Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
2.
Mol Cell ; 84(6): 1101-1119.e9, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38428433

ABSTRACT

Mitochondrial outer membrane ⍺-helical proteins play critical roles in mitochondrial-cytoplasmic communication, but the rules governing the targeting and insertion of these biophysically diverse proteins remain unknown. Here, we first defined the complement of required mammalian biogenesis machinery through genome-wide CRISPRi screens using topologically distinct membrane proteins. Systematic analysis of nine identified factors across 21 diverse ⍺-helical substrates reveals that these components are organized into distinct targeting pathways that act on substrates based on their topology. NAC is required for the efficient targeting of polytopic proteins, whereas signal-anchored proteins require TTC1, a cytosolic chaperone that physically engages substrates. Biochemical and mutational studies reveal that TTC1 employs a conserved TPR domain and a hydrophobic groove in its C-terminal domain to support substrate solubilization and insertion into mitochondria. Thus, the targeting of diverse mitochondrial membrane proteins is achieved through topological triaging in the cytosol using principles with similarities to ER membrane protein biogenesis systems.


Subject(s)
Mitochondrial Membranes , Saccharomyces cerevisiae Proteins , Animals , Mitochondrial Membranes/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Protein Transport , Saccharomyces cerevisiae Proteins/metabolism , Mammals/metabolism
3.
Nat Protoc ; 19(1): 127-158, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37974029

ABSTRACT

The isolation of proteins in high yield and purity is a major bottleneck for the analysis of their three-dimensional structure, function and interactome. Here, we present a streamlined workflow for the rapid production of proteins or protein complexes using lentiviral transduction of human suspension cells, combined with highly specific nanobody-mediated purification and proteolytic elution. Application of the method requires prior generation of a plasmid coding for a protein of interest (POI) fused to an N- or C-terminal GFP or ALFA peptide tag using a lentiviral plasmid toolkit we have designed. The plasmid is then used to generate human suspension cell lines stably expressing the tagged fusion protein by lentiviral transduction. By leveraging the picomolar affinity of the GFP and ALFA nanobodies for their respective tags, the POI can be specifically captured from the resulting cell lysate even when expressed at low levels and under a variety of conditions, including detergents and mild denaturants. Finally, rapid and specific elution of the POI (in its tagged or untagged form) under native conditions is achieved within minutes at 4 °C, using the engineered SUMO protease SENPEuB. We demonstrate the wide applicability of the method by purifying multiple challenging soluble and membrane protein complexes to high purity from human cells. Our strategy is also directly compatible with many widely used GFP-expression plasmids, cell lines and transgenic model organisms. Finally, our method is faster than alternative approaches, requiring only 8 d from plasmid to purified protein, and results in substantially improved yields and purity.


Subject(s)
Peptides , Proteins , Humans , Proteolysis , Recombinant Fusion Proteins , Chromatography, Affinity/methods
4.
bioRxiv ; 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38076791

ABSTRACT

Mammalian membrane proteins perform essential physiologic functions that rely on their accurate insertion and folding at the endoplasmic reticulum (ER). Using forward and arrayed genetic screens, we systematically studied the biogenesis of a panel of membrane proteins, including several G-protein coupled receptors (GPCRs). We observed a central role for the insertase, the ER membrane protein complex (EMC), and developed a dual-guide approach to identify genetic modifiers of the EMC. We found that the back of sec61 (BOS) complex, a component of the 'multipass translocon', was a physical and genetic interactor of the EMC. Functional and structural analysis of the EMC•BOS holocomplex showed that characteristics of a GPCR's soluble domain determine its biogenesis pathway. In contrast to prevailing models, no single insertase handles all substrates. We instead propose a unifying model for coordination between the EMC, multipass translocon, and Sec61 for biogenesis of diverse membrane proteins in human cells.

5.
BMC Genomics ; 24(1): 651, 2023 Oct 30.
Article in English | MEDLINE | ID: mdl-37904134

ABSTRACT

Mapping genetic interactions is essential for determining gene function and defining novel biological pathways. We report a simple to use CRISPR interference (CRISPRi) based platform, compatible with Fluorescence Activated Cell Sorting (FACS)-based reporter screens, to query epistatic relationships at scale. This is enabled by a flexible dual-sgRNA library design that allows for the simultaneous delivery and selection of a fixed sgRNA and a second randomized guide, comprised of a genome-wide library, with a single transduction. We use this approach to identify epistatic relationships for a defined biological pathway, showing both increased sensitivity and specificity than traditional growth screening approaches.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , RNA, Guide, CRISPR-Cas Systems , Gene Library , Genome , CRISPR-Cas Systems
6.
Nature ; 623(7988): 842-852, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37853127

ABSTRACT

Optimum protein function and biochemical activity critically depends on water availability because solvent thermodynamics drive protein folding and macromolecular interactions1. Reciprocally, macromolecules restrict the movement of 'structured' water molecules within their hydration layers, reducing the available 'free' bulk solvent and therefore the total thermodynamic potential energy of water, or water potential. Here, within concentrated macromolecular solutions such as the cytosol, we found that modest changes in temperature greatly affect the water potential, and are counteracted by opposing changes in osmotic strength. This duality of temperature and osmotic strength enables simple manipulations of solvent thermodynamics to prevent cell death after extreme cold or heat shock. Physiologically, cells must sustain their activity against fluctuating temperature, pressure and osmotic strength, which impact water availability within seconds. Yet, established mechanisms of water homeostasis act over much slower timescales2,3; we therefore postulated the existence of a rapid compensatory response. We find that this function is performed by water potential-driven changes in macromolecular assembly, particularly biomolecular condensation of intrinsically disordered proteins. The formation and dissolution of biomolecular condensates liberates and captures free water, respectively, quickly counteracting thermal or osmotic perturbations of water potential, which is consequently robustly buffered in the cytoplasm. Our results indicate that biomolecular condensation constitutes an intrinsic biophysical feedback response that rapidly compensates for intracellular osmotic and thermal fluctuations. We suggest that preserving water availability within the concentrated cytosol is an overlooked evolutionary driver of protein (dis)order and function.


Subject(s)
Macromolecular Substances , Proteins , Solvents , Thermodynamics , Water , Cell Death , Cytosol/chemistry , Cytosol/metabolism , Homeostasis , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Osmolar Concentration , Pressure , Proteins/chemistry , Proteins/metabolism , Solvents/chemistry , Solvents/metabolism , Temperature , Time Factors , Water/chemistry , Water/metabolism
7.
bioRxiv ; 2023 Aug 17.
Article in English | MEDLINE | ID: mdl-37645817

ABSTRACT

Mitochondrial outer membrane α-helical proteins play critical roles in mitochondrial-cytoplasmic communication, but the rules governing the targeting and insertion of these biophysically diverse substrates remain unknown. Here, we first defined the complement of required mammalian biogenesis machinery through genome-wide CRISPRi screens using topologically distinct membrane proteins. Systematic analysis of nine identified factors across 21 diverse α-helical substrates reveals that these components are organized into distinct targeting pathways which act on substrates based on their topology. NAC is required for efficient targeting of polytopic proteins whereas signal-anchored proteins require TTC1, a novel cytosolic chaperone which physically engages substrates. Biochemical and mutational studies reveal that TTC1 employs a conserved TPR domain and a hydrophobic groove in its C-terminal domain to support substrate solubilization and insertion into mitochondria. Thus, targeting of diverse mitochondrial membrane proteins is achieved through topological triaging in the cytosol using principles with similarities to ER membrane protein biogenesis systems.

8.
J Cell Biol ; 222(8)2023 08 07.
Article in English | MEDLINE | ID: mdl-37199759

ABSTRACT

Tail-anchored (TA) proteins play essential roles in mammalian cells, and their accurate localization is critical for proteostasis. Biophysical similarities lead to mistargeting of mitochondrial TA proteins to the ER, where they are delivered to the insertase, the ER membrane protein complex (EMC). Leveraging an improved structural model of the human EMC, we used mutagenesis and site-specific crosslinking to map the path of a TA protein from its cytosolic capture by methionine-rich loops to its membrane insertion through a hydrophilic vestibule. Positively charged residues at the entrance to the vestibule function as a selectivity filter that uses charge-repulsion to reject mitochondrial TA proteins. Similarly, this selectivity filter retains the positively charged soluble domains of multipass substrates in the cytosol, thereby ensuring they adopt the correct topology and enforcing the "positive-inside" rule. Substrate discrimination by the EMC provides a biochemical explanation for one role of charge in TA protein sorting and protects compartment integrity by limiting protein misinsertion.


Subject(s)
Endoplasmic Reticulum , Membrane Proteins , Mitochondrial Proteins , Humans , Endoplasmic Reticulum/metabolism , Membrane Proteins/genetics , Membrane Proteins/chemistry , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Protein Transport , Protein Conformation
9.
J Cell Sci ; 136(10)2023 05 15.
Article in English | MEDLINE | ID: mdl-37218462

ABSTRACT

Translation of mRNAs containing premature termination codons (PTCs) results in truncated protein products with deleterious effects. Nonsense-mediated decay (NMD) is a surveillance pathway responsible for detecting PTC containing transcripts. Although the molecular mechanisms governing mRNA degradation have been extensively studied, the fate of the nascent protein product remains largely uncharacterized. Here, we use a fluorescent reporter system in mammalian cells to reveal a selective degradation pathway specifically targeting the protein product of an NMD mRNA. We show that this process is post-translational and dependent on the ubiquitin proteasome system. To systematically uncover factors involved in NMD-linked protein quality control, we conducted genome-wide flow cytometry-based screens. Our screens recovered known NMD factors but suggested that protein degradation did not depend on the canonical ribosome-quality control (RQC) pathway. A subsequent arrayed screen demonstrated that protein and mRNA branches of NMD rely on a shared recognition event. Our results establish the existence of a targeted pathway for nascent protein degradation from PTC containing mRNAs, and provide a reference for the field to identify and characterize required factors.


Subject(s)
Mammals , Nonsense Mediated mRNA Decay , Animals , Nonsense Mediated mRNA Decay/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Mammals/metabolism
10.
bioRxiv ; 2023 Aug 14.
Article in English | MEDLINE | ID: mdl-36711738

ABSTRACT

Mapping genetic interactions is essential for determining gene function and defining novel biological pathways. We report a simple to use CRISPR interference (CRISPRi) based platform, compatible with Fluorescence Activated Cell Sorting (FACS)-based reporter screens, to query epistatic relationships at scale. This is enabled by a flexible dual-sgRNA library design that allows for the simultaneous delivery and selection of a fixed sgRNA and a second randomized guide, comprised of a genome-wide library, with a single transduction. We use this approach to identify epistatic relationships for a defined biological pathway, showing both increased sensitivity and specificity than traditional growth screening approaches.

11.
Article in English | MEDLINE | ID: mdl-36041783

ABSTRACT

Tail-anchored (TA) proteins are an essential class of integral membrane proteins required for many aspects of cellular physiology. TA proteins contain a single carboxy-terminal transmembrane domain that must be post-translationally recognized, guided to, and ultimately inserted into the correct cellular compartment. The majority of TA proteins begin their biogenesis in the endoplasmic reticulum (ER) and utilize two parallel strategies for targeting and insertion: the guided-entry of tail-anchored proteins (GET) and ER-membrane protein complex (EMC) pathways. Here we focus on how these two sets of machinery target, transfer, and insert TAs into the lipid bilayer in close collaboration with quality control machinery. Additionally, we highlight the unifying features of the insertion process as revealed by recent structures of the GET and EMC membrane protein complexes.


Subject(s)
Endoplasmic Reticulum , Membrane Proteins , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Protein Binding , Protein Transport
12.
Science ; 378(6617): 317-322, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36264797

ABSTRACT

In the mitochondrial outer membrane, α-helical transmembrane proteins play critical roles in cytoplasmic-mitochondrial communication. Using genome-wide CRISPR screens, we identified mitochondrial carrier homolog 2 (MTCH2), and its paralog MTCH1, and showed that it is required for insertion of biophysically diverse tail-anchored (TA), signal-anchored, and multipass proteins, but not outer membrane ß-barrel proteins. Purified MTCH2 was sufficient to mediate insertion into reconstituted proteoliposomes. Functional and mutational studies suggested that MTCH2 has evolved from a solute carrier transporter. MTCH2 uses membrane-embedded hydrophilic residues to function as a gatekeeper for the outer membrane, controlling mislocalization of TAs into the endoplasmic reticulum and modulating the sensitivity of leukemia cells to apoptosis. Our identification of MTCH2 as an insertase provides a mechanistic explanation for the diverse phenotypes and disease states associated with MTCH2 dysfunction.


Subject(s)
Apoptosis , Mitochondrial Membrane Transport Proteins , Mitochondrial Membranes , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membranes/metabolism , Humans , Endoplasmic Reticulum/metabolism , K562 Cells
14.
Nat Commun ; 12(1): 6035, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34654800

ABSTRACT

Between 6-20% of the cellular proteome is under circadian control and tunes mammalian cell function with daily environmental cycles. For cell viability, and to maintain volume within narrow limits, the daily variation in osmotic potential exerted by changes in the soluble proteome must be counterbalanced. The mechanisms and consequences of this osmotic compensation have not been investigated before. In cultured cells and in tissue we find that compensation involves electroneutral active transport of Na+, K+, and Cl- through differential activity of SLC12A family cotransporters. In cardiomyocytes ex vivo and in vivo, compensatory ion fluxes confer daily variation in electrical activity. Perturbation of soluble protein abundance has commensurate effects on ion composition and cellular function across the circadian cycle. Thus, circadian regulation of the proteome impacts ion homeostasis with substantial consequences for the physiology of electrically active cells such as cardiomyocytes.


Subject(s)
Cell Physiological Phenomena , Circadian Rhythm/physiology , Ion Transport/physiology , Osmosis , Animals , Cardiovascular System/pathology , Cells, Cultured , Chlorides/metabolism , Fibroblasts , Homeostasis , Lung , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Potassium/metabolism , Proteome , Sodium/metabolism , Solute Carrier Family 12, Member 2/genetics
15.
Mol Cell ; 81(13): 2693-2704.e12, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33964204

ABSTRACT

The assembly of nascent proteins into multi-subunit complexes is a tightly regulated process that must occur at high fidelity to maintain cellular homeostasis. The ER membrane protein complex (EMC) is an essential insertase that requires seven membrane-spanning and two soluble cytosolic subunits to function. Here, we show that the kinase with no lysine 1 (WNK1), known for its role in hypertension and neuropathy, functions as an assembly factor for the human EMC. WNK1 uses a conserved amphipathic helix to stabilize the soluble subunit, EMC2, by binding to the EMC2-8 interface. Shielding this hydrophobic surface prevents promiscuous interactions of unassembled EMC2 and directly competes for binding of E3 ubiquitin ligases, permitting assembly. Depletion of WNK1 thus destabilizes both the EMC and its membrane protein clients. This work describes an unexpected role for WNK1 in protein biogenesis and defines the general requirements of an assembly factor that will apply across the proteome.


Subject(s)
Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Multiprotein Complexes/metabolism , WNK Lysine-Deficient Protein Kinase 1/metabolism , Endoplasmic Reticulum/genetics , HeLa Cells , Humans , Multiprotein Complexes/genetics , WNK Lysine-Deficient Protein Kinase 1/genetics
16.
Cell ; 183(5): 1325-1339.e21, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33080218

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a recently identified coronavirus that causes the respiratory disease known as coronavirus disease 2019 (COVID-19). Despite the urgent need, we still do not fully understand the molecular basis of SARS-CoV-2 pathogenesis. Here, we comprehensively define the interactions between SARS-CoV-2 proteins and human RNAs. NSP16 binds to the mRNA recognition domains of the U1 and U2 splicing RNAs and acts to suppress global mRNA splicing upon SARS-CoV-2 infection. NSP1 binds to 18S ribosomal RNA in the mRNA entry channel of the ribosome and leads to global inhibition of mRNA translation upon infection. Finally, NSP8 and NSP9 bind to the 7SL RNA in the signal recognition particle and interfere with protein trafficking to the cell membrane upon infection. Disruption of each of these essential cellular functions acts to suppress the interferon response to viral infection. Our results uncover a multipronged strategy utilized by SARS-CoV-2 to antagonize essential cellular processes to suppress host defenses.


Subject(s)
COVID-19/metabolism , Host-Pathogen Interactions , Protein Biosynthesis , RNA Splicing , SARS-CoV-2/metabolism , Viral Nonstructural Proteins/metabolism , A549 Cells , Animals , COVID-19/virology , Chlorocebus aethiops , HEK293 Cells , Humans , Interferons/metabolism , Protein Transport , RNA, Messenger/metabolism , RNA, Ribosomal, 18S/metabolism , RNA, Small Cytoplasmic/chemistry , RNA, Small Cytoplasmic/metabolism , Signal Recognition Particle/chemistry , Signal Recognition Particle/metabolism , Vero Cells , Viral Nonstructural Proteins/chemistry
17.
Science ; 369(6502): 433-436, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32439656

ABSTRACT

A defining step in the biogenesis of a membrane protein is the insertion of its hydrophobic transmembrane helices into the lipid bilayer. The nine-subunit endoplasmic reticulum (ER) membrane protein complex (EMC) is a conserved co- and posttranslational insertase at the ER. We determined the structure of the human EMC in a lipid nanodisc to an overall resolution of 3.4 angstroms by cryo-electron microscopy, permitting building of a nearly complete atomic model. We used structure-guided mutagenesis to demonstrate that substrate insertion requires a methionine-rich cytosolic loop and occurs via an enclosed hydrophilic vestibule within the membrane formed by the subunits EMC3 and EMC6. We propose that the EMC uses local membrane thinning and a positively charged patch to decrease the energetic barrier for insertion into the bilayer.


Subject(s)
Endoplasmic Reticulum/chemistry , Intracellular Membranes/chemistry , Membrane Proteins/chemistry , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Humans , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure
18.
Cell Rep ; 30(11): 3691-3698.e5, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32187542

ABSTRACT

A large proportion of membrane proteins must be assembled into oligomeric complexes for function. How this process occurs is poorly understood, but it is clear that complex assembly must be tightly regulated to avoid accumulation of orphan subunits with potential cytotoxic effects. We interrogated assembly in mammalian cells by using the WRB/CAML complex, an essential insertase for tail-anchored proteins in the endoplasmic reticulum (ER), as a model system. Our data suggest that the stability of each subunit is differentially regulated. In WRB's absence, CAML folds incorrectly, causing aberrant exposure of a hydrophobic transmembrane domain to the ER lumen. When present, WRB can correct the topology of CAML both in vitro and in cells. In contrast, WRB can independently fold correctly but is still degraded in the absence of CAML. We therefore propose that there are at least two distinct regulatory pathways for the surveillance of orphan subunits in the mammalian ER.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Protein Subunits/metabolism , Animals , Dogs , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Protein Binding , Protein Stability , Proteolysis , Rabbits , Ribosomes/metabolism
19.
Curr Opin Cell Biol ; 41: 91-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27155805

ABSTRACT

The translocation of most eukaryotic secreted and integral membrane proteins occurs co-translationally at the endoplasmic reticulum (ER). These nascent polypeptides are recognized on the ribosome by the signal recognition particle (SRP), targeted to the ER, and translocated across or inserted into the membrane by the Sec61 translocation channel. Structural analysis of these co-translational processes has been challenging due to the size, complexity, and flexibility of the targeting and translocation machinery. Recent technological advances in cryo-electron microscopy (cryo-EM) have resulted in increasingly powerful tools to study large, heterogeneous, and low-abundance samples. These advances are being utilized to obtain near-atomic resolution reconstructions of functional translation, targeting, and translocation intermediates, paving the way to a mechanistic understanding of protein biogenesis.


Subject(s)
Proteins/chemistry , Translocation, Genetic , Animals , Intracellular Membranes/metabolism , Protein Transport , Proteins/metabolism , Secretory Pathway , Signal Recognition Particle/chemistry , Signal Recognition Particle/metabolism
20.
Science ; 351(6268): 88-91, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26721998

ABSTRACT

Secreted and integral membrane proteins compose up to one-third of the biological proteome. These proteins contain hydrophobic signals that direct their translocation across or insertion into the lipid bilayer by the Sec61 protein-conducting channel. The molecular basis of how hydrophobic signals within a nascent polypeptide trigger channel opening is not understood. Here, we used cryo-electron microscopy to determine the structure of an active Sec61 channel that has been opened by a signal sequence. The signal supplants helix 2 of Sec61α, which triggers a rotation that opens the central pore both axially across the membrane and laterally toward the lipid bilayer. Comparisons with structures of Sec61 in other states suggest a pathway for how hydrophobic signals engage the channel to gain access to the lipid bilayer.


Subject(s)
Membrane Proteins/chemistry , Animals , Cryoelectron Microscopy , Crystallography, X-Ray , Dogs , Hydrophobic and Hydrophilic Interactions , Lipid Bilayers/chemistry , Protein Sorting Signals , Protein Structure, Secondary , Ribosomes/chemistry , SEC Translocation Channels
SELECTION OF CITATIONS
SEARCH DETAIL
...