Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Hum Mutat ; 41(5): 998-1011, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31999394

RESUMO

Inactivating variants in the centrosomal CEP78 gene have been found in cone-rod dystrophy with hearing loss (CRDHL), a particular phenotype distinct from Usher syndrome. Here, we identified and functionally characterized the first CEP78 missense variant c.449T>C, p.(Leu150Ser) in three CRDHL families. The variant was found in a biallelic state in two Belgian families and in a compound heterozygous state-in trans with c.1462-1G>T-in a third German family. Haplotype reconstruction showed a founder effect. Homology modeling revealed a detrimental effect of p.(Leu150Ser) on protein stability, which was corroborated in patients' fibroblasts. Elongated primary cilia without clear ultrastructural abnormalities in sperm or nasal brushes suggest impaired cilia assembly. Two affected males from different families displayed sperm abnormalities causing infertility. One of these is a heterozygous carrier of a complex allele in SPAG17, a ciliary gene previously associated with autosomal recessive male infertility. Taken together, our data indicate that a missense founder allele in CEP78 underlies the same sensorineural CRDHL phenotype previously associated with inactivating variants. Interestingly, the CEP78 phenotype has been possibly expanded with male infertility. Finally, CEP78 loss-of-function variants may have an underestimated role in misdiagnosed Usher syndrome, with or without sperm abnormalities.


Assuntos
Alelos , Proteínas de Ciclo Celular/genética , Distrofias de Cones e Bastonetes/genética , Efeito Fundador , Perda Auditiva/genética , Infertilidade Masculina/genética , Mutação de Sentido Incorreto , Adolescente , Proteínas de Ciclo Celular/química , Cílios/metabolismo , Cílios/ultraestrutura , Distrofias de Cones e Bastonetes/diagnóstico , Análise Mutacional de DNA , Feminino , Fibroblastos/metabolismo , Genótipo , Perda Auditiva/diagnóstico , Humanos , Infertilidade Masculina/diagnóstico , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Linhagem , Fenótipo , Conformação Proteica , Relação Estrutura-Atividade , Síndrome , Sequenciamento do Exoma
2.
Hum Mutat ; 40(5): 539-551, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30668888

RESUMO

Sorsby fundus dystrophy (SFD) is a macular degeneration caused by mutations in TIMP3, the majority of which introduce a novel cysteine. However, the exact molecular mechanisms underlying SFD remain unknown. We aimed to provide novel insights into the functional consequences of a distinct N-terminal mutation. Haplotype reconstruction in three SFD families revealed that the identified c.113C>G, p.(Ser38Cys) mutation is a founder in Belgian and northern French families with a late-onset SFD phenotype. Functional consequences of the p.(Ser38Cys) mutation were investigated by high-resolution Western blot analysis of wild type and mutant TIMP3 using patient fibroblasts and in vitro generated proteins, and by molecular modeling of TIMP3 and its interaction partners. We could not confirm a previous hypothesis on dimerization of mutant TIMP3 proteins. However, we identified aberrant intramolecular disulfide bonding. Our data provide evidence for disruption of the established Cys36-Cys143 disulfide bond and formation of a novel Cys36-Cys38 bond, possibly associated with increased glycosylation of the protein. In conclusion, we propose a novel pathogenetic mechanism underlying the p.(Ser38Cys) TIMP3 founder mutation involving intramolecular disulfide bonding. These results provide new insights into the pathogenesis of SFD and other retinopathies linked to mutations in TIMP3, such as age-related macular degeneration.


Assuntos
Efeito Fundador , Degeneração Macular/diagnóstico , Degeneração Macular/genética , Mutação , Domínios e Motivos de Interação entre Proteínas , Inibidor Tecidual de Metaloproteinase-3/química , Inibidor Tecidual de Metaloproteinase-3/genética , Idoso , Dissulfetos , Feminino , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Haplótipos , Humanos , Masculino , Metaloproteinase 2 da Matriz/química , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Peso Molecular , Linhagem , Conformação Proteica , Relação Estrutura-Atividade , Inibidor Tecidual de Metaloproteinase-3/metabolismo
3.
Am J Hum Genet ; 99(2): 470-80, 2016 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-27486781

RESUMO

Inherited retinal dystrophies (iRDs) are a group of genetically and clinically heterogeneous conditions resulting from mutations in over 250 genes. Here, homozygosity mapping and whole-exome sequencing (WES) in a consanguineous family revealed a homozygous missense mutation, c.973C>T (p.His325Tyr), in RCBTB1. In affected individuals, it was found to segregate with retinitis pigmentosa (RP), goiter, primary ovarian insufficiency, and mild intellectual disability. Subsequent analysis of WES data in different cohorts uncovered four additional homozygous missense mutations in five unrelated families in whom iRD segregates with or without syndromic features. Ocular phenotypes ranged from typical RP starting in the second decade to chorioretinal dystrophy with a later age of onset. The five missense mutations affect highly conserved residues either in the sixth repeat of the RCC1 domain or in the BTB1 domain. A founder haplotype was identified for mutation c.919G>A (p.Val307Met), occurring in two families of Mediterranean origin. We showed ubiquitous mRNA expression of RCBTB1 and demonstrated predominant RCBTB1 localization in human inner retina. RCBTB1 was very recently shown to be involved in ubiquitination, more specifically as a CUL3 substrate adaptor. Therefore, the effect on different components of the CUL3 and NFE2L2 (NRF2) pathway was assessed in affected individuals' lymphocytes, revealing decreased mRNA expression of NFE2L2 and several NFE2L2 target genes. In conclusion, our study puts forward mutations in RCBTB1 as a cause of autosomal-recessive non-syndromic and syndromic iRD. Finally, our data support a role for impaired ubiquitination in the pathogenetic mechanism of RCBTB1 mutations.


Assuntos
Alelos , Fatores de Troca do Nucleotídeo Guanina/genética , Mutação de Sentido Incorreto/genética , Distrofias Retinianas/genética , Ubiquitinação/genética , Adolescente , Adulto , Idade de Início , Criança , Consanguinidade , Proteínas Culina/metabolismo , Exoma/genética , Feminino , Efeito Fundador , Genes Recessivos , Haplótipos/genética , Homozigoto , Humanos , Linfócitos/metabolismo , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Linhagem , Fenótipo , RNA Mensageiro/genética , Retina/metabolismo , Síndrome , Turquia
4.
Genet Med ; 21(4): 1028, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30607024

RESUMO

The original version of this Article contained an incorrect version of Fig. 3, which included two variants initially shown in black text in Fig. 3a that the authors removed from the final manuscript. The correct version of Fig. 3 without the two variants now appears in the PDF and HTML versions of the Article.

5.
Genet Med ; 21(6): 1319-1329, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30377383

RESUMO

PURPOSE: RAX2 encodes a homeobox-containing transcription factor, in which four monoallelic pathogenic variants have been described in autosomal dominant cone-dominated retinal disease. METHODS: Exome sequencing in a European cohort with inherited retinal disease (IRD) (n = 2086) was combined with protein structure modeling of RAX2 missense variants, bioinformatics analysis of deletion breakpoints, haplotyping of RAX2 variant c.335dup, and clinical assessment of biallelic RAX2-positive cases and carrier family members. RESULTS: Biallelic RAX2 sequence and structural variants were found in five unrelated European index cases, displaying nonsyndromic autosomal recessive retinitis pigmentosa (ARRP) with an age of onset ranging from childhood to the mid-40s (average mid-30s). Protein structure modeling points to loss of function of the novel recessive missense variants and to a dominant-negative effect of the reported dominant RAX2 alleles. Structural variants were fine-mapped to disentangle their underlying mechanisms. Haplotyping of c.335dup in two cases suggests a common ancestry. CONCLUSION: This study supports a role for RAX2 as a novel disease gene for recessive IRD, broadening the mutation spectrum from sequence to structural variants and revealing a founder effect. The identification of biallelic RAX2 pathogenic variants in five unrelated families shows that RAX2 loss of function may be a nonnegligible cause of IRD in unsolved ARRP cases.


Assuntos
Proteínas do Olho/genética , Proteínas de Homeodomínio/genética , Retinose Pigmentar/genética , Fatores de Transcrição/genética , Adulto , Análise Mutacional de DNA/métodos , Proteínas do Olho/metabolismo , Proteínas do Olho/fisiologia , Feminino , Genes Recessivos/genética , Estudos de Associação Genética/métodos , Genótipo , Haplótipos/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Mutação/genética , Mutação de Sentido Incorreto/genética , Linhagem , Fenótipo , Retina/metabolismo , Retina/patologia , Doenças Retinianas/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , População Branca/genética
6.
Int J Mol Sci ; 20(9)2019 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-31060328

RESUMO

The small GTase Arf6 has several important functions in intracellular vesicular trafficking and regulates the recycling of different types of cargo internalized via clathrin-dependent or -independent endocytosis. It activates the lipid modifying enzymes PIP 5-kinase and phospholipase D, promotes actin polymerization, and affects several functionally distinct processes in the cell. Arf6 is used for the phagocytosis of pathogens and can be directly or indirectly targeted by various pathogens to block phagocytosis or induce the uptake of intracellular pathogens. Arf6 is also used in the signaling of Toll-like receptors and in the activation of NADPH oxidases. In this review, we first give an overview of the different roles and mechanisms of action of Arf6 and then focus on its role in innate immunity and host-pathogen interactions.


Assuntos
Fatores de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/metabolismo , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Fator 6 de Ribosilação do ADP , Animais , Biomarcadores , Ativação Enzimática , Interações Hospedeiro-Parasita/genética , Interações Hospedeiro-Parasita/imunologia , Humanos , Fagocitose/genética , Fagocitose/imunologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Receptores Toll-Like/metabolismo , Virulência
7.
Int J Mol Sci ; 20(9)2019 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-31027327

RESUMO

The MAPPIT (mammalian protein protein interaction trap) method allows high-throughput detection of protein interactions by very simple co-transfection of three plasmids in HEK293T cells, followed by a luciferase readout. MAPPIT detects a large percentage of all protein interactions, including those requiring posttranslational modifications and endogenous or exogenous ligands. Here, we present a straightforward method that allows detailed mapping of interaction interfaces via MAPPIT. The method provides insight into the interaction mechanism and reveals how this is affected by disease-associated mutations. By combining error-prone polymerase chain reaction (PCR) for random mutagenesis, 96-well DNA prepping, Sanger sequencing, and MAPPIT via 384-well transfections, we test the effects of a large number of mutations of a selected protein on its protein interactions. The entire screen takes less than three months and interactions with multiple partners can be studied in parallel. The effect of mutations on the MAPPIT readout is mapped on the protein structure, allowing unbiased identification of all putative interaction sites. We have thus far analysed 6 proteins and mapped their interfaces for 16 different interaction partners. Our method is broadly applicable as the required tools are simple and widely available.


Assuntos
Mutagênese/genética , Mapeamento de Interação de Proteínas/métodos , Animais , Humanos , Ligação Proteica
8.
Genet Med ; 20(7): 717-727, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29261182

RESUMO

PURPOSE: Disorders or differences of sex development (DSDs) are rare congenital conditions characterized by atypical sex development. Despite advances in genomic technologies, the molecular cause remains unknown in 50% of cases. METHODS: Homozygosity mapping and whole-exome sequencing revealed an ESR2 variant in an individual with syndromic 46,XY DSD. Additional cases with 46,XY DSD underwent whole-exome sequencing and targeted next-generation sequencing of ESR2. Functional characterization of the identified variants included luciferase assays and protein structure analysis. Gonadal ESR2 expression was assessed in human embryonic data sets and immunostaining of estrogen receptor-ß (ER-ß) was performed in an 8-week-old human male embryo. RESULTS: We identified a homozygous ESR2 variant, c.541_543del p.(Asn181del), located in the highly conserved DNA-binding domain of ER-ß, in an individual with syndromic 46,XY DSD. Two additional heterozygous missense variants, c.251G>T p.(Gly84Val) and c.1277T>G p.(Leu426Arg), located in the N-terminus and the ligand-binding domain of ER-ß, were found in unrelated, nonsyndromic 46,XY DSD cases. Significantly increased transcriptional activation and an impact on protein conformation were shown for the p.(Asn181del) and p.(Leu426Arg) variants. Testicular ESR2 expression was previously documented and ER-ß immunostaining was positive in the developing intestine and eyes. CONCLUSION: Our study supports a role for ESR2 as a novel candidate gene for 46,XY DSD.


Assuntos
Transtorno 46,XY do Desenvolvimento Sexual/genética , Receptor beta de Estrogênio/genética , Adolescente , Alelos , Substituição de Aminoácidos/genética , Criança , Mapeamento Cromossômico/métodos , Receptor beta de Estrogênio/metabolismo , Feminino , Frequência do Gene/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Masculino , Mutação/genética , Conformação Proteica , Relação Estrutura-Atividade , Sequenciamento do Exoma/métodos , Adulto Jovem
9.
Genet Med ; 19(4): 367-376, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27490115

RESUMO

PURPOSE: We aimed to identify the genetic cause in a cohort of 11 unrelated cases and two sisters with 46,XX SRY-negative (ovo)testicular disorders of sex development (DSD). METHODS: Whole-exome sequencing (n = 9), targeted resequencing (n = 4), and haplotyping were performed. Immunohistochemistry of sex-specific markers was performed on patients' gonads. The consequences of mutation were investigated using luciferase assays, localization studies, and RNA-seq. RESULTS: We identified a novel heterozygous NR5A1 mutation, c.274C>T p.(Arg92Trp), in three unrelated patients. The Arg92 residue is highly conserved and located in the Ftz-F1 region, probably involved in DNA-binding specificity and stability. There were no consistent changes in transcriptional activation or subcellular localization. Transcriptomics in patient-derived lymphocytes showed upregulation of MAMLD1, a direct NR5A1 target previously associated with 46,XY DSD. In gonads of affected individuals, ovarian FOXL2 and testicular SRY-independent SOX9 expression observed. CONCLUSIONS: We propose NR5A1, previously associated with 46,XY DSD and 46,XX primary ovarian insufficiency, as a novel gene for 46,XX (ovo)testicular DSD. We hypothesize that p.(Arg92Trp) results in decreased inhibition of the male developmental pathway through downregulation of female antitestis genes, thereby tipping the balance toward testicular differentiation in 46,XX individuals. In conclusion, our study supports a role for NR5A1 in testis differentiation in the XX gonad.Genet Med 19 4, 367-376.


Assuntos
Proteínas de Ligação a DNA/genética , Sequenciamento do Exoma/métodos , Perfilação da Expressão Gênica/métodos , Proteínas Nucleares/genética , Transtornos Ovotesticulares do Desenvolvimento Sexual/genética , Análise de Sequência de RNA/métodos , Fator Esteroidogênico 1/genética , Fatores de Transcrição/genética , Feminino , Predisposição Genética para Doença , Haplótipos , Humanos , Masculino , Modelos Moleculares , Mutação de Sentido Incorreto , Ovário/metabolismo , Transtornos Ovotesticulares do Desenvolvimento Sexual/metabolismo , Linhagem , Polimorfismo de Nucleotídeo Único , Fator Esteroidogênico 1/química , Fator Esteroidogênico 1/metabolismo , Testículo/metabolismo , Regulação para Cima , Adulto Jovem
10.
Cell Mol Life Sci ; 72(3): 629-644, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25098352

RESUMO

The adipocyte-derived cytokine leptin acts as a metabolic switch, connecting the body's metabolism to high-energy consuming processes such as reproduction and immune responses. We here provide genetic and biochemical evidence that the metabolic and immune functions of leptin can be uncoupled at the receptor level. First, homozygous mutant fatt/fatt mice carry a spontaneous splice mutation causing deletion of the leptin receptor (LR) immunoglobulin-like domain (IGD) in all LR isoforms. These mice are hyperphagic and morbidly obese, but display only minimal changes in size and cellularity of the thymus, and cellular immune responses are unaffected. These animals also displayed liver damage in response to concavalin A comparable to wild-type and heterozygous littermates. Second, treatment of healthy mice with a neutralizing nanobody targeting IGD induced weight gain and hyperinsulinaemia, but completely failed to block development of experimentally induced autoimmune diseases. These data indicate that leptin receptor deficiency or antagonism profoundly affects metabolism, with little concomitant effects on immune functions.


Assuntos
Leptina/imunologia , Leptina/metabolismo , Receptores para Leptina/metabolismo , Análise de Variância , Animais , Artrite Experimental/patologia , Sequência de Bases , Western Blotting , Doença Hepática Induzida por Substâncias e Drogas/patologia , Primers do DNA/genética , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/patologia , Citometria de Fluxo , Células HEK293 , Humanos , Células MCF-7 , Masculino , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Glicoproteína Mielina-Oligodendrócito/toxicidade , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Receptores para Leptina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Deleção de Sequência/genética
11.
J Biol Chem ; 289(3): 1364-76, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24297182

RESUMO

Recognition of lipopolysaccharides (LPS) by Toll-like receptor 4 (TLR4) at the plasma membrane triggers NF-κB activation through recruitment of the adaptor proteins Mal and MyD88. Endocytosis of the activated TLR4 allows recruitment of the adaptors Tram and Trif, leading to activation of the transcription factor IRF3 and interferon production. The small GTPase ADP-ribosylation factor 6 (Arf6) was shown to regulate the plasma membrane association of Mal. Here we demonstrate that inhibition of Arf6 also markedly reduced LPS-induced cytokine production in Mal(-/-) mouse macrophages. In this article, we focus on a novel role for Arf6 in the MyD88-independent TLR4 pathway. MyD88-independent IRF3 activation and IRF3-dependent gene transcription were strictly dependent on Arf6. Arf6 was involved in transport of Tram to the endocytic recycling compartment and internalization of LPS, possibly explaining its requirement for LPS-induced IRF3 activation. Together, these results show a critical role for Arf6 in regulating Tram/Trif-dependent TLR4 signaling.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Macrófagos/metabolismo , Receptores de Interleucina/metabolismo , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/metabolismo , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/genética , Animais , Linhagem Celular , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Camundongos , Camundongos Knockout , Proteínas Proteolipídicas Associadas a Linfócitos e Mielina/genética , Proteínas Proteolipídicas Associadas a Linfócitos e Mielina/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Receptores de Interleucina/genética , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia
12.
J Cell Sci ; 126(Pt 16): 3770-81, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23750007

RESUMO

The mechanisms controlling the steady-state cell surface levels of cytokine receptors, and consequently the cellular response to cytokines, remain poorly understood. The number of surface-exposed receptors is a dynamic balance of de novo synthesis, transport to the plasma membrane, internalization, recycling, degradation and ectodomain shedding. We previously reported that the E3 ubiquitin ligase RING finger protein 41 (RNF41) inhibits basal lysosomal degradation and enhances ectodomain shedding of JAK2-associated cytokine receptors. Ubiquitin-specific protease 8 (USP8), an RNF41-interacting deubiquitylating enzyme (DUB) stabilizes RNF41 and is involved in trafficking of various transmembrane proteins. The present study identifies USP8 as a substrate of RNF41 and reveals that loss of USP8 explains the aforementioned RNF41 effects. RNF41 redistributes and ubiquitylates USP8, and reduces USP8 levels. In addition, USP8 knockdown functionally matches the effects of RNF41 ectopic expression on the model leptin and leukemia inhibitory factor (LIF) receptors. Moreover, RNF41 indirectly destabilizes the ESCRT-0 complex through suppression of USP8. Collectively, our findings demonstrate that RNF41 controls JAK2-associated cytokine receptor trafficking by acting as a key regulator of USP8 and ESCRT-0 stability. Balanced reciprocal cross-regulation of RNF41 and USP8 thus determines whether receptors are sorted for lysosomal degradation or recycling, this way regulating basal cytokine receptor levels.


Assuntos
Endopeptidases/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Receptores de Citocinas/metabolismo , Ubiquitina Tiolesterase/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Movimento Celular/fisiologia , Endopeptidases/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Humanos , Ligação Proteica , Transporte Proteico , Receptores de Citocinas/genética , Transdução de Sinais , Transfecção , Ubiquitina Tiolesterase/genética , Ubiquitina-Proteína Ligases/genética
13.
J Biol Chem ; 288(17): 12054-66, 2013 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-23460645

RESUMO

Upon activation, Toll-like receptor 4 (TLR4) binds adapter proteins, including MyD88 (myeloid differentiation primary response gene 88) and Mal (MyD88 adapter-like) for its signal transduction. TLR4 and the adapter proteins each contain a Toll/Il-1 receptor domain (TIR domain). In this study we used random mutagenesis and the mammalian two-hybrid method MAPPIT (mammalian protein-protein interaction trap) to identify mutations in Mal that disrupt its interaction with TLR4 and/or MyD88. Our study shows that four potential binding sites and the AB-loop in the Mal TIR domain all contribute to formation of the TLR4-Mal-MyD88 complex. Mutations in the symmetrical back-to-back Mal homodimer interface affect Mal homodimerization and interaction with MyD88 and TLR4. Our data suggest that Mal dimerization may lead to formation of potential binding platforms on the top and the side of the Mal dimer that bind MyD88 or TLR4. Mutations that affect the interaction of Mal with MyD88 also affect NF-κB activation induced by Mal overexpression. In MAPPIT, co-expression of the MyD88 TIR domain enhances Mal dimerization and Mal binding to TLR4. Similarly, co-expression of Mal and the MyD88 TIR domain strongly promotes dimerization of the TLR4 intracellular domain in MAPPIT. The different types of TIR-TIR interactions in the TLR4-Mal-MyD88 complex thus show cooperative binding in MAPPIT. We present plausible models for the TIR-TIR interactions in the TLR4-Mal-MyD88 complex.


Assuntos
Sítios de Ligação , Modelos Biológicos , Fator 88 de Diferenciação Mieloide , Multimerização Proteica/fisiologia , Receptor 4 Toll-Like , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mutação , Fator 88 de Diferenciação Mieloide/química , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Mapeamento de Peptídeos/métodos , Ligação Proteica , Receptores de Interleucina-1/química , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Receptor 4 Toll-Like/química , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
14.
Biol Chem ; 395(5): 499-514, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24523306

RESUMO

The adipocyte-derived hormone/cytokine leptin acts as a metabolic switch, connecting the body's nutritional status to high energy consuming processes such as reproduction and immune responses. Inappropriate leptin responses can promote autoimmune diseases and tumorigenesis. In this review we discuss the current strategies to modulate leptin signaling and the possibilities for their use in research and therapy.


Assuntos
Leptina/antagonistas & inibidores , Obesidade/metabolismo , Adipócitos/metabolismo , Humanos , Leptina/imunologia , Transdução de Sinais
16.
Mol Metab ; 84: 101938, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38631478

RESUMO

OBJECTIVE: The peroxisome proliferator-activated receptor α (PPARα) is a transcription factor driving target genes involved in fatty acid ß-oxidation. To what extent various PPARα interacting proteins may assist its function as a transcription factor is incompletely understood. An ORFeome-wide unbiased mammalian protein-protein interaction trap (MAPPIT) using PPARα as bait revealed a PPARα-ligand-dependent interaction with the orphan nuclear receptor estrogen-related receptor α (ERRα). The goal of this study was to characterize the nature of the interaction in depth and to explore whether it was of physiological relevance. METHODS: We used orthogonal protein-protein interaction assays and pharmacological inhibitors of ERRα in various systems to confirm a functional interaction and study the impact of crosstalk mechanisms. To characterize the interaction surfaces and contact points we applied a random mutagenesis screen and structural overlays. We pinpointed the extent of reciprocal ligand effects of both nuclear receptors via coregulator peptide recruitment assays. On PPARα targets revealed from a genome-wide transcriptome analysis, we performed an ERRα chromatin immunoprecipitation analysis on both fast and fed mouse livers. RESULTS: Random mutagenesis scanning of PPARα's ligand-binding domain and coregulator profiling experiments supported the involvement of (a) bridging coregulator(s), while recapitulation of the interaction in vitro indicated the possibility of a trimeric interaction with RXRα. The PPARα·ERRα interaction depends on 3 C-terminal residues within helix 12 of ERRα and is strengthened by both PGC1α and serum deprivation. Pharmacological inhibition of ERRα decreased the interaction of ERRα to ligand-activated PPARα and revealed a transcriptome in line with enhanced mRNA expression of prototypical PPARα target genes, suggesting a role for ERRα as a transcriptional repressor. Strikingly, on other PPARα targets, including the isolated PDK4 enhancer, ERRα behaved oppositely. Chromatin immunoprecipitation analyses demonstrate a PPARα ligand-dependent ERRα recruitment onto chromatin at PPARα-binding regions, which is lost following ERRα inhibition in fed mouse livers. CONCLUSIONS: Our data support the coexistence of multiple layers of transcriptional crosstalk mechanisms between PPARα and ERRα, which may serve to finetune the activity of PPARα as a nutrient-sensing transcription factor.


Assuntos
Receptor ERRalfa Relacionado ao Estrogênio , PPAR alfa , Receptores de Estrogênio , PPAR alfa/metabolismo , PPAR alfa/genética , Animais , Camundongos , Receptores de Estrogênio/metabolismo , Receptores de Estrogênio/genética , Humanos , Regulação da Expressão Gênica , Células HEK293 , Masculino , Camundongos Endogâmicos C57BL , Ligação Proteica , Fígado/metabolismo
17.
J Biol Chem ; 287(6): 4088-98, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22139835

RESUMO

Toll-like receptor signaling requires interactions of the Toll/IL-1 receptor (TIR) domains of the receptor and adapter proteins. Using the mammalian protein-protein interaction trap strategy, homology modeling, and site-directed mutagenesis, we identify the interaction surfaces in the TLR4 TIR domain for the TLR4-TLR4, TLR4-MyD88 adapter-like (MAL), and TLR4-TRIF-related adapter molecule (TRAM) interaction. Two binding sites are equally important for TLR4 dimerization and adapter recruitment. In a model based on the crystal structure of the dimeric TLR10 TIR domain, the first binding site mediates TLR4-TLR4 TIR-TIR interaction. Upon dimerization, two identical second binding sites of the TLR4 TIR domain are juxtaposed and form an extended binding platform for both MAL and TRAM. In our mammalian protein-protein interaction trap assay, MAL and TRAM compete for binding to this platform. Our data suggest that adapter binding can stabilize the TLR4 TIR dimerization.


Assuntos
Modelos Moleculares , Multimerização Proteica/fisiologia , Receptor 4 Toll-Like/química , Animais , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Fator 88 de Diferenciação Mieloide/química , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Mapeamento de Peptídeos , Estrutura Terciária de Proteína , Homologia Estrutural de Proteína , Receptor 10 Toll-Like/química , Receptor 10 Toll-Like/genética , Receptor 10 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
18.
Biochem J ; 441(1): 425-34, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21851341

RESUMO

The adipocyte-derived cytokine leptin acts as a metabolic switch, connecting the body's metabolism to high-energy consuming processes such as reproduction and immune responses. Accumulating evidence suggests that leptin plays a role in human pathologies, such as autoimmune diseases and cancer, thus providing a rationale for the development of leptin antagonists. In the present study, we generated and evaluated a panel of neutralizing nanobodies targeting the LR (leptin receptor). A nanobody comprises the variable domain of the naturally occurring single-chain antibodies found in members of the Camelidae family. We identified three classes of neutralizing nanobodies targeting different LR subdomains: i.e. the CRH2 (cytokine receptor homology 2), Ig-like and FNIII (fibronectin type III) domains. Only nanobodies directed against the CRH2 domain inhibited leptin binding. We could show that a nanobody that targets the Ig-like domain potently interfered with leptin-dependent regulation of hypothalamic NPY (neuropeptide Y) expression. As a consequence, daily intraperitoneal injection increased body weight, body fat content, food intake, liver size and serum insulin levels. All of these characteristics resemble the phenotype of leptin and LR-deficient animals. The results of the present study support proposed models of the activated LR complex, and demonstrate that it is possible to block LR signalling without affecting ligand binding. These nanobodies form new tools to study the mechanisms of BBB (blood-brain barrier) leptin transport and the effect of LR inhibition in disease models.


Assuntos
Anticorpos/farmacologia , Leptina/antagonistas & inibidores , Nanoestruturas/química , Receptores para Leptina/antagonistas & inibidores , Tecido Adiposo , Animais , Anticorpos/química , Camelídeos Americanos , Regulação da Expressão Gênica , Células HEK293 , Humanos , Hiperinsulinismo , Fígado/anatomia & histologia , Camundongos , Camundongos Endogâmicos C57BL , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Tamanho do Órgão , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais , Aumento de Peso
19.
Cell Rep Methods ; 3(11): 100641, 2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-37963462

RESUMO

Protein mutagenesis is essential for unveiling the molecular mechanisms underlying protein function in health, disease, and evolution. In the past decade, deep mutational scanning methods have evolved to support the functional analysis of nearly all possible single-amino acid changes in a protein of interest. While historically these methods were developed in lower organisms such as E. coli and yeast, recent technological advancements have resulted in the increased use of mammalian cells, particularly for studying proteins involved in human disease. These advancements will aid significantly in the classification and interpretation of variants of unknown significance, which are being discovered at large scale due to the current surge in the use of whole-genome sequencing in clinical contexts. Here, we explore the experimental aspects of deep mutational scanning studies in mammalian cells and report the different methods used in each step of the workflow, ultimately providing a useful guide toward the design of such studies.


Assuntos
Escherichia coli , Proteínas , Animais , Humanos , Mutação , Proteínas/genética , Mutagênese , Aminoácidos , Mamíferos/genética
20.
Cells ; 12(12)2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37371105

RESUMO

The glucocorticoid receptor α (GRα) is a member of the nuclear receptor superfamily and functions as a glucocorticoid (GC)-responsive transcription factor. GR can halt inflammation and kill off cancer cells, thus explaining the widespread use of glucocorticoids in the clinic. However, side effects and therapy resistance limit GR's therapeutic potential, emphasizing the importance of resolving all of GR's context-specific action mechanisms. Fortunately, the understanding of GR structure, conformation, and stoichiometry in the different GR-controlled biological pathways is now gradually increasing. This information will be crucial to close knowledge gaps on GR function. In this review, we focus on the various domains and mechanisms of action of GR, all from a structural perspective.


Assuntos
Receptores de Glucocorticoides , Humanos , Glucocorticoides/farmacologia , Glucocorticoides/metabolismo , Receptores de Glucocorticoides/metabolismo , Fatores de Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA