Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 6021, 2022 10 12.
Article in English | MEDLINE | ID: mdl-36224199

ABSTRACT

Drug-induced cytopenias are a prevalent and significant issue that worsens clinical outcomes and hinders the effective treatment of cancer. While reductions in blood cell numbers are classically associated with traditional cytotoxic chemotherapies, they also occur with newer targeted small molecules and the factors that determine the hematotoxicity profiles of oncologic drugs are not fully understood. Here, we explore why some Aurora kinase inhibitors cause preferential neutropenia. By studying drug responses of healthy human hematopoietic cells in vitro and analyzing existing gene expression datasets, we provide evidence that the enhanced vulnerability of neutrophil-lineage cells to Aurora kinase inhibition is caused by early developmental changes in ATP-binding cassette (ABC) transporter expression. These data show that hematopoietic cell-intrinsic expression of ABC transporters may be an important factor that determines how some Aurora kinase inhibitors affect the bone marrow.


Subject(s)
ATP-Binding Cassette Transporters , Neutrophils , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate , Aurora Kinases/metabolism , Hematopoiesis/genetics , Humans , Neoplasm Proteins/metabolism , Neutrophils/metabolism , Protein Kinase Inhibitors/pharmacology
2.
Biochim Biophys Acta Gen Subj ; 1865(1): 129754, 2021 01.
Article in English | MEDLINE | ID: mdl-33010352

ABSTRACT

BACKGROUND: Ubiquitously eXpressed Transcript isoform 2 (UXTV2) is a prefoldin-like protein involved in NF-κB signaling, apoptosis, and the androgen and estrogen response. UXT-V2 is a cofactor in the NF-κB transcriptional enhanceosome, and its knockdown inhibits TNF-α -induced NF-κB activation. Fbxo7 is an F-box protein that interacts with SKP1, Cullin1 and RBX1 proteins to form an SCF(Fbxo7) E3 ubiquitin ligase complex. Fbxo7 negatively regulates NF-κB signaling through TRAF2 and cIAP1 ubiquitination. METHODS: We combine co-immunoprecipitation, ubiquitination in vitro and in vivo, cycloheximide chase assay, ubiquitin chain restriction analysis and microscopy to investigate interaction between Fbxo7 and overexpressed UXT-V2-HA. RESULTS: The Ubl domain of Fbxo7 contributes to interaction with UXTV2. This substrate is polyubiquitinated by SCF(Fbxo7) with K48 and K63 ubiquitin chain linkages in vitro and in vivo. This post-translational modification decreases UXT-V2 stability and promotes its proteasomal degradation. We further show that UXTV1, an alternatively spliced isoform of UXT, containing 12 additional amino acids at the N-terminus as compared to UXTV2, also interacts with and is ubiquitinated by Fbxo7. Moreover, FBXO7 knockdown promotes UXT-V2 accumulation, and the overexpression of Fbxo7-ΔF-box protects UXT-V2 from proteasomal degradation and enhances the responsiveness of NF-κB reporter. We find that UXT-V2 colocalizes with Fbxo7 in the cell nucleus. CONCLUSIONS: Together, our study reveals that SCF(Fbxo7) mediates the proteasomal degradation of UXT-V2 causing the inhibition of the NF-κB signaling pathway. GENERAL SIGNIFICANCE: Discovering new substrates of E3 ubiquitin-ligase SCF(Fbxo7) contributes to understand its function in different diseases such as cancer and Parkinson.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Molecular Chaperones/metabolism , NF-kappa B/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , Cell Line, Tumor , HEK293 Cells , Humans , Proteasome Endopeptidase Complex/metabolism , Protein Isoforms/metabolism , Proteolysis , Ubiquitination
3.
Front Physiol ; 10: 1278, 2019.
Article in English | MEDLINE | ID: mdl-31649556

ABSTRACT

Fbxo7 is the substrate-recognition subunit of an SCF-type ubiquitin E3 ligase complex. It has physiologically important functions in regulating mitophagy, proteasome activity and the cell cycle in multiple cell types, like neurons, lymphocytes and erythrocytes. Here, we show that in addition to the previously known Parkinsonian and hematopoietic phenotypes, male mice with reduced Fbxo7 expression are sterile. In these males, despite successful meiosis, nuclear elongation and eviction of histones from chromatin, the developing spermatids are phagocytosed by Sertoli cells during late spermiogenesis, as the spermatids undergo cytoplasmic remodeling. Surprisingly, despite the loss of all germ cells, there was no evidence of the symplast formation and cell sloughing that is typically associated with spermatid death in other mouse sterility models, suggesting that novel cell death and/or cell disposal mechanisms may be engaged in Fbxo7 mutant males. Mutation of the Drosophila Fbxo7 ortholog, nutcracker (ntc) also leads to sterility with germ cell death during cytoplasmic remodeling, indicating that the requirement for Fbxo7 at this stage is conserved. The ntc phenotype was attributed to decreased levels of the proteasome regulator, DmPI31 and reduced proteasome activity. Consistent with the fly model, we observe a reduction in PI31 levels in mutant mice; however, there is no alteration in proteasome activity in whole mouse testes. Our results are consistent with findings that Fbxo7 regulates PI31 protein levels, and indicates that a defect at the late stages of spermiogenesis, possibly due to faulty spatial dynamics of proteasomes during cytoplasmic remodeling, may underlie the fertility phenotype in mice.

4.
J Pathol ; 249(2): 241-254, 2019 10.
Article in English | MEDLINE | ID: mdl-31144295

ABSTRACT

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.


Subject(s)
Dopaminergic Neurons/metabolism , F-Box Proteins/metabolism , Mesencephalon/metabolism , Nerve Degeneration , Parkinson Disease/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Ribosomal Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Behavior, Animal , Disease Models, Animal , Dopaminergic Neurons/pathology , F-Box Proteins/genetics , Female , Locomotion , Male , Mesencephalon/pathology , Mesencephalon/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Parkinson Disease/genetics , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Proto-Oncogene Proteins c-mdm2/genetics , Ribosomal Proteins/genetics , Signal Transduction , Tumor Suppressor Protein p53/genetics
5.
Brain Res ; 1693(Pt A): 11-23, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29723523

ABSTRACT

Many RNA binding proteins, including FUS, contain moderately repetitive, low complexity, intrinsically disordered domains. These sequence motifs have recently been found to underpin reversible liquid: liquid phase separation and gelation of these proteins, permitting them to reversibly transition from a monodispersed state to liquid droplet- or hydrogel-like states. This function allows the proteins to serve as scaffolds for the formation of reversible membraneless intracellular organelles such as nucleoli, stress granules and neuronal transport granules. Using FUS as an example, this review examines the biophysics of this physiological process, and reports on how mutations and changes in post-translational state alter phase behaviour, and lead to neurodegenerative diseases such as amyotrophic lateral sclerosis and frontotemporal lobar degeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/physiopathology , Amyotrophic Lateral Sclerosis/physiopathology , Biophysics/methods , Cytoplasmic Granules/metabolism , DNA-Binding Proteins/metabolism , Frontotemporal Dementia/genetics , Humans , Mutation , Neurodegenerative Diseases/pathology , Protein Domains , Protein Processing, Post-Translational , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , RNA-Binding Proteins/metabolism , Temporal Lobe/metabolism
6.
Cell ; 173(3): 720-734.e15, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677515

ABSTRACT

Reversible phase separation underpins the role of FUS in ribonucleoprotein granules and other membrane-free organelles and is, in part, driven by the intrinsically disordered low-complexity (LC) domain of FUS. Here, we report that cooperative cation-π interactions between tyrosines in the LC domain and arginines in structured C-terminal domains also contribute to phase separation. These interactions are modulated by post-translational arginine methylation, wherein arginine hypomethylation strongly promotes phase separation and gelation. Indeed, significant hypomethylation, which occurs in FUS-associated frontotemporal lobar degeneration (FTLD), induces FUS condensation into stable intermolecular ß-sheet-rich hydrogels that disrupt RNP granule function and impair new protein synthesis in neuron terminals. We show that transportin acts as a physiological molecular chaperone of FUS in neuron terminals, reducing phase separation and gelation of methylated and hypomethylated FUS and rescuing protein synthesis. These results demonstrate how FUS condensation is physiologically regulated and how perturbations in these mechanisms can lead to disease.


Subject(s)
Arginine/chemistry , Molecular Chaperones/chemistry , RNA-Binding Protein FUS/chemistry , Amyotrophic Lateral Sclerosis/metabolism , Animals , Cations , DNA Methylation , Frontotemporal Dementia/metabolism , Frontotemporal Lobar Degeneration/metabolism , Humans , Microscopy, Atomic Force , Microscopy, Fluorescence , Protein Binding , Protein Domains , Protein Processing, Post-Translational , Protein Structure, Secondary , RNA-Binding Protein FUS/metabolism , Tyrosine/chemistry , Xenopus laevis
7.
Curr Protein Pept Sci ; 18(7): 715-724, 2017.
Article in English | MEDLINE | ID: mdl-26965690

ABSTRACT

Fbxo7/PARK15 has well-defined roles, acting as part of a Skp1-Cul1-F box protein (SCF)- type E3 ubiquitin ligase and also having SCF-independent activities. Mutations within FBXO7 have been found to cause an early-onset Parkinson's disease, and these are found within or near to its functional domains, including its F-box domain (FBD), its proline rich region (PRR), and its ubiquitinlike domain (Ubl). We highlight recent advances in our understanding of Fbxo7 function in Parkinson's disease, with respect to these mutations and where they occur in the Fbxo7 protein. We hypothesize that many of Fbxo7 functions contribute to its role in PD pathogenesis.


Subject(s)
F-Box Proteins/genetics , Mutation , Parkinson Disease/genetics , SKP Cullin F-Box Protein Ligases/genetics , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , F-Box Proteins/chemistry , F-Box Proteins/metabolism , Gene Expression , Humans , Neurons/metabolism , Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Domains , SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/metabolism , Structure-Activity Relationship , Ubiquitination
8.
Cell Mol Life Sci ; 74(8): 1553-1566, 2017 04.
Article in English | MEDLINE | ID: mdl-27915416

ABSTRACT

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.


Subject(s)
Cyclin-Dependent Kinase 6/immunology , Cyclin-Dependent Kinase Inhibitor p27/immunology , F-Box Proteins/immunology , T-Lymphocytes/cytology , Animals , Apoptosis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Cycle , Cell Differentiation , Cell Proliferation , Down-Regulation , F-Box Proteins/genetics , Female , Gene Deletion , Lymphocyte Activation , Male , Mice, Inbred C57BL , Mutation , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology
9.
Biochem J ; 473(20): 3563-3580, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27503909

ABSTRACT

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.


Subject(s)
F-Box Proteins/metabolism , Membrane Transport Proteins/metabolism , Parkinson Disease/enzymology , Parkinson Disease/metabolism , Receptors, Cell Surface/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , F-Box Proteins/genetics , Fluorescent Antibody Technique , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Immunoprecipitation , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Membrane Transport Proteins/genetics , Mitochondrial Precursor Protein Import Complex Proteins , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Parkinson Disease/genetics , Point Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Cell Surface/genetics , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Ubiquitination/genetics , Ubiquitination/physiology
10.
Semin Cancer Biol ; 36: 3-17, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26416465

ABSTRACT

F-box proteins (FBP) are the substrate specifying subunit of Skp1-Cul1-FBP (SCF)-type E3 ubiquitin ligases and are responsible for directing the ubiquitination of numerous proteins essential for cellular function. Due to their ability to regulate the expression and activity of oncogenes and tumour suppressor genes, FBPs themselves play important roles in cancer development and progression. In this review, we provide a comprehensive overview of FBPs and their targets in relation to their interaction with the hallmarks of cancer cell biology, including the regulation of proliferation, epigenetics, migration and invasion, metabolism, angiogenesis, cell death and DNA damage responses. Each cancer hallmark is revealed to have multiple FBPs which converge on common signalling hubs or response pathways. We also highlight the complex regulatory interplay between SCF-type ligases and other ubiquitin ligases. We suggest six highly interconnected FBPs affecting multiple cancer hallmarks, which may prove sensible candidates for therapeutic intervention.


Subject(s)
F-Box Proteins/metabolism , Neoplasms/metabolism , Signal Transduction , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Movement/genetics , Cell Proliferation , DNA Damage , Epigenesis, Genetic , F-Box Proteins/genetics , Gene Expression Regulation, Neoplastic , Genomic Instability , Humans , Mitochondria/metabolism , Neoplasm Invasiveness , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Oxidative Stress , Protein Binding , Receptors, Cell Surface/metabolism
11.
J Pathol ; 237(2): 263-72, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26095538

ABSTRACT

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.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Erythrocytes/metabolism , Erythropoiesis , F-Box Proteins/metabolism , Anemia/blood , Anemia/genetics , Anemia/pathology , Animals , Cell Cycle Checkpoints , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/genetics , Down-Regulation , Erythrocytes/pathology , Erythropoiesis/genetics , F-Box Proteins/genetics , Genotype , Hemoglobins/metabolism , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Mitophagy , Phenotype , Protein Stability , RNA Interference , Signal Transduction , Time Factors , Transfection
12.
Open Biol ; 3(10): 130131, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-24107298

ABSTRACT

F-box proteins (FBPs) are substrate-recruiting subunits of Skp1-cullin1-FBP (SCF)-type E3 ubiquitin ligases. To date, 69 FBPs have been identified in humans, but ubiquitinated substrates have only been identified for a few, with the majority of FBPs remaining 'orphans'. In recent years, a growing body of work has identified non-canonical, SCF-independent roles for about 12% of the human FBPs. These atypical FBPs affect processes as diverse as transcription, cell cycle regulation, mitochondrial dynamics and intracellular trafficking. Here, we provide a general review of FBPs, with a particular emphasis on these expanded functions. We review Fbxo7 as an exemplar of this special group as it has well-defined roles in both SCF and non-SCF complexes. We review its function as a cell cycle regulator, via its ability to stabilize p27 protein and Cdk6 complexes, and as a proteasome regulator, owing to its high affinity binding to PI31. We also highlight recent advances in our understanding of Fbxo7 function in Parkinson's disease, where it functions in the regulation of mitophagy with PINK1 and Parkin. We postulate that a few extraordinary FBPs act as platforms that seamlessly segue their canonical and non-canonical functions to integrate different cellular pathways and link their regulation.


Subject(s)
F-Box Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Animals , Cell Cycle , Cyclin-Dependent Kinase 6/metabolism , Evolution, Molecular , F-Box Proteins/genetics , Gene Expression Regulation , Humans , Mitophagy , Mutation , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Proteasome Endopeptidase Complex/metabolism , Protein Kinases/metabolism , Proteins/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
13.
Nat Neurosci ; 16(9): 1257-65, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23933751

ABSTRACT

Compelling evidence indicates that two autosomal recessive Parkinson's disease genes, PINK1 (PARK6) and Parkin (PARK2), cooperate to mediate the autophagic clearance of damaged mitochondria (mitophagy). Mutations in the F-box domain-containing protein Fbxo7 (encoded by PARK15) also cause early-onset autosomal recessive Parkinson's disease, by an unknown mechanism. Here we show that Fbxo7 participates in mitochondrial maintenance through direct interaction with PINK1 and Parkin and acts in Parkin-mediated mitophagy. Cells with reduced Fbxo7 expression showed deficiencies in translocation of Parkin to mitochondria, ubiquitination of mitofusin 1 and mitophagy. In Drosophila, ectopic overexpression of Fbxo7 rescued loss of Parkin, supporting a functional relationship between the two proteins. Parkinson's disease-causing mutations in Fbxo7 interfered with this process, emphasizing the importance of mitochondrial dysfunction in Parkinson's disease pathogenesis.


Subject(s)
F-Box Proteins/metabolism , Mitophagy/genetics , Parkinson Disease/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , Animals, Genetically Modified , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Cell Line, Tumor , Cells, Cultured , Drosophila , F-Box Proteins/genetics , Female , Fertility/genetics , Fibroblasts/drug effects , Fibroblasts/pathology , Fibroblasts/ultrastructure , Humans , Male , Microtubule-Associated Proteins/metabolism , Mitochondria/pathology , Mitophagy/drug effects , Mutation/genetics , Parkinson Disease/pathology , Protein Transport/drug effects , Protein Transport/genetics , Proton Ionophores/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Time Factors , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects , Ubiquitination/genetics
14.
PLoS One ; 6(6): e21165, 2011.
Article in English | MEDLINE | ID: mdl-21695055

ABSTRACT

Fbxo7 is an unusual F box protein that augments D-type cyclin complex formation with Cdk6, but not Cdk4 or Cdk2, and its over-expression has been demonstrated to transform immortalised fibroblasts in a Cdk6-dependent manner. Here we present new evidence in vitro and in vivo on the oncogenic potential of this regulatory protein in primary haematopoietic stem and progenitor cells (HSPCs). Increasing Fbxo7 expression in HSPCs suppressed their colony forming ability in vitro, specifically decreasing CD11b (Mac1) expression, and these effects were dependent on an intact p53 pathway. Furthermore, increased Fbxo7 levels enhanced the proliferative capacity of p53 null HSPCs when they were grown in reduced concentrations of stem cell factor. Finally, irradiated mice reconstituted with p53 null, but not wild-type, HSPCs expressing Fbxo7 showed a statistically significant increase in the incidence of T cell lymphoma in vivo. These data argue that Fbxo7 negatively regulates the proliferation and differentiation of HSPCs in a p53-dependent manner, and that in the absence of p53, Fbxo7 expression can promote T cell lymphomagenesis.


Subject(s)
F-Box Proteins/metabolism , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Lymphoma/pathology , Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/deficiency , Animals , Cell Differentiation/genetics , Cell Division/genetics , Cell Proliferation , Gene Deletion , Gene Expression Regulation/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Humans , Mice , Stem Cell Factor/metabolism , Tumor Suppressor Protein p53/genetics
15.
J Cell Sci ; 124(Pt 13): 2175-86, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21652635

ABSTRACT

Fbxo7 is an unusual F-box protein because most of its interacting proteins are not substrates for ubiquitin-mediated degradation. Fbxo7 directly binds p27 and Cdk6, enhances the level of cyclin D-Cdk6 complexes, and its overexpression causes Cdk6-dependent transformation of immortalised fibroblasts. Here, we test the ability of Fbxo7 to transform haematopoietic pro-B (Ba/F3) cells which, unexpectedly, it was unable to do despite high levels of Cdk6. Instead, reduction of Fbxo7 expression increased proliferation, decreased cell size and shortened G1 phase. Analysis of cell cycle regulators showed that cells had decreased levels of p27, and increased levels of S phase cyclins and Cdk2 activity. Also, Fbxo7 protein levels correlated inversely with those of CD43, suggesting direct regulation of its expression and, therefore, of B cell maturation. Alterations to Cdk6 protein levels did not affect the cell cycle, indicating that Cdk6 is neither rate-limiting nor essential in Ba/F3 cells; however, decreased expression of Cdk6 also enhanced levels of CD43, indicating that expression of CD43 is independent of cell cycle regulation. The physiological effect of reduced levels of Fbxo7 was assessed by creating a transgenic mouse with a LacZ insertion into the Fbxo7 locus. Homozygous Fbxo7(LacZ) mice showed significantly increased pro-B cell and pro-erythroblast populations, consistent with Fbxo7 having an anti-proliferative function and/or a role in promoting maturation of precursor cells.


Subject(s)
Cell Differentiation , Cell Proliferation , F-Box Proteins/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , B-Lymphocytes/metabolism , Cell Cycle/genetics , Cell Cycle/physiology , Cell Differentiation/genetics , Cell Line , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclins/biosynthesis , G1 Phase/genetics , G1 Phase/physiology , Humans , Leukosialin/biosynthesis , Mice , Mice, Transgenic , Signal Transduction/genetics
16.
Am J Pathol ; 175(4): 1598-609, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19717642

ABSTRACT

Here, we describe the generation and characterization of a novel tau transgenic mouse model (mTau) that overexpresses wild-type murine tau protein by twofold compared with endogenous levels. Transgenic tau expression was driven by a BAC transgene containing the entire wild-type mouse tau locus, including the endogenous promoter and the regulatory elements associated with the tau gene. The mTau model therefore differs from other tau models in that regulation of the genomic mouse transgene mimics that of the endogenous gene, including normal exon splicing regulation. Biochemical data from the mTau mice demonstrated that modest elevation of mouse tau leads to tau hyperphosphorylation at multiple pathologically relevant epitopes and accumulation of sarkosyl-insoluble tau. The mTau mice show a progressive increase in hyperphosphorylated tau pathology with age up to 15 to 18 months, which is accompanied by gliosis and vacuolization. In contrast, older mice show a decrease in tau pathology levels, which may represent hippocampal neuronal loss occurring in this wild-type model. Collectively, these results describe a novel model of tauopathy that develops pathological changes reminiscent of early stage Alzheimer's disease and other related neurodegenerative diseases, achieved without overexpression of a mutant human tau transgene. This model will provide an important tool for understanding the early events leading to the development of tau pathology and a model for analysis of potential therapeutic targets for sporadic tauopathies.


Subject(s)
Nerve Degeneration/complications , Nerve Degeneration/pathology , Tauopathies/complications , Tauopathies/pathology , tau Proteins/metabolism , Aging/pathology , Animals , Axons/pathology , Axons/ultrastructure , Breeding , Chromosomes, Artificial, Bacterial/genetics , Gene Expression Regulation , Genetic Vectors/genetics , Genome/genetics , Mice , Mice, Transgenic , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Phosphorylation , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...