Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Mol Cell ; 82(6): 1123-1139.e8, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35182481

RESUMEN

A mesenchymal tumor phenotype associates with immunotherapy resistance, although the mechanism is unclear. Here, we identified FBXO7 as a maintenance regulator of mesenchymal and immune evasion phenotypes of cancer cells. FBXO7 bound and stabilized SIX1 co-transcriptional regulator EYA2, stimulating mesenchymal gene expression and suppressing IFNα/ß, chemokines CXCL9/10, and antigen presentation machinery, driven by AXL extracellular ligand GAS6. Ubiquitin ligase SCFFBXW7 antagonized this pathway by promoting EYA2 degradation. Targeting EYA2 Tyr phosphatase activity decreased mesenchymal phenotypes and enhanced cancer cell immunogenicity, resulting in attenuated tumor growth and metastasis, increased infiltration of cytotoxic T and NK cells, and enhanced anti-PD-1 therapy response in mouse tumor models. FBXO7 expression correlated with mesenchymal and immune-suppressive signatures in patients with cancer. An FBXO7-immune gene signature predicted immunotherapy responses. Collectively, the FBXO7/EYA2-SCFFBXW7 axis maintains mesenchymal and immune evasion phenotypes of cancer cells, providing rationale to evaluate FBXO7/EYA2 inhibitors in combination with immune-based therapies to enhance onco-immunotherapy responses.


Asunto(s)
Proteínas F-Box , Proteína 7 que Contiene Repeticiones F-Box-WD , Neoplasias , Animales , Línea Celular Tumoral , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Proteínas de Homeodominio/genética , Humanos , Evasión Inmune , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Neoplasias/genética , Proteínas Nucleares/metabolismo , Fenotipo , Proteínas Tirosina Fosfatasas/genética , Ubiquitina/metabolismo
2.
EMBO Rep ; 24(8): e56399, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37334901

RESUMEN

The protein kinase PINK1 and ubiquitin ligase Parkin promote removal of damaged mitochondria via a feed-forward mechanism involving ubiquitin (Ub) phosphorylation (pUb), Parkin activation, and ubiquitylation of mitochondrial outer membrane proteins to support the recruitment of mitophagy receptors. The ubiquitin ligase substrate receptor FBXO7/PARK15 is mutated in an early-onset parkinsonian-pyramidal syndrome. Previous studies have proposed a role for FBXO7 in promoting Parkin-dependent mitophagy. Here, we systematically examine the involvement of FBXO7 in depolarization and mt UPR-dependent mitophagy in the well-established HeLa and induced-neurons cell systems. We find that FBXO7-/- cells have no demonstrable defect in: (i) kinetics of pUb accumulation, (ii) pUb puncta on mitochondria by super-resolution imaging, (iii) recruitment of Parkin and autophagy machinery to damaged mitochondria, (iv) mitophagic flux, and (v) mitochondrial clearance as quantified by global proteomics. Moreover, global proteomics of neurogenesis in the absence of FBXO7 reveals no obvious alterations in mitochondria or other organelles. These results argue against a general role for FBXO7 in Parkin-dependent mitophagy and point to the need for additional studies to define how FBXO7 mutations promote parkinsonian-pyramidal syndrome.


Asunto(s)
Proteínas F-Box , Mitofagia , Humanos , Células HeLa , Mitofagia/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Ubiquitina/metabolismo , Proteínas F-Box/genética , Proteínas F-Box/metabolismo
3.
J Biol Chem ; 299(3): 102909, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36646384

RESUMEN

Parkinson's disease (PD) is a degenerative disorder of the central nervous system that affects 1% of the population over the age of 60. Although aging is one of the main risk factors for PD, the pathogenic mechanism of this disease remains unclear. Mutations in the F-box-only protein 7 (FBXO7) gene have been previously found to cause early onset autosomal recessive familial PD. FBXO7 is an adaptor protein in the SKP1-Cullin-1-F-box (SCF) E3 ligase complex that facilitates the ubiquitination of substrates. Sirtuin 7 (SIRT7) is an NAD+-dependent histone deacetylase that regulates aging and stress responses. In this study, we identified FBXO7 as a novel E3 ligase for SIRT7 that negatively regulates intracellular SIRT7 levels through SCF-dependent Lys-48-linked polyubiquitination and proteasomal degradation. Consequently, we show that FBXO7 promoted the blockade of SIRT7 deacetylase activity, causing an increase in acetylated histone 3 levels at the Lys-18 and Lys-36 residues and the repression of downstream RPS20 gene transcription. Moreover, we demonstrate that treatment with hydrogen peroxide triggered the FBXO7-mediated degradation of SIRT7, leading to mammalian cell death. In particular, the PD-linked FBXO7-R498X mutant, which reduced SCF-dependent E3 ligase activity, did not affect the stability of SIRT7. Collectively, these findings suggest that FBXO7 negatively regulates SIRT7 stability and may suppress the cytoprotective effects of SIRT7 during hydrogen peroxide-induced mammalian cell death.


Asunto(s)
Proteínas F-Box , Enfermedad de Parkinson , Sirtuinas , Animales , Humanos , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Peróxido de Hidrógeno/metabolismo , Proteínas F-Box/metabolismo , Ubiquitinación , Enfermedad de Parkinson/metabolismo , Muerte Celular , Mamíferos/metabolismo , Sirtuinas/genética , Sirtuinas/metabolismo
4.
J Neurochem ; 167(2): 296-317, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37753846

RESUMEN

Mutations in PARK15, which encodes for the F-box protein FBXO7 have been associated with Parkinsonian Pyramidal syndrome, a rare and complex movement disorder with Parkinsonian symptoms, pyramidal tract signs and juvenile onset. Our previous study showed that systemic loss of Fbxo7 in mice causes motor defects and premature death. We have also demonstrated that FBXO7 has a crucial role in neurons as the specific deletion in tyrosine hydroxylase-positive or glutamatergic forebrain neurons leads to late-onset or early-onset motor dysfunction, respectively. In this study, we examined NEX-Cre;Fbxo7fl/fl mice, in which Fbxo7 was specifically deleted in glutamatergic projection neurons. The effects of FBXO7 deficiency on striatal integrity were investigated with HPLC and histological analyses. NEX-Cre;Fbxo7fl/fl mice revealed an increase in striatal dopamine concentrations, changes in the glutamatergic, GABAergic and dopaminergic pathways, astrogliosis and microgliosis and little or no neuronal loss in the striatum. To determine the effects on the integrity of the synapse, we purified synaptic membranes, subjected them to quantitative mass spectrometry analysis and found alterations in the complement system, endocytosis and exocytosis pathways. These neuropathological changes coincide with alterations in spontaneous home cage behavior. Taken together, our findings suggest that FBXO7 is crucial for corticostriatal projections and the synaptic integrity of the striatum, and consequently for proper motor control.

5.
J Biol Chem ; 297(6): 101426, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34800438

RESUMEN

Parkinson's disease (PD) is characterized by the progressive loss of midbrain dopamine neurons in the substantia nigra. Mutations in the F-box only protein 7 gene (Fbxo7) have been reported to cause an autosomal recessive form of early-onset familial PD. FBXO7 is a part of the SKP1-Cullin1-F-box (SCF) E3 ubiquitin ligase complex, which mediates ubiquitination of numerous substrates. FBXO7 also regulates mitophagy, cell growth, and proteasome activity. A member of the FOXO family, the transcription factor FOXO4, is also known to modulate several cellular responses, including cell cycle progression and apoptosis; however, the relationship between FBXO7 and FOXO4 has not been investigated. In this study, we determined that FBXO7 binds to FOXO4 and negatively regulates intracellular FOXO4 levels. Interestingly, we also found that FBXO7-mediated degradation of FOXO4 did not occur through either of two major proteolysis systems, the ubiquitin-proteasome system or the lysosome-autophagy pathway, although it was blocked by a caspase 8-specific inhibitor and caspase 8-knockdown. Moreover, intracellular FOXO4 levels were greatly reduced in dopaminergic MN9D cells following treatment with neurotoxic 6-hydroxydopamine (6-OHDA), which was produced upon FBXO7-mediated and caspase 8-mediated proteolysis. Taken together, these results suggest that FOXO4 is negatively regulated in FBXO7-linked PD through caspase 8 activation, suppressing the cytoprotective effect of FOXO4 during 6-OHDA-induced neuronal cell death.


Asunto(s)
Caspasa 8/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Factores de Transcripción Forkhead/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Proteolisis , Animales , Caspasa 8/genética , Proteínas de Ciclo Celular/genética , Proteínas F-Box/genética , Factores de Transcripción Forkhead/genética , Células HEK293 , Humanos , Células MCF-7 , Masculino , Ratones , Enfermedad de Parkinson/genética
6.
Biochem Biophys Res Commun ; 558: 224-230, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32933748

RESUMEN

The NF-κB transcription factor is involved in inflammation and cell proliferation, survival, and transformation. It is a heterodimer made of p50 or p52 and a member of the Rel family of proteins. p50 and p52 are derived from limited ubiquitin- and proteasome-mediated proteolytic processing of the larger precursors p105 and p100, respectively. Both precursors can be either processed or completely degraded by the ubiquitin-proteasome system. Previous work in our laboratory identified KPC1 as a ubiquitin ligase that mediates processing of p105 to the p50 subunit. Overexpression of the ligase leads to increased level of p50 with a resultant marked tumor-suppressive effect. In the present study, we identify FBXO7, a known ubiquitin ligase that binds to p105 and ubiquitinates it, but surprisingly, leads to its accumulation and to that of p65 - the Rel partner of p50 - and to increased cell proliferation. Importantly, a ΔF-Box mutant of FBXO7 which is inactive has similar effects on accumulation of p105 and cell proliferation, strongly suggesting that p105 is a pseudo substrate of FBXO7.


Asunto(s)
Proteínas F-Box/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Secuencia de Aminoácidos , Línea Celular , Proliferación Celular/fisiología , Estabilidad de Enzimas , Proteínas F-Box/antagonistas & inhibidores , Proteínas F-Box/genética , Células HEK293 , Células HeLa , Humanos , Células K562 , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Subunidad p50 de NF-kappa B/antagonistas & inhibidores , Subunidad p50 de NF-kappa B/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Proteolisis , ARN Interferente Pequeño/genética , Especificidad por Sustrato , Factor de Transcripción ReIA/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinación
7.
Biochem Biophys Res Commun ; 554: 41-48, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33774278

RESUMEN

Fbxo7 is a key player in the differentiation and function of numerous blood cell types, and in neurons, oligodendrocytes and spermatocytes. In an effort to gain insight into the physiological and pathological settings where Fbxo7 is likely to play a key role, we sought to define the transcription factors which direct FBXO7 expression. Using sequence alignments across 28 species, we defined the human FBXO7 promoter and found that it contains two conserved regions enriched for multiple transcription factor binding sites. Many of these have roles in either neuronal or haematopoietic development. Using various FBXO7 promoter reporters, we found ELF4, Pax5 and c-Myb have functional binding sites that activate transcription. We find endogenous Pax5 is bound to the FBXO7 promoter in pre-B cells, and that the exogenous expression of Pax5 represses Fbxo7 transcription in early pro-B cells.


Asunto(s)
Linfocitos B/metabolismo , Proteínas F-Box/genética , Factor de Transcripción PAX5/metabolismo , Proteínas Proto-Oncogénicas c-myb/metabolismo , Linfocitos B/citología , Secuencia de Bases , Sitios de Unión , Diferenciación Celular/fisiología , Células Cultivadas , Secuencia Conservada , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas F-Box/metabolismo , Humanos , Factor de Transcripción PAX5/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myb/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
J Neurosci ; 39(28): 5606-5626, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31085610

RESUMEN

Myelination of axons facilitates the rapid propagation of electrical signals and the long-term integrity of axons. The ubiquitin-proteasome system is essential for proper protein homeostasis, which is particularly crucial for interactions of postmitotic cells. In our study, we examined how the E3 ubiquitin ligase FBXO7-SCF (SKP1, Cul1, F-box protein) expressed in myelinating cells affects the axon-myelin unit. Deletion of Fbxo7 in oligodendrocytes and Schwann cells in mice using the Cnp1-Cre driver line led to motor impairment due to hindlimb paresis. It did not result in apoptosis of myelinating cells, nor did it affect the proper myelination of axons or lead to demyelination. It however triggered axonal degeneration in the CNS and resulted in the severe degeneration of axons in the PNS, inducing a full-blown neuropathy. Both the CNS and PNS displayed inflammation, while the PNS was also characterized by fibrosis, massive infiltration of macrophages, and edema. Tamoxifen-induced deletion of Fbxo7, after myelination using the Plp1-CreERT2 line, led to a small number of degenerated axons and hence a very mild peripheral neuropathy. Interestingly, loss of Fbxo7 also resulted in reduced proteasome activity in Schwann cells but not in cerebellar granule neurons, indicating a specific sensitivity of the former cell type. Together, our results demonstrate an essential role for FBXO7 in myelinating cells to support associated axons, which is fundamental to the proper developmental establishment and the long-term integrity of the axon-myelin unit.SIGNIFICANCE STATEMENT The myelination of axons facilitates the fast propagation of electrical signals and the trophic support of the myelin-axon unit. Here, we report that deletion of Fbxo7 in myelinating cells in mice triggered motor impairment but had no effect on myelin biogenesis. Loss of Fbxo7 in myelinating glia, however, led to axonal degeneration in the CNS and peripheral neuropathy of the axonal type. In addition, we found that Schwann cells were particularly sensitive to Fbxo7 deficiency reflected by reduced proteasome activity. Based on these findings, we conclude that Fbxo7 is essential for the support of the axon-myelin unit and long-term axonal health.


Asunto(s)
Axones/metabolismo , Proteínas F-Box/genética , Vaina de Mielina/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo , Animales , Apoptosis , Axones/patología , Células Cultivadas , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Proteínas F-Box/metabolismo , Femenino , Eliminación de Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/patología , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/patología , Complejo de la Endopetidasa Proteasomal/metabolismo
9.
EMBO J ; 35(18): 2008-25, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27497298

RESUMEN

Mutations in the FBXO7 (PARK15) gene have been implicated in a juvenile form of parkinsonism termed parkinsonian pyramidal syndrome (PPS), characterized by Parkinsonian symptoms and pyramidal tract signs. FBXO7 (F-box protein only 7) is a subunit of the SCF (SKP1/cullin-1/F-box protein) E3 ubiquitin ligase complex, but its relevance and function in neurons remain to be elucidated. Here, we report that the E3 ligase FBXO7-SCF binds to and ubiquitinates the proteasomal subunit PSMA2. In addition, we show that FBXO7 is a proteasome-associated protein involved in proteasome assembly. In FBXO7 knockout mice, we find reduced proteasome activity and early-onset motor deficits together with premature death. In addition, we demonstrate that NEX (neuronal helix-loop-helix protein-1)-Cre-induced deletion of the FBXO7 gene in forebrain neurons or the loss of FBXO7 in tyrosine hydroxylase (TH)-positive neurons results in motor defects, reminiscent of the phenotype in PARK15 patients. Taken together, our study establishes a vital role for FBXO7 in neurons, which is required for proper motor control and accentuates the importance of FBXO7 in proteasome function.


Asunto(s)
Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Técnicas de Inactivación de Genes , Trastornos Parkinsonianos/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Ratones Noqueados , Procesamiento Proteico-Postraduccional , Ubiquitinación
10.
J Pathol ; 249(2): 241-254, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31144295

RESUMEN

The field of Parkinson's disease research has been impeded by the absence of animal models that clearly phenocopy the features of this neurodegenerative condition. Mutations in FBXO7/PARK15 are associated with both sporadic Parkinson's disease and a severe form of autosomal recessive early-onset Parkinsonism. Here we report that conditional deletion of Fbxo7 in the midbrain dopamine neurons results in an early reduction in striatal dopamine levels, together with a slow, progressive loss of midbrain dopamine neurons and onset of locomotor defects. Unexpectedly, a later compensatory response led to a near-full restoration of dopaminergic fibre innervation in the striatum, but nigral cell loss was irreversible. Mechanistically, there was increased expression in the dopamine neurons of FBXO7-interacting protein, RPL23, which is a sensor of ribosomal stress that inhibits MDM2, the negative regulator of p53. A corresponding activated p53 transcriptional signature biased towards pro-apoptotic genes was also observed. These data suggest that the neuroprotective role of FBXO7 involves its suppression of the RPL23-MDM2-p53 axis that promotes cell death in dopaminergic midbrain neurons. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Proteínas F-Box/metabolismo , Mesencéfalo/metabolismo , Degeneración Nerviosa , Enfermedad de Parkinson/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Ribosómicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Conducta Animal , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/patología , Proteínas F-Box/genética , Femenino , Locomoción , Masculino , Mesencéfalo/patología , Mesencéfalo/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Ribosómicas/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/genética
11.
J Clin Lab Anal ; 34(8): e23324, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32274857

RESUMEN

BACKGROUND: Mutations in the FBXO7 gene can cause a rare chromosomal recessive neurodegenerative disease, Parkinsonian-pyramidal syndrome (PPS). Patients with this syndrome mainly show early-onset Parkinson's syndrome. Here, we present a Chinese family with infantile-onset PPS caused by FBXO7 mutations. METHODS: The clinical phenotypes and medical records of the proband and his family members were collected. The proband, his sibling, and his parents underwent whole-exome sequencing (WES) by next-generation sequencing. RESULTS: The proband and his sibling had a typical PPS phenotype with onset during infancy. WES identified compound heterozygous variants in the FBXO7 gene, including a nonsense mutation, p. Trp134*, and a splicing mutation, IVS5-1G > A, which were shared by both siblings and inherited from each of the parents. These variants have not been reported in literatures or databases. According to the American College of Medical Genetics and Genomics guidelines, the p. Trp134* and IVS5-1G > A mutations were classified as pathogenic variants. CONCLUSIONS: We report a case of siblings in a Chinese family with infantile-onset PPS caused by FBXO7 gene mutations determined by WES. These findings will contribute to the in-depth study of the pathogenesis of PPS among patients with FBXO7 gene mutations.


Asunto(s)
Blefaroespasmo , Proteínas F-Box/genética , Mutación/genética , Enfermedad de Parkinson Secundaria , Adulto , Pueblo Asiatico/genética , Blefaroespasmo/genética , Blefaroespasmo/patología , Encéfalo/patología , Niño , Preescolar , China , Análisis Mutacional de ADN , Femenino , Globo Pálido/patología , Humanos , Masculino , Enfermedad de Parkinson Secundaria/genética , Enfermedad de Parkinson Secundaria/patología , Linaje , Hermanos , Secuenciación del Exoma
12.
J Neurochem ; 144(2): 118-127, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29134665

RESUMEN

Parkinson disease (PD) is, without doubt, a burden on modern society as the prevalence increases significantly with age. Owing to this growing number of PD cases, it is more critical than ever to understand the pathogenic mechanisms underlying PD to identify therapeutic targets. The discovery of genetic mutations associated with PD and parkinsonism paves the way toward this goal. Even though, familial forms of the disease represent the minority of PD cases and some forms are so rare that there are only a few affected families, the research on the associated genes is invaluable. Recent additions to PARK mutations are those in PARK15 that encodes the F-box protein O-type 7 (FBXO7). In this review, we highlight the recent research on FBXO7, which advances our knowledge of the etiopathological pathways and fills unexpected gaps therein, justifying the dedicated study of rare variants of PD.


Asunto(s)
Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Animales , Blefaroespasmo/genética , Globo Pálido , Humanos , Ratones , Enfermedad de Parkinson Secundaria/genética
13.
Cell Mol Life Sci ; 74(8): 1553-1566, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27915416

RESUMEN

G1 phase cell cycle proteins, such as cyclin-dependent kinase 6 (Cdk6) and its activating partners, the D-type cyclins, are important regulators of T-cell development and function. An F-box protein, called F-box only protein 7 (Fbxo7), acts as a cell cycle regulator by enhancing cyclin D-Cdk6 complex formation and stabilising levels of p27, a cyclin-dependent kinase inhibitor. We generated a murine model of reduced Fbxo7 expression to test its physiological role in multiple tissues and found that these mice displayed a pronounced thymic hypoplasia. Further analysis revealed that Fbxo7 differentially affected proliferation and apoptosis of thymocytes at various stages of differentiation in the thymus and also mature T-cell function and proliferation in the periphery. Paradoxically, Fbxo7-deficient immature thymocytes failed to undergo expansion in the thymus due to a lack of Cdk6 activity, while mature T cells showed enhanced proliferative capacity upon T-cell receptor engagement due to reduced p27 levels. Our studies reveal differential cell cycle regulation by Fbxo7 at different stages in T-cell development.


Asunto(s)
Quinasa 6 Dependiente de la Ciclina/inmunología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/inmunología , Proteínas F-Box/inmunología , Linfocitos T/citología , Animales , Apoptosis , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Regulación hacia Abajo , Proteínas F-Box/genética , Femenino , Eliminación de Gen , Activación de Linfocitos , Masculino , Ratones Endogámicos C57BL , Mutación , Linfocitos T/inmunología , Timo/citología , Timo/inmunología
14.
Genome ; 60(1): 46-54, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27936908

RESUMEN

Parkinsonian-pyramidal syndrome (PPS) is an early onset form of Parkinson's disease (PD) that shows degeneration of the extrapyramidal region of the brain to result in a severe form of PD. The toxic protein build-up has been implicated in the onset of PPS. Protein removal is mediated by an intracellular proteasome complex: an E3 ubiquitin ligase, the targeting component, is essential for function. FBXO7 encodes the F-box component of the SCF E3 ubiquitin ligase linked to familial forms of PPS. The Drosophila melanogaster homologue nutcracker (ntc) and a binding partner, PI31, have been shown to be active in proteasome function. We show that altered expression of either ntc or PI31 in dopaminergic neurons leads to a decrease in longevity and locomotor ability, phenotypes both associated with models of PD. Furthermore, expression of ntc-RNAi in an established α-synuclein-dependent model of PD rescues the phenotypes of diminished longevity and locomotor control.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas F-Box/metabolismo , Enfermedad de Parkinson/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Portadoras/química , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Ojo/embriología , Ojo/metabolismo , Proteínas F-Box/química , Proteínas F-Box/genética , Femenino , Expresión Génica , Longevidad/genética , Masculino , Especificidad de Órganos/genética , Organogénesis/genética , Enfermedad de Parkinson/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
15.
Biochem J ; 473(20): 3563-3580, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27503909

RESUMEN

Fbxo7 is a clinically relevant F-box protein, associated with both cancer and Parkinson's disease (PD). Additionally, SNPs within FBXO7 are correlated with alterations in red blood cell parameters. Point mutations within FBXO7 map within specific functional domains, including near its F-box domain and its substrate recruiting domains, suggesting that deficiencies in SCFFbxo7/PARK15 ubiquitin ligase activity are mechanistically linked to early-onset PD. To date, relatively few substrates of the ligase have been identified. These include HURP (hepatoma up-regulated protein), whose ubiquitination results in proteasome-mediated degradation, and c-IAP1 (inhibitor of apoptosis protein 1), TNF receptor-associated factor 2 (TRAF2), and NRAGE, which are not destabilized as a result of ubiquitination. None of these substrates have been linked directly to PD, nor has it been determined whether they would directly engage neuronal cell death pathways. To discover ubiquitinated substrates of SCFFbxo7 implicated more directly in PD aetiology, we conducted a high-throughput screen using protein arrays to identify new candidates. A total of 338 new targets were identified and from these we validated glycogen synthase kinase 3ß (Gsk3ß), which can phosphorylate α-synuclein, and translocase of outer mitochondrial membrane 20 (Tomm20), a mitochondrial translocase that, when ubiquitinated, promotes mitophagy, as SCFFbxo7 substrates both in vitro and in vivo Ubiquitin chain restriction analyses revealed that Fbxo7 modified Gsk3ß using K63 linkages. Our results indicate that Fbxo7 negatively regulates Gsk3ß activity, rather than its levels or localization. In addition, Fbxo7 ubiquitinated Tomm20, and its levels correlated with Fbxo7 expression, indicating a stabilizing effect. None of the PD-associated mutations in Fbxo7 impaired Tomm20 ubiquitination. Our findings demonstrate that SCFFbxo7 has an impact directly on two proteins implicated in pathological processes leading to PD.


Asunto(s)
Proteínas F-Box/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/metabolismo , Receptores de Superficie Celular/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas F-Box/genética , Técnica del Anticuerpo Fluorescente , Glucógeno Sintasa Quinasa 3 beta , Células HEK293 , Humanos , Inmunoprecipitación , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Enfermedad de Parkinson/genética , Mutación Puntual/genética , Polimorfismo de Nucleótido Simple/genética , Receptores de Superficie Celular/genética , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Factor 2 Asociado a Receptor de TNF/genética , Factor 2 Asociado a Receptor de TNF/metabolismo , Ubiquitinación/genética , Ubiquitinación/fisiología
16.
J Pathol ; 237(2): 263-72, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26095538

RESUMEN

During the final stages of erythropoiesis, lineage-restricted progenitors mature over three to five cell divisions, culminating with withdrawal from the cell cycle and the loss of most organelles, including mitochondria and nuclei. Recent genome-wide association studies in human populations have associated several SNPs near or within FBXO7 with erythrocyte phenotypes. Fbxo7 encodes a multi-functional F-box protein known to bind p27 and participate in selective mitophagy. One SNP causes an amino acid substitution (Met115Ile) and is associated with smaller erythrocytes. We find that the less common IIe115 allele of Fbxo7 binds less efficiently to p27, and cells expressing this allele proliferate faster than cells expressing Met115. We show that an erythroleukaemic cell line with reduced Fbxo7 expression fails to stabilize p27 levels, exit the cell cycle, and produce haemoglobin. In addition, mice deficient in Fbxo7 expression are anaemic due to a reduction in erythrocyte numbers, and this is associated with lower p27 levels, increased numbers of late-stage erythroblasts with greater than 2N DNA content, and delayed mitophagy during terminal differentiation. Collectively, these data support an important physiological, cell cycle regulatory role for Fbxo7 during erythropoiesis.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Eritrocitos/metabolismo , Eritropoyesis , Proteínas F-Box/metabolismo , Anemia/sangre , Anemia/genética , Anemia/patología , Animales , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Regulación hacia Abajo , Eritrocitos/patología , Eritropoyesis/genética , Proteínas F-Box/genética , Genotipo , Hemoglobinas/metabolismo , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Mitofagia , Fenotipo , Estabilidad Proteica , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección
17.
Mov Disord ; 30(8): 1130-3, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26010069

RESUMEN

BACKGROUND: Recessive mutations in the F-box protein 7 gene (FBXO7; PARK15) have been identified as a cause of the parkinsonian-pyramidal syndrome. Here, we report clinical and genetic findings in a Turkish family with novel FBXO7 mutations. METHODS: Whole exome and targeted Sanger sequencing were performed for genetic analysis in a family with two members affected by Parkinson's disease (PD). All family members underwent detailed clinical, mental, and neurological examination. RESULTS: The new p.L34R (c.101 T>G) FBXO7 mutation was detected in a homozygous state in two Turkish sibs with typical levodopa-responsive PD. CONCLUSION: This is the first time a FBXO7 mutation has been identified that causes a phenotype compatible with typical idiopathic PD and presents with some of its common nonmotor features, such as rapid eye movement sleep behavior disorder, depression, and anxiety.


Asunto(s)
Proteínas F-Box/genética , Enfermedad de Parkinson/genética , Anciano de 80 o más Años , Consanguinidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Enfermedad de Parkinson/fisiopatología , Linaje , Turquía
18.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 1): 155-64, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24419388

RESUMEN

The FP (Fbxo7/PI31) domains found in the F-box protein Fbxo7 and the proteasome inhibitor PI31 mediate the homodimerization and heterodimerization of Fbxo7 and PI31. Fbxo7 is the substrate-recognition subunit of the SCF(Fbxo7) (Skp1-Cul1-F-box protein) E3 ubiquitin ligase that catalyzes the ubiquitination of hepatoma up-regulated protein (HURP) and inhibitor of apoptosis protein (IAP). Fbxo7 also interacts with proteins that are not substrates of the ubiquitin proteasome system, such as Cdk6 and PI31. Here, the crystal structure of the Fbxo7 FP domain is reported at 2.0 Šresolution. The Fbxo7 FP domain adopts an α/ß-fold similar to that of the PI31 FP domain. However, an α-helix and three ß-strands in the Fbxo7 FP domain are longer than their counterparts in the PI31 FP domain. The differences in these secondary-structural elements are spatially clustered to define a more structured and extended C-terminal end of the Fbxo7 FP domain. The two FP domains also differ substantially in the length and conformation of the longest connecting loop. More importantly, structural differences between the two FP domains lead to drastically different modes of inter-domain protein-protein interaction. The inter-domain interface of the Fbxo7 FP domain is defined by the α-helical surface in one protomer and the ß-sheet surface in the other protomer, whereas for the PI31 domain it is defined by either the α-helical surfaces or the ß-sheet surfaces in both protomers. The inter-domain interaction of the Fbxo7 FP domain is much more extensive, featuring a larger contact surface area, better shape complementarity and more hydrophobic and hydrogen-bonding interactions. The Fbxo7 FP domain also has the potential to bind two protein partners simultaneously using the α-helical and ß-sheet surfaces. The results of this structural study provide critical insights into how Fbxo7 may dimerize (or multimerize) and interact with other regulatory proteins via the FP domain.


Asunto(s)
Proteínas F-Box/química , Proteínas F-Box/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Dominios y Motivos de Interacción de Proteínas , Mapas de Interacción de Proteínas , Estructura Terciaria de Proteína
19.
FEBS J ; 291(12): 2565-2589, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38466799

RESUMEN

Mutations in FBXO7 have been discovered to be associated with an atypical parkinsonism. We report here a new homozygous missense mutation in a paediatric patient that causes an L250P substitution in the dimerisation domain of Fbxo7. This alteration selectively ablates the Fbxo7-PI31 interaction and causes a significant reduction in Fbxo7 and PI31 levels in patient cells. Consistent with their association with proteasomes, patient fibroblasts have reduced proteasome activity and proteasome subunits. We also show PI31 interacts with the MiD49/51 fission adaptor proteins, and unexpectedly, PI31 acts to facilitate SCFFbxo7-mediated ubiquitination of MiD49. The L250P mutation reduces the SCFFbxo7 ligase-mediated ubiquitination of a subset of its known substrates. Although MiD49/51 expression was reduced in patient cells, there was no effect on the mitochondrial network. However, patient cells show reduced levels of mitochondrial function and mitophagy, higher levels of ROS and are less viable under stress. Our study demonstrates that Fbxo7 and PI31 regulate proteasomes and mitochondria and reveals a new function for PI31 in enhancing the SCFFbxo7 E3 ubiquitin ligase activity.


Asunto(s)
Proteínas F-Box , Mitocondrias , Complejo de la Endopetidasa Proteasomal , Ubiquitinación , Humanos , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Mitocondrias/metabolismo , Mitocondrias/genética , Mutación Missense , Mitofagia/genética , Fibroblastos/metabolismo , Masculino , Células HEK293 , Femenino
20.
FEBS J ; 291(12): 2562-2564, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38708447

RESUMEN

Around 10% of Parkinson's disease (PD) cases are associated with mutations in various genes, including FBXO7, which encodes the substrate-recognition component for the Skp1-Cullin-F-box (SCF) class of ubiquitin E3 ligases that target proteins for proteasomal degradation. In their recent study, Al Rawi et al. characterized a new mutation in FBXO7, L250P, in a pediatric patient. Their findings reveal that the L250P mutation abolishes Fbxo7 interaction with the proteasome regulator, proteasome inhibitor 31kD (PI31), affecting proteasomal activity and the ubiquitination of some of the ligase's targets. Furthermore, the authors show that this previously undescribed mutation impairs mitochondrial function and mitophagy, emphasizing the importance of mitochondrial and proteasomal dysfunction in PD pathogenesis.


Asunto(s)
Proteínas F-Box , Mitocondrias , Enfermedad de Parkinson , Complejo de la Endopetidasa Proteasomal , Humanos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Mutación , Mitofagia/genética , Ubiquitinación
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda