCL de Células B , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteína Adaptadora de Sinalização CRADD/deficiência , Proteína Adaptadora de Sinalização CRADD/genética , Permeabilidade Capilar , Peptídeos Penetradores de Células/genética , Peptídeos Penetradores de Células/metabolismo , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana , Humanos , Mediadores da Inflamação/metabolismo , Pulmão/irrigação sanguínea , Camundongos , Camundongos da Linhagem 129 , Microvasos/citologia , Microvasos/metabolismo , RNA Interferente Pequeno/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais
17.
BMB Rep ; 46(9): 471-6, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24064063

RESUMO

The PIDDosome, which is an oligomeric signaling complex composed of PIDD, RAIDD and caspase-2, can induce proximity-based dimerization and activation of caspase-2. In the PIDDosome assembly, the adaptor protein RAIDD interacts with PIDD and caspase-2 via CARD:CARD and DD:DD, respectively. To analyze the PIDDosome assembly, we purified all of the DD superfamily members and performed biochemical analyses. The results revealed that caspase-2 CARD is an insoluble protein that can be solubilized by its binding partner, RAIDD CARD, but not by full-length RAIDD; this indicates that full-length RAIDD in closed states cannot interact with caspase-2 CARD. Moreover, we found that caspase-2 CARD can be solubilized and interact with full-length RAIDD in the presence of PIDD DD, indicating that PIDD DD initially binds to RAIDD, after which caspase-2 can be recruited to RAIDD via a CARD:CARD interaction. Our study will be useful in determining the order of assembly of the PIDDosome.


Assuntos
Proteína Adaptadora de Sinalização CRADD/metabolismo , Caspase 2/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Animais , Proteína Adaptadora de Sinalização CRADD/genética , Caspase 2/genética , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Camundongos , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína
18.
Biochem J ; 455(1): 15-25, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23815625

RESUMO

Neuronal apoptotic death generally requires de novo transcription, and activation of the transcription factor c-Jun has been shown to be necessary in multiple neuronal death paradigms. Caspase-2 has been implicated in death of neuronal and non-neuronal cells, but its relationship to transcriptional activation has not been clearly elucidated. In the present study, using two different neuronal apoptotic paradigms, ß-amyloid treatment and NGF (nerve growth factor) withdrawal, we examined the hierarchical role of caspase-2 activation in the transcriptional control of neuron death. Both paradigms induce rapid activation of caspase-2 as well as activation of the transcription factor c-Jun and subsequent induction of the pro-apoptotic BH3 (Bcl-homology domain 3)-only protein Bim (Bcl-2-interacting mediator of cell death). Caspase-2 activation is dependent on the adaptor protein RAIDD {RIP (receptor-interacting protein)-associated ICH-1 [ICE (interleukin-1ß-converting enzyme)/CED-3 (cell-death determining 3) homologue 1] protein with a death domain}, and both caspase-2 and RAIDD are required for c-Jun activation and Bim induction. The present study thus shows that rapid caspase-2 activation is essential for c-Jun activation and Bim induction in neurons subjected to apoptotic stimuli. This places caspase-2 at an apical position in the apoptotic cascade and demonstrates for the first time that caspase-2 can regulate transcription.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Proteína Adaptadora de Sinalização CRADD/genética , Caspase 2/genética , Proteínas de Membrana/genética , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas/genética , Ativação Transcricional/efeitos dos fármacos , Peptídeos beta-Amiloides/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Proteína Adaptadora de Sinalização CRADD/metabolismo , Caspase 2/metabolismo , Feto , Proteínas de Membrana/metabolismo , Fator de Crescimento Neural/deficiência , Neurônios/citologia , Neurônios/efeitos dos fármacos , Cultura Primária de Células , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
19.
Appl Biochem Biotechnol ; 170(1): 210-8, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23494218

RESUMO

PIDDosome is a recently-identified caspase-2-activating molecular complex formed by genotoxic stress that leads to caspase-2-dependent apoptosis. PIDD, RAIDD, and caspase-2 are three protein components of PIDDosome. The core portion of PIDDosome is formed by the unique screw rotation of seven RAIDD DD and five PIDD DD. In the current study, we found that two mutations generated during structural-based mutagenesis studies, Q169E and R170A on RAIDD DD, were dominant negative. Because the discovery of dominant-negative mutants might implicate the disease and therapeutic intervention, newly identified dominant-negative mutants could lead to new potential applications for treatment of human diseases caused by excessive or reduced apoptosis.


Assuntos
Proteína Adaptadora de Sinalização CRADD/química , Caspase 2/química , Cisteína Endopeptidases/química , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/química , Mutação , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Apoptose/genética , Proteína Adaptadora de Sinalização CRADD/genética , Caspase 2/genética , Cisteína Endopeptidases/genética , Dano ao DNA , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Escherichia coli/genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Multimerização Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Homologia de Sequência de Aminoácidos
20.
Cell Death Differ ; 19(10): 1722-32, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22595758

RESUMO

The PIDDosome, a multiprotein complex constituted of the 'p53-induced protein with a death domain (PIDD), 'receptor-interacting protein (RIP)-associated ICH-1/CED-3 homologous protein with a death domain' (RAIDD) and pro-Caspase-2 has been defined as an activating platform for this apoptosis-related protease. PIDD has been implicated in p53-mediated cell death in response to DNA damage but also in DNA repair and nuclear factor kappa-light-chain enhancer (NF-κB) activation upon genotoxic stress, together with RIP-1 kinase and Nemo/IKKγ. As all these cellular responses are critical for tumor suppression and deregulated expression of individual PIDDosome components has been noted in human cancer, we investigated their role in oncogenesis induced by DNA damage or oncogenic stress in gene-ablated mice. We observed that Pidd or Caspase-2 failed to suppress lymphoma formation triggered by γ-irradiation or 3-methylcholanthrene-driven fibrosarcoma development. In contrast, Caspase-2 showed tumor suppressive capacity in response to aberrant c-Myc expression, which did not rely on PIDD, the BH3-only protein Bid (BH3 interacting domain death agonist) or the death receptor ligand Trail (TNF-related apoptosis-inducing ligand), but associated with reduced rates of p53 loss and increased extranodal dissemination of tumor cells. In contrast, Pidd deficiency associated with abnormal M-phase progression and delayed disease onset, indicating that both proteins are differentially engaged upon oncogenic stress triggered by c-Myc, leading to opposing effects on tumor-free survival.


Assuntos
Proteína Adaptadora de Sinalização CRADD/metabolismo , Caspase 2/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Proteína Adaptadora de Sinalização CRADD/antagonistas & inibidores , Proteína Adaptadora de Sinalização CRADD/genética , Caspase 2/deficiência , Caspase 2/genética , Linhagem Celular , Dano ao DNA , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/antagonistas & inibidores , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Proteínas Ativadoras de GTPase/metabolismo , Raios gama , Células HCT116 , Humanos , Quinase I-kappa B/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Metilcolantreno/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...