Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
1.
Front Immunol ; 12: 631271, 2021.
Article in English | MEDLINE | ID: mdl-33763075

ABSTRACT

Phosphoinositide 3-kinases (PI3Ks) play a central role in adaptive immunity by transducing signals from the T cell antigen receptor (TCR) via production of PIP3. PI3Kδ is a heterodimer composed of a p110δ catalytic subunit associated with a p85α or p85ß regulatory subunit and is preferentially engaged by the TCR upon T cell activation. The molecular mechanisms leading to PI3Kδ recruitment and activation at the TCR signalosome remain unclear. In this study, we have used quantitative mass spectrometry, biochemical approaches and CRISPR-Cas9 gene editing to uncover the p110δ interactome in primary CD4+ T cells. Moreover, we have determined how the PI3Kδ interactome changes upon the differentiation of small naïve T cells into T cell blasts expanded in the presence of IL-2. Our interactomic analyses identified multiple constitutive and inducible PI3Kδ-interacting proteins, some of which were common to naïve and previously-activated T cells. Our data reveals that PI3Kδ rapidly interacts with as many as seven adaptor proteins upon TCR engagement, including the Gab-family proteins, GAB2 and GAB3, a CD5-CBL signalosome and the transmembrane proteins ICOS and TRIM. Our results also suggest that PI3Kδ pre-forms complexes with the adaptors SH3KBP1 and CRKL in resting cells that could facilitate the localization and activation of p110δ at the plasma membrane by forming ternary complexes during early TCR signalling. Furthermore, we identify interactions that were not previously known to occur in CD4+ T cells, involving BCAP, GAB3, IQGAP3 and JAML. We used CRISPR-Cas9-mediated gene knockout in primary T cells to confirm that BCAP is a positive regulator of PI3K-AKT signalling in CD4+ T cell blasts. Overall, our results provide evidence for a large protein network that regulates the recruitment and activation of PI3Kδ in T cells. Finally, this work shows how the PI3Kδ interactome is remodeled as CD4+ T cells differentiate from naïve T cells to activated T cell blasts. These activated T cells upregulate additional PI3Kδ adaptor proteins, including BCAP, GAB2, IQGAP3 and ICOS. This rewiring of TCR-PI3K signalling that occurs upon T cell differentiation may serve to reduce the threshold of activation and diversify the inputs for the PI3K pathway in effector T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/immunology , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/immunology , Receptors, Antigen, T-Cell/immunology , Animals , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/drug effects , CRISPR-Cas Systems , Gene Editing , Gene Knockout Techniques , Interleukin-2/pharmacology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/genetics , Signal Transduction , Specific Pathogen-Free Organisms
2.
Methods Mol Biol ; 2247: 3-16, 2021.
Article in English | MEDLINE | ID: mdl-33301109

ABSTRACT

Membrane proteins constitute an important class of proteins for medical, pharmaceutical, and biotechnological reasons. Understanding the structure and function of membrane proteins and their complexes is of key importance, but the progress in this area is slow because of the difficulties to produce them in sufficient quality and quantity. Overexpression of membrane proteins is often restricted by the limited capability of translocation systems to integrate proteins into the membrane and to fold them properly. Purification of membrane proteins requires their isolation from the membrane, which is a further challenge. The choice of expression system, detergents, and purification tags is therefore an important decision. Here, we present a protocol for expression in bacteria and isolation of a seven-subunit membrane protein complex, the bacterial holo-translocon, which can serve as a starting point for the production of other membrane protein complexes for structural and functional studies.


Subject(s)
Membrane Proteins/biosynthesis , Membrane Proteins/isolation & purification , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/isolation & purification , Protein Subunits/biosynthesis , Protein Subunits/isolation & purification , Recombinant Proteins , Chromatography, Affinity , Chromatography, Gel , Escherichia coli/genetics , Gene Expression , Membrane Proteins/chemistry , Plasmids , Promoter Regions, Genetic , Protein Multimerization , Protein Subunits/chemistry
3.
Methods Mol Biol ; 2247: 17-38, 2021.
Article in English | MEDLINE | ID: mdl-33301110

ABSTRACT

Most cellular processes are mediated by multi-subunit protein complexes which have attracted major interest in both academia and industry. Recombinant production of such entities in quantity and quality sufficient for functional and structural investigations may be extremely challenging and necessitate specific technologies. The baculovirus expression vector system is widely used for the production of eukaryotic multiprotein complexes, and a variety of strategies are available to assemble transfer vectors for the generation of recombinant baculoviruses. Here we detail applications of homology-based cloning techniques for one-step construction of dual promoter baculovirus transfer plasmids and of restriction-free (RF) cloning for the modification of existing constructs.


Subject(s)
Baculoviridae/genetics , Gene Expression , Genetic Vectors/genetics , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/genetics , Recombinant Proteins , Base Sequence , Cell Line , Cells, Cultured , Cloning, Molecular/methods , Gene Order , Multiprotein Complexes/chemistry , Plasmids/genetics , Promoter Regions, Genetic , Recombinant Fusion Proteins
4.
Nat Nanotechnol ; 15(9): 783-791, 2020 09.
Article in English | MEDLINE | ID: mdl-32690886

ABSTRACT

The assembly of protein machines in cells is precise, rapid, and coupled to protein synthesis with regulation in space and time. The assembly of natural and synthetic nanomachines could be similarly controlled by genetic programming outside the cell. Here, we present quasi-two-dimensional (2D) silicon compartments that enable programming of protein assembly lines by local synthesis from surface-immobilized DNA brushes. Using this platform, we studied the autonomous synthesis and assembly of a structural complex from a bacteriophage and a bacterial RNA-synthesizing machine. Local synthesis and surface capture of complexes provided high assembly yield and sensitive detection of spatially resolved assembly intermediates, with the 3D geometry of the compartment and the 2D pattern of brushes dictating the yield and mode of assembly steps. Localized synthesis of proteins in a single gene brush enhances their interactions, and displacement of their genes in separated brushes leads to step-by-step surface assembly. This methodology enables spatial regulation of protein synthesis, and deciphering, reconstruction and design of biological machine assembly lines.


Subject(s)
Bacteriophage T4/genetics , Immobilized Nucleic Acids/genetics , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/genetics , Protein Engineering/instrumentation , Protein Engineering/methods , Cell-Free System , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Equipment Design , Escherichia coli/genetics , Gene Silencing , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Promoter Regions, Genetic , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Silicon , Viral Proteins/genetics , Viral Proteins/metabolism
5.
Viruses ; 11(3)2019 02 26.
Article in English | MEDLINE | ID: mdl-30813511

ABSTRACT

The baculovirus/insect cell system (BICS) is widely used in academia and industry to produce eukaryotic proteins for many applications, ranging from structure analysis to drug screening and the provision of protein biologics and therapeutics. Multi-protein complexes have emerged as vital catalysts of cellular function. In order to unlock the structure and mechanism of these essential molecular machines and decipher their function, we developed MultiBac, a BICS particularly tailored for heterologous multigene transfer and multi-protein complex production. Baculovirus is unique among common viral vectors in its capacity to accommodate very large quantities of heterologous DNA and to faithfully deliver this cargo to a host cell of choice. We exploited this beneficial feature to outfit insect cells with synthetic DNA circuitry conferring new functionality during heterologous protein expression, and developing customized MultiBac baculovirus variants in the process. By altering its tropism, recombinant baculovirions can be used for the highly efficient delivery of a customized DNA cargo in mammalian cells and tissues. Current advances in synthetic biology greatly facilitate the construction or recombinant baculoviral genomes for gene editing and genome engineering, mediated by a MultiBac baculovirus tailored to this purpose. Here, recent developments and exploits of the MultiBac system are presented and discussed.


Subject(s)
Baculoviridae/genetics , DNA , Gene Transfer Techniques , Insecta/cytology , Recombinant Proteins/biosynthesis , Animals , Baculoviridae/physiology , Cloning, Molecular , Gene Editing , Genetic Vectors , Humans , Mammals , Multiprotein Complexes/biosynthesis , Viral Tropism
6.
Nature ; 563(7730): 209-213, 2018 11.
Article in English | MEDLINE | ID: mdl-30232455

ABSTRACT

Tc toxins secrete toxic enzymes into host cells using a unique syringe-like injection mechanism. They are composed of three subunits, TcA, TcB and TcC. TcA forms the translocation channel and the TcB-TcC heterodimer functions as a cocoon that shields the toxic enzyme. Binding of the cocoon to the channel triggers opening of the cocoon and translocation of the toxic enzyme into the channel. Here we show in atomic detail how the assembly of the three components activates the toxin. We find that part of the cocoon completely unfolds and refolds into an alternative conformation upon binding. The presence of the toxic enzyme inside the cocoon is essential for its subnanomolar binding affinity for the TcA subunit. The enzyme passes through a narrow negatively charged constriction site inside the cocoon, probably acting as an extruder that releases the unfolded protein with its C terminus first into the translocation channel.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Cryoelectron Microscopy , Multiprotein Complexes/ultrastructure , Photorhabdus/ultrastructure , Protein Refolding , Protein Unfolding , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/metabolism , ADP Ribose Transferases/ultrastructure , Bacterial Toxins/biosynthesis , Cytotoxins/biosynthesis , Cytotoxins/chemistry , Cytotoxins/metabolism , Models, Biological , Models, Molecular , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Photorhabdus/chemistry , Protein Conformation , Protein Transport
7.
Proc Natl Acad Sci U S A ; 115(33): E7748-E7757, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30065114

ABSTRACT

Approximately one-third of the mammalian proteome is transported from the endoplasmic reticulum-to-Golgi via COPII-coated vesicles. SEC23, a core component of coat protein-complex II (COPII), is encoded by two paralogous genes in vertebrates (Sec23a and Sec23b). In humans, SEC23B deficiency results in congenital dyserythropoietic anemia type-II (CDAII), while SEC23A deficiency results in a skeletal phenotype (with normal red blood cells). These distinct clinical disorders, together with previous biochemical studies, suggest unique functions for SEC23A and SEC23B. Here we show indistinguishable intracellular protein interactomes for human SEC23A and SEC23B, complementation of yeast Sec23 by both human and murine SEC23A/B, and rescue of the lethality of sec23b deficiency in zebrafish by a sec23a-expressing transgene. We next demonstrate that a Sec23a coding sequence inserted into the murine Sec23b locus completely rescues the lethal SEC23B-deficient pancreatic phenotype. We show that SEC23B is the predominantly expressed paralog in human bone marrow, but not in the mouse, with the reciprocal pattern observed in the pancreas. Taken together, these data demonstrate an equivalent function for SEC23A/B, with evolutionary shifts in the transcription program likely accounting for the distinct phenotypes of SEC23A/B deficiency within and across species, a paradigm potentially applicable to other sets of paralogous genes. These findings also suggest that enhanced erythroid expression of the normal SEC23A gene could offer an effective therapeutic approach for CDAII patients.


Subject(s)
COP-Coated Vesicles/metabolism , Erythrocytes/metabolism , Multiprotein Complexes/biosynthesis , Vesicular Transport Proteins/biosynthesis , Anemia, Dyserythropoietic, Congenital/genetics , Anemia, Dyserythropoietic, Congenital/metabolism , Bone Marrow/metabolism , Bone Marrow/pathology , COP-Coated Vesicles/genetics , Erythrocytes/pathology , Gene Expression Regulation , HEK293 Cells , Humans , Multiprotein Complexes/genetics , Species Specificity , Vesicular Transport Proteins/genetics
8.
Nature ; 561(7722): 268-272, 2018 09.
Article in English | MEDLINE | ID: mdl-30158700

ABSTRACT

The folding of newly synthesized proteins to the native state is a major challenge within the crowded cellular environment, as non-productive interactions can lead to misfolding, aggregation and degradation1. Cells cope with this challenge by coupling synthesis with polypeptide folding and by using molecular chaperones to safeguard folding cotranslationally2. However, although most of the cellular proteome forms oligomeric assemblies3, little is known about the final step of folding: the assembly of polypeptides into complexes. In prokaryotes, a proof-of-concept study showed that the assembly of heterodimeric luciferase is an organized cotranslational process that is facilitated by spatially confined translation of the subunits encoded on a polycistronic mRNA4. In eukaryotes, however, fundamental differences-such as the rarity of polycistronic mRNAs and different chaperone constellations-raise the question of whether assembly is also coordinated with translation. Here we provide a systematic and mechanistic analysis of the assembly of protein complexes in eukaryotes using ribosome profiling. We determined the in vivo interactions of the nascent subunits from twelve hetero-oligomeric protein complexes of Saccharomyces cerevisiae at near-residue resolution. We find nine complexes assemble cotranslationally; the three complexes that do not show cotranslational interactions are regulated by dedicated assembly chaperones5-7. Cotranslational assembly often occurs uni-directionally, with one fully synthesized subunit engaging its nascent partner subunit, thereby counteracting its propensity for aggregation. The onset of cotranslational subunit association coincides directly with the full exposure of the nascent interaction domain at the ribosomal tunnel exit. The action of the ribosome-associated Hsp70 chaperone Ssb8 is coordinated with assembly. Ssb transiently engages partially synthesized interaction domains and then dissociates before the onset of partner subunit association, presumably to prevent premature assembly interactions. Our study shows that cotranslational subunit association is a prevalent mechanism for the assembly of hetero-oligomers in yeast and indicates that translation, folding and the assembly of protein complexes are integrated processes in eukaryotes.


Subject(s)
Amino Acyl-tRNA Synthetases/biosynthesis , Fatty Acid Synthases/biosynthesis , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/chemistry , Protein Biosynthesis , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/genetics , Fatty Acid Synthases/chemistry , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , HSP70 Heat-Shock Proteins/metabolism , Models, Molecular , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Binding , Protein Subunits/biosynthesis , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism
9.
J Biol Chem ; 293(36): 14155-14164, 2018 09 07.
Article in English | MEDLINE | ID: mdl-30012887

ABSTRACT

Endosomal transport represents the primary mode for intracellular trafficking and signaling transduction and thus has to be tightly controlled. The molecular processes controlling the endosomal positioning utilize several large protein complexes, one of which contains the small GTPase Rab7, Rab-interacting lysosomal protein (RILP), and oxysterol-binding protein-related protein 1 (ORP1L). Rab7 is known to interact with RILP through a canonical binding site termed the effector-interacting switch region, but it is not clear how Rab7 interacts with ORP1L, limiting our understanding of the overall process. Here, we report structural and biochemical investigation of the Rab7-ORP1L interaction. We found that, contrary to prior studies, the interaction between Rab7 and the N-terminal ankyrin repeat domain (ARDN) of ORP1L is independent of Rab7's GTP- or GDP-binding state. Moreover, we show that Rab7 interacts with ORP1L ARDN via a unique region consisting of helix3 (α3) and 310-helix 2 (η2). This architecture leaves the canonical effector-interacting switch regions available for RILP binding and thus allows formation of the ORP1L-Rab7-RILP tripartite complex. Mutational disruption of the interacting interface between ORP1L and Rab7 compromised the ability of ORP1L-Rab7-RILP to regulate the late endosome positioning. Collectively, our results again manifested the versatility in the interaction between GTPase and its effector.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Endosomes/metabolism , Multiprotein Complexes/biosynthesis , Receptors, Steroid/metabolism , rab GTP-Binding Proteins/metabolism , Binding Sites , Biological Transport , HeLa Cells , Humans , Multiprotein Complexes/chemistry , Protein Binding , Protein Interaction Domains and Motifs , rab7 GTP-Binding Proteins
10.
Exp Hematol ; 62: 39-44.e2, 2018 06.
Article in English | MEDLINE | ID: mdl-29596882

ABSTRACT

Acquired aplastic anemia (AA) is a bone marrow (BM) failure associated with autoimmune destruction of hematopoietic stem cells (HSCs). Although somatic mutations have been identified in AA patients, mutations alone do not explain AA pathophysiology. SWI/SNF is an evolutionarily conserved, multi-subunit, ATP-dependent chromatin-remodeling protein complex that plays an important role in mammalian hematopoiesis. Herein, gene expression analysis identified a significant loss of the SWI/SNF core component SMARCC1, along with ARID1B, ACTL6A, and SMARCD1, in human AA BM CD34+ HSCs and hematopoietic stem and progenitor cells (HSPCs) compared with normal HSPCs. However, expression of SMARCA4, SMARCB1, SMARCD3, and DPF2 remained intact in our AA cohort. PBRM1, BRD7, and SMARCA2 expression were significantly upregulated in both untreated and follow-up AA patients. Clonal hematopoiesis in AA is associated with evolution to late clonal disorders, including myelodysplastic syndromes (MDS). Apart from SMARCD1 loss, we did not observe significant alteration of SWI/SNF expression in MDS HSPCs, indicating SWI/SNF differential expression in AA and MDS. In addition, except for ACTL6A, SWI/SNF expression was unaltered in aged HSPCs. Importantly, our results provide evidence for loss of SWI/SNF in AA, and may implicate AA HSPC-autonomous defective SWI/SNF regulation as an integral component of BM failure, in addition to autoimmune destruction of AA HSCs. These findings illustrate for the first time SWI/SNF subunit expression heterogeneity in human AA HSPCs and require prognostic validation in a larger cohort.


Subject(s)
Anemia, Aplastic/genetics , Chromatin Assembly and Disassembly/genetics , DNA-Binding Proteins/deficiency , Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Multiprotein Complexes/genetics , Myelodysplastic Syndromes/genetics , Transcription Factors/deficiency , Adolescent , Adult , Aged , Anemia, Aplastic/metabolism , Anemia, Aplastic/pathology , Bone Marrow/pathology , Clone Cells/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Female , Follow-Up Studies , Gene Expression Regulation , Hematopoiesis , Humans , Male , Middle Aged , Multiprotein Complexes/biosynthesis , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Protein Subunits , Transcription Factors/biosynthesis , Transcription Factors/genetics , Young Adult
11.
Trends Biochem Sci ; 43(1): 4-9, 2018 01.
Article in English | MEDLINE | ID: mdl-29203338

ABSTRACT

The Rvb1-Rvb2-Tah1-Pih1/prefoldin-like (R2TP/PFDL) complex is a unique chaperone that provides a platform for the assembly and maturation of many key multiprotein complexes in mammalian cells. Here, we propose to rename R2TP/PFDL as PAQosome (particle for arrangement of quaternary structure) to more accurately represent its unique function.


Subject(s)
Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Structure, Quaternary , ATPases Associated with Diverse Cellular Activities/metabolism , Apoptosis Regulatory Proteins/metabolism , Carrier Proteins/metabolism , DNA Helicases/metabolism , Humans , Multiprotein Complexes/biosynthesis
12.
Mol Reprod Dev ; 84(12): 1238-1249, 2017 12.
Article in English | MEDLINE | ID: mdl-29024220

ABSTRACT

In vitro embryo production is an established method for both humans and animals, but is fraught with inferior development and health issues in offspring born after in vitro fertilization procedures. Analysis of epigenetic changes caused by exposure to in vitro conditions should shed light on potential sources of these phenotypes. Using immunocytochemistry, we investigated the localization and relative abundance of components associated with the SWI/SNF (Switch/Sucrose non-fermentable) chromatin-remodeling complex-including BAF155, BAF170, BAF180, BAF53A, BAF57, BAF60A, BAF45D, ARID1A, ARID1B, ARID2, SNF5, and BRD7-in oocytes and in in vitro-produced and in vivo-derived porcine embryos. Differences in the localization of BAF155, BAF170, BAF60A, and ARID1B among these sources indicate that improper timing of chromatin remodeling and cellular differentiation might occur in early preimplantation embryos produced and cultured in vitro.


Subject(s)
Blastocyst/metabolism , Chromatin Assembly and Disassembly/physiology , Gene Expression Regulation, Developmental/physiology , Multiprotein Complexes/biosynthesis , Animals , Blastocyst/cytology , Swine
13.
Mol Cell Biol ; 37(18)2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28674186

ABSTRACT

Transforming growth factor ß (TGF-ß)-induced migration of triple-negative breast cancer (TNBC) cells is dependent on nuclear export of the orphan receptor NR4A1, which plays a role in proteasome-dependent degradation of SMAD7. In this study, we show that TGF-ß induces p38α (mitogen-activated protein kinase 14 [MAPK14]), which in turn phosphorylates NR4A1, resulting in nuclear export of the receptor. TGF-ß/p38α and NR4A1 also play essential roles in the induction of epithelial-to-mesenchymal transition (EMT) and induction of ß-catenin in TNBC cells, and these TGF-ß-induced responses and nuclear export of NR4A1 are blocked by NR4A1 antagonists, the p38 inhibitor SB202190, and kinase-dead [p38(KD)] and dominant-negative [p38(DN)] forms of p38α. Inhibition of NR4A1 nuclear export results in nuclear export of TGF-ß-induced ß-catenin, which then undergoes proteasome-dependent degradation. TGF-ß-induced ß-catenin also regulates NR4A1 expression through formation of the ß-catenin-TCF-3/TCF-4/LEF-1 complex on the NR4A1 promoter. Thus, TGF-ß-induced nuclear export of NR4A1 in TNBC cells plays an essential role in cell migration, SMAD7 degradation, EMT, and induction of ß-catenin, and all of these pathways are inhibited by bis-indole-derived NR4A1 antagonists that inhibit nuclear export of the receptor and thereby block TGF-ß-induced migration and EMT.


Subject(s)
Active Transport, Cell Nucleus/physiology , Epithelial-Mesenchymal Transition/physiology , Mitogen-Activated Protein Kinase 14/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Transforming Growth Factor beta/genetics , Triple Negative Breast Neoplasms/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Cell Movement , Female , Humans , Imidazoles/pharmacology , Lymphoid Enhancer-Binding Factor 1/metabolism , Mitogen-Activated Protein Kinase 14/biosynthesis , Multiprotein Complexes/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phosphorylation , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Smad7 Protein/metabolism , Transcription Factor 4 , Transcription Factor 7-Like 1 Protein/metabolism , Transcription Factors/metabolism , beta Catenin/metabolism
14.
Nature ; 545(7654): 365-369, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28489822

ABSTRACT

The mechanistic target of rapamycin (mTOR) has a key role in the integration of various physiological stimuli to regulate several cell growth and metabolic pathways. mTOR primarily functions as a catalytic subunit in two structurally related but functionally distinct multi-component kinase complexes, mTOR complex 1 (mTORC1) and mTORC2 (refs 1, 2). Dysregulation of mTOR signalling is associated with a variety of human diseases, including metabolic disorders and cancer. Thus, both mTORC1 and mTORC2 kinase activity is tightly controlled in cells. mTORC1 is activated by both nutrients and growth factors, whereas mTORC2 responds primarily to extracellular cues such as growth-factor-triggered activation of PI3K signalling. Although both mTOR and GßL (also known as MLST8) assemble into mTORC1 and mTORC2 (refs 11, 12, 13, 14, 15), it remains largely unclear what drives the dynamic assembly of these two functionally distinct complexes. Here we show, in humans and mice, that the K63-linked polyubiquitination status of GßL dictates the homeostasis of mTORC2 formation and activation. Mechanistically, the TRAF2 E3 ubiquitin ligase promotes K63-linked polyubiquitination of GßL, which disrupts its interaction with the unique mTORC2 component SIN1 (refs 12, 13, 14) to favour mTORC1 formation. By contrast, the OTUD7B deubiquitinase removes polyubiquitin chains from GßL to promote GßL interaction with SIN1, facilitating mTORC2 formation in response to various growth signals. Moreover, loss of critical ubiquitination residues in GßL, by either K305R/K313R mutations or a melanoma-associated GßL(ΔW297) truncation, leads to elevated mTORC2 formation, which facilitates tumorigenesis, in part by activating AKT oncogenic signalling. In support of a physiologically pivotal role for OTUD7B in the activation of mTORC2/AKT signalling, genetic deletion of Otud7b in mice suppresses Akt activation and Kras-driven lung tumorigenesis in vivo. Collectively, our study reveals a GßL-ubiquitination-dependent switch that fine-tunes the dynamic organization and activation of the mTORC2 kinase under both physiological and pathological conditions.


Subject(s)
Carcinogenesis , Endopeptidases/metabolism , Multiprotein Complexes/metabolism , Signal Transduction , TNF Receptor-Associated Factor 2/metabolism , TOR Serine-Threonine Kinases/metabolism , Ubiquitin/metabolism , Ubiquitination , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Endopeptidases/deficiency , Endopeptidases/genetics , Enzyme Activation , Female , Homeostasis , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/chemistry , Phosphorylation , Polyubiquitin/metabolism , Protein Binding , Protein Subunits/chemistry , Protein Subunits/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/biosynthesis , TOR Serine-Threonine Kinases/chemistry , mTOR Associated Protein, LST8 Homolog
15.
Hum Mol Genet ; 26(3): 509-518, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28053051

ABSTRACT

Mutations in peripherin 2 (PRPH2), also known as retinal degeneration slow/RDS, lead to various retinal degenerations including retinitis pigmentosa (RP) and macular/pattern dystrophy (MD/PD). PRPH2-associated disease is often characterized by a phenotypic variability even within families carrying the same mutation, raising interest in potential modifiers. PRPH2 oligomerizes with its homologue rod outer segment (OS) membrane protein 1 (ROM1), and non-pathogenic PRPH2/ROM1 mutations, when present together, lead to digenic RP. We asked whether ROM1 could modify the phenotype of a PRPH2 mutation associated with a high degree of intrafamilial phenotypic heterogeneity: Y141C. In vitro, Y141C-Prph2 showed signs of retention in the endoplasmic reticulum (ER), however co-expression with Rom1 rescued this phenotype. In the heterozygous Y141C knockin mouse model (Prph2Y/+), Y141C-Prph2 and Rom1 formed abnormal complexes but were present at normal levels. Abnormal complexes were eliminated in the absence of Rom1 (Prph2Y/+/Rom1-/-) and total Prph2 levels were reduced to those found in the haploinsufficient Prph2+/- RP model. The biochemical changes had functional and structural consequences; while Prph2Y/+ animals exhibited a cone-rod electroretinogram defect, Prph2Y/+/Rom1-/- animals displayed a rod-dominant phenotype and OSs similar to those seen in the Prph2+/-. These data show that ablation of Rom1 results in the conversion of an MD/PD phenotype characterized by cone functional defects and the formation of abnormal Prph2/Rom1 complexes to an RP phenotype characterized by rod-dominant functional defects and reductions in total Prph2 protein. Thus one method by which ROM1 may act as a disease modifier is by contributing to the large variability in PRPH2-associated disease phenotypes.


Subject(s)
Peripherins/genetics , Retinal Degeneration/genetics , Retinitis Pigmentosa/genetics , Tetraspanins/genetics , Animals , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Eye Proteins , Gene Expression Regulation , Gene Knock-In Techniques , Humans , Macular Degeneration/genetics , Macular Degeneration/pathology , Mice , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Mutation , Pedigree , Peripherins/biosynthesis , Peripherins/chemistry , Phenotype , Photoreceptor Cells, Vertebrate/chemistry , Photoreceptor Cells, Vertebrate/metabolism , Protein Multimerization , Retinal Degeneration/pathology , Retinitis Pigmentosa/pathology , Tetraspanins/biosynthesis , Tetraspanins/chemistry
16.
J Cell Biochem ; 118(4): 924-933, 2017 04.
Article in English | MEDLINE | ID: mdl-27862254

ABSTRACT

Renal proximal tubule epithelial cells (PTECs) are known to reabsorb salts and small plasma proteins filtered through Bowman's capsule. Following acute kidney injury, PTECs assume some characteristics of hepatocytes in producing various plasma proteins. We now demonstrate that even at a resting state, a PTEC cell line, HK2 expresses mRNAs for and synthesizes and secretes plasma proteins in a complex with complement C3, an α2 -macroglobulin family chaperone, including albumin, transferrin, α1 -antitrypsin, α1 -antichymotrypsin, α2 -HS-glycoprotein, ceruloplasmin, haptoglobin, C1-inhibitor, secreted phosphoprotein-24, and insulin-like growth factor-1. When grown on transwell inserts, HK2 cells predominantly secrete (∼90%) plasma proteins into the apical side and a smaller fraction into the basolateral side as determined by ELISA assays. When cultured in the presence of exogenous cytokines such as IL1ß, IL6, TNFα, BMP2, or TGFß1, HK2 cell mRNA expressions for plasma proteins were variably affected whereas basolateral secretions were elevated to or in excess of those of the apical level. In addition, HK2 cells produce proTGFß1 with its intact N-terminal latency associated peptide and latent-TGF-ß-binding proteins. The complex cannot be dissociated under conditions of SDS, heating, and electrophoresis. Moreover, HK2 cells maintain their ability to quickly uptake exogenously added serum proteins from the culture medium, as if they are recognized differently by the endocytic receptors. These results provide new insight into the hepatization of PTECs. In addition to their unique uptake of plasma proteins and salts from the filtrate, they are a source of urinary proteins under normal conditions as wells as in chronic and acute kidney diseases. J. Cell. Biochem. 118: 924-933, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Blood Proteins/biosynthesis , Kidney Tubules, Proximal/metabolism , Biological Transport, Active , Blood Proteins/genetics , Cell Line , Cell Membrane/metabolism , Cell Polarity , Complement C3/biosynthesis , Complement C3/genetics , Cytokines/metabolism , Cytokines/pharmacology , Epithelial Cells/metabolism , Gene Expression , Hep G2 Cells , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/pharmacology , Kidney Tubules, Proximal/cytology , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/genetics , Proteolysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/genetics
17.
PLoS Genet ; 12(11): e1006409, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27812096

ABSTRACT

Polyploidization has crucial impacts on the evolution of different eukaryotic lineages including fungi, plants and animals. Recent genome data suggest that, for many polyploidization events, all duplicated chromosomes are maintained and genome reorganizations occur much later during evolution. However, newly-formed polyploid genomes are intrinsically unstable and often quickly degenerate into aneuploidy or diploidy. The transition between these two states remains enigmatic. In this study, laboratory evolution experiments were conducted to investigate this phenomenon. We show that robust tetraploidy is achieved in evolved yeast cells by increasing the abundance of Sch9-a protein kinase activated by the TORC1 (Target of Rapamycin Complex 1) and other signaling pathways. Overexpressing SCH9, but not TOR1, allows newly-formed tetraploids to exhibit evolved phenotypes and knocking out SCH9 diminishes the evolved phenotypes. Furthermore, when cells were challenged with conditions causing ancestral cells to evolve aneuploidy, tetraploidy was maintained in the evolved lines. Our results reveal a determinant role for Sch9 during the early stage of polyploid evolution.


Subject(s)
Multiprotein Complexes/genetics , Polyploidy , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , TOR Serine-Threonine Kinases/genetics , Aneuploidy , Diploidy , Directed Molecular Evolution , Gene Expression Regulation, Fungal , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Saccharomyces cerevisiae Proteins/biosynthesis , Signal Transduction/genetics , TOR Serine-Threonine Kinases/biosynthesis , Tetraploidy
18.
Nat Methods ; 13(12): 997-1000, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27749839

ABSTRACT

We present a baculovirus-based protein engineering method that enables site-specific introduction of unique functionalities in a eukaryotic protein complex recombinantly produced in insect cells. We demonstrate the versatility of this efficient and robust protein production platform, 'MultiBacTAG', (i) for the fluorescent labeling of target proteins and biologics using click chemistries, (ii) for glycoengineering of antibodies, and (iii) for structure-function studies of novel eukaryotic complexes using single-molecule Förster resonance energy transfer as well as site-specific crosslinking strategies.


Subject(s)
Green Fluorescent Proteins/biosynthesis , Multiprotein Complexes/biosynthesis , Protein Engineering/methods , Recombinant Proteins/biosynthesis , Viral Proteins/biosynthesis , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Cell Culture Techniques , Fluorescence Resonance Energy Transfer/methods , Genetic Code , Genetic Vectors , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Plasmids , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sf9 Cells , Spodoptera , Viral Proteins/chemistry , Viral Proteins/genetics
19.
Genes Dev ; 30(21): 2341-2344, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27881598

ABSTRACT

Macromolecular complexes, rather than individual biopolymers, perform many cellular activities. Faithful assembly of these complexes in vivo is therefore a vital challenge of all cells, and its failure can have fatal consequences. To form functional complexes, cells use elaborate measures to select the "right" components and combine them into working entities. How assembly is achieved at the molecular level is unclear in many cases. Three groups (Jin and colleagues, pp. 2391-2403; Xu and colleagues, pp. 2376-2390; and Tang and colleagues in Cell Research) have now provided insights into how an assembly factor specifically recognizes substrate RNA molecules and enables their usage for assembly of Sm-class uridine-rich small nuclear RNA-protein complexes.


Subject(s)
Models, Molecular , Multiprotein Complexes/biosynthesis , RNA, Small Nuclear/metabolism , SMN Complex Proteins/chemistry , SMN Complex Proteins/metabolism , Base Sequence , Multiprotein Complexes/chemistry , Protein Binding , Protein Domains , Protein Structure, Tertiary , RNA, Small Nuclear/chemistry , Sequence Alignment
20.
Anal Biochem ; 512: 110-113, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27558914

ABSTRACT

We report the construction of a versatile Gateway-based co-expression vector set for producing multiprotein complexes in Escherichia coli. The set consists of two groups of three vectors (pCoGW and pCo0GW), each having a specific antibiotic resistance gene, a compatible origin of replication and allowing cloning of up to two genes, each under control of its own T7 promoter. To validate the set, 33 (co-)expression plasmids encoding fluorescent protein (GFP, DsRed and ECFP) have been generated. Protein expression levels were quantified and (co-)expression visualized by fluorescent microscopy. The results illustrate the applicability of these vectors in co-expression studies.


Subject(s)
Escherichia coli , Gene Expression , Gene Library , Genetic Vectors/genetics , Green Fluorescent Proteins , Multiprotein Complexes , Escherichia coli/genetics , Escherichia coli/metabolism , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL