Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
FEBS Lett ; 597(20): 2534-2545, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37620293

ABSTRACT

The nuclear pore complex (NPC) is among the most elaborate protein complexes in eukaryotes. While ribosomes and proteasomes are known to require dedicated assembly machinery, our understanding of NPC assembly is at a relatively early stage. Defects in NPC assembly or homeostasis are tied to movement disorders, including dystonia and amyotrophic lateral sclerosis (ALS), as well as aging, requiring a better understanding of these processes to enable therapeutic intervention. Here, we discuss recent progress in the understanding of NPC assembly and highlight how related defects in human disorders can shed light on NPC biogenesis. We propose that the condensation of phenylalanine-glycine repeat nucleoporins needs to be carefully controlled during NPC assembly to prevent aberrant condensation, aggregation, or amyloid formation.

2.
Cell Rep ; 41(8): 111675, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36417855

ABSTRACT

Many human diseases are caused by mutations in nuclear envelope (NE) proteins. How protein homeostasis and disease etiology are interconnected at the NE is poorly understood. Specifically, the identity of local ubiquitin ligases that facilitate ubiquitin-proteasome-dependent NE protein turnover is presently unknown. Here, we employ a short-lived, Lamin B receptor disease variant as a model substrate in a genetic screen to uncover key elements of NE protein turnover. We identify the ubiquitin-conjugating enzymes (E2s) Ube2G2 and Ube2D3, the membrane-resident ubiquitin ligases (E3s) RNF5 and HRD1, and the poorly understood protein TMEM33. RNF5, but not HRD1, requires TMEM33 both for efficient biosynthesis and function. Once synthesized, RNF5 responds dynamically to increased substrate levels at the NE by departing from the endoplasmic reticulum, where HRD1 remains confined. Thus, mammalian protein quality control machinery partitions between distinct cellular compartments to address locally changing substrate loads, establishing a robust cellular quality control system.


Subject(s)
Membrane Proteins , Ubiquitin-Protein Ligases , Animals , Humans , Ubiquitin-Protein Ligases/metabolism , Membrane Proteins/metabolism , Endoplasmic Reticulum/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin/metabolism , Mammals/metabolism
3.
Nat Cell Biol ; 24(11): 1630-1641, 2022 11.
Article in English | MEDLINE | ID: mdl-36302970

ABSTRACT

DYT1 dystonia is a debilitating neurological movement disorder arising from mutation in the AAA+ ATPase TorsinA. The hallmark of Torsin dysfunction is nuclear envelope blebbing resulting from defects in nuclear pore complex biogenesis. Whether blebs actively contribute to disease manifestation is unknown. We report that FG-nucleoporins in the bleb lumen form aberrant condensates and contribute to DYT1 dystonia by provoking two proteotoxic insults. Short-lived ubiquitylated proteins that are normally rapidly degraded partition into the bleb lumen and become stabilized. In addition, blebs selectively sequester a specific HSP40-HSP70 chaperone network that is modulated by the bleb component MLF2. MLF2 suppresses the ectopic accumulation of FG-nucleoporins and modulates the selective properties and size of condensates in vitro. Our study identifies dual mechanisms of proteotoxicity in the context of condensate formation and establishes FG-nucleoporin-directed activities for a nuclear chaperone network.


Subject(s)
Dystonia , Nuclear Envelope , Humans , Dystonia/metabolism , Nuclear Envelope/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism
4.
Sci Rep ; 12(1): 11233, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35787659

ABSTRACT

How does the mind organize thoughts? The hippocampal-entorhinal complex is thought to support domain-general representation and processing of structural knowledge of arbitrary state, feature and concept spaces. In particular, it enables the formation of cognitive maps, and navigation on these maps, thereby broadly contributing to cognition. It has been proposed that the concept of multi-scale successor representations provides an explanation of the underlying computations performed by place and grid cells. Here, we present a neural network based approach to learn such representations, and its application to different scenarios: a spatial exploration task based on supervised learning, a spatial navigation task based on reinforcement learning, and a non-spatial task where linguistic constructions have to be inferred by observing sample sentences. In all scenarios, the neural network correctly learns and approximates the underlying structure by building successor representations. Furthermore, the resulting neural firing patterns are strikingly similar to experimentally observed place and grid cell firing patterns. We conclude that cognitive maps and neural network-based successor representations of structured knowledge provide a promising way to overcome some of the short comings of deep learning towards artificial general intelligence.


Subject(s)
Grid Cells , Language , Cognition , Models, Neurological , Neural Networks, Computer
5.
Int J Mol Sci ; 23(9)2022 Apr 21.
Article in English | MEDLINE | ID: mdl-35563018

ABSTRACT

DYT1 dystonia is a debilitating neurological movement disorder that arises upon Torsin ATPase deficiency. Nuclear envelope (NE) blebs that contain FG-nucleoporins (FG-Nups) and K48-linked ubiquitin are the hallmark phenotype of Torsin manipulation across disease models of DYT1 dystonia. While the aberrant deposition of FG-Nups is caused by defective nuclear pore complex assembly, the source of K48-ubiquitylated proteins inside NE blebs is not known. Here, we demonstrate that the characteristic K48-ubiquitin accumulation inside blebs requires p97 activity. This activity is highly dependent on the p97 adaptor UBXD1. We show that p97 does not significantly depend on the Ufd1/Npl4 heterodimer to generate the K48-ubiquitylated proteins inside blebs, nor does inhibiting translation affect the ubiquitin sequestration in blebs. However, stimulating global ubiquitylation by heat shock greatly increases the amount of K48-ubiquitin sequestered inside blebs. These results suggest that blebs have an extraordinarily high capacity for sequestering ubiquitylated protein generated in a p97-dependent manner. The p97/UBXD1 axis is thus a major factor contributing to cellular DYT1 dystonia pathology and its modulation represents an unexplored potential for therapeutic development.


Subject(s)
Adaptor Proteins, Vesicular Transport , Adenosine Triphosphatases , Autophagy-Related Proteins , Dystonia , Nuclear Envelope , Nuclear Proteins , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Adenosine Triphosphatases/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Cell Membrane Structures/metabolism , Dystonia/genetics , Dystonia/metabolism , Dystonia Musculorum Deformans , Humans , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Nuclear Envelope/metabolism , Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ubiquitin/metabolism
6.
J Biol Chem ; 297(2): 100937, 2021 08.
Article in English | MEDLINE | ID: mdl-34224731

ABSTRACT

The endoplasmic reticulum (ER) is a membrane-bound organelle responsible for protein folding, lipid synthesis, and calcium homeostasis. Maintenance of ER structural integrity is crucial for proper function, but much remains to be learned about the molecular players involved. To identify proteins that support the structure of the ER, we performed a proteomic screen and identified nodal modulator (NOMO), a widely conserved type I transmembrane protein of unknown function, with three nearly identical orthologs specified in the human genome. We found that overexpression of NOMO1 imposes a sheet morphology on the ER, whereas depletion of NOMO1 and its orthologs causes a collapse of ER morphology concomitant with the formation of membrane-delineated holes in the ER network positive for the lysosomal marker lysosomal-associated protein 1. In addition, the levels of key players of autophagy including microtubule-associated protein light chain 3 and autophagy cargo receptor p62/sequestosome 1 strongly increase upon NOMO depletion. In vitro reconstitution of NOMO1 revealed a "beads on a string" structure likely representing consecutive immunoglobulin-like domains. Extending NOMO1 by insertion of additional immunoglobulin folds results in a correlative increase in the ER intermembrane distance. Based on these observations and a genetic epistasis analysis including the known ER-shaping proteins Atlastin2 and Climp63, we propose a role for NOMO1 in the functional network of ER-shaping proteins.


Subject(s)
Endoplasmic Reticulum , Proteomics , Sequestosome-1 Protein , Autophagy , Endoplasmic Reticulum Stress , Homeostasis , Humans , Lysosomes/metabolism
7.
Mol Biol Cell ; 31(13): 1315-1323, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32530796

ABSTRACT

The nuclear envelope (NE) is continuous with the endoplasmic reticulum (ER), yet the NE carries out many functions distinct from those of bulk ER. This functional specialization depends on a unique protein composition that defines NE identity and must be both established and actively maintained. The NE undergoes extensive remodeling in interphase and mitosis, so mechanisms that seal NE holes and protect its unique composition are critical for maintaining its functions. New evidence shows that closure of NE holes relies on regulated de novo lipid synthesis, providing a link between lipid metabolism and generating and maintaining NE identity. Here, we review regulation of the lipid bilayers of the NE and suggest ways to generate lipid asymmetry across the NE despite its direct continuity with the ER. We also discuss the elusive mechanism of membrane fusion during nuclear pore complex (NPC) biogenesis. We propose a model in which NPC biogenesis is carefully controlled to ensure that a permeability barrier has been established before membrane fusion, thereby avoiding a major threat to compartmentalization.


Subject(s)
Lipid Bilayers/metabolism , Lipid Metabolism , Nuclear Envelope/metabolism , Animals , Cell Cycle , Humans , Membrane Fusion , Mitosis , Nuclear Envelope/physiology
8.
J Cell Biol ; 219(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32342107

ABSTRACT

Nuclear envelope herniations (blebs) containing FG-nucleoporins and ubiquitin are the phenotypic hallmark of Torsin ATPase manipulation. Both the dynamics of blebbing and the connection to nuclear pore biogenesis remain poorly understood. We employ a proteomics-based approach to identify myeloid leukemia factor 2 (MLF2) as a luminal component of the bleb. Using an MLF2-based live-cell imaging platform, we demonstrate that nuclear envelope blebbing occurs rapidly and synchronously immediately after nuclear envelope reformation during mitosis. Bleb formation is independent of ubiquitin conjugation within the bleb, but strictly dependent on POM121, a transmembrane nucleoporin essential for interphase nuclear pore biogenesis. Nup358, a late marker for interphase nuclear pore complex (NPC) biogenesis, is underrepresented relative to FG-nucleoporins in nuclear envelopes of Torsin-deficient cells. The kinetics of bleb formation, its dependence on POM121, and a reduction of mature NPCs in Torsin-deficient cells lead us to conclude that the hallmark phenotype of Torsin manipulation represents aberrant NPC intermediates.


Subject(s)
Molecular Chaperones/metabolism , Nuclear Envelope/metabolism , Nuclear Pore/metabolism , Nuclear Proteins/metabolism , Gene Knockout Techniques , HeLa Cells , Humans , Interphase/genetics , Interphase/physiology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/metabolism , Microscopy, Electron, Transmission , Mitosis/genetics , Mitosis/physiology , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Nuclear Envelope/genetics , Nuclear Envelope/ultrastructure , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins/genetics , RNA, Small Interfering , Ubiquitin/chemistry , Ubiquitin/metabolism
9.
Biomolecules ; 10(3)2020 03 19.
Article in English | MEDLINE | ID: mdl-32204310

ABSTRACT

Torsin ATPases are members of the AAA+ (ATPases associated with various cellular activities) superfamily of proteins, which participate in essential cellular processes. While AAA+ proteins are ubiquitously expressed and demonstrate distinct subcellular localizations, Torsins are the only AAA+ to reside within the nuclear envelope (NE) and endoplasmic reticulum (ER) network. Moreover, due to the absence of integral catalytic features, Torsins require the NE- and ER-specific regulatory cofactors, lamina-associated polypeptide 1 (LAP1) and luminal domain like LAP1 (LULL1), to efficiently trigger their atypical mode of ATP hydrolysis. Despite their implication in an ever-growing list of diverse processes, the specific contributions of Torsin/cofactor assemblies in maintaining normal cellular physiology remain largely enigmatic. Resolving gaps in the functional and mechanistic principles of Torsins and their cofactors are of considerable medical importance, as aberrant Torsin behavior is the principal cause of the movement disorder DYT1 early-onset dystonia. In this review, we examine recent findings regarding the phenotypic consequences of compromised Torsin and cofactor activities. In particular, we focus on the molecular features underlying NE defects and the contributions of Torsins to nuclear pore complex biogenesis, as well as the growing implications of Torsins in cellular lipid metabolism. Additionally, we discuss how understanding Torsins may facilitate the study of essential but poorly understood processes at the NE and ER, and aid in the development of therapeutic strategies for dystonia.


Subject(s)
Dystonia/metabolism , Endoplasmic Reticulum/metabolism , HSC70 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Nuclear Envelope/metabolism , Animals , Dystonia/genetics , Dystonia/pathology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , HSC70 Heat-Shock Proteins/genetics , Humans , Molecular Chaperones/genetics , Nuclear Envelope/genetics , Nuclear Envelope/pathology
10.
J Clin Invest ; 129(11): 4576-4579, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31589164

ABSTRACT

Mutations affecting the integrity of the essential torsin ATPase/cofactor system have been identified in a steadily increasing number of congenital disorders. Since most of these mutations affect brain function, much of the research has focused on deciphering disease etiology in the brain. However, torsin is expressed in a wide variety of nonneural tissues and is strictly conserved across species, including the lowest metazoans, suggesting that it plays roles extending beyond neurons. In this issue of the JCI, Shin et al. explored torsin function in the mammalian liver. The group reports major defects in hepatic lipid metabolism when the torsin system is compromised in mice. Remarkably, conditional deletion of either torsinA or its cofactor, lamina-associated polypeptide 1 (LAP1), resulted in fatty liver disease and steatohepatitis, likely from a secretion defect of VLDLs. This study considerably expands our understanding of torsin biology, while providing defined opportunities for future investigations of torsin function and dysfunction in human pathologies.


Subject(s)
Fatty Liver , Nuclear Envelope , Animals , Humans , Membrane Proteins , Mice , Peptides
11.
Methods Enzymol ; 619: 47-69, 2019.
Article in English | MEDLINE | ID: mdl-30910029

ABSTRACT

Lamin B receptor (LBR) is an inner nuclear membrane protein that associates with the nuclear lamina and harbors sterol reductase activity essential for cholesterol biosynthesis. Several LBR mutations implicated in human congenital disorders give rise to C-terminal truncations which render LBR metabolically unstable, resulting in their rapid turnover in the nucleus. These LBR variants serve as model substrates for investigating the poorly understood protein quality control pathways in the mammalian nuclear envelope (NE). Here we describe a split-GFP-based method for tagging these model substrates to enable live cell imaging and flow cytometry for the identification and characterization of NE-resident protein turnover machinery. Furthermore, we describe a facile subcellular fractionation method to isolate a soluble LBR degradation intermediate, allowing the deconvolution of the membrane extraction and proteasomal turnover steps. The combination of imaging-based and biochemical approaches described here facilitates detailed mechanistic studies to dissect protein turnover in the nuclear compartment.


Subject(s)
Nuclear Envelope/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cell Fractionation/methods , Cell Line , Cell Nucleus/metabolism , Flow Cytometry/methods , Fluorescent Antibody Technique/methods , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/metabolism , Humans , Optical Imaging/methods , Receptors, Cytoplasmic and Nuclear/analysis , Lamin B Receptor
12.
Mol Biol Cell ; 28(21): 2765-2772, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28814508

ABSTRACT

TorsinA is an essential AAA+ ATPase requiring LAP1 or LULL1 as cofactors. The dynamics of the Torsin/cofactor system remain poorly understood, with previous models invoking Torsin/cofactor assemblies with fixed stoichiometries. Here we demonstrate that TorsinA assembles into homotypic oligomers in the presence of ATP. Torsin variants mutated at the "back" interface disrupt homo-oligomerization but still show robust ATPase activity in the presence of its cofactors. These Torsin mutants are severely compromised in their ability to rescue nuclear envelope defects in Torsin-deficient cells, suggesting that TorsinA homo-oligomers play a key role in vivo. Engagement of the oligomer by LAP1 triggers ATP hydrolysis and rapid complex disassembly. Thus the Torsin complex is a highly dynamic assembly whose oligomeric state is tightly controlled by distinctively localized cellular cofactors. Our discovery that LAP1 serves as a modulator of the oligomeric state of an AAA+ protein establishes a novel means of regulating this important class of oligomeric ATPases.


Subject(s)
HSC70 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Adenosine Triphosphatases/metabolism , Carrier Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Hydrolysis , Membrane Proteins/metabolism , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Mutation , Nuclear Envelope/metabolism
13.
Front Mol Biosci ; 4: 29, 2017.
Article in English | MEDLINE | ID: mdl-28553638

ABSTRACT

Torsins are essential, disease-relevant AAA+ (ATPases associated with various cellular activities) proteins residing in the endoplasmic reticulum and perinuclear space, where they are implicated in a variety of cellular functions. Recently, new structural and functional details about Torsins have emerged that will have a profound influence on unraveling the precise mechanistic details of their yet-unknown mode of action in the cell. While Torsins are phylogenetically related to Clp/HSP100 proteins, they exhibit comparatively weak ATPase activities, which are tightly controlled by virtue of an active site complementation through accessory cofactors. This control mechanism is offset by a TorsinA mutation implicated in the severe movement disorder DYT1 dystonia, suggesting a critical role for the functional Torsin-cofactor interplay in vivo. Notably, TorsinA lacks aromatic pore loops that are both conserved and critical for the processive unfolding activity of Clp/HSP100 proteins. Based on these distinctive yet defining features, we discuss how the apparent dynamic nature of the Torsin-cofactor system can inform emerging models and hypotheses for Torsin complex formation and function. Specifically, we propose that the dynamic assembly and disassembly of the Torsin/cofactor system is a critical property that is required for Torsins' functional roles in nuclear trafficking and nuclear pore complex assembly or homeostasis that merit further exploration. Insights obtained from these future studies will be a valuable addition to our understanding of disease etiology of DYT1 dystonia.

14.
Rare Dis ; 4(1): e1241363, 2016.
Article in English | MEDLINE | ID: mdl-27830109

ABSTRACT

Lamin B Receptor (LBR) is an inner nuclear membrane protein associated with the rare human diseases Pelger-Huët anomaly and Greenberg skeletal dysplasia. A new study has used CRISPR/Cas9-mediated genetic manipulations in a human cell system to determine that the molecular etiology of these previously poorly understood disorders is a defect in cholesterol synthesis due to loss of LBR-associated sterol C14 reductase activity. The study furthermore determined that disease-associated LBR point mutations reduce sterol C14 reductase activity by decreasing the affinity of LBR for the reducing agent NADPH. Moreover, two disease-associated LBR truncation mutants were found to be highly unstable at the protein level and are rapidly turned over by a novel nuclear membrane-based protein quality control pathway. Thus, truncated LBR variants can now be used as model substrates for further investigations of nuclear protein quality control to uncover possible implications for other disease-associated nuclear envelopathies.

15.
Mol Biol Cell ; 27(25): 3964-3971, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27798237

ABSTRACT

The human genome encodes four Torsin ATPases, the functions of which are poorly understood. In this study, we use CRISPR/Cas9 engineering to delete all four Torsin ATPases individually and in combination. Using nuclear envelope (NE) blebbing as a phenotypic measure, we establish a direct correlation between the number of inactivated Torsin alleles and the occurrence of omega-shaped herniations within the lumen of the NE. A similar, although not identical, redundancy is observed for LAP1 and LULL1, which serve as regulatory cofactors for a subset of Torsin ATPases. Unexpectedly, deletion of Tor2A in a TorA/B/3A-deficient background results in a stark increase of bleb formation, even though Tor2A does not respond to LAP1/LULL1 stimulation. The robustness of the observed phenotype in Torsin-deficient cells enables a structural analysis via electron microscopy tomography and a compositional analysis via immunogold labeling. Ubiquitin and nucleoporins were identified as distinctively localizing components of the omega-shaped bleb structure. These findings suggest a functional link between the Torsin/cofactor system and NE/nuclear pore complex biogenesis or homeostasis and establish a Torsin-deficient cell line as a valuable experimental platform with which to decipher Torsin function.


Subject(s)
Intercellular Signaling Peptides and Proteins/genetics , Molecular Chaperones/genetics , Nuclear Envelope/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Molecular Chaperones/metabolism , Nuclear Envelope/physiology , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/metabolism
16.
Elife ; 52016 06 23.
Article in English | MEDLINE | ID: mdl-27336722

ABSTRACT

Lamin B receptor (LBR) is a polytopic membrane protein residing in the inner nuclear membrane in association with the nuclear lamina. We demonstrate that human LBR is essential for cholesterol synthesis. LBR mutant derivatives implicated in Greenberg skeletal dysplasia or Pelger-Huët anomaly fail to rescue the cholesterol auxotrophy of a LBR-deficient human cell line, consistent with a loss-of-function mechanism for these congenital disorders. These disease-causing variants fall into two classes: point mutations in the sterol reductase domain perturb enzymatic activity by reducing the affinity for the essential cofactor NADPH, while LBR truncations render the mutant protein metabolically unstable, leading to its rapid degradation at the inner nuclear membrane. Thus, metabolically unstable LBR variants may serve as long-sought-after model substrates enabling previously impossible investigations of poorly understood protein turnover mechanisms at the inner nuclear membrane of higher eukaryotes.


Subject(s)
Cholesterol/metabolism , Mutation , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Cells, Cultured , Humans , Osteochondrodysplasias/physiopathology , Pelger-Huet Anomaly/physiopathology , Lamin B Receptor
17.
J Biol Chem ; 291(18): 9469-81, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-26953341

ABSTRACT

Torsin ATPases are the only representatives of the AAA+ ATPase family that reside in the lumen of the endoplasmic reticulum (ER) and nuclear envelope. Two of these, TorsinA and TorsinB, are anchored to the ER membrane by virtue of an N-terminal hydrophobic domain. Here we demonstrate that the imposition of ER stress leads to a proteolytic cleavage event that selectively removes the hydrophobic domain from the AAA+ domain of TorsinA, which retains catalytic activity. Both the pharmacological inhibition profile and the identified cleavage site between two juxtaposed cysteine residues are distinct from those of presently known proteases, suggesting that a hitherto uncharacterized, membrane-associated protease accounts for TorsinA processing. This processing occurs not only in stress-exposed cell lines but also in primary cells from distinct organisms including stimulated B cells, indicating that Torsin conversion in response to physiologically relevant stimuli is an evolutionarily conserved process. By establishing 5-nitroisatin as a cell-permeable inhibitor for Torsin processing, we provide the methodological framework for interfering with Torsin processing in a wide range of primary cells without the need for genetic manipulation.


Subject(s)
B-Lymphocytes/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum/metabolism , Lymphocyte Activation/physiology , Molecular Chaperones/metabolism , Proteolysis , B-Lymphocytes/cytology , Cell Membrane/genetics , Endoplasmic Reticulum/genetics , HEK293 Cells , HeLa Cells , Humans , Molecular Chaperones/genetics , Protein Structure, Tertiary , Protein Transport/physiology
18.
Curr Opin Cell Biol ; 40: 1-7, 2016 06.
Article in English | MEDLINE | ID: mdl-26803745

ABSTRACT

Torsin ATPases are the only members of the AAA+ ATPase family that localize to the endoplasmic reticulum and contiguous perinuclear space. Accordingly, they are well positioned to perform essential work in these compartments, but their precise functions remain elusive. Recent studies have deciphered an unusual ATPase activation mechanism relying on Torsin-associated transmembrane cofactors, LAP1 or LULL1. These findings profoundly change our molecular view of the Torsin machinery and rationalize several human mutations in TorsinA or LAP1 leading to congenital disorders, symptoms of which have recently been recapitulated in mouse models. Here, we review these recent advances in the Torsin field and discuss the most pressing questions in relation to nuclear envelope dynamics.


Subject(s)
Adenosine Triphosphatases/metabolism , Dystonia/genetics , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphate/metabolism , Animals , Disease Models, Animal , Dystonia/enzymology , Endoplasmic Reticulum/metabolism , HSC70 Heat-Shock Proteins/genetics , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Nuclear Envelope/metabolism
19.
Crit Rev Biochem Mol Biol ; 50(6): 532-49, 2015.
Article in English | MEDLINE | ID: mdl-26592310

ABSTRACT

Torsin ATPases (Torsins) belong to the widespread AAA+ (ATPases associated with a variety of cellular activities) family of ATPases, which share structural similarity but have diverse cellular functions. Torsins are outliers in this family because they lack many characteristics of typical AAA+ proteins, and they are the only members of the AAA+ family located in the endoplasmic reticulum and contiguous perinuclear space. While it is clear that Torsins have essential roles in many, if not all metazoans, their precise cellular functions remain elusive. Studying Torsins has significant medical relevance since mutations in Torsins or Torsin-associated proteins result in a variety of congenital human disorders, the most frequent of which is early-onset torsion (DYT1) dystonia, a severe movement disorder. A better understanding of the Torsin system is needed to define the molecular etiology of these diseases, potentially enabling corrective therapy. Here, we provide a comprehensive overview of the Torsin system in metazoans, discuss functional clues obtained from various model systems and organisms and provide a phylogenetic and structural analysis of Torsins and their regulatory cofactors in relation to disease-causative mutations. Moreover, we review recent data that have led to a dramatically improved understanding of these machines at a molecular level, providing a foundation for investigating the molecular defects underlying the associated movement disorders. Lastly, we discuss our ideas on how recent progress may be utilized to inform future studies aimed at determining the cellular role(s) of these atypical molecular machines and their implications for dystonia treatment options.


Subject(s)
Molecular Chaperones/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , Disease Models, Animal , Dystonia Musculorum Deformans/genetics , Dystonia Musculorum Deformans/metabolism , HSC70 Heat-Shock Proteins/metabolism , Humans , Membrane Proteins/metabolism , Molecular Chaperones/analysis , Molecular Chaperones/genetics , Molecular Sequence Data , Mutation , Protein Transport , Sequence Alignment
20.
Biochem Soc Trans ; 43(5): 874-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26517897

ABSTRACT

Repeat proteins are an attractive target for protein engineering and design. We have focused our attention on the design and engineering of one particular class: tetratricopeptide repeat (TPR) proteins. In previous work, we have shown that the structure and stability of TPR proteins can be manipulated in a rational fashion [Cortajarena (2011) Prot. Sci. 20: , 1042-1047; Main (2003) Structure 11: , 497-508]. Building on those studies, we have designed and characterized a number of different peptide-binding TPR modules and we have also assembled these modules into supramolecular arrays [Cortajarena (2009) ACS Chem. Biol. 5: , 545-552; Cortajarena (2008) ACS Chem. Biol. 3: , 161-166; Jackrel (2009) Prot. Sci. 18: , 762-774; Kajander (2007) Acta Crystallogr. D Biol. Crystallogr. 63: , 800-811]. Here we focus on the development of one such TPR-peptide interaction for a practical application, affinity purification. We illustrate the general utility of our designed protein interaction. Furthermore, this example highlights how basic research on protein-peptide interactions can lead to the development of novel reagents with important practical applications.


Subject(s)
Indicators and Reagents/chemistry , Protein Engineering , Recombinant Fusion Proteins/chemistry , Repetitive Sequences, Amino Acid , Animals , Cell Line , Chromatography, Affinity , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/genetics , Immobilized Proteins/metabolism , Indicators and Reagents/metabolism , Ligands , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...