Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Am J Hum Genet ; 106(5): 623-631, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32275884

RESUMO

Nucleoporins (NUPs) are an essential component of the nuclear-pore complex, which regulates nucleocytoplasmic transport of macromolecules. Pathogenic variants in NUP genes have been linked to several inherited human diseases, including a number with progressive neurological degeneration. We present six affected individuals with bi-allelic truncating variants in NUP188 and strikingly similar phenotypes and clinical courses, representing a recognizable genetic syndrome; the individuals are from four unrelated families. Key clinical features include congenital cataracts, hypotonia, prenatal-onset ventriculomegaly, white-matter abnormalities, hypoplastic corpus callosum, congenital heart defects, and central hypoventilation. Characteristic dysmorphic features include small palpebral fissures, a wide nasal bridge and nose, micrognathia, and digital anomalies. All affected individuals died as a result of respiratory failure, and five of them died within the first year of life. Nuclear import of proteins was decreased in affected individuals' fibroblasts, supporting a possible disease mechanism. CRISPR-mediated knockout of NUP188 in Drosophila revealed motor deficits and seizure susceptibility, partially recapitulating the neurological phenotype seen in affected individuals. Removal of NUP188 also resulted in aberrant dendrite tiling, suggesting a potential role of NUP188 in dendritic development. Two of the NUP188 pathogenic variants are enriched in the Ashkenazi Jewish population in gnomAD, a finding we confirmed with a separate targeted population screen of an international sampling of 3,225 healthy Ashkenazi Jewish individuals. Taken together, our results implicate bi-allelic loss-of-function NUP188 variants in a recessive syndrome characterized by a distinct neurologic, ophthalmologic, and facial phenotype.


Assuntos
Alelos , Encéfalo/anormalidades , Proteínas de Drosophila/genética , Anormalidades do Olho/genética , Cardiopatias Congênitas/genética , Mutação com Perda de Função/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Transporte Ativo do Núcleo Celular , Animais , Núcleo Celular/metabolismo , Pré-Escolar , Dendritos/metabolismo , Dendritos/patologia , Drosophila melanogaster , Anormalidades do Olho/mortalidade , Feminino , Fibroblastos , Genes Recessivos , Cardiopatias Congênitas/mortalidade , Humanos , Lactente , Recém-Nascido , Judeus/genética , Masculino , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Convulsões/metabolismo , Síndrome , beta Carioferinas/metabolismo
2.
Hum Mol Genet ; 28(23): 3921-3927, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31600784

RESUMO

Allgrove syndrome (AS) is a rare disease with broad neurological involvement. Neurodegeneration can affect spinal motor neurons, Purkinje cells, striatal neurons and the autonomic system. The mechanisms that lead to neuronal loss are still unclear. Recessive mutations in the AAAS gene affect the encoded protein Aladin, which would normally localize to the cytoplasmic face of the nuclear membrane as part of the nuclear pore complex (NPC). While the NPC is known to be a key factor for nucleocytoplasmic transport, the precise role of Aladin has not been elucidated yet. Here, we explored the consequences of the homozygous AAAS mutation c.464G>A (p.R155H) in central nervous system tissues and fibroblasts of a novel AS patient presenting motor neuron disease, cerebellar ataxia and autonomic dysfunction. Neuropathological analyses showed severe loss of motor neurons and Purkinje cells, with significant reduction in the perinuclear expression of Aladin. A reduced amount of protein was detected in the nuclear membrane fraction of the patient's brain. RNA analysis revealed a significant reduction of the transcript AAAS-1, while the AAAS-2 transcript was upregulated in fibroblasts. To our knowledge, this is the first study to demonstrate the effects of AAAS mutations in the human central nervous system.


Assuntos
Insuficiência Adrenal/genética , Acalasia Esofágica/genética , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Insuficiência Adrenal/metabolismo , Idade de Início , Idoso , Substituição de Aminoácidos , Sistema Nervoso Central/metabolismo , Regulação para Baixo , Acalasia Esofágica/metabolismo , Fibroblastos/metabolismo , Humanos , Masculino , Mutação Puntual , Análise de Sequência de DNA
3.
Nucleic Acids Res ; 47(8): 4054-4067, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-30715474

RESUMO

DNA lesions interfere with cellular processes such as transcription and replication and need to be adequately resolved to warrant genome integrity. Beyond their primary role in molecule transport, nuclear pore complexes (NPCs) function in other processes such as transcription, nuclear organization and DNA double strand break (DSB) repair. Here we found that the removal of UV-induced DNA lesions by nucleotide excision repair (NER) is compromised in the absence of the Nup84 nuclear pore component. Importantly, nup84Δ cells show an exacerbated sensitivity to UV in early S phase and delayed replication fork progression, suggesting that unrepaired spontaneous DNA lesions persist during S phase. In addition, nup84Δ cells are defective in the repair of replication-born DSBs by sister chromatid recombination (SCR) and rely on post-replicative repair functions for normal proliferation, indicating dysfunctions in the cellular pathways that enable replication on damaged DNA templates. Altogether, our data reveal a central role of the NPC in the DNA damage response to facilitate replication progression through damaged DNA templates by promoting efficient NER and SCR and preventing chromosomal rearrangements.


Assuntos
Reparo do DNA , DNA Fúngico/genética , Genoma Fúngico , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Pontos de Checagem da Fase S do Ciclo Celular/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Replicação do DNA/efeitos da radiação , DNA Fúngico/metabolismo , Instabilidade Genômica , Poro Nuclear/metabolismo , Poro Nuclear/efeitos da radiação , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Pontos de Checagem da Fase S do Ciclo Celular/efeitos da radiação , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efeitos da radiação , Proteínas de Saccharomyces cerevisiae/metabolismo , Troca de Cromátide Irmã , Raios Ultravioleta
4.
PLoS Genet ; 14(12): e1007845, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30543681

RESUMO

Nucleoporins build the nuclear pore complex (NPC), which, as sole gate for nuclear-cytoplasmic exchange, is of outmost importance for normal cell function. Defects in the process of nucleocytoplasmic transport or in its machinery have been frequently described in human diseases, such as cancer and neurodegenerative disorders, but only in a few cases of developmental disorders. Here we report biallelic mutations in the nucleoporin NUP88 as a novel cause of lethal fetal akinesia deformation sequence (FADS) in two families. FADS comprises a spectrum of clinically and genetically heterogeneous disorders with congenital malformations related to impaired fetal movement. We show that genetic disruption of nup88 in zebrafish results in pleiotropic developmental defects reminiscent of those seen in affected human fetuses, including locomotor defects as well as defects at neuromuscular junctions. Phenotypic alterations become visible at distinct developmental stages, both in affected human fetuses and in zebrafish, whereas early stages of development are apparently normal. The zebrafish phenotypes caused by nup88 deficiency are rescued by expressing wild-type Nup88 but not the disease-linked mutant forms of Nup88. Furthermore, using human and mouse cell lines as well as immunohistochemistry on fetal muscle tissue, we demonstrate that NUP88 depletion affects rapsyn, a key regulator of the muscle nicotinic acetylcholine receptor at the neuromuscular junction. Together, our studies provide the first characterization of NUP88 in vertebrate development, expand our understanding of the molecular events causing FADS, and suggest that variants in NUP88 should be investigated in cases of FADS.


Assuntos
Artrogripose/genética , Genes Letais , Mutação , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Alelos , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Artrogripose/embriologia , Artrogripose/fisiopatologia , Consanguinidade , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Modelos Moleculares , Proteínas Musculares/metabolismo , Junção Neuromuscular/fisiopatologia , Complexo de Proteínas Formadoras de Poros Nucleares/química , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Linhagem , Gravidez , Conformação Proteica , Receptores Nicotínicos/metabolismo , Homologia de Sequência de Aminoácidos , Peixe-Zebra/anormalidades , Peixe-Zebra/genética , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
5.
Biochem Biophys Res Commun ; 521(4): 1077-1082, 2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31733835

RESUMO

RIG-I-like receptors (RLRs) are cytoplasmic sensors for viral RNA that elicit antiviral innate immune responses. RLR signaling culminates in the activation of the protein kinase TBK1, which mediates phosphorylation and nuclear translocation of IRF3 that regulates expression of type I interferon genes. Here, we found that Nucleoporin 93 (Nup93), components of nuclear pore complex (NPC), plays an important role in RLR-mediated antiviral responses. Nup93-deficient RAW264.7 macrophage cells exhibited decreased expression of Ifnb1 and Cxcl10 genes after treatment with a synthetic RLR agonist stimulation as well as Newcastle Disease Virus infection. Silencing Nup93 in murine primary macrophages and embryonic fibroblasts also resulted in reduced expression of these genes. IRF3 nuclear translocation during RLR signaling was impaired in Nup93-deficient RAW264.7 cells. Notably, the activation of TBK1 during RLR signaling was also decreased in Nup93-deficient cells. We found that Nup93 formed a complex with TBK1, and Nup93 overexpression enhanced TBK1-mediated IFNß promoter activation. Taken together, our findings suggest that Nup93 regulates antiviral innate immunity by enhancing TBK1 activity and IRF3 nuclear translocation.


Assuntos
Antivirais/metabolismo , Imunidade Inata , Vírus da Doença de Newcastle/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Imunidade Inata/efeitos dos fármacos , Fator Regulador 3 de Interferon/metabolismo , Camundongos , Vírus da Doença de Newcastle/efeitos dos fármacos , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Poli I-C/farmacologia , Ligação Proteica/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico/efeitos dos fármacos , Células RAW 264.7
6.
Cell Mol Life Sci ; 76(17): 3407-3432, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30944974

RESUMO

Nucleocytoplasmic transport is dysregulated in sporadic and familial amyotrophic lateral sclerosis (ALS) and retinal ganglion neurons (RGNs) are purportedly involved in ALS. The Ran-binding protein 2 (Ranbp2) controls rate-limiting steps of nucleocytoplasmic transport. Mice with Ranbp2 loss in Thy1+-motoneurons develop cardinal ALS-like motor traits, but the impairments in RGNs and the degree of dysfunctional consonance between RGNs and motoneurons caused by Ranbp2 loss are unknown. This will help to understand the role of nucleocytoplasmic transport in the differential vulnerability of neuronal cell types to ALS and to uncover non-motor endophenotypes with pathognomonic signs of ALS. Here, we ascertain Ranbp2's function and endophenotypes in RGNs of an ALS-like mouse model lacking Ranbp2 in motoneurons and RGNs. Thy1+-RGNs lacking Ranbp2 shared with motoneurons the dysregulation of nucleocytoplasmic transport. RGN abnormalities were comprised morphologically by soma hypertrophy and optic nerve axonopathy and physiologically by a delay of the visual pathway's evoked potentials. Whole-transcriptome analysis showed restricted transcriptional changes in optic nerves that were distinct from those found in sciatic nerves. Specifically, the level and nucleocytoplasmic partition of the anti-apoptotic and novel substrate of Ranbp2, Pttg1/securin, were dysregulated. Further, acetyl-CoA carboxylase 1, which modulates de novo synthesis of fatty acids and T-cell immunity, showed the highest up-regulation (35-fold). This effect was reflected by the activation of ramified CD11b+ and CD45+-microglia, increase of F4\80+-microglia and a shift from pseudopodial/lamellipodial to amoeboidal F4\80+-microglia intermingled between RGNs of naive mice. Further, there was the intracellular sequestration in RGNs of metalloproteinase-28, which regulates macrophage recruitment and polarization in inflammation. Hence, Ranbp2 genetic insults in RGNs and motoneurons trigger distinct paracrine signaling likely by the dysregulation of nucleocytoplasmic transport of neuronal-type selective substrates. Immune-modulators underpinning RGN-to-microglial signaling are regulated by Ranbp2, and this neuronal-glial system manifests endophenotypes that are likely useful in the prognosis and diagnosis of motoneuron diseases, such as ALS.


Assuntos
Microglia/metabolismo , Chaperonas Moleculares/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Células Ganglionares da Retina/metabolismo , Acetil-CoA Carboxilase/genética , Acetil-CoA Carboxilase/metabolismo , Transporte Ativo do Núcleo Celular , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Modelos Animais de Doenças , Potenciais Evocados/efeitos dos fármacos , Regulação da Expressão Gênica , Metabolismo dos Lipídeos , Metaloproteinases da Matriz Secretadas/genética , Metaloproteinases da Matriz Secretadas/metabolismo , Camundongos , Camundongos Knockout , Chaperonas Moleculares/genética , Neurônios Motores/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Nervo Óptico/anormalidades , Nervo Óptico/patologia , Comunicação Parácrina , Tamoxifeno/farmacologia , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Transcriptoma
7.
Hum Genet ; 138(3): 221-229, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30758658

RESUMO

Nuclear pore complex (NPC) is a fundamental component of the nuclear envelope and is key to the nucleocytoplasmic transport. Mutations in several NUP genes that encode individual components of NPC known as nucleoporins have been identified in recent years among patients with static encephalopathies characterized by developmental delay and microcephaly. We describe a multiplex consanguineous family in which four affected members presented with severe neonatal hypotonia, profound global developmental delay, progressive microcephaly and early death. Autozygome and linkage analysis revealed that this phenotype is linked to a founder disease haplotype (chr9:127,113,732-135,288,807) in which whole exome sequencing revealed the presence of a novel homozygous missense variant in NUP214. Functional analysis of patient-derived fibroblasts recapitulated the dysmorphic phenotype of nuclei that was previously described in NUP214 knockdown cells. In addition, the typical rim staining of NUP214 is largely displaced, further supporting the deleterious effect of the variant. Our data expand the list of NUP genes that are mutated in encephalopathy disorders in humans.


Assuntos
Encefalopatias/diagnóstico , Encefalopatias/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Microcefalia/diagnóstico , Microcefalia/genética , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Sequência de Aminoácidos , Consanguinidade , Feminino , Genes Recessivos , Ligação Genética , Homozigoto , Humanos , Lactente , Mutação , Linhagem , Fenótipo , Índice de Gravidade de Doença , Sequenciamento do Exoma
8.
RNA ; 23(3): 365-377, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27932586

RESUMO

Nuclear pore complexes (NPCs), which are composed of nucleoporins (Nups) and regulate transport between the nucleus and cytoplasm, significantly impact the replicative life span (RLS) of Saccharomyces cerevisiae We previously reported that deletion of the nonessential gene NUP100 increases RLS, although the molecular basis for this effect was unknown. In this study, we find that nuclear tRNA accumulation contributes to increased longevity in nup100Δ cells. Fluorescence in situ hybridization (FISH) experiments demonstrate that several specific tRNAs accumulate in the nuclei of nup100Δ mutants. Protein levels of the transcription factor Gcn4 are increased when NUP100 is deleted, and GCN4 is required for the elevated life spans of nup100Δ mutants, similar to other previously described tRNA export and ribosomal mutants. Northern blots indicate that tRNA splicing and aminoacylation are not significantly affected in nup100Δ cells, suggesting that Nup100 is largely required for nuclear export of mature, processed tRNAs. Distinct tRNAs accumulate in the nuclei of nup100Δ and msn5Δ mutants, while Los1-GFP nucleocytoplasmic shuttling is unaffected by Nup100. Thus, we conclude that Nup100 regulates tRNA export in a manner distinct from Los1 or Msn5. Together, these experiments reveal a novel Nup100 role in the tRNA life cycle that impacts the S. cerevisiae life span.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Núcleo Celular/metabolismo , Regulação Fúngica da Expressão Gênica , Complexo de Proteínas Formadoras de Poros Nucleares/genética , RNA Fúngico/genética , RNA de Transferência/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Transporte Ativo do Núcleo Celular/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Northern Blotting , Divisão Celular , Meios de Cultura/química , Hibridização in Situ Fluorescente , Carioferinas/deficiência , Carioferinas/genética , Poro Nuclear/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , RNA Fúngico/metabolismo , RNA de Transferência/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Tempo
9.
Mol Pharmacol ; 92(1): 30-47, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28461585

RESUMO

Pannexin-1 (Panx1) channels mediate the efflux of ATP and AMP from cancer cells in response to induction of extrinsic apoptosis by death receptors or intrinsic apoptosis by chemotherapeutic agents. We previously described the accumulation of extracellular ATP /AMP during chemotherapy-induced apoptosis in Jurkat human leukemia cells. In this study, we compared how different signaling pathways determine extracellular nucleotide pools in control Jurkat cells versus Jurkat lines that lack the Fas-associated death domain (FADD) or receptor-interacting protein kinase 1 (RIP1) cell death regulatory proteins. Tumor necrosis factor-α induced extrinsic apoptosis in control Jurkat cells and necroptosis in FADD-deficient cells; treatment of both lines with chemotherapeutic drugs elicited similar intrinsic apoptosis. Robust extracellular ATP/AMP accumulation was observed in the FADD-deficient cells during necroptosis, but not during apoptotic activation of Panx1 channels. Accumulation of extracellular ATP/AMP was similarly absent in RIP1-deficient Jurkat cells during apoptotic responses to chemotherapeutic agents. Apoptotic activation triggered equivalent proteolytic gating of Panx1 channels in all three Jurkat cell lines. The differences in extracellular ATP/AMP accumulation correlated with cell-line-specific expression of ectonucleotidases that metabolized the released ATP/AMP. CD73 mRNA, and αß-methylene-ADP-inhibitable ecto-AMPase activity were elevated in the FADD-deficient cells. In contrast, the RIP1-deficient cells were defined by increased expression of tartrate-sensitive prostatic acid phosphatase as a broadly acting ectonucleotidase. Thus, extracellular nucleotide accumulation during regulated tumor cell death involves interplay between ATP/AMP efflux pathways and different cell-autonomous ectonucleotidases. Differential expression of particular ectonucleotidases in tumor cell variants will determine whether chemotherapy-induced activation of Panx1 channels drives accumulation of immunostimulatory ATP versus immunosuppressive adenosine within the tumor microenvironment.


Assuntos
Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Antineoplásicos/toxicidade , Conexinas/metabolismo , Proteína de Domínio de Morte Associada a Fas/deficiência , Proteínas do Tecido Nervoso/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Líquido Extracelular/efeitos dos fármacos , Líquido Extracelular/metabolismo , Humanos , Células Jurkat , Proteínas de Ligação a RNA , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
10.
Reproduction ; 151(1): 51-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26515778

RESUMO

Nucleoporin 50 kDa (NUP50), a component of the nuclear pore complex, is highly expressed in male germ cells, but its role in germ cells is largely unknown. In this study, we analyzed the expression and function of NUP50 during the embryonic development of germ cells using NUP50-deficient mice. NUP50 was expressed in germ cells of both sexes at embryonic day 15.5 (E15.5), E13.5, and E12.5. In addition, NUP50 expression was also detected in primordial germ cells (PGCs) migrating into the genital ridges at E9.5. The gonads of Nup50-/- embryos of both sexes contained few PGCs at both E11.5 and E12.5 and no developing germ cells at E15.5. The migratory PGCs in Nup50-/- embryos at E9.5 showed increased apoptosis but a normal rate of proliferation, resulting in the progressive loss of germ cells at later stages. Taken together, these results suggest that NUP50 plays an essential role in the survival of PGCs during embryonic development.


Assuntos
Células Germinativas Embrionárias/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Animais , Apoptose , Proliferação de Células , Sobrevivência Celular/fisiologia , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Feminino , Imunofluorescência , Idade Gestacional , Gônadas/química , Gônadas/embriologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexo de Proteínas Formadoras de Poros Nucleares/análise
11.
J Biol Chem ; 289(43): 29767-89, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25187515

RESUMO

Retinal pigment epithelium (RPE) degeneration underpins diseases triggered by disparate genetic lesions, noxious insults, or both. The pleiotropic Ranbp2 controls the expression of intrinsic and extrinsic pathological stressors impinging on cellular viability. However, the physiological targets and mechanisms controlled by Ranbp2 in tissue homeostasis, such as RPE, are ill defined. We show that mice, RPE-cre::Ranbp2(-/-), with selective Ranbp2 ablation in RPE develop pigmentary changes, syncytia, hypoplasia, age-dependent centrifugal and non-apoptotic degeneration of the RPE, and secondary leakage of choriocapillaris. These manifestations are accompanied by the development of F-actin clouds, metalloproteinase-11 activation, deregulation of expression or subcellular localization of critical RPE proteins, atrophic cell extrusions into the subretinal space, and compensatory proliferation of peripheral RPE. To gain mechanistic insights into what Ranbp2 activities are vital to the RPE, we performed genetic complementation analyses of transgenic lines of bacterial artificial chromosomes of Ranbp2 harboring loss of function of selective Ranbp2 domains expressed in a Ranbp2(-/-) background. Among the transgenic lines produced, only Tg(RBD2/3*-HA)::RPE-cre::Ranbp2(-/-)-expressing mutations, which selectively impair binding of RBD2/3 (Ran-binding domains 2 and 3) of Ranbp2 to Ran-GTP, recapitulate RPE degeneration, as observed with RPE-cre::Ranbp2(-/-). By contrast, Tg(RBD2/3*-HA) expression rescues the degeneration of cone photoreceptors lacking Ranbp2. The RPE of RPE-cre::Ranbp2(-/-) and Tg(RBD2/3*-HA)::RPE-cre::Ranbp2(-/-) share proteostatic deregulation of Ran GTPase, serotransferrin, and γ-tubulin and suppression of light-evoked electrophysiological responses. These studies unravel selective roles of Ranbp2 and its RBD2 and RBD3 in RPE survival and functions. We posit that the control of Ran GTPase by Ranbp2 emerges as a novel therapeutic target in diseases promoting RPE degeneration.


Assuntos
Deleção de Genes , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/química , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Animais , Capilares/patologia , Proliferação de Células , Sobrevivência Celular , Cromossomos Artificiais Bacterianos/metabolismo , Progressão da Doença , Fenômenos Eletrofisiológicos , Integrases/metabolismo , Camundongos , Camundongos Transgênicos , Mutação/genética , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Estrutura Terciária de Proteína , Epitélio Pigmentado da Retina/fisiopatologia , Epitélio Pigmentado da Retina/ultraestrutura , Relação Estrutura-Atividade
12.
J Biol Chem ; 289(8): 4600-25, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24403063

RESUMO

The immunophilins, cyclophilins, catalyze peptidyl cis-trans prolyl-isomerization (PPIase), a rate-limiting step in protein folding and a conformational switch in protein function. Cyclophilins are also chaperones. Noncatalytic mutations affecting the only cyclophilins with known but distinct physiological substrates, the Drosophila NinaA and its mammalian homolog, cyclophilin-B, impair opsin biogenesis and cause osteogenesis imperfecta, respectively. However, the physiological roles and substrates of most cyclophilins remain unknown. It is also unclear if PPIase and chaperone activities reflect distinct cyclophilin properties. To elucidate the physiological idiosyncrasy stemming from potential cyclophilin functions, we generated mice lacking endogenous Ran-binding protein-2 (Ranbp2) and expressing bacterial artificial chromosomes of Ranbp2 with impaired C-terminal chaperone and with (Tg-Ranbp2(WT-HA)) or without PPIase activities (Tg-Ranbp2(R2944A-HA)). The transgenic lines exhibit unique effects in proteostasis. Either line presents selective deficits in M-opsin biogenesis with its accumulation and aggregation in cone photoreceptors but without proteostatic impairment of two novel Ranbp2 cyclophilin partners, the cytokine-responsive effectors, STAT3/STAT5. Stress-induced STAT3 activation is also unaffected in Tg-Ranbp2(R2944A-HA)::Ranbp2(-/-). Conversely, proteomic analyses found that the multisystem proteinopathy/amyotrophic lateral sclerosis proteins, heterogeneous nuclear ribonucleoproteins A2/B1, are down-regulated post-transcriptionally only in Tg-Ranbp2(R2944A-HA)::Ranbp2(-/-). This is accompanied by the age- and tissue-dependent reductions of diubiquitin and ubiquitylated proteins, increased deubiquitylation activity, and accumulation of the 26 S proteasome subunits S1 and S5b. These manifestations are absent in another line, Tg-Ranbp2(CLDm-HA)::Ranbp2(-/-), harboring SUMO-1 and S1-binding mutations in the Ranbp2 cyclophilin-like domain. These results unveil distinct mechanistic and biological links between PPIase and chaperone activities of Ranbp2 cyclophilin toward proteostasis of selective substrates and with novel therapeutic potential.


Assuntos
Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/química , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Peptidilprolil Isomerase/química , Peptidilprolil Isomerase/metabolismo , Dobramento de Proteína , Envelhecimento/metabolismo , Animais , Biocatálise , Regulação para Baixo , Potenciais Evocados Visuais , Proteínas Ativadoras de GTPase/metabolismo , Células HeLa , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Histona Desacetilases/metabolismo , Humanos , Camundongos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutação/genética , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Opsinas/metabolismo , Especificidade de Órgãos , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Transporte Proteico , Células Fotorreceptoras Retinianas Cones/citologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Fatores de Transcrição STAT/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Relação Estrutura-Atividade , Ubiquitina/metabolismo
13.
Life Sci Alliance ; 5(3)2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34911810

RESUMO

Nuclear pore complexes, the channels connecting the nucleus with the cytoplasm, are built by multiple copies of ∼30 proteins called nucleoporins. Recent evidence has exposed that nucleoporins can play cell type-specific functions. Despite novel discoveries into the cellular functions of nucleoporins, their role in the regulation of mammalian tissue physiology remains mostly unexplored because of a limited number of nucleoporin mouse models. Here we show that ablation of Nup210/Gp210, a nucleoporin previously identified to play a role in myoblast differentiation and Zebrafish muscle maturation, is dispensable for skeletal muscle formation and growth in mice. We found that although primary satellite cells from Nup210 knockout mice can differentiate, these animals show delayed muscle repair after injury. Moreover, Nup210 knockout mice display an increased percentage of centrally nucleated fibers and abnormal fiber type distribution as they age. Muscle function experiments also exposed that Nup210 is required for muscle endurance during voluntary running. Our findings indicate that in mammals, Nup210 is important for the maintenance of skeletal muscle integrity and for proper muscle function providing novel insights into the in vivo roles of nuclear pore complex components.


Assuntos
Músculos/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Fenótipo , Regeneração/genética , Fatores Etários , Animais , Diferenciação Celular , Imunofluorescência , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Desenvolvimento Muscular/genética , Músculos/patologia , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo
14.
Yeast ; 28(6): 453-65, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21446055

RESUMO

Proteins exit the endoplasmic reticulum (ER) in vesicles pinching off from the membrane at sites covered by the COPII coat, which consists of Sec23/24p and Sec13/31p. We have shown that the glycoprotein Hsp150 exits the ER in the absence of Sec13p or any member of the Sec24p family. The determinant responsible for this resides in the C-terminal domain of Hsp150 (CTD). Here, A- and B-type Walker motifs were identified in the CTD. Authentic Hsp150 from the yeast culture medium, as well as Hsp150 and the CTD fragment produced in Escherichia coli, exhibited ATPase activity nearly three times higher than the published activity of the ER chaperone Kar2p/BiP. Deletion of the Walker motif, and a K335A mutation in it, abolished the ATPase activity. Hsp150 homologues Pir3p and Pir4p, differing in critical amino acids of the Walker motif, also lacked ATPase activity. Unexpectedly, inactivation of the ATPase activity blocked ER exit of Hsp150 in the absence of Sec24p or Sec13p function, whereas secretion in normal cells was not compromised. To our knowledge this is the first documentation of the ATPase activity of a protein serving an intracellular transport function.


Assuntos
Adenosina Trifosfatases/metabolismo , Retículo Endoplasmático/metabolismo , Glicoproteínas/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas de Membrana/deficiência , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Escherichia coli/genética , Expressão Gênica , Glicoproteínas/genética , Glicoproteínas/isolamento & purificação , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/isolamento & purificação , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/isolamento & purificação , Deleção de Sequência
15.
Nat Cell Biol ; 6(3): 197-206, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15039779

RESUMO

Translocation through nuclear pore complexes (NPCs) requires interactions between receptor-cargo complexes and phenylalanine-glycine (FG) repeats in multiple FG domain-containing NPC proteins (FG-Nups). We have systematically deleted the FG domains of 11 Saccharomyces cerevisiae FG-Nups in various combinations. All five asymmetrically localized FG domains deleted together were non-essential. However, specific combinations of symmetrically localized FG domains were essential. Over half the total mass of FG domains could be deleted without loss of viability or the NPC's normal permeability barrier. Significantly, symmetric deletions caused mild reductions in Kap95-Kap60-mediated import rates, but virtually abolished Kap104 import. These results suggest the existence of multiple translocation pathways.


Assuntos
Transporte Ativo do Núcleo Celular/genética , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Poro Nuclear/metabolismo , Transporte Proteico/genética , Sequências Repetitivas de Aminoácidos/fisiologia , Saccharomyces cerevisiae/metabolismo , Permeabilidade da Membrana Celular/genética , Sobrevivência Celular/genética , Células Cultivadas , Deleção de Genes , Carioferinas/genética , Carioferinas/metabolismo , Mutação/genética , Poro Nuclear/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Estrutura Terciária de Proteína/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , beta Carioferinas/genética , beta Carioferinas/metabolismo
16.
J Immunol ; 183(5): 3278-84, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19641134

RESUMO

The serine-threonine kinase RIP1 was originally identified through its ability to bind to the death domain of Fas (CD95). RIP1 has been shown to be recruited to the Fas death-inducing signaling complex (DISC) and is required for the induction of necrotic cell death. In this study, we show that in Jurkat T lymphocytes, RIP1 is also necessary for the most efficient activation of downstream caspases by Fas when treated with membrane-bound Fas ligand, but not with agonistic Abs or cross-linked soluble Fas ligand. RIP1 participates in the Fas-associated death domain protein-mediated recruitment of caspase-8 to the Fas receptor complex in a manner that promotes caspase-8 activation. Cross-linking Abs, such as CH11, bypass the requirement for RIP1 in caspase activation by initiating larger, though less efficient, DISC complexes, while membrane-bound Fas ligand initiates a smaller but more efficient DISC that functions, in part, by effectively incorporating more RIP1 into the complex. Consequently, RIP1 is likely a more integral part of physiological signaling through the Fas/CD95 receptor complex than previously recognized; at least when the signal is mediated by full-length membrane-bound FasL. Cross-linked soluble FasL, which also occurs physiologically, behaves similarly to the CH11 Ab, and may therefore be more likely to initiate nonapoptotic Fas signaling due to less RIP1 in the receptor complex. Thus, agonists that bind the same Fas receptor initiate mechanistically distinct pathways resulting in differential cytotoxicity.


Assuntos
Caspase 8/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Proteína Ligante Fas/metabolismo , Proteínas de Membrana/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Proteínas de Ligação a RNA/fisiologia , Anticorpos/metabolismo , Morte Celular/imunologia , Reagentes de Ligações Cruzadas/metabolismo , Citotoxicidade Imunológica , Ativação Enzimática/imunologia , Proteína Ligante Fas/imunologia , Proteína Ligante Fas/toxicidade , Humanos , Imunidade Inata , Imunoglobulina M/metabolismo , Células Jurkat , Proteínas de Membrana/imunologia , Proteínas de Membrana/toxicidade , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Oligopeptídeos , Peptídeos/imunologia , Proteínas de Ligação a RNA/metabolismo , Receptor fas/agonistas , Receptor fas/imunologia , Receptor fas/metabolismo
17.
Nat Commun ; 11(1): 2606, 2020 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-32451376

RESUMO

Nucleoporin proteins (Nups) have been proposed to mediate spatial and temporal chromatin organization during gene regulation. Nevertheless, the molecular mechanisms in mammalian cells are not well understood. Here, we report that Nucleoporin 153 (NUP153) interacts with the chromatin architectural proteins, CTCF and cohesin, and mediates their binding across cis-regulatory elements and TAD boundaries in mouse embryonic stem (ES) cells. NUP153 depletion results in altered CTCF and cohesin binding and differential gene expression - specifically at the bivalent developmental genes. To investigate the molecular mechanism, we utilize epidermal growth factor (EGF)-inducible immediate early genes (IEGs). We find that NUP153 controls CTCF and cohesin binding at the cis-regulatory elements and POL II pausing during the basal state. Furthermore, efficient IEG transcription relies on NUP153. We propose that NUP153 links the nuclear pore complex (NPC) to chromatin architecture allowing genes that are poised to respond rapidly to developmental cues to be properly modulated.


Assuntos
Fator de Ligação a CCCTC/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Animais , Fator de Ligação a CCCTC/química , Proteínas de Ciclo Celular/química , Linhagem Celular , Cromatina/química , Cromatina/genética , Proteínas Cromossômicas não Histona/química , Genes Precoces , Células HeLa , Humanos , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Poro Nuclear/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , RNA Polimerase II/metabolismo , Elementos Reguladores de Transcrição , Coesinas
18.
Science ; 294(5546): 1531-3, 2001 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-11711676

RESUMO

The sperm acrosome is essential for sperm-egg fusion and is often defective in men with nonobstructive infertility. Here we report that male mice with a null mutation in Hrb are infertile and display round-headed spermatozoa that lack an acrosome. In wild-type spermatids, Hrb is associated with the cytosolic surface of proacrosomic transport vesicles that fuse to create a single large acrosomic vesicle at step 3 of spermiogenesis. Although proacrosomic vesicles form in spermatids that lack Hrb, the vesicles are unable to fuse, blocking acrosome development at step 2. We conclude that Hrb is required for docking and/or fusion of proacrosomic vesicles during acrosome biogenesis.


Assuntos
Acrossomo/fisiologia , Acrossomo/ultraestrutura , Proteínas de Transporte/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Proteínas de Ligação a RNA , Espermátides/fisiologia , Espermatogênese , Vesículas Transportadoras/fisiologia , Acrossomo/química , Subunidades alfa do Complexo de Proteínas Adaptadoras , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Animais , Proteínas de Ligação ao Cálcio/análise , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Transporte/análise , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Feminino , Fertilização in vitro , Marcação de Genes , Complexo de Golgi/química , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Microscopia Confocal , Microscopia Eletrônica , Microscopia Imunoeletrônica , Mutação , Complexo de Proteínas Formadoras de Poros Nucleares/análise , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Fosfoproteínas/análise , Fosfoproteínas/metabolismo , Contagem de Espermatozoides , Motilidade dos Espermatozoides , Interações Espermatozoide-Óvulo , Espermátides/química , Espermátides/ultraestrutura , Espermatozoides/fisiologia , Espermatozoides/ultraestrutura , Vesículas Transportadoras/química
19.
Mol Cell Biol ; 26(18): 6772-85, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16943420

RESUMO

Nucleoporins mediate transport of macromolecules across the nuclear pore complex, yet the function of many individual nucleoporins is largely unresolved. To address this question, we depleted cells of the cytoplasmic nucleoporins Nup214/CAN and Nup358/RanBP2 by RNA interference. Depletion of Nup214 resulted in codepletion of its binding partner, Nup88. Nuclear pore complexes assembled in the absence of Nup214/Nup88 or Nup358 were fully functional in nuclear protein import, whereas nuclear mRNA export was slightly impaired. Depletion of Nup358 had only a minor effect on nuclear protein export. In contrast, depletion of Nup214/Nup88 led to strongly reduced CRM1-mediated export of the shuttling transcription factor NFAT as well as a human immunodeficiency virus-Rev derivative. A specific role of Nup214 in protein export is furthered by the biochemical properties of a high-affinity complex containing Nup214, CRM1, RanGTP, and an export cargo. Our results show that the Nup214/Nup88 complex is required for efficient CRM1-mediated transport, supporting a model involving a high-affinity binding site for CRM1 at Nup214 in the terminal steps of export.


Assuntos
Transporte Ativo do Núcleo Celular , Carioferinas/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Poro Nuclear/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Processos de Crescimento Celular , Citoplasma/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Células HeLa , Humanos , Chaperonas Moleculares/metabolismo , Fatores de Transcrição NFATC/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Poli A/metabolismo , Ligação Proteica , Interferência de RNA , Transporte de RNA , Proteína Exportina 1
20.
BMC Syst Biol ; 12(1): 62, 2018 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-29848314

RESUMO

BACKGROUND: Atrial fibrillation is a cardiac disease driven by numerous idiopathic etiologies. NUP155 is a nuclear pore complex protein that has been identified as a clinical driver of atrial fibrillation, yet the precise mechanism is unknown. The present study employs a systems biology algorithm to identify effects of NUP155 disruption on cardiogenicity in a model of stem cell-derived differentiation. METHODS: Embryonic stem (ES) cell lines (n = 5) with truncated NUP155 were cultured in parallel with wild type (WT) ES cells (n = 5), and then harvested for RNAseq. Samples were run on an Illumina HiSeq 2000. Reads were analyzed using Strand NGS, Cytoscape, DAVID and Ingenuity Pathways Analysis to deconvolute the NUP155-disrupted transcriptome. Network topological analysis identified key features that controlled framework architecture and functional enrichment. RESULTS: In NUP155 truncated ES cells, significant expression changes were detected in 326 genes compared to WT. These genes segregated into clusters that enriched for specific gene ontologies. Deconvolution of the collective framework into discrete sub-networks identified a module with the highest score that enriched for Cardiovascular System Development, and revealed NTRK1/TRKA and SRSF2/SC35 as critical hubs within this cardiogenic module. CONCLUSIONS: The strategy of pluripotent transcriptome deconvolution used in the current study identified a novel association of NUP155 with potential drivers of arrhythmogenic AF. Here, NUP155 regulates cardioplasticity of a sub-network embedded within a larger framework of genome integrity, and exemplifies how transcriptome cardiogenicity in an embryonic stem cell genome is recalibrated by nucleoporin dysfunction.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Miocárdio/citologia , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Transdução de Sinais/genética , Animais , Corpos Embrioides/citologia , Camundongos , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa