Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
J Cell Biol ; 223(1)2024 01 01.
Article in English | MEDLINE | ID: mdl-38032389

ABSTRACT

Nedd4-2 is an E3 ubiquitin ligase in which missense mutation is related to familial epilepsy, indicating its critical role in regulating neuronal network activity. However, Nedd4-2 substrates involved in neuronal network function have yet to be identified. Using mouse lines lacking Nedd4-1 and Nedd4-2, we identified astrocytic channel proteins inwardly rectifying K+ channel 4.1 (Kir4.1) and Connexin43 as Nedd4-2 substrates. We found that the expression of Kir4.1 and Connexin43 is increased upon conditional deletion of Nedd4-2 in astrocytes, leading to an elevation of astrocytic membrane ion permeability and gap junction activity, with a consequent reduction of γ-oscillatory neuronal network activity. Interestingly, our biochemical data demonstrate that missense mutations found in familial epileptic patients produce gain-of-function of the Nedd4-2 gene product. Our data reveal a process of coordinated astrocytic ion channel proteostasis that controls astrocyte function and astrocyte-dependent neuronal network activity and elucidate a potential mechanism by which aberrant Nedd4-2 function leads to epilepsy.


Subject(s)
Astrocytes , Cell Membrane Permeability , Connexin 43 , Nedd4 Ubiquitin Protein Ligases , Potassium Channels, Inwardly Rectifying , Animals , Humans , Mice , Connexin 43/genetics , Mutation, Missense , Proteostasis , Potassium Channels, Inwardly Rectifying/genetics , Nedd4 Ubiquitin Protein Ligases/genetics , Epilepsy
2.
Sci Adv ; 9(31): eadh2073, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37531430

ABSTRACT

Ubiquitin and ubiquitin-like conjugation cascades consist of dedicated E1, E2, and E3 enzymes with E3s providing substrate specificity. Mass spectrometry-based approaches have enabled the identification of more than 6500 SUMO2/3 target proteins. The limited number of SUMO E3s provides the unique opportunity to systematically study E3 substrate wiring. We developed SUMO-activated target traps (SATTs) and systematically identified substrates for eight different SUMO E3s, PIAS1, PIAS2, PIAS3, PIAS4, NSMCE2, ZNF451, LAZSUL (ZNF451-3), and ZMIZ2. SATTs enabled us to identify 427 SUMO1 and 961 SUMO2/3 targets in an E3-specific manner. We found pronounced E3 substrate preference. Quantitative proteomics enabled us to measure substrate specificity of E3s, quantified using the SATT index. Furthermore, we developed the Polar SATTs web-based tool to browse the dataset in an interactive manner. Overall, we uncover E3-to-target wiring of 1388 SUMO substrates, highlighting unique and overlapping sets of substrates for eight different SUMO E3 ligases.


Subject(s)
Proteome , Ubiquitin-Protein Ligases , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism
3.
Immunity ; 56(7): 1631-1648.e10, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37392737

ABSTRACT

CD137 (4-1BB)-activating receptor represents a promising cancer immunotherapeutic target. Yet, the cellular program driven by CD137 and its role in cancer immune surveillance remain unresolved. Using T cell-specific deletion and agonist antibodies, we found that CD137 modulates tumor infiltration of CD8+-exhausted T (Tex) cells expressing PD1, Lag-3, and Tim-3 inhibitory receptors. T cell-intrinsic, TCR-independent CD137 signaling stimulated the proliferation and the terminal differentiation of Tex precursor cells through a mechanism involving the RelA and cRel canonical NF-κB subunits and Tox-dependent chromatin remodeling. While Tex cell accumulation induced by prophylactic CD137 agonists favored tumor growth, anti-PD1 efficacy was improved with subsequent CD137 stimulation in pre-clinical mouse models. Better understanding of T cell exhaustion has crucial implications for the treatment of cancer and infectious diseases. Our results identify CD137 as a critical regulator of Tex cell expansion and differentiation that holds potential for broad therapeutic applications.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Mice , Animals , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Cell Differentiation , Cell Proliferation , Receptors, Antigen, T-Cell
4.
Front Immunol ; 13: 926714, 2022.
Article in English | MEDLINE | ID: mdl-35874734

ABSTRACT

Exhausted CD8+ T (Tex) cells are a distinct cell population that arise during persistent antigen exposure in the context of chronic infections and cancers. Although characterized by progressive loss of effector functions, high and sustained inhibitory receptor expression and distinct transcriptional and epigenetic programs, Tex cells are heterogeneous. Among these, a self-renewing TCF-1+ Tex population, having unique characteristics and the ability to respond to immune-checkpoint blockade, gives rise to TCF-1- terminally Tex cells. These TCF-1+ cells have stem cell-like properties similar to memory T cell populations, but the signals that regulate the developmental pathways and relationships among exhausted cell populations are still unclear. Here, we review our current understanding of Tex cell biology, and discuss some less appreciated molecules and pathways affecting T cell exhaustion. We highlight two co-stimulatory receptors, CD226 and CD137, and their role in inducing or restraining T cell exhaustion, as well as signaling pathways that may be amenable to pharmacological inhibition with a focus on Phosphoinositide-3 Kinase and IL-2 partial agonists. Finally, we discuss novel methods that may increase TCF-1+ populations and therefore improve immunotherapy responsiveness. Understanding features of and pathways to exhaustion has important implications for the success of immunotherapy, including checkpoint blockade and adoptive T-cell transfer therapies.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy , Humans , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy
5.
Genes Dev ; 35(15-16): 1142-1160, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34244292

ABSTRACT

The establishment of cell fates involves alterations of transcription factor repertoires and repurposing of transcription factors by post-translational modifications. In embryonic stem cells (ESCs), the chromatin organizers SATB2 and SATB1 balance pluripotency and differentiation by activating and repressing pluripotency genes, respectively. Here, we show that conditional Satb2 gene inactivation weakens ESC pluripotency, and we identify SUMO2 modification of SATB2 by the E3 ligase ZFP451 as a potential driver of ESC differentiation. Mutations of two SUMO-acceptor lysines of Satb2 (Satb2K →R ) or knockout of Zfp451 impair the ability of ESCs to silence pluripotency genes and activate differentiation-associated genes in response to retinoic acid (RA) treatment. Notably, the forced expression of a SUMO2-SATB2 fusion protein in either Satb2K →R or Zfp451-/- ESCs rescues, in part, their impaired differentiation potential and enhances the down-regulation of Nanog The differentiation defect of Satb2K →R ESCs correlates with altered higher-order chromatin interactions relative to Satb2wt ESCs. Upon RA treatment of Satb2wt ESCs, SATB2 interacts with ZFP451 and the LSD1/CoREST complex and gains binding at differentiation genes, which is not observed in RA-treated Satb2K →R cells. Thus, SATB2 SUMOylation may contribute to the rewiring of transcriptional networks and the chromatin interactome of ESCs in the transition of pluripotency to differentiation.


Subject(s)
Embryonic Stem Cells , Sumoylation , Ubiquitin-Protein Ligases/metabolism , Cell Differentiation/genetics , Chromatin/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Mol Cell ; 81(5): 1013-1026.e11, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33548202

ABSTRACT

In response to stress, human cells coordinately downregulate transcription and translation of housekeeping genes. To downregulate transcription, the negative elongation factor (NELF) is recruited to gene promoters impairing RNA polymerase II elongation. Here we report that NELF rapidly forms nuclear condensates upon stress in human cells. Condensate formation requires NELF dephosphorylation and SUMOylation induced by stress. The intrinsically disordered region (IDR) in NELFA is necessary for nuclear NELF condensation and can be functionally replaced by the IDR of FUS or EWSR1 protein. We find that biomolecular condensation facilitates enhanced recruitment of NELF to promoters upon stress to drive transcriptional downregulation. Importantly, NELF condensation is required for cellular viability under stressful conditions. We propose that stress-induced NELF condensates reported here are nuclear counterparts of cytosolic stress granules. These two stress-inducible condensates may drive the coordinated downregulation of transcription and translation, likely forming a critical node of the stress survival strategy.


Subject(s)
Heat-Shock Response/genetics , Intrinsically Disordered Proteins/genetics , Protein Processing, Post-Translational , RNA Polymerase II/genetics , Transcription, Genetic , Transcriptional Elongation Factors/genetics , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Chromatin/chemistry , Chromatin/metabolism , Clone Cells , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Genes, Reporter , HEK293 Cells , HeLa Cells , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Phosphorylation , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Signal Transduction , Stress, Physiological , Sumoylation , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/metabolism , Red Fluorescent Protein
8.
Immunity ; 53(4): 824-839.e10, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053331

ABSTRACT

CD8+ T cells within the tumor microenvironment (TME) are exposed to various signals that ultimately determine functional outcomes. Here, we examined the role of the co-activating receptor CD226 (DNAM-1) in CD8+ T cell function. The absence of CD226 expression identified a subset of dysfunctional CD8+ T cells present in peripheral blood of healthy individuals. These cells exhibited reduced LFA-1 activation, altered TCR signaling, and a distinct transcriptomic program upon stimulation. CD226neg CD8+ T cells accumulated in human and mouse tumors of diverse origin through an antigen-specific mechanism involving the transcriptional regulator Eomesodermin (Eomes). Despite similar expression of co-inhibitory receptors, CD8+ tumor-infiltrating lymphocyte failed to respond to anti-PD-1 in the absence of CD226. Immune checkpoint blockade efficacy was hampered in Cd226-/- mice. Anti-CD137 (4-1BB) agonists also stimulated Eomes-dependent CD226 loss that limited the anti-tumor efficacy of this treatment. Thus, CD226 loss restrains CD8+ T cell function and limits the efficacy of cancer immunotherapy.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , T-Box Domain Proteins/immunology , Animals , Humans , Immune Checkpoint Inhibitors/immunology , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Transcriptome/immunology , Tumor Microenvironment/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
9.
Methods Mol Biol ; 1934: 223-233, 2019.
Article in English | MEDLINE | ID: mdl-31256382

ABSTRACT

Protein regulation by reversible attachment of SUMO (small ubiquitin-related modifier) plays an important role in several cellular processes such as transcriptional regulation, nucleo-cytoplasmic transport, cell-cycle progression, meiosis, and DNA repair. However, most sumoylated proteins are of marginal abundance at steady state levels, which is due to strict regulation and/or rapid turnover of modification and de-modification. Consequently, analysis of protein sumoylation in vivo is very challenging. Nonetheless, a novel method was established that allows detection of sumoylated proteins at endogenous levels from vertebrate cells and tissues. This approach involves the enrichment of sumoylated proteins by immunoprecipitation followed by peptide elution. After endogenous substrate sumoylation is verified, addressing its functional consequences is the next logical step. This requires SUMO site mapping that benefits from larger quantities of modified protein. Here, we shortly describe strategies to achieve efficient in vitro sumoylation of many substrates.


Subject(s)
Small Ubiquitin-Related Modifier Proteins/metabolism , Chromatography, Affinity , Humans , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/chemistry , Small Ubiquitin-Related Modifier Proteins/isolation & purification , Substrate Specificity , Sumoylation
10.
JCI Insight ; 52019 06 13.
Article in English | MEDLINE | ID: mdl-31194697

ABSTRACT

Immunotherapy holds promise for multiple myeloma (MM) patients but little is known about how MM-induced immunosuppression influences response to therapy. Here, we investigated the impact of disease progression on immunotherapy efficacy in the Vk*MYC mouse model. Treatment with agonistic anti-CD137 (4-1BB) mAbs efficiently protected mice when administered early but failed to contain MM growth when delayed more than three weeks after Vk*MYC tumor cell challenge. The quality of CD8+ T cell response to CD137 stimulation was not altered by the presence of MM, but CD8+ T cell numbers were profoundly reduced at the time of treatment. Our data suggest that an insufficient ratio of CD8+ T cells over MM cells (CD8/MM) accounts for the loss of anti-CD137 mAb efficacy. We established serum M-protein levels prior to therapy as a predictive factor of response. Moreover, we developed an in silico model to capture the dynamic interactions between CD8+ T cells and MM cells. Finally, we explored two methods to improve the CD8/MM ratio: anti-CD137 mAb immunotherapy combined with Treg-depletion or administered after chemotherapy treatment with cyclophosphamide or melphalan efficiently reduced MM burden and prolonged survival. Altogether, our data indicate that consolidation treatment with anti-CD137 mAbs might prevent MM relapse.


Subject(s)
4-1BB Ligand/metabolism , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Immunotherapy/methods , Multiple Myeloma/drug therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Myeloma/pathology , T-Lymphocytes, Regulatory
11.
Methods Enzymol ; 618: 167-185, 2019.
Article in English | MEDLINE | ID: mdl-30850051

ABSTRACT

The small ubiquitin-related modifier (SUMO) is a protein of ~10kDa that is covalently conjugated to its substrate proteins in an enzymatic process called sumoylation. This posttranslational modification is an essential regulatory mechanism that plays crucial roles in many cellular pathways. It allows rapid adaptation to environmental changes by switching protein functions due to alternate complex assemblies, changes in intracellular localization, enzymatic activity, or stability. SUMO conjugation is executed by the hierarchical action of E1, E2, and E3 enzymes. Both E2 and E3 enzymes contribute to substrate specificity but with E3 ligases being the more important for this. E1 and E2 activities are essential for all sumoylation reactions but usually-with a few exceptions-modify substrates only inefficiently. Hence, most substrates require the additional action of an E3 ligase or a cofactor. Here, we describe methods to distinguish a bona fide E3 ligase from a cofactor activity by using in vitro sumoylation assays.


Subject(s)
Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Enzyme Assays/methods , Humans , Sumoylation
13.
Biomol Concepts ; 8(1): 13-36, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28284030

ABSTRACT

The regulation of protein fate by modification with the small ubiquitin-related modifier (SUMO) plays an essential and crucial role in most cellular pathways. Sumoylation is highly dynamic due to the opposing activities of SUMO conjugation and SUMO deconjugation. SUMO conjugation is performed by the hierarchical action of E1, E2 and E3 enzymes, while its deconjugation involves SUMO-specific proteases. In this review, we summarize and compare the mechanistic principles of how SUMO gets conjugated to its substrate. We focus on the interplay of the E1, E2 and E3 enzymes and discuss how specificity could be achieved given the limited number of conjugating enzymes and the thousands of substrates.


Subject(s)
Sumoylation , SUMO-1 Protein/metabolism , Substrate Specificity , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
15.
Methods Mol Biol ; 1475: 67-78, 2016.
Article in English | MEDLINE | ID: mdl-27631798

ABSTRACT

Protein modification with the small ubiquitin-related modifier SUMO is a potent regulatory mechanism implicated in a variety of biological pathways. In vitro sumoylation reactions have emerged as a versatile tool to identify and characterize novel SUMO enzymes as well as their substrates. Here, we present detailed protocols for the purification and fluorescent labeling of mammalian SUMO paralogs for their application in sumoylation assays. These assays provide a fast readout for in vitro SUMO chain formation activity of E3 ligases in a paralog-specific manner. Finally, we critically analyze the application of fluorescent SUMO proteins to study substrate modification in vitro revealing also the drawbacks of the system.


Subject(s)
Biological Assay , Protein Processing, Post-Translational , SUMO-1 Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitins/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Fluorescent Dyes/chemistry , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Protein Binding , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , SUMO-1 Protein/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Spectrometry, Fluorescence , Staining and Labeling/methods , Sumoylation , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitins/genetics
16.
Int J Biochem Cell Biol ; 79: 478-487, 2016 10.
Article in English | MEDLINE | ID: mdl-27343429

ABSTRACT

The small ubiquitin related modifier SUMO regulates protein functions to maintain cell homeostasis. SUMO attachment is executed by the hierarchical action of E1, E2 and E3 enzymes of which E3 ligases ensure substrate specificity. We recently identified the ZNF451 family as novel class of SUMO2/3 specific E3 ligases and characterized their function in SUMO chain formation. The founding member, ZNF451isoform1 (ZNF451-1) partially resides in PML bodies, nuclear structures organized by the promyelocytic leukemia gene product PML. As PML and diverse PML components are well known SUMO substrates the question arises whether ZNF451-1 is involved in their sumoylation. Here, we show that ZNF451-1 indeed functions as SUMO2/3 specific E3 ligase for PML and selected PML components in vitro. Mutational analysis indicates that substrate sumoylation employs an identical biochemical mechanism as we described for SUMO chain formation. In vivo, ZNF451-1 RNAi depletion leads to PML stabilization and an increased number of PML bodies. By contrast, PML degradation upon arsenic trioxide treatment is not ZNF451-1 dependent. Our data suggest a regulatory role of ZNF451-1 in fine-tuning physiological PML levels in a RNF4 cooperative manner in the mouse neuroblastoma N2a cell-line.


Subject(s)
Cell Nucleus/metabolism , Promyelocytic Leukemia Protein/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism , Amino Acid Sequence , Aminoacyltransferases , Animals , Cell Line, Tumor , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Substrate Specificity , Transcription Factors/chemistry , Transcription Factors/deficiency , Transcription Factors/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Zinc Fingers
17.
Cell Rep ; 15(7): 1481-1492, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27160902

ABSTRACT

The HECT domain E3 ligase HACE1 has been identified as a tumor suppressor in multiple cancers. Here, we report that HACE1 is a central gatekeeper of TNFR1-induced cell fate. Genetic inactivation of HACE1 inhibits TNF-stimulated NF-κB activation and TNFR1-NF-κB-dependent pathogen clearance in vivo. Moreover, TNF-induced apoptosis was impaired in hace1 mutant cells and knockout mice in vivo. Mechanistically, HACE1 is essential for the ubiquitylation of the adaptor protein TRAF2 and formation of the apoptotic caspase-8 effector complex. Intriguingly, loss of HACE1 does not impair TNFR1-mediated necroptotic cell fate via RIP1 and RIP3 kinases. Loss of HACE1 predisposes animals to colonic inflammation and carcinogenesis in vivo, which is markedly alleviated by genetic inactivation of RIP3 kinase and TNFR1. Thus, HACE1 controls TNF-elicited cell fate decisions and exerts tumor suppressor and anti-inflammatory activities via a TNFR1-RIP3 kinase-necroptosis pathway.


Subject(s)
Cell Lineage , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/drug effects , Caspase 8/metabolism , Cell Lineage/drug effects , Colitis/metabolism , Colitis/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dextran Sulfate , Embryo, Mammalian/cytology , Enzyme Activation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Deletion , Mice, Inbred C57BL , Mutation/genetics , NF-kappa B/metabolism , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Factor 2/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitination/drug effects
18.
Nat Struct Mol Biol ; 22(12): 959-67, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26524493

ABSTRACT

SUMO chains act as stress-induced degradation tags or repair factor-recruiting signals at DNA lesions. Although E1 activating, E2 conjugating and E3 ligating enzymes efficiently assemble SUMO chains, specific chain-elongation mechanisms are unknown. E4 elongases are specialized E3 ligases that extend a chain but are inefficient in the initial conjugation of the modifier. We identified ZNF451, a representative member of a new class of SUMO2 and SUMO3 (SUMO2/3)-specific enzymes that execute catalysis via a tandem SUMO-interaction motif (SIM) region. One SIM positions the donor SUMO while a second SIM binds SUMO on the back side of the E2 enzyme. This tandem-SIM region is sufficient to extend a back side-anchored SUMO chain (E4 elongase activity), whereas efficient chain initiation also requires a zinc-finger region to recruit the initial acceptor SUMO (E3 ligase activity). Finally, we describe four human proteins sharing E4 elongase activities and their function in stress-induced SUMO2/3 conjugation.


Subject(s)
Protein Multimerization , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Humans , Vertebrates
19.
Nat Struct Mol Biol ; 22(12): 968-75, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26524494

ABSTRACT

E3 protein ligases enhance transfer of ubiquitin-like (Ubl) proteins from E2 conjugating enzymes to substrates by stabilizing the thioester-charged E2~Ubl in a closed configuration optimally aligned for nucleophilic attack. Here, we report biochemical and structural data that define the N-terminal domain of the Homo sapiens ZNF451 as the catalytic module for SUMO E3 ligase activity. The ZNF451 catalytic module contains tandem SUMO-interaction motifs (SIMs) bridged by a Pro-Leu-Arg-Pro (PLRP) motif. The first SIM and PLRP motif engage thioester-charged E2~SUMO while the next SIM binds a second molecule of SUMO bound to the back side of E2. We show that ZNF451 is SUMO2 specific and that SUMO modification of ZNF451 may contribute to activity by providing a second molecule of SUMO that interacts with E2. Our results are consistent with ZNF451 functioning as a bona fide SUMO E3 ligase.


Subject(s)
SUMO-1 Protein/chemistry , SUMO-1 Protein/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Aminoacyltransferases , Binding Sites , Crystallography, X-Ray , Humans , Protein Binding , Protein Conformation
20.
Neuromolecular Med ; 15(4): 639-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23990202

ABSTRACT

Posttranslational modification of proteins by the small ubiquitin-like modifier (SUMO) is a potent regulator of various cellular events. Hundreds of substrates have been identified, many of them involved in vital processes like transcriptional regulation, signal transduction, protein degradation, cell cycle regulation, DNA repair, chromatin organization, and nuclear transport. In recent years, protein sumoylation increasingly attracted attention, as it could be linked to heart failure, cancer, and neurodegeneration. However, underlying mechanisms involving how modification by SUMO contributes to disease development are still scarce thus necessitating further research. This review aims to critically discuss currently available concepts of the SUMO pathway, thereby highlighting regulation in the healthy versus diseased organism, focusing on neurologic aspects. Better understanding of differential regulation in health and disease may finally allow to uncover pathogenic mechanisms and contribute to the development of disease-specific therapies.


Subject(s)
Nerve Tissue Proteins/physiology , Nervous System Physiological Phenomena/physiology , Small Ubiquitin-Related Modifier Proteins/physiology , Sumoylation/physiology , Amino Acid Motifs , Animals , Cell Hypoxia , Consensus Sequence , Cysteine Endopeptidases/physiology , DNA Repair , Gene Expression Regulation , Humans , Metabolic Networks and Pathways , Models, Molecular , Nerve Degeneration , Nervous System Diseases/metabolism , Protein Conformation , Ubiquitin-Protein Ligase Complexes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL