Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Invest Dermatol ; 144(2): 284-295.e16, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37716648

RESUMO

Desmosomes are dynamic complex protein structures involved in cellular adhesion. Disruption of these structures by loss-of-function variants in desmosomal genes leads to a variety of skin- and heart-related phenotypes. In this study, we report TUFT1 as a desmosome-associated protein, implicated in epidermal integrity. In two siblings with mild skin fragility, woolly hair, and mild palmoplantar keratoderma but without a cardiac phenotype, we identified a homozygous splice-site variant in the TUFT1 gene, leading to aberrant mRNA splicing and loss of TUFT1 protein. Patients' skin and keratinocytes showed acantholysis, perinuclear retraction of intermediate filaments, and reduced mechanical stress resistance. Immunolabeling and transfection studies showed that TUFT1 is positioned within the desmosome and that its location is dependent on the presence of the desmoplakin carboxy-terminal tail. A Tuft1-knockout mouse model mimicked the patients' phenotypes. Altogether, this study reveals TUFT1 as a desmosome-associated protein, whose absence causes skin fragility, woolly hair, and palmoplantar keratoderma.


Assuntos
Doenças do Cabelo , Ceratodermia Palmar e Plantar , Anormalidades da Pele , Animais , Humanos , Camundongos , Desmoplaquinas/genética , Desmoplaquinas/metabolismo , Desmossomos/metabolismo , Cabelo/metabolismo , Doenças do Cabelo/genética , Doenças do Cabelo/metabolismo , Ceratodermia Palmar e Plantar/genética , Ceratodermia Palmar e Plantar/metabolismo , Pele/metabolismo , Anormalidades da Pele/metabolismo
2.
Acta Neuropathol ; 146(2): 353-368, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37119330

RESUMO

Hereditary spastic paraplegias (HSP) are rare, inherited neurodegenerative or neurodevelopmental disorders that mainly present with lower limb spasticity and muscle weakness due to motor neuron dysfunction. Whole genome sequencing identified bi-allelic truncating variants in AMFR, encoding a RING-H2 finger E3 ubiquitin ligase anchored at the membrane of the endoplasmic reticulum (ER), in two previously genetically unexplained HSP-affected siblings. Subsequently, international collaboration recognized additional HSP-affected individuals with similar bi-allelic truncating AMFR variants, resulting in a cohort of 20 individuals from 8 unrelated, consanguineous families. Variants segregated with a phenotype of mainly pure but also complex HSP consisting of global developmental delay, mild intellectual disability, motor dysfunction, and progressive spasticity. Patient-derived fibroblasts, neural stem cells (NSCs), and in vivo zebrafish modeling were used to investigate pathomechanisms, including initial preclinical therapy assessment. The absence of AMFR disturbs lipid homeostasis, causing lipid droplet accumulation in NSCs and patient-derived fibroblasts which is rescued upon AMFR re-expression. Electron microscopy indicates ER morphology alterations in the absence of AMFR. Similar findings are seen in amfra-/- zebrafish larvae, in addition to altered touch-evoked escape response and defects in motor neuron branching, phenocopying the HSP observed in patients. Interestingly, administration of FDA-approved statins improves touch-evoked escape response and motor neuron branching defects in amfra-/- zebrafish larvae, suggesting potential therapeutic implications. Our genetic and functional studies identify bi-allelic truncating variants in AMFR as a cause of a novel autosomal recessive HSP by altering lipid metabolism, which may potentially be therapeutically modulated using precision medicine with statins.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Paraplegia Espástica Hereditária , Animais , Humanos , Paraplegia Espástica Hereditária/tratamento farmacológico , Paraplegia Espástica Hereditária/genética , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Peixe-Zebra , Mutação , Neurônios Motores , Receptores do Fator Autócrino de Motilidade/genética
3.
PLoS One ; 13(6): e0199737, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29953513

RESUMO

INTRODUCTION: The gene encoding isocitrate dehydrogenase 1 (IDH1) is frequently mutated in several tumor types including gliomas. The most prevalent mutation in gliomas is a missense mutation leading to a substitution of arginine with histidine at the residue 132 (R132H). Wild type IDH1 catalyzes oxidative decarboxylation of isocitrate to α-ketoglutarate (α-KG) whereas mutant IDH1 converts α-KG into D2-hydroxyglutarate (D2HG). Unfortunately, there are few in vivo model systems for IDH-mutated tumors to study the effects of IDH1 mutations in tumor development. We have therefore created transgenic zebrafish lines that express various IDH1 mutants. MATERIALS AND METHODS: IDH1 mutations (IDH1R132H, IDH1R132C and loss-of-function mutation IDH1G70D), IDH1wildtype or eGFP were cloned into constructs with several brain-specific promoters (Nestin, Gfap or Gata2). These constructs were injected into fertilized zebrafish eggs at the one-cell stage. RESULTS: In total more than ten transgenic zebrafish lines expressing various brain-specific IDH1 mutations were created. A significant increase in the level of D2HG was observed in all transgenic lines expressing IDH1R132C or IDH1R132H, but not in any of the lines expressing IDH1wildtype, IDH1G70D or eGFP. No differences in 5-hydroxymethyl cytosine and mature collagen IV levels were observed between wildtype and mutant IDH1 transgenic fish. To our surprise, we failed to identify any strong phenotype, despite increased levels of the oncometabolite D2HG. No tumors were observed, even when backcrossing with tp53-mutant fish which suggests that additional transforming events are required for tumor formation. Elevated D2HG levels could be lowered by treatment of the transgenic zebrafish with an inhibitor of mutant IDH1 activity. CONCLUSIONS: We have generated a transgenic zebrafish model system for mutations in IDH1 that can be used for functional analysis and drug screening. Our model systems help understand the biology of IDH1 mutations and its role in tumor formation.


Assuntos
Animais Geneticamente Modificados , Glioma , Isocitrato Desidrogenase , Mutação de Sentido Incorreto , Neoplasias Experimentais , Proteínas de Peixe-Zebra , Peixe-Zebra , Substituição de Aminoácidos , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Glioma/tratamento farmacológico , Glioma/enzimologia , Glioma/genética , Glioma/patologia , Humanos , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
4.
Nat Commun ; 9(1): 299, 2018 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-29352114

RESUMO

The ubiquitously expressed RNA-binding proteins Roquin-1 and Roquin-2 are essential for appropriate immune cell function and postnatal survival of mice. Roquin proteins repress target mRNAs by recognizing secondary structures in their 3'-UTRs and by inducing mRNA decay. However, it is unknown if other cellular proteins contribute to target control. To identify cofactors of Roquin, we used RNA interference to screen ~1500 genes involved in RNA-binding or mRNA degradation, and identified NUFIP2 as a cofactor of Roquin-induced mRNA decay. NUFIP2 binds directly and with high affinity to Roquin, which stabilizes NUFIP2 in cells. Post-transcriptional repression of human ICOS by endogenous Roquin proteins requires two neighboring non-canonical stem-loops in the ICOS 3'-UTR. This unconventional cis-element as well as another tandem loop known to confer Roquin-mediated regulation of the Ox40 3'-UTR, are bound cooperatively by Roquin and NUFIP2. NUFIP2 therefore emerges as a cofactor that contributes to mRNA target recognition by Roquin.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Proteínas Nucleares/genética , Proteínas de Ligação a RNA/genética , Receptores OX40/genética , Proteínas Repressoras/genética , Ubiquitina-Proteína Ligases/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linfócitos T CD4-Positivos/citologia , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Proteína Coestimuladora de Linfócitos T Induzíveis/antagonistas & inibidores , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Sequências Repetidas Invertidas , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/imunologia , Conformação de Ácido Nucleico , Cultura Primária de Células , Ligação Proteica , Estabilidade de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/imunologia , Receptores OX40/antagonistas & inibidores , Receptores OX40/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Repressoras/imunologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Ubiquitina-Proteína Ligases/imunologia
5.
Neuron ; 93(2): 331-347, 2017 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-28065649

RESUMO

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a neurodegenerative disorder caused by a limited expansion of CGG repeats in the 5' UTR of FMR1. Two mechanisms are proposed to cause FXTAS: RNA gain-of-function, where CGG RNA sequesters specific proteins, and translation of CGG repeats into a polyglycine-containing protein, FMRpolyG. Here we developed transgenic mice expressing CGG repeat RNA with or without FMRpolyG. Expression of FMRpolyG is pathogenic, while the sole expression of CGG RNA is not. FMRpolyG interacts with the nuclear lamina protein LAP2ß and disorganizes the nuclear lamina architecture in neurons differentiated from FXTAS iPS cells. Finally, expression of LAP2ß rescues neuronal death induced by FMRpolyG. Overall, these results suggest that translation of expanded CGG repeats into FMRpolyG alters nuclear lamina architecture and drives pathogenesis in FXTAS.


Assuntos
Ataxia/genética , Proteínas de Ligação a DNA/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Proteínas de Membrana/metabolismo , Lâmina Nuclear/metabolismo , Peptídeos/genética , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Tremor/genética , Expansão das Repetições de Trinucleotídeos/genética , Animais , Ataxia/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Lâmina Nuclear/patologia , Peptídeos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Tremor/metabolismo
6.
J Exp Med ; 213(7): 1163-74, 2016 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-27325888

RESUMO

Pseudo-TORCH syndrome (PTS) is characterized by microcephaly, enlarged ventricles, cerebral calcification, and, occasionally, by systemic features at birth resembling the sequelae of congenital infection but in the absence of an infectious agent. Genetic defects resulting in activation of type 1 interferon (IFN) responses have been documented to cause Aicardi-Goutières syndrome, which is a cause of PTS. Ubiquitin-specific peptidase 18 (USP18) is a key negative regulator of type I IFN signaling. In this study, we identified loss-of-function recessive mutations of USP18 in five PTS patients from two unrelated families. Ex vivo brain autopsy material demonstrated innate immune inflammation with calcification and polymicrogyria. In vitro, patient fibroblasts displayed severely enhanced IFN-induced inflammation, which was completely rescued by lentiviral transduction of USP18. These findings add USP18 deficiency to the list of genetic disorders collectively termed type I interferonopathies. Moreover, USP18 deficiency represents the first genetic disorder of PTS caused by dysregulation of the response to type I IFNs. Therapeutically, this places USP18 as a promising target not only for genetic but also acquired IFN-mediated CNS disorders.


Assuntos
Doenças Autoimunes do Sistema Nervoso , Encéfalo/imunologia , Calcinose , Endopeptidases/deficiência , Imunidade Inata , Interferon Tipo I/imunologia , Microglia/imunologia , Malformações do Sistema Nervoso , Transdução de Sinais , Doenças Autoimunes do Sistema Nervoso/genética , Doenças Autoimunes do Sistema Nervoso/imunologia , Doenças Autoimunes do Sistema Nervoso/patologia , Encéfalo/patologia , Calcinose/genética , Calcinose/imunologia , Calcinose/patologia , Endopeptidases/imunologia , Feminino , Humanos , Interferon Tipo I/genética , Masculino , Microglia/patologia , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/imunologia , Malformações do Sistema Nervoso/patologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Ubiquitina Tiolesterase
7.
Front Cell Neurosci ; 9: 234, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26190969

RESUMO

Defects in the rat sarcoma viral oncogene homolog (Ras)/extracellular-signal-regulated kinase and the phosphatidylinositol 3-kinase-mammalian target of rapamycin (mTOR) signaling pathways are responsible for several neurodevelopmental disorders. These disorders are an important cause for intellectual disability; additional manifestations include autism spectrum disorder, seizures, and brain malformations. Changes in synaptic function are thought to underlie the neurological conditions associated with these syndromes. We therefore studied morphology and in vivo synaptic transmission of the calyx of Held synapse, a relay synapse in the medial nucleus of the trapezoid body (MNTB) of the auditory brainstem, in mouse models of tuberous sclerosis complex (TSC), Fragile X syndrome (FXS), Neurofibromatosis type 1 (NF1), and Costello syndrome. Calyces from both Tsc1(+/-) and from Fmr1 knock-out (KO) mice showed increased volume and surface area compared to wild-type (WT) controls. In addition, in Fmr1 KO animals a larger fraction of calyces showed complex morphology. In MNTB principal neurons of Nf1 (+/) (-) mice the average delay between EPSPs and APs was slightly smaller compared to WT controls, which could indicate an increased excitability. Otherwise, no obvious changes in synaptic transmission, or short-term plasticity were observed during juxtacellular recordings in any of the four lines. Our results in these four mutants thus indicate that abnormalities of mTOR or Ras signaling do not necessarily result in changes in in vivo synaptic transmission.

8.
Hum Mol Genet ; 24(17): 4948-57, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26060190

RESUMO

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder affecting carriers of the fragile X-premutation, who have an expanded CGG repeat in the 5'-UTR of the FMR1 gene. FXTAS is characterized by progressive development of intention tremor, ataxia, parkinsonism and neuropsychological problems. The disease is thought to be caused by a toxic RNA gain-of-function mechanism, and the major hallmark of the disease is ubiquitin-positive intranuclear inclusions in neurons and astrocytes. We have developed a new transgenic mouse model in which we can induce expression of an expanded repeat in the brain upon doxycycline (dox) exposure (i.e. Tet-On mice). This Tet-On model makes use of the PrP-rtTA driver and allows us to study disease progression and possibilities of reversibility. In these mice, 8 weeks of dox exposure was sufficient to induce the formation of ubiquitin-positive intranuclear inclusions, which also stain positive for the RAN translation product FMRpolyG. Formation of these inclusions is reversible after stopping expression of the expanded CGG RNA at an early developmental stage. Furthermore, we observed a deficit in the compensatory eye movements of mice with inclusions, a functional phenotype that could be reduced by stopping expression of the expanded CGG RNA early in the disease development. Taken together, this study shows, for the first time, the potential of disease reversibility and suggests that early intervention might be beneficial for FXTAS patients.


Assuntos
Ataxia/genética , Ataxia/fisiopatologia , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/fisiopatologia , Tremor/genética , Tremor/fisiopatologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Movimentos Oculares/genética , Expressão Gênica , Genes Reporter , Humanos , Corpos de Inclusão Intranuclear/patologia , Camundongos , Camundongos Transgênicos , Peptídeos/metabolismo , Ligação Proteica , Transporte Proteico , Expansão das Repetições de Trinucleotídeos , Ubiquitina/metabolismo
9.
EMBO Mol Med ; 7(4): 423-37, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25693964

RESUMO

Loss of function of the FMR1 gene leads to fragile X syndrome (FXS), the most common form of intellectual disability. The loss of FMR1 function is usually caused by epigenetic silencing of the FMR1 promoter leading to expansion and subsequent methylation of a CGG repeat in the 5' untranslated region. Very few coding sequence variations have been experimentally characterized and shown to be causal to the disease. Here, we describe a novel FMR1 mutation and reveal an unexpected nuclear export function for the C-terminus of FMRP. We screened a cohort of patients with typical FXS symptoms who tested negative for CGG repeat expansion in the FMR1 locus. In one patient, we identified a guanine insertion in FMR1 exon 15. This mutation alters the open reading frame creating a short novel C-terminal sequence, followed by a stop codon. We find that this novel peptide encodes a functional nuclear localization signal (NLS) targeting the patient FMRP to the nucleolus in human cells. We also reveal an evolutionarily conserved nuclear export function associated with the endogenous C-terminus of FMRP. In vivo analyses in Drosophila demonstrate that a patient-mimetic mutation alters the localization and function of Dfmrp in neurons, leading to neomorphic neuronal phenotypes.


Assuntos
Núcleo Celular , Proteína do X Frágil da Deficiência Intelectual , Síndrome do Cromossomo X Frágil , Mutação , Sinais de Localização Nuclear , Expansão das Repetições de Trinucleotídeos , Animais , Linhagem Celular Transformada , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Drosophila melanogaster , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/metabolismo , Síndrome do Cromossomo X Frágil/patologia , Humanos , Masculino , Camundongos , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico/genética
10.
Stem Cell Reports ; 3(5): 892-904, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25418731

RESUMO

The scarcity of primordial germ cells (PGCs) in the developing mammalian embryo hampers robust biochemical analysis of the processes that underlie early germ cell formation. Here, we demonstrate that DAZL, a germ cell-specific RNA binding protein, is a robust PGC marker during in vitro germ cell development. Using Dazl-GFP reporter ESCs, we demonstrate that DAZL plays a central role in a large mRNA/protein interactive network that blocks the translation of core pluripotency factors, including Sox2 and Sall4, as well as of Suz12, a polycomb family member required for differentiation of pluripotent cells. Thus, DAZL limits both pluripotency and somatic differentiation in nascent PGCs. In addition, we observed that DAZL associates with mRNAs of key Caspases and similarly inhibits their translation. This elegant fail-safe mechanism ensures that, whereas loss of DAZL results in prolonged expression of pluripotency factors, teratoma formation is avoided due to the concomitant activation of the apoptotic cascade.


Assuntos
Apoptose/genética , Diferenciação Celular/genética , Embrião de Mamíferos/metabolismo , Células Germinativas/metabolismo , Proteínas de Ligação a RNA/genética , Animais , Animais Geneticamente Modificados , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Immunoblotting , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Microscopia Confocal , Análise de Sequência com Séries de Oligonucleotídeos , Células-Tronco Pluripotentes/metabolismo , Interferência de RNA , Proteínas de Ligação a RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Neurobiol Dis ; 65: 43-54, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24418349

RESUMO

Male premutation carriers presenting between 55 and 200 CGG repeats in the Fragile-X-associated (FMR1) gene are at risk of developing Fragile X Tremor/Ataxia Syndrome (FXTAS), and females undergo Premature Ovarian Failure (POF1). Here, we have evaluated gene expression profiles from blood in male FMR1 premutation carriers and detected a strong deregulation of genes enriched in FXTAS relevant biological pathways, including inflammation, neuronal homeostasis and viability. Gene expression profiling distinguished between control individuals, carriers with FXTAS and carriers without FXTAS, with levels of expanded FMR1 mRNA being increased in FXTAS patients. In vitro studies in a neuronal cell model indicate that expression levels of expanded FMR1 5'-UTR are relevant in modulating the transcriptome. Thus, perturbations of the transcriptome may be an interplay between the CGG expansion size and FMR1 expression levels. Several deregulated genes (DFFA, BCL2L11, BCL2L1, APP, SOD1, RNF10, HDAC5, KCNC3, ATXN7, ATXN3 and EAP1) were validated in brain samples of a FXTAS mouse model. Downregulation of EAP1, a gene involved in the female reproductive system physiology, was confirmed in female carriers. Decreased levels were detected in female carriers with POF1 compared to those without POF1, suggesting that EAP1 levels contribute to ovarian insufficiency. In summary, gene expression profiling in blood has uncovered mechanisms that may underlie different pathological aspects of the premutation. A better understanding of the transcriptome dynamics in relation with expanded FMR1 mRNA expression levels and CGG expansion size may provide mechanistic insights into the disease process and a more accurate FXTAS diagnosis to the myriad of phenotypes associated with the premutation.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/genética , Regulação da Expressão Gênica/genética , Infertilidade/genética , Doenças Neurodegenerativas/sangue , Expansão das Repetições de Trinucleotídeos/genética , Adulto , Idoso , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Análise em Microsséries , Proteínas dos Microfilamentos , Pessoa de Meia-Idade , Neuroblastoma/patologia , Doenças Neurodegenerativas/genética , Fenótipo , Proteínas/genética , Proteínas/metabolismo , Securina/genética , Securina/metabolismo
12.
Proc Natl Acad Sci U S A ; 110(32): 13109-13, 2013 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-23878230

RESUMO

Recent evidence indicates there is a role for small membrane vesicles, including exosomes, as vehicles for intercellular communication. Exosomes secreted by most cell types can mediate transfer of proteins, mRNAs, and microRNAs, but their role in the transmission of infectious agents is less established. Recent studies have shown that hepatocyte-derived exosomes containing hepatitis C virus (HCV) RNA can activate innate immune cells, but the role of exosomes in the transmission of HCV between hepatocytes remains unknown. In this study, we investigated whether exosomes transfer HCV in the presence of neutralizing antibodies. Purified exosomes isolated from HCV-infected human hepatoma Huh7.5.1 cells were shown to contain full-length viral RNA, viral protein, and particles, as determined by RT-PCR, mass spectrometry, and transmission electron microscopy. Exosomes from HCV-infected cells were capable of transmitting infection to naive human hepatoma Huh7.5.1 cells and establishing a productive infection. Even with subgenomic replicons, lacking structural viral proteins, exosome-mediated transmission of HCV RNA was observed. Treatment with patient-derived IgGs showed a variable degree of neutralization of exosome-mediated infection compared with free virus. In conclusion, this study showed that hepatic exosomes can transmit productive HCV infection in vitro and are partially resistant to antibody neutralization. This discovery sheds light on neutralizing antibodies resistant to HCV transmission by exosomes as a potential immune evasion mechanism.


Assuntos
Exossomos/virologia , Hepacivirus/genética , RNA Viral/genética , Vírion/genética , Anticorpos Neutralizantes/imunologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Claudina-1/imunologia , Claudina-1/metabolismo , Exossomos/metabolismo , Exossomos/ultraestrutura , Hepacivirus/imunologia , Hepacivirus/fisiologia , Hepatite C/imunologia , Hepatite C/virologia , Interações Hospedeiro-Patógeno , Humanos , Imunoglobulina G/imunologia , Espectrometria de Massas , Microscopia Confocal , Microscopia Eletrônica de Transmissão , RNA Viral/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Depuradores Classe B/imunologia , Receptores Depuradores Classe B/metabolismo , Tetraspanina 28/imunologia , Tetraspanina 28/metabolismo , Vírion/fisiologia , Vírion/ultraestrutura
13.
Brain Res Bull ; 90: 118-26, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23123961

RESUMO

ATP13A2 is a lysosome-specific transmembrane ATPase protein of unknown function. This protein was initially linked to Kufor-Rakeb syndrome where it is absent or mutated. More recently, point mutations in ATP13A2 were linked to familial cases of Parkinson's disease. Zebrafish is commonly used as a vertebrate model for the study of different neurodegenerative diseases and has homologues of several Parkinson's disease associated proteins. Here, we describe for the first time the zebrafish homologue of human ATP13A2, demonstrating the homology between the protein sequences, which supports a conserved biological role. Furthermore, the spatial pattern of protein expression was studied and the lethality of the knockdown of ATP13A2 suggests it plays a crucial role during embryonic development. Our findings bring new insight into the biology of ATP13A2 and open novel opportunities for its study using zebrafish as a model organism.


Assuntos
Desenvolvimento Embrionário/genética , ATPases Translocadoras de Prótons/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Animais , Biologia Computacional , Embrião não Mamífero , Desenvolvimento Embrionário/efeitos dos fármacos , Humanos , Locomoção/efeitos dos fármacos , Locomoção/genética , Morfolinos/farmacologia , Mutação/genética , ATPases Translocadoras de Prótons/química , ATPases Translocadoras de Prótons/metabolismo , Homologia de Sequência , Proteínas de Peixe-Zebra/metabolismo
14.
PLoS One ; 8(12): e82589, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24391718

RESUMO

BACKGROUND: Current markers for prostate cancer, such as PSA lack specificity. Therefore, novel biomarkers are needed. Unfortunately, the complexity of body fluids often hampers biomarker discovery. An attractive alternative approach is the isolation of small vesicles, i.e. exosomes, ∼100 nm, which contain proteins that are specific to the tissue from which they are derived and therefore can be considered as treasure chests for disease-specific biomarker discovery. MATERIALS AND METHODS: Exosomes were isolated from 2 immortalized primary prostate epithelial cells (PNT2C2 and RWPE-1) and 2 PCa cell lines (PC346C and VCaP) by ultracentrifugation. After tryptic digestion, proteomic analyses utilized a nanoLC coupled with an LTQ-Orbitrap operated in tandem MS (MS/MS) mode. Accurate Mass and Time (AMT) tag approach was employed for peptide identification and quantitation. Candidate biomarkers were validated by Western blotting and Immunohistochemistry. RESULTS: Proteomic characterization resulted in the identification of 248, 233, 169, and 216 proteins by at least 2 peptides in exosomes from PNT2C2, RWPE-1, PC346C, and VCaP, respectively. Statistical analyses revealed 52 proteins differently abundant between PCa and control cells, 9 of which were more abundant in PCa. Validation by Western blotting confirmed a higher abundance of FASN, XPO1 and PDCD6IP (ALIX) in PCa exosomes. CONCLUSIONS: Identification of exosomal proteins using high performance LC-FTMS resulted in the discovery of PDCD6IP, FASN, XPO1 and ENO1 as new candidate biomarkers for prostate cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias da Próstata/metabolismo , Western Blotting , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Exossomos/metabolismo , Exossomos/ultraestrutura , Ácido Graxo Sintase Tipo I/metabolismo , Humanos , Carioferinas/metabolismo , Masculino , Microscopia Eletrônica de Transmissão , Fosfopiruvato Hidratase/metabolismo , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/ultraestrutura , Proteômica/métodos , Receptores Citoplasmáticos e Nucleares/metabolismo , Espectrometria de Massas em Tandem , Proteínas Supressoras de Tumor/metabolismo , Proteína Exportina 1
15.
Am J Hum Genet ; 90(3): 467-77, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22341971

RESUMO

Manganese is essential for several metabolic pathways but becomes toxic in excessive amounts. Manganese levels in the body are therefore tightly regulated, but the responsible protein(s) remain incompletely known. We studied two consanguineous families with neurologic disorders including juvenile-onset dystonia, adult-onset parkinsonism, severe hypermanganesemia, polycythemia, and chronic hepatic disease, including steatosis and cirrhosis. We localized the genetic defect by homozygosity mapping and then identified two different homozygous frameshift SLC30A10 mutations, segregating with disease. SLC30A10 is highly expressed in the liver and brain, including in the basal ganglia. Its encoded protein belongs to a large family of membrane transporters, mediating the efflux of divalent cations from the cytosol. We show the localization of SLC30A10 in normal human liver and nervous system, and its depletion in liver from one affected individual. Our in silico analyses suggest that SLC30A10 possesses substrate specificity different from its closest (zinc-transporting) homologs. We also show that the expression of SLC30A10 and the levels of the encoded protein are markedly induced by manganese in vitro. The phenotype associated with SLC30A10 mutations is broad, including neurologic, hepatic, and hematologic disturbances. Intrafamilial phenotypic variability is also present. Chelation therapy can normalize the manganesemia, leading to marked clinical improvements. In conclusion, we show that SLC30A10 mutations cause a treatable recessive disease with pleomorphic phenotype, and provide compelling evidence that SLC30A10 plays a pivotal role in manganese transport. This work has broad implications for understanding of the manganese biology and pathophysiology in multiple human organs.


Assuntos
Proteínas de Transporte de Cátions/genética , Intoxicação por Manganês/genética , Proteínas de Membrana Transportadoras/genética , Doenças Metabólicas/genética , Transtornos Parkinsonianos/genética , Idoso , Sequência de Aminoácidos , Encéfalo/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Mapeamento Cromossômico/métodos , Feminino , Mutação da Fase de Leitura/genética , Genes Recessivos , Predisposição Genética para Doença , Células Hep G2 , Homozigoto , Humanos , Imuno-Histoquímica/métodos , Fígado/metabolismo , Masculino , Manganês/metabolismo , Intoxicação por Manganês/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Doenças Metabólicas/metabolismo , Pessoa de Meia-Idade , Dados de Sequência Molecular , Fenótipo , Alinhamento de Sequência/métodos , Células Tumorais Cultivadas , Transportador 8 de Zinco
16.
Hum Mol Genet ; 20(1): 64-79, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20935171

RESUMO

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late adult-onset neurodegenerative disorder caused by a premutation CGG-trinucleotide repeat expansion (55-200 CGG repeats) within the 5'-untranslated region of the FMR1 gene. Although FXTAS generally affects premutation carriers over 50 years of age, cognitive and psychological symptoms can appear in carriers during childhood, suggesting that the FMR1 premutation affects brain function early in life. Recent work with cultured hippocampal neurons from a premutation (Fmr1 CGG knock-in) mouse model revealed impaired development of early postnatal neurons, consistent with the developmental clinical involvement of premutation carriers. In the current work, we show that the presence of premutation CGG-repeat expansions in the mouse Fmr1 gene alters embryonic neocortical development. Specifically, embryonic premutation mice display migration defects in the neocortex and altered expression of neuronal lineage markers. The current data demonstrate that premutation alleles of the Fmr1 gene are associated with defects in developmental programs operating during prenatal stages of brain formation and provide further evidence that the FMR1 premutation has a neurodevelopmental component.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/genética , Neocórtex/crescimento & desenvolvimento , Expansão das Repetições de Trinucleotídeos/genética , Regiões 5' não Traduzidas/genética , Animais , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Vetores Genéticos , Camundongos , Retroviridae
17.
Ann Neurol ; 68(6): 835-44, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21194153

RESUMO

OBJECTIVE: The overall permeability of the blood-brain barrier (BBB) is regulated by specialized cerebral endothelial cells and their junctional complexes, consisting of adherens junctions (AJs) and tight junctions (TJs). Among the members of the glucose transporters (Glut), Glut1 is a unique molecule expressed in the cerebral endothelial cells. Glut1 and the junctional proteins are concomitantly downregulated in situations in which breakdown of the BBB has taken place. We hypothesized that the expression of Glut1 may play a significant role in the development of the cerebral microvasculature with BBB properties. To date, there is no information on the role of Glut1 during the development of BBB. In the present study, the in vivo effects of Glut1 knockdown on the cerebral vascular development were investigated. METHODS: Zebrafish was used as a model organism. We confirmed that the structure of the zebrafish homologue of Glut1 is highly similar to the human Glut1 and that the function of the Glut1-mediated cerebral uptake of glucose is evolutionally conserved. RESULTS: In the Glut1 knockdown model, we observed loss of the cerebral endothelial cells, with concomitant downregulation of the junctional proteins important for intactness of the AJs/TJs and impaired cerebral circulation. The resulting leaky BBB caused vasogenic brain edema. INTERPRETATION: The data suggest a crucial role of Glut1 in the development of the cerebral endothelial cells with BBB properties in vivo. The findings suggest that modulation of Glut1 expression and function may open new directions of research for therapeutic strategies to prevent vasogenic brain edema.


Assuntos
Barreira Hematoencefálica/fisiologia , Transportador de Glucose Tipo 1/fisiologia , Animais , Animais Geneticamente Modificados , Aquaporina 4/metabolismo , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/efeitos dos fármacos , Claudina-5 , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Ensaio de Imunoadsorção Enzimática/métodos , Fibronectinas/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patologia , Glucose/metabolismo , Transportador de Glucose Tipo 1/genética , Humanos , Imunoprecipitação/métodos , Proteínas de Membrana/metabolismo , Microscopia Confocal/métodos , Modelos Animais , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Gravação em Vídeo , Peixe-Zebra
18.
Gastroenterology ; 137(5): 1757-67.e1, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19698716

RESUMO

BACKGROUND & AIMS: The pathogenesis of inflammatory bowel disease involves dysfunctional mucosal immune responses to commensal bacteria in genetically predisposed hosts. Interactions between host cells and bacteria are complicated, making it a challenge to assess their relative contribution to intestinal pathology. We developed a zebrafish model of enterocolitis to study these interactions. METHODS: Enterocolitis was induced by intrarectal administration of the hapten oxazolone in adult wild-type and myeloperoxidase-reporter transgenic zebrafish in the presence or absence of antibiotics. Intestinal inflammation was evaluated by histological and flow cytometry analyses and cytokine profiling with quantitative real-time polymerase chain reaction. Changes in the composition of the intestinal microbiota following antibiotic administration were assessed by 16SrRNA sequencing and bacterial load was quantified by culture on nonselective media (colony-forming units). RESULTS: In zebrafish, the infiltrate and severity of oxazolone-induced enterocolitis are influenced by the composition of the microbiota. Inflammation is characterized by granulocyte influx; epithelial damage; goblet cell depletion; and increased expression of interleukin-1beta, tumor necrosis factor-alpha, and interleukin-10. Zebrafish given vancomycin had bacterial populations dominated by Fusobacteria and reduced enterocolitis scores, intestinal damage, and percentages of infiltrating neutrophils and eosinophils. In contrast, zebrafish given colistin sulphate had a predominance of proteobacteria and reduced eosinophil and lymphocyte infiltration, but enterocolitis scores were not reduced. CONCLUSIONS: In zebrafish with oxazolone-induced enterocolitis, components of the intestinal microbiota affect the severity and composition of the intestinal infiltrate.


Assuntos
Adjuvantes Imunológicos , Enterocolite/microbiologia , Enterocolite/patologia , Intestinos/microbiologia , Oxazolona , Adjuvantes Imunológicos/farmacologia , Animais , Antibacterianos/uso terapêutico , Colistina/uso terapêutico , Modelos Animais de Doenças , Enterocolite/tratamento farmacológico , Intestinos/efeitos dos fármacos , Intestinos/patologia , Oxazolona/farmacologia , Vancomicina/uso terapêutico , Peixe-Zebra
19.
Mol Cell Proteomics ; 8(6): 1192-205, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19204029

RESUMO

Novel markers for prostate cancer (PCa) are needed because current established markers such as prostate-specific antigen lack diagnostic specificity and prognostic value. Proteomics analysis of serum from mice grafted with human PCa xenografts resulted in the identification of 44 tumor-derived proteins. Besides secreted proteins we identified several cytoplasmic proteins, among which were most subunits of the proteasome. Native gel electrophoresis and sandwich ELISA showed that these subunits are present as proteasome complexes in the serum from xenograft-bearing mice. We hypothesized that the presence of proteasome subunits and other cytoplasmic proteins in serum of xenografted mice could be explained by the secretion of small vesicles by cancer cells, so-called exosomes. Therefore, mass spectrometry and Western blotting analyses of the protein content of exosomes isolated from PCa cell lines was performed. This resulted in the identification of mainly cytoplasmic proteins of which several had previously been identified in the serum of xenografted mice, including proteasome subunits. The isolated exosomes also contained RNA, including the gene fusion TMPRSS2-ERG product. These observations suggest that although their function is not clearly defined cancer-derived exosomes offer possibilities for the identification of novel biomarkers for PCa.


Assuntos
Citoplasma/metabolismo , Exossomos/metabolismo , Proteínas de Neoplasias/metabolismo , Transplante Heterólogo , Animais , Western Blotting , Cromatografia Líquida , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Humanos , Masculino , Espectrometria de Massas , Camundongos , Camundongos Nus , Espectroscopia de Infravermelho com Transformada de Fourier
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA