Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Viruses ; 13(7)2021 07 15.
Article in English | MEDLINE | ID: mdl-34372579

ABSTRACT

Numerous viruses have evolved sophisticated countermeasures to hijack the early programmed cell death of host cells in response to infection, including the use of proteins homologous in sequence or structure to Bcl-2. Orf virus, a member of the parapoxviridae, encodes for the Bcl-2 homolog ORFV125, a potent inhibitor of Bcl-2-mediated apoptosis in the host. ORFV125 acts by directly engaging host proapoptotic Bcl-2 proteins including Bak and Bax as well as the BH3-only proteins Hrk and Puma. Here, we determined the crystal structures of ORFV125 bound to the BH3 motif of proapoptotic proteins Puma and Hrk. The structures reveal that ORFV125 engages proapoptotic BH3 motif peptides using the canonical ligand binding groove. An Arg located in the structurally equivalent BH1 region of ORFV125 forms an ionic interaction with the conserved Asp in the BH3 motif in a manner that mimics the canonical ionic interaction seen in host Bcl-2:BH3 motif complexes. These findings provide a structural basis for Orf virus-mediated inhibition of host cell apoptosis and reveal the flexibility of virus encoded Bcl-2 proteins to mimic key interactions from endogenous host signalling pathways.


Subject(s)
Orf virus/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/ultrastructure , Apoptosis/genetics , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/ultrastructure , Crystallography, X-Ray/methods , Humans , Orf virus/metabolism , Parapoxvirus/genetics , Parapoxvirus/metabolism , Protein Binding/genetics , Protein Conformation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/ultrastructure , Proto-Oncogene Proteins c-bcl-2/metabolism , Viral Proteins/metabolism
2.
Cell ; 179(6): 1319-1329.e8, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31704029

ABSTRACT

mTORC1 controls anabolic and catabolic processes in response to nutrients through the Rag GTPase heterodimer, which is regulated by multiple upstream protein complexes. One such regulator, FLCN-FNIP2, is a GTPase activating protein (GAP) for RagC/D, but despite its important role, how it activates the Rag GTPase heterodimer remains unknown. We used cryo-EM to determine the structure of FLCN-FNIP2 in a complex with the Rag GTPases and Ragulator. FLCN-FNIP2 adopts an extended conformation with two pairs of heterodimerized domains. The Longin domains heterodimerize and contact both nucleotide binding domains of the Rag heterodimer, while the DENN domains interact at the distal end of the structure. Biochemical analyses reveal a conserved arginine on FLCN as the catalytic arginine finger and lead us to interpret our structure as an on-pathway intermediate. These data reveal features of a GAP-GTPase interaction and the structure of a critical component of the nutrient-sensing mTORC1 pathway.


Subject(s)
Carrier Proteins/ultrastructure , Cryoelectron Microscopy , Monomeric GTP-Binding Proteins/ultrastructure , Multiprotein Complexes/ultrastructure , Proto-Oncogene Proteins/ultrastructure , Tumor Suppressor Proteins/ultrastructure , Arginine/metabolism , Biocatalysis , Carrier Proteins/chemistry , GTPase-Activating Proteins/metabolism , HEK293 Cells , Humans , Hydrolysis , Models, Molecular , Monomeric GTP-Binding Proteins/chemistry , Multiprotein Complexes/chemistry , Protein Conformation , Protein Multimerization , Proto-Oncogene Proteins/chemistry , Tumor Suppressor Proteins/chemistry
3.
Mol Genet Genomic Med ; 7(7): e00772, 2019 07.
Article in English | MEDLINE | ID: mdl-31187595

ABSTRACT

BACKGROUND: The human TET2 gene plays a pivotal role in the epigenetic regulation of normal and malignant hematopoiesis. Somatic TET2 mutations have been repeatedly identified in age-related clonal hematopoiesis and in myeloid neoplasms ranging from acute myeloid leukemia (AML) to myeloproliferative neoplasms. However, there have been no attempts to systematically explore the structural and functional consequences of the hundreds of TET2 missense variants reported to date. METHODS: We have sequenced the TET2 gene in 189 Spanish AML patients using Sanger sequencing and NGS protocols. Next, we performed a thorough bioinformatics analysis of TET2 protein and of the expected impact of all reported TET2 missense variants on protein structure and function, exploiting available structure-and-function information as well as 3D structure prediction tools. RESULTS: We have identified 38 TET2 allelic variants in the studied patients, including two frequent SNPs: p.G355D (10 cases) and p.I1762V (28 cases). Four of the detected mutations are reported here for the first time: c.122C>T (p.P41L), c.4535C>G (p.A1512G), c.4760A>G (p.D1587G), and c.5087A>T (p.Y1696F). We predict a complex multidomain architecture for the noncatalytic regions of TET2, and in particular the presence of well-conserved α+ß globular domains immediately preceding and following the actual catalytic unit. Further, we provide a rigorous interpretation of over 430 missense SNVs that affect the TET2 catalytic domain, and we hypothesize explanations for ~700 additional variants found within the regulatory regions of the protein. Finally, we propose a systematic classification of all missense mutants and SNPs reported to date into three major categories (severe, moderate, and mild), based on their predicted structural and functional impact. CONCLUSIONS: The proposed classification of missense TET2 variants would help to assess their clinical impact on human neoplasia and may guide future structure-and-function investigations of TET family members.


Subject(s)
DNA-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Adult , Alleles , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/ultrastructure , Dioxygenases , Epigenesis, Genetic , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Male , Middle Aged , Mutation , Mutation, Missense/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/ultrastructure
4.
Mol Cell ; 67(2): 322-333.e6, 2017 Jul 20.
Article in English | MEDLINE | ID: mdl-28689658

ABSTRACT

The proteasome holoenzyme is activated by its regulatory particle (RP) consisting of two subcomplexes, the lid and the base. A key event in base assembly is the formation of a heterohexameric ring of AAA-ATPases, which is guided by at least four RP assembly chaperones in mammals: PAAF1, p28/gankyrin, p27/PSMD9, and S5b. Using cryogenic electron microscopy, we analyzed the non-AAA structure of the p28-bound human RP at 4.5 Å resolution and determined seven distinct conformations of the Rpn1-p28-AAA subcomplex within the p28-bound RP at subnanometer resolutions. Remarkably, the p28-bound AAA ring does not form a channel in the free RP and spontaneously samples multiple "open" and "closed" topologies at the Rpt2-Rpt6 and Rpt3-Rpt4 interfaces. Our analysis suggests that p28 assists the proteolytic core particle to select a specific conformation of the ATPase ring for RP engagement and is released in a shoehorn-like fashion in the last step of the chaperone-mediated proteasome assembly.


Subject(s)
Molecular Chaperones/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins/metabolism , ATPases Associated with Diverse Cellular Activities , Adaptor Proteins, Signal Transducing/metabolism , Cryoelectron Microscopy , HEK293 Cells , Humans , LIM Domain Proteins/metabolism , LIM Domain Proteins/ultrastructure , Models, Molecular , Molecular Chaperones/ultrastructure , Proteasome Endopeptidase Complex/ultrastructure , Protein Binding , Protein Structure, Quaternary , Protein Subunits , Proto-Oncogene Proteins/ultrastructure , Structure-Activity Relationship , Transcription Factors/metabolism , Transcription Factors/ultrastructure , Transfection
5.
J Struct Biol ; 192(3): 449-456, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26458359

ABSTRACT

Cerebral cavernous malformations (CCM) are vascular dysplasias that usually occur in the brain and are associated with mutations in the KRIT1/CCM1, CCM2/MGC4607/OSM/Malcavernin, and PDCD10/CCM3/TFAR15 genes. Here we report the 2.9 Å crystal structure of the ankyrin repeat domain (ARD) and FERM domain of the protein product of KRIT1 (KRIT1; Krev interaction trapped 1). The crystal structure reveals that the KRIT1 ARD contains 4 ankyrin repeats. There is an unusual conformation in the ANK4 repeat that is stabilized by Trp-404, and the structure reveals a solvent exposed ankyrin groove. Domain orientations of the three copies within the asymmetric unit suggest a stable interaction between KRIT1 ARD and FERM domains, indicating a globular ARD-FERM module. This resembles the additional F0 domain found N-terminal to the FERM domain of talin. Structural analysis of KRIT1 ARD-FERM highlights surface regions of high evolutionary conservation, and suggests potential sites that could mediate interaction with binding partners. The structure therefore provides a better understanding of KRIT1 at the molecular level.


Subject(s)
Ankyrin Repeat/physiology , Microtubule-Associated Proteins/ultrastructure , Proto-Oncogene Proteins/ultrastructure , Amino Acid Sequence , Brain/pathology , Crystallography, X-Ray , Hemangioma, Cavernous, Central Nervous System/genetics , Humans , KRIT1 Protein , Microtubule-Associated Proteins/genetics , Molecular Sequence Data , Protein Binding/genetics , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Sequence Alignment
6.
J Cell Biol ; 208(7): 987-1001, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25825518

ABSTRACT

Mutations in the essential adaptor proteins CCM2 or CCM3 lead to cerebral cavernous malformations (CCM), vascular lesions that most frequently occur in the brain and are strongly associated with hemorrhagic stroke, seizures, and other neurological disorders. CCM2 binds CCM3, but the molecular basis of this interaction, and its functional significance, have not been elucidated. Here, we used x-ray crystallography and structure-guided mutagenesis to show that an α-helical LD-like motif within CCM2 binds the highly conserved "HP1" pocket of the CCM3 focal adhesion targeting (FAT) homology domain. By knocking down CCM2 or CCM3 and rescuing with binding-deficient mutants, we establish that CCM2-CCM3 interactions protect CCM2 and CCM3 proteins from proteasomal degradation and show that both CCM2 and CCM3 are required for normal endothelial cell network formation. However, CCM3 expression in the absence of CCM2 is sufficient to support normal cell growth, revealing complex-independent roles for CCM3.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/metabolism , Cell Proliferation/genetics , Central Nervous System/blood supply , Membrane Proteins/metabolism , Neovascularization, Physiologic/genetics , Proto-Oncogene Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/ultrastructure , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/ultrastructure , Cell Line , Crystallography, X-Ray , Gene Expression , Hemangioma, Cavernous, Central Nervous System/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/ultrastructure , Mutagenesis , Paxillin/metabolism , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Proteolysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/ultrastructure , RNA Interference , RNA, Small Interfering , Sequence Alignment
7.
Nucleus ; 5(6): 601-12, 2014.
Article in English | MEDLINE | ID: mdl-25485891

ABSTRACT

Nuclear pore complexes (NPCs) span the 2 membranes of the nuclear envelope (NE) and facilitate nucleocytoplasmic exchange of macromolecules. NPCs have a roughly tripartite structural organization with the so-called nuclear basket emanating from the NPC scaffold into the nucleoplasm. The nuclear basket is composed of the 3 nucleoporins Nup153, Nup50, and Tpr, but their specific role for the structural organization of this NPC substructure is, however, not well established. In this study, we have used thin-section transmission electron microscopy to determine the structural consequences of altering the expression of Nup153 in human cells. We show that the assembly and integrity of the nuclear basket is not affected by Nup153 depletion, whereas its integrity is perturbed in cells expressing high concentrations of the zinc-finger domain of Nup153. Moreover, even mild over-expression of Nup153 is coinciding with massive changes in nuclear organization and it is the excess of the zinc-finger domain of Nup153 that is sufficient to induce these rearrangements. Our data indicate a central function of Nup153 in the organization of the nucleus, not only at the periphery, but throughout the entire nuclear interior.


Subject(s)
Cell Nucleolus/ultrastructure , Nuclear Pore Complex Proteins/genetics , Cell Nucleolus/genetics , Gene Expression Regulation , HeLa Cells , Humans , Microscopy, Electron , Nuclear Pore/genetics , Nuclear Pore/ultrastructure , Nuclear Pore Complex Proteins/biosynthesis , Nuclear Pore Complex Proteins/ultrastructure , Nuclear Proteins/biosynthesis , Nuclear Proteins/ultrastructure , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/ultrastructure
8.
Biochem Biophys Res Commun ; 450(1): 741-5, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24950407

ABSTRACT

Hepatitis B virus X protein (HBx) is a multifunctional protein, which is considered to be an essential molecule for viral replication and the development of liver diseases. Recently, it has been demonstrated that HBx can directly interact with Bcl-2 and Bcl-xL through a sequence (termed the BH3-like motif) that is related to the BH3 motif of pro-apoptotic BH3-only proteins. Here, we present the first structural characterization of the HBx BH3-like motif by circular dichroism and NMR spectroscopies. Our results demonstrated that the HBx BH3-like motif has the ability to form an α-helix, and the potential helical region involves residues L108-L134. This is a common characteristic among the BH3 peptides of pro-apoptotic BH3-only proteins, implying that HBx may interact with Bcl-2 and Bcl-xL, by forming an α-helix, similar to the interaction mode of other BH3 peptides with Bcl-2 and Bcl-xL.


Subject(s)
Peptide Fragments/chemistry , Peptide Fragments/ultrastructure , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/ultrastructure , Trans-Activators/chemistry , Trans-Activators/ultrastructure , Water/chemistry , Amino Acid Motifs , Amino Acid Sequence , Molecular Sequence Data , Protein Conformation , Viral Regulatory and Accessory Proteins
9.
Biophys J ; 97(4): L8-L10, 2009 Aug 19.
Article in English | MEDLINE | ID: mdl-19686639

ABSTRACT

The inactive-to-active conformational transition of the catalytic domain of human c-Src tyrosine kinase is characterized using the string method with swarms-of-trajectories with all-atom explicit solvent molecular dynamics simulations. The activation process occurs in two main steps in which the activation loop (A-loop) opens first, followed by the rotation of the alphaC helix. The computed potential of mean force energy along the activation pathway displays a local minimum, which allows the identification of an intermediate state. These results show that the string method with swarms-of-trajectories is an effective technique to characterize complex and slow conformational transitions in large biomolecular systems.


Subject(s)
Models, Chemical , Models, Molecular , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/ultrastructure , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/ultrastructure , Binding Sites , CSK Tyrosine-Protein Kinase , Catalysis , Computer Simulation , Enzyme Activation , Protein Binding , Protein Conformation , Protein Structure, Tertiary , src-Family Kinases
10.
Methods ; 39(4): 309-15, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16935003

ABSTRACT

Electron microscopy (EM) has been used for several decades to study the mechanisms of nuclear transport. In early studies of nuclear import, gold-conjugated nuclear proteins were microinjected into cells and followed by EM. As the components of the nuclear pore complex (NPC) and soluble mediators of nuclear import were cloned and characterized, gold-conjugated antibodies were utilized to sublocalize the components of the nuclear transport machinery by immuno-EM. Further, gold-conjugated recombinant proteins were used to probe permeabilized cells or isolated nuclear envelopes and characterize binding sites for these proteins at the NPC. More recently, recombinant gold-conjugated nuclear proteins were used in in vitro nuclear import assays to help dissect the mechanisms of nuclear import. We have used this ultrastructural nuclear import assay to study the nuclear import of the transcription factor PU.1. The results showed that this import requires energy but is carrier-independent. In the presence of energy, gold-conjugated PU.1 shifted to the nuclear side of the NPC and the inside of the nucleus. In conjunction with biochemical assays, these results indicated that this shift involved Ran-dependent binding of PU.1 to NUP153, a nucleoporin situated at the nuclear side of the NPC. Here we describe in detail the methods used in the ultrastructural nuclear import assay including preparation of recombinant protein, gold conjugation, in vitro nuclear import assay, electron microscopy, and data analysis.


Subject(s)
Active Transport, Cell Nucleus/physiology , Biological Assay , Cell Nucleus/metabolism , Nuclear Proteins/metabolism , Cell Nucleus/ultrastructure , HeLa Cells , Humans , Immunohistochemistry , In Vitro Techniques , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Nuclear Pore/ultrastructure , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore Complex Proteins/ultrastructure , Nuclear Proteins/ultrastructure , Protein Transport/physiology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/ultrastructure , Trans-Activators/metabolism , Trans-Activators/ultrastructure
13.
Mol Biol Cell ; 15(9): 4261-77, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15229283

ABSTRACT

The vertebrate nuclear pore complex (NPC) is a macromolecular assembly of protein subcomplexes forming a structure of eightfold radial symmetry. The NPC core consists of globular subunits sandwiched between two coaxial ring-like structures of which the ring facing the nuclear interior is capped by a fibrous structure called the nuclear basket. By postembedding immunoelectron microscopy, we have mapped the positions of several human NPC proteins relative to the NPC core and its associated basket, including Nup93, Nup96, Nup98, Nup107, Nup153, Nup205, and the coiled coil-dominated 267-kDa protein Tpr. To further assess their contributions to NPC and basket architecture, the genes encoding Nup93, Nup96, Nup107, and Nup205 were posttranscriptionally silenced by RNA interference (RNAi) in HeLa cells, complementing recent RNAi experiments on Nup153 and Tpr. We show that Nup96 and Nup107 are core elements of the NPC proper that are essential for NPC assembly and docking of Nup153 and Tpr to the NPC. Nup93 and Nup205 are other NPC core elements that are important for long-term maintenance of NPCs but initially dispensable for the anchoring of Nup153 and Tpr. Immunogold-labeling for Nup98 also results in preferential labeling of NPC core regions, whereas Nup153 is shown to bind via its amino-terminal domain to the nuclear coaxial ring linking the NPC core structures and Tpr. The position of Tpr in turn is shown to coincide with that of the nuclear basket, with different Tpr protein domains corresponding to distinct basket segments. We propose a model in which Tpr constitutes the central architectural element that forms the scaffold of the nuclear basket.


Subject(s)
Nuclear Pore Complex Proteins/metabolism , Nuclear Pore/metabolism , Proto-Oncogene Proteins/metabolism , HeLa Cells , Humans , Microscopy, Immunoelectron , Models, Biological , Multiprotein Complexes , Nuclear Pore/ultrastructure , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/ultrastructure , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/ultrastructure , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/ultrastructure , RNA Interference
14.
Mol Cancer Res ; 1(1): 79-87, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12496371

ABSTRACT

The EphA2 receptor protein tyrosine kinase is overexpressed and functionally altered in a large number of human carcinomas. Despite its elevated levels in cancer, the EphA2 on the surface of malignant cells demonstrates lower levels of ligand binding and tyrosine phosphorylation than the EphA2 on non-transformed epithelial cells. In our present study, we demonstrate that ligand-mediated stimulation causes EphA2 to be internalized and degraded. The mechanism of this response involves ligand-mediated autophosphorylation of EphA2, which promotes an association between EphA2 and the c-Cbl adaptor protein. We also show that c-Cbl promotes stimulation-dependent EphA2 degradation. These findings are important for understanding the causes of EphA2 overexpression in malignant cells and provide a foundation for investigating EphA2 as a potential target for therapeutic intervention.


Subject(s)
Breast Neoplasms/enzymology , Epithelial Cells/enzymology , Proto-Oncogene Proteins/metabolism , Receptor, EphA2/metabolism , Ubiquitin-Protein Ligases , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/ultrastructure , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Endosomes/metabolism , Endosomes/ultrastructure , Ephrin-A1/metabolism , Ephrin-A1/pharmacology , Ephrin-A1/ultrastructure , Epithelial Cells/pathology , Humans , Ligands , Proto-Oncogene Proteins/ultrastructure , Proto-Oncogene Proteins c-cbl , Receptor, EphA2/ultrastructure , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Tumor Cells, Cultured
15.
Biochem Biophys Res Commun ; 298(5): 744-9, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12419316

ABSTRACT

Direct imaging of the interaction of the apoptotic protein, Bax, with membrane bilayers shows the presence of toroidal-shaped pores using atomic force microscopy. These pores are sufficiently large to allow passage of proteins from the intermitochondrial space. Both the perturbation of the membrane and the amount of protein bound to the bilayer are increased in the presence of calcium. The results from the imaging are consistent with leakage studies from liposomes of the same composition. The work shows that Bax by itself can form pores in membrane bilayers.


Subject(s)
Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Apoptosis/physiology , Calcium/metabolism , In Vitro Techniques , Lipid Bilayers , Liposomes , Membranes/metabolism , Membranes/ultrastructure , Microscopy, Atomic Force , Mitochondria/metabolism , Mitochondria/ultrastructure , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/ultrastructure , bcl-2-Associated X Protein
16.
Exp Cell Res ; 276(2): 223-32, 2002 Jun 10.
Article in English | MEDLINE | ID: mdl-12027452

ABSTRACT

Here we report immunofluorescence localizations of the Drosophila Tpr protein which are inconsistent with a filament-forming protein statically associated with nuclear pore complex-associated intranuclear filaments. Using tissues from throughout the Drosophila life cycle, we observe that Tpr is often localized to discontinuous, likely granular or particulate structures in the deep nuclear interior. These apparent granules have no obvious connectivity to pore complexes in the nuclear periphery, and are often localized on the surfaces of chromosomes and to the perinucleolar region. Most strikingly, after 1 h of heat shock, the great majority of the Tpr in the deep nuclear interior accumulates at a single heat shock puff, while Tpr in the nuclear periphery appears unchanged. This heat shock puff, 93D, is a known repository for many components of pre-mRNA metabolism during heat shock. Although we do not observe Tpr at sites of transcription under normal conditions, the 93D heat shock result leads us to favor a role for Tpr in mRNA metabolism, such as the transport of mRNA through the nuclear interior to nuclear pore complexes. Consistent with this, we observe networks of Tpr containing granules spanning between the nucleolus and the nuclear periphery which are also decorated by an anti-SR protein antibody. Since we also observe Drosophila Tpr in reticular or fibrous structures in other nuclei, such as salivary gland polytene nuclei, these results indicate that Tpr can exist in at least two structural forms, and suggest that Tpr may relocalize or even change structural forms in response to cellular needs.


Subject(s)
Cell Nucleus Structures/metabolism , Cytoskeleton/metabolism , Drosophila melanogaster/metabolism , Nuclear Pore Complex Proteins/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Active Transport, Cell Nucleus/genetics , Animals , Binding Sites/genetics , Cell Compartmentation/genetics , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , Cell Nucleus Structures/ultrastructure , Chromosomes/genetics , Cytoplasmic Granules/genetics , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Cytoskeleton/genetics , Cytoskeleton/ultrastructure , DNA-Binding Proteins/genetics , Drosophila melanogaster/cytology , Female , Heat-Shock Response/genetics , Male , Molecular Weight , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/ultrastructure , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/ultrastructure , RNA, Messenger/genetics
17.
Eur J Cell Biol ; 81(1): 26-35, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11893076

ABSTRACT

Cbl is an adaptor protein that is phosphorylated and recruited to several receptor and non-receptor tyrosine kinases upon their activation. After binding to the activated receptor, Cbl plays a key role as a kinase inhibitor and as an E3 ubiquitin ligase, thereby contributing to receptor down-regulation and internalization. In addition, Cbl translocates to intracellular vesicular compartments following receptor activation. We report here that Cbl also associates with Golgi membranes. Confocal immunofluorescence staining of Cbl in a variety of unstimulated cells, including CHO cells, revealed a prominent perinuclear colocalization of Cbl and a Golgi marker. Both the prominent Cbl staining and the Golgi marker were dispersed by brefeldin A. Subcellular fractionation of CHO cells demonstrated that about 10% of Cbl is stably associated with membranes, and that Golgi-enriched membrane fractions produced by isopycnic density centrifugation and free-flow electrophoresis are also enriched in Cbl, relative to other membrane fractions. The membrane-bound Cbl was hyperphosphorylated and it co-immunoprecipitated with endogenous Src. By immunofluorescence, some Src colocalized with Cbl and Golgi markers, and Src, like Cbl, was present in the Golgi-enriched fraction prepared by sequential density centrifugation and free-flow electrophoresis. Transfection of an activated form of Src, but not wild-type Src, increased the amount of Src that co-immunoprecipitated with Cbl, and increased the intensity of Cbl staining on the Golgi. This result, together with the increased tyrosine phosphorylation of the membrane-associated Cbl, suggests that Golgi-associated Cbl could be part of a molecular complex that contains activated Src. The localization and interaction of Src and Cbl at the Golgi and the regulation of the interaction of Cbl with Golgi membrane suggest that this complex may contribute to the regulation of Golgi function.


Subject(s)
Cell Compartmentation/physiology , Eukaryotic Cells/metabolism , Golgi Apparatus/metabolism , Intracellular Membranes/metabolism , Protein Transport/physiology , Proto-Oncogene Proteins/metabolism , Ubiquitin-Protein Ligases , src-Family Kinases/metabolism , Animals , Biomarkers/analysis , CHO Cells , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cricetinae , Endosomes/metabolism , Endosomes/ultrastructure , Eukaryotic Cells/cytology , Gene Expression Regulation/physiology , Golgi Apparatus/ultrastructure , Intracellular Membranes/ultrastructure , Macromolecular Substances , Membrane Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins/ultrastructure , Proto-Oncogene Proteins c-cbl , Subcellular Fractions/metabolism , src-Family Kinases/genetics , src-Family Kinases/ultrastructure
18.
Nucleic Acids Res ; 26(23): 5425-31, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9826768

ABSTRACT

c-myb antisense oligonucleotides (AS ODNs) were reversibly immobilized to a novel polymeric core shell nanosphere and their cellular uptake and inhibitory effect on HL60 leukemia cell proliferation studied. The nanosphere surface was so designed as to directly bind ODNs via ionic interactions and reversibly release them inside the cells. Compared with the cellular uptake of free oligonucleotide, the use of AS ODN (immobilized to the nanospheres) produced a 50-fold increase in the intracellular concentration. Specifically, a single dose of 320 nM of AS ODN immobilized to the nanospheres was capable of inhibiting HL60 cell proliferation with the same degree of efficiency obtained using a 50-fold higher dose of free AS ODN. Flow cytometric experiments with fluoresceinated ODNs showed a temperature-dependent uptake, which was detectable as early as 2 h after the beginning of treatment. The inhibitory effect on cell proliferation was maintained for up to 8 days of culture. Moreover, the level of c-Myb protein decreased by 24% after 2 days and by 60% after 4 days of treatment, thus indicating a continuous and sustained release of non-degraded AS ODN from the nanospheres inside the cells.


Subject(s)
Oligonucleotides, Antisense/chemical synthesis , Oligonucleotides, Antisense/metabolism , Polymers/chemical synthesis , Polymers/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Blotting, Western , Chromatography, High Pressure Liquid , Culture Media/metabolism , Flow Cytometry , HL-60 Cells , Humans , Microspheres , Oligonucleotides, Antisense/ultrastructure , Proto-Oncogene Proteins/ultrastructure , Proto-Oncogene Proteins c-myb , Receptors, Complement/metabolism , Recombinant Proteins/metabolism , Trans-Activators/ultrastructure , Tumor Cells, Cultured
19.
J Cell Biol ; 132(5): 945-53, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8603925

ABSTRACT

The high-affinity NGF receptor is thought to be a complex of two receptors , gp75 and the tyrosine kinase TrkA, but direct biochemical evidence for such an association had been lacking. In this report, we demonstrate the existence of such a gp75-TrkA complex by a copatching technique. Gp75 on the surface of intact cells is patched with an anti-gp75 antibody and fluorescent secondary antibody, the cells are then fixed to prevent further antibody-induced redistributions, and the distribution of TrkA is probed with and anti-TrkA antibody and fluorescent secondary antibody. We utilize a baculovirus-insect cell expression of wild-type and mutated NGF receptors. TrkA and gp75 copatch in both the absence and presence of NGF. The association is specific, since gp75 does not copatch with other tyrosine kinase receptors, including TrkB, platelet-derived growth factor receptor-beta, and Torso (Tor). To determine which domains of TrkA are required for copatching, we used a series of TrkA-Tor chimeric receptors and show that the extracellular domain of TrkA is sufficient for copatching with gp75. A chimeric receptor with TrkA transmembrane and intracellular domains show partial copatching with gp75. Deletion of the intracellular domain of gp75 decreases but does not eliminate copatching. A point mutation which inactivates the TrkA kinase has no effect on copatching, indicating that this enzymatic activity is not required for association with gp75. Hence, although interactions between the gp75 and TrkA extracellular domains are sufficient for complex formation, interactions involving other receptor domains also play a role.


Subject(s)
Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Cell Compartmentation , Cells, Cultured , Fluorescent Antibody Technique , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/ultrastructure , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/ultrastructure , Receptor, Nerve Growth Factor , Receptor, trkA , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/ultrastructure , Recombinant Proteins/metabolism , Spodoptera/cytology
20.
Int J Cancer ; 53(3): 401-8, 1993 Feb 01.
Article in English | MEDLINE | ID: mdl-7679090

ABSTRACT

Over-expression of the c-erbB-2 (HER-2/neu) gene product p185 occurs in 30% of breast and ovarian cancers. The p185 protein might serve as a target for serotherapy in that antibodies against different epitopes on the extracellular domain of p185 can inhibit growth of tumor cells in the absence of cellular or humoral effector mechanisms. To define epitopes of functional relevance, 11 monoclonal antibodies (MAbs) were evaluated for their ability to bind to the extracellular domain of p185. Results of competition studies with 125I-labeled and non-labeled antibodies indicated that 10 of 11 epitopes were grouped in a linear array. Antibodies against 7 epitopes inhibited anchorage-independent growth and antibodies against 2 of these epitopes also inhibited anchorage-dependent growth of SKBr3 breast-cancer cells that over-expressed p185. Treatment with antibodies exerted cytotoxic rather than cytostatic effects. When antibodies were used in combination, additive or supra-additive inhibition of anchorage-independent and anchorage-dependent growth was observed between pairs of antibodies. Growth inhibition did not relate to the affinity of the antibody or its isotype. Two antibodies that inhibited both anchorage-dependent and anchorage-independent growth also blocked binding of the HER-2/neu ligand, whereas 5 antibodies that inhibited only anchorage-independent growth had no effect on ligand binding. Inhibition of cell growth did not correlate with internalization of p185 or down-regulation of p185 on the cell surface. Fab fragments of active antibodies could also inhibit anchorage-independent growth of SKBr3. Thus, murine MAbs and their fragments recognized both immunochemically distinct and functionally distinct epitopes on the p185 molecule. Whereas inhibition of anchorage-dependent growth correlated with the ability of antibodies to block ligand binding, inhibition of anchorage-independent growth did not correlate with effects on ligand binding, internalization, cell-surface expression or cross-linking of p185.


Subject(s)
Proto-Oncogene Proteins/immunology , Antibodies, Monoclonal/immunology , Antigens, Surface/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Adhesion , Cell Division , Epitopes , Extracellular Space , Growth Inhibitors , Humans , In Vitro Techniques , Proto-Oncogene Proteins/ultrastructure , Receptor, ErbB-2 , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...