Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.884
Filter
Add more filters

Publication year range
1.
Cell ; 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39106863

ABSTRACT

It is currently not known whether mRNAs fulfill structural roles in the cytoplasm. Here, we report the fragile X-related protein 1 (FXR1) network, an mRNA-protein (mRNP) network present throughout the cytoplasm, formed by FXR1-mediated packaging of exceptionally long mRNAs. These mRNAs serve as an underlying condensate scaffold and concentrate FXR1 molecules. The FXR1 network contains multiple protein binding sites and functions as a signaling scaffold for interacting proteins. We show that it is necessary for RhoA signaling-induced actomyosin reorganization to provide spatial proximity between kinases and their substrates. Point mutations in FXR1, found in its homolog FMR1, where they cause fragile X syndrome, disrupt the network. FXR1 network disruption prevents actomyosin remodeling-an essential and ubiquitous process for the regulation of cell shape, migration, and synaptic function. Our findings uncover a structural role for cytoplasmic mRNA and show how the FXR1 RNA-binding protein as part of the FXR1 network acts as an organizer of signaling reactions.

2.
Cell ; 183(6): 1682-1698.e24, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33232692

ABSTRACT

In order to analyze how a signal transduction network converts cellular inputs into cellular outputs, ideally one would measure the dynamics of many signals within the network simultaneously. We found that, by fusing a fluorescent reporter to a pair of self-assembling peptides, it could be stably clustered within cells at random points, distant enough to be resolved by a microscope but close enough to spatially sample the relevant biology. Because such clusters, which we call signaling reporter islands (SiRIs), can be modularly designed, they permit a set of fluorescent reporters to be efficiently adapted for simultaneous measurement of multiple nodes of a signal transduction network within single cells. We created SiRIs for indicators of second messengers and kinases and used them, in hippocampal neurons in culture and intact brain slices, to discover relationships between the speed of calcium signaling, and the amplitude of PKA signaling, upon receiving a cAMP-driving stimulus.


Subject(s)
Fluorescent Dyes/metabolism , Genes, Reporter , Optical Imaging , Signal Transduction , Animals , Calcium/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Green Fluorescent Proteins/metabolism , HeLa Cells , Hippocampus/metabolism , Humans , Mice , Neurons/metabolism , Peptides/metabolism , Proteins/metabolism , Pyramidal Cells/metabolism
3.
Cell ; 179(4): 923-936.e11, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31675499

ABSTRACT

Tight junctions are cell-adhesion complexes that seal tissues and are involved in cell polarity and signaling. Supra-molecular assembly and positioning of tight junctions as continuous networks of adhesion strands are dependent on the membrane-associated scaffolding proteins ZO1 and ZO2. To understand how zona occludens (ZO) proteins organize junction assembly, we performed quantitative cell biology and in vitro reconstitution experiments. We discovered that ZO proteins self-organize membrane-attached compartments via phase separation. We identified the multivalent interactions of the conserved PDZ-SH3-GuK supra-domain as the driver of phase separation. These interactions are regulated by phosphorylation and intra-molecular binding. Formation of condensed ZO protein compartments is sufficient to specifically enrich and localize tight-junction proteins, including adhesion receptors, cytoskeletal adapters, and transcription factors. Our results suggest that an active-phase transition of ZO proteins into a condensed membrane-bound compartment drives claudin polymerization and coalescence of a continuous tight-junction belt.


Subject(s)
Tight Junctions/genetics , Zonula Occludens Proteins/genetics , Zonula Occludens-1 Protein/genetics , Zonula Occludens-2 Protein/genetics , Animals , Binding Sites/genetics , Cell Adhesion/genetics , Cell Polarity/genetics , Dogs , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Membrane Proteins/genetics , PDZ Domains/genetics , Phosphoproteins/genetics , Phosphorylation/genetics , Protein Binding/genetics , Signal Transduction/genetics , Tight Junctions/metabolism , Zonula Occludens Proteins/chemistry , Zonula Occludens Proteins/ultrastructure , Zonula Occludens-1 Protein/chemistry , Zonula Occludens-1 Protein/ultrastructure , Zonula Occludens-2 Protein/chemistry , Zonula Occludens-2 Protein/ultrastructure , src Homology Domains/genetics
4.
Cell ; 174(5): 1172-1187.e16, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30078712

ABSTRACT

Synapses are semi-membraneless, protein-dense, sub-micron chemical reaction compartments responsible for signal processing in each and every neuron. Proper formation and dynamic responses to stimulations of synapses, both during development and in adult, are fundamental to functions of mammalian brains, although the molecular basis governing formation and modulation of compartmentalized synaptic assemblies is unclear. Here, we used a biochemical reconstitution approach to show that, both in solution and on supported membrane bilayers, multivalent interaction networks formed by major excitatory postsynaptic density (PSD) scaffold proteins led to formation of PSD-like assemblies via phase separation. The reconstituted PSD-like assemblies can cluster receptors, selectively concentrate enzymes, promote actin bundle formation, and expel inhibitory postsynaptic proteins. Additionally, the condensed phase PSD assemblies have features that are distinct from those in homogeneous solutions and fit for synaptic functions. Thus, we have built a molecular platform for understanding how neuronal synapses are formed and dynamically regulated.


Subject(s)
Neurogenesis , Neuronal Plasticity , Post-Synaptic Density , Synapses/physiology , Animals , Brain/physiology , Disks Large Homolog 4 Protein/physiology , Hippocampus/physiology , Light , Mice , Microscopy, Confocal , Neurons/physiology , Scattering, Radiation , Signal Transduction , Synaptic Transmission
5.
Annu Rev Cell Dev Biol ; 35: 111-129, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31340125

ABSTRACT

Many cellular processes rely on precise and timely deformation of the cell membrane. While many proteins participate in membrane reshaping and scission, usually in highly specialized ways, Bin/amphiphysin/Rvs (BAR) domain proteins play a pervasive role, as they not only participate in many aspects of cell trafficking but also are highly versatile membrane remodelers. Subtle changes in the shape and size of the BAR domain can greatly impact the way in which BAR domain proteins interact with the membrane. Furthermore, the activity of BAR domain proteins can be tuned by external physical parameters, and so they behave differently depending on protein surface density, membrane tension, or membrane shape. These proteins can form 3D structures that mold the membrane and alter its liquid properties, even promoting scission under various circumstances.As such, BAR domain proteins have numerous roles within the cell. Endocytosis is among the most highly studied processes in which BAR domain proteins take on important roles. Over the years, a more complete picture has emerged in which BAR domain proteins are tied to almost all intracellular compartments; examples include endosomal sorting and tubular networks in the endoplasmic reticulum and T-tubules. These proteins also have a role in autophagy, and their activity has been linked with cancer. Here, we briefly review the history of BAR domain protein discovery, discuss the mechanisms by which BAR domain proteins induce curvature, and attempt to settle important controversies in the field. Finally, we review BAR domain proteins in the context of a cell, highlighting their emerging roles in cell signaling and organelle shaping.


Subject(s)
Carrier Proteins/metabolism , Cell Membrane Structures/chemistry , Membrane Proteins/metabolism , Animals , Carrier Proteins/chemistry , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane Structures/metabolism , Cell Shape , Humans , Membrane Proteins/chemistry , Neoplasms/pathology , Organelles/chemistry , Organelles/metabolism , Protein Domains
6.
Cell ; 169(6): 1066-1077.e10, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575670

ABSTRACT

Centrosomes are non-membrane-bound compartments that nucleate microtubule arrays. They consist of nanometer-scale centrioles surrounded by a micron-scale, dynamic assembly of protein called the pericentriolar material (PCM). To study how PCM forms a spherical compartment that nucleates microtubules, we reconstituted PCM-dependent microtubule nucleation in vitro using recombinant C. elegans proteins. We found that macromolecular crowding drives assembly of the key PCM scaffold protein SPD-5 into spherical condensates that morphologically and dynamically resemble in vivo PCM. These SPD-5 condensates recruited the microtubule polymerase ZYG-9 (XMAP215 homolog) and the microtubule-stabilizing protein TPXL-1 (TPX2 homolog). Together, these three proteins concentrated tubulin ∼4-fold over background, which was sufficient to reconstitute nucleation of microtubule asters in vitro. Our results suggest that in vivo PCM is a selective phase that organizes microtubule arrays through localized concentration of tubulin by microtubule effector proteins.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Cell Cycle Proteins/metabolism , Centrosome/chemistry , Microtubules/metabolism , Tubulin/metabolism , Animals , Caenorhabditis elegans/cytology , Carrier Proteins/metabolism , Centrosome/metabolism , Protein Serine-Threonine Kinases/metabolism
7.
Cell ; 170(1): 172-184.e11, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28648660

ABSTRACT

Membrane scission is essential for intracellular trafficking. While BAR domain proteins such as endophilin have been reported in dynamin-independent scission of tubular membrane necks, the cutting mechanism has yet to be deciphered. Here, we combine a theoretical model, in vitro, and in vivo experiments revealing how protein scaffolds may cut tubular membranes. We demonstrate that the protein scaffold bound to the underlying tube creates a frictional barrier for lipid diffusion; tube elongation thus builds local membrane tension until the membrane undergoes scission through lysis. We call this mechanism friction-driven scission (FDS). In cells, motors pull tubes, particularly during endocytosis. Through reconstitution, we show that motors not only can pull out and extend protein-scaffolded tubes but also can cut them by FDS. FDS is generic, operating even in the absence of amphipathic helices in the BAR domain, and could in principle apply to any high-friction protein and membrane assembly.


Subject(s)
Endocytosis , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Acyltransferases/chemistry , Acyltransferases/metabolism , Animals , Biomechanical Phenomena , Friction , Humans , Lipid Metabolism , Protein Domains , Rats
8.
Mol Cell ; 83(17): 3140-3154.e7, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37572670

ABSTRACT

Peroxiredoxins (Prdxs) utilize reversibly oxidized cysteine residues to reduce peroxides and promote H2O2 signal transduction, including H2O2-induced activation of P38 MAPK. Prdxs form H2O2-induced disulfide complexes with many proteins, including multiple kinases involved in P38 MAPK signaling. Here, we show that a genetically encoded fusion between a Prdx and P38 MAPK is sufficient to hyperactivate the kinase in yeast and human cells by a mechanism that does not require the H2O2-sensing cysteine of the Prdx. We demonstrate that a P38-Prdx fusion protein compensates for loss of the yeast scaffold protein Mcs4 and MAP3K activity, driving yeast into mitosis. Based on our findings, we propose that the H2O2-induced formation of Prdx-MAPK disulfide complexes provides an alternative scaffold and signaling platform for MAPKK-MAPK signaling. The demonstration that formation of a complex with a Prdx is sufficient to modify the activity of a kinase has broad implications for peroxide-based signal transduction in eukaryotes.


Subject(s)
Peroxiredoxins , p38 Mitogen-Activated Protein Kinases , Humans , Cysteine/metabolism , Disulfides , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Oxidation-Reduction , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
9.
Mol Cell ; 81(17): 3509-3525.e5, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34320406

ABSTRACT

Nuclear chromosomes transcribe far more RNA than required to encode protein. Here we investigate whether non-coding RNA broadly contributes to cytological-scale chromosome territory architecture. We develop a procedure that depletes soluble proteins, chromatin, and most nuclear RNA from the nucleus but does not delocalize XIST, a known architectural RNA, from an insoluble chromosome "scaffold." RNA-seq analysis reveals that most RNA in the nuclear scaffold is repeat-rich, non-coding, and derived predominantly from introns of nascent transcripts. Insoluble, repeat-rich (C0T-1) RNA co-distributes with known scaffold proteins including scaffold attachment factor A (SAF-A), and distribution of these components inversely correlates with chromatin compaction in normal and experimentally manipulated nuclei. We further show that RNA is required for SAF-A to interact with chromatin and for enrichment of structurally embedded "scaffold attachment regions" prevalent in euchromatin. Collectively, the results indicate that long nascent transcripts contribute a dynamic structural role that promotes the open architecture of active chromosome territories.


Subject(s)
Chromatin/metabolism , Nuclear Matrix/metabolism , RNA, Untranslated/metabolism , Animals , Cell Line , Cell Nucleus/physiology , Chromatin/genetics , Chromosomes/genetics , Chromosomes/metabolism , Euchromatin/metabolism , Humans , Mice , Nuclear Matrix/genetics , RNA/genetics , RNA/metabolism , RNA, Long Noncoding/genetics , RNA, Untranslated/genetics , Transcription, Genetic/genetics
10.
Mol Cell ; 73(5): 971-984.e5, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30661983

ABSTRACT

Both the timing and kinetics of neurotransmitter release depend on the positioning of clustered Ca2+ channels in active zones to docked synaptic vesicles on presynaptic plasma membranes. However, how active zones form is not known. Here, we show that RIM and RIM-BP, via specific multivalent bindings, form dynamic and condensed assemblies through liquid-liquid phase separation. Voltage-gated Ca2+ channels (VGCCs), via C-terminal-tail-mediated direct binding to both RIM and RIM-BP, can be enriched to the RIM and RIM-BP condensates. We further show that RIM and RIM-BP, together with VGCCs, form dense clusters on the supported lipid membrane bilayers via phase separation. Therefore, RIMs and RIM-BPs are plausible organizers of active zones, and the formation of RIM and RIM-BP condensates may cluster VGCCs into nano- or microdomains and position the clustered Ca2+ channels with Ca2+ sensors on docked vesicles for efficient and precise synaptic transmissions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium Channels, N-Type/metabolism , GTP-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Presynaptic Terminals/metabolism , Synaptic Membranes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding Sites , Calcium Channels, N-Type/genetics , GTP-Binding Proteins/genetics , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Kinetics , Membrane Microdomains/genetics , Membrane Microdomains/metabolism , Mice , Protein Binding , Protein Interaction Domains and Motifs , Rats , SNARE Proteins/genetics , SNARE Proteins/metabolism , Solubility , Synaptic Membranes/genetics , Synaptic Transmission
11.
Proc Natl Acad Sci U S A ; 121(35): e2408554121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39172789

ABSTRACT

Biomolecules can be sequestered into membrane-less compartments, referred to as biomolecular condensates. Experimental and computational methods have helped define the physical-chemical properties of condensates. Less is known about how the high macromolecule concentrations in condensed phases contribute "solvent" interactions that can remodel the free-energy landscape of other condensate-resident proteins, altering thermally accessible conformations and, in turn, modulating function. Here, we use solution NMR spectroscopy to obtain atomic resolution insights into the interactions between the immature form of superoxide dismutase 1 (SOD1), which can mislocalize and aggregate in stress granules, and the RNA-binding protein CAPRIN1, a component of stress granules. NMR studies of CAPRIN1:SOD1 interactions, focused on both unfolded and folded SOD1 states in mixed phase and demixed CAPRIN1-based condensates, establish that CAPRIN1 shifts the SOD1 folding equilibrium toward the unfolded state through preferential interactions with the unfolded ensemble, with little change to the structure of the folded conformation. Key contacts between CAPRIN1 and the H80-H120 region of unfolded SOD1 are identified, as well as SOD1 interaction sites near both the arginine-rich and aromatic-rich regions of CAPRIN1. Unfolding of immature SOD1 in the CAPRIN1 condensed phase is shown to be coupled to aggregation, while a more stable zinc-bound, dimeric form of SOD1 is less susceptible to unfolding when solvated by CAPRIN1. Our work underscores the impact of the condensate solvent environment on the conformational states of resident proteins and supports the hypothesis that ALS mutations that decrease metal binding or dimerization function as drivers of aggregation in condensates.


Subject(s)
Solvents , Superoxide Dismutase-1 , Superoxide Dismutase-1/chemistry , Superoxide Dismutase-1/metabolism , Superoxide Dismutase-1/genetics , Humans , Solvents/chemistry , Protein Unfolding , Protein Binding , Protein Folding , Models, Molecular , Stress Granules/metabolism , Stress Granules/chemistry , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/chemistry , Protein Conformation , Magnetic Resonance Spectroscopy
12.
Trends Biochem Sci ; 47(7): 561-569, 2022 07.
Article in English | MEDLINE | ID: mdl-35331611

ABSTRACT

Advances in cryogenic electron microscopy (cryo-EM) enabled routine near-atomic structure determination of membrane proteins, while nanodisc technology has provided a way to provide membrane proteins with a native or native-like lipid environment. After giving a brief history of membrane mimetics, we present example structures of membrane proteins in nanodiscs that revealed information not provided by structures obtained in detergent. We describe how the lipid environment surrounding the membrane protein can be custom designed during nanodisc assembly and how it can be modified after assembly to test functional hypotheses. Because nanodiscs most closely replicate the physiologic environment of membrane proteins and often afford novel mechanistic insights, we propose that nanodiscs ought to become the standard for structural studies on membrane proteins.


Subject(s)
Membrane Proteins , Nanostructures , Lipid Bilayers/chemistry , Lipids , Membrane Proteins/metabolism , Microscopy, Electron , Models, Molecular , Nanostructures/chemistry
13.
Hum Mol Genet ; 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38881369

ABSTRACT

The Shoc2 scaffold protein is crucial in transmitting signals within the Epidermal Growth Factor Receptor (EGFR)-mediated Extracellular signal-Regulated Kinase (ERK1/2) pathway. While the significance of Shoc2 in this pathway is well-established, the precise mechanisms through which Shoc2 governs signal transmission remain to be fully elucidated. Hereditary variants in Shoc2 are responsible for Noonan Syndrome with Loose anagen Hair (NSLH). However, due to the absence of known enzymatic activity in Shoc2, directly assessing how these variants affect its function is challenging. ERK1/2 phosphorylation is used as a primary parameter of Shoc2 function, but the impact of Shoc2 mutants on the pathway activation is unclear. This study investigates how the NSLH-associated Shoc2 variants influence EGFR signals in the context of the ERK1/2 and AKT downstream signaling pathways. We show that when the ERK1/2 pathway is a primary signaling pathway activated downstream of EGFR, Shoc2 variants cannot upregulate ERK1/2 phosphorylation to the level of the WT Shoc2. Yet, when the AKT and ERK1/2 pathways were activated, in cells expressing Shoc2 variants, ERK1/2 phosphorylation was higher than in cells expressing WT Shoc2. In cells expressing the Shoc2 NSLH mutants, we found that the AKT signaling pathway triggers the PAK activation, followed by phosphorylation of Raf-1/MEK1/2 and activation of the ERK1/2 signaling axis. Hence, our studies reveal a previously unrecognized feedback regulation downstream of the EGFR and provide additional evidence for the role of Shoc2 as a "gatekeeper" in controlling the selection of downstream effectors within the EGFR signaling network.

14.
Proc Natl Acad Sci U S A ; 120(43): e2303794120, 2023 10 24.
Article in English | MEDLINE | ID: mdl-37844230

ABSTRACT

ß-arrestins are multivalent adaptor proteins that bind active phosphorylated G protein-coupled receptors (GPCRs) to inhibit G protein signaling, mediate receptor internalization, and initiate alternative signaling events. ß-arrestins link agonist-stimulated GPCRs to downstream signaling partners, such as the c-Raf-MEK1-ERK1/2 cascade leading to ERK1/2 activation. ß-arrestins have been thought to transduce signals solely via passive scaffolding by facilitating the assembly of multiprotein signaling complexes. Recently, however, ß-arrestin 1 and 2 were shown to activate two downstream signaling effectors, c-Src and c-Raf, allosterically. Over the last two decades, ERK1/2 have been the most intensely studied signaling proteins scaffolded by ß-arrestins. Here, we demonstrate that ß-arrestins play an active role in allosterically modulating ERK kinase activity in vitro and within intact cells. Specifically, we show that ß-arrestins and their GPCR-mediated active states allosterically enhance ERK2 autophosphorylation and phosphorylation of a downstream ERK2 substrate, and we elucidate the mechanism by which ß-arrestins do so. Furthermore, we find that allosteric stimulation of dually phosphorylated ERK2 by active-state ß-arrestin 2 is more robust than by active-state ß-arrestin 1, highlighting differential capacities of ß-arrestin isoforms to regulate effector signaling pathways downstream of GPCRs. In summary, our study provides strong evidence for a new paradigm in which ß-arrestins function as active "catalytic" scaffolds to allosterically unlock the enzymatic activity of signaling components downstream of GPCR activation.


Subject(s)
Arrestins , Signal Transduction , beta-Arrestins/metabolism , beta-Arrestin 1/genetics , beta-Arrestin 1/metabolism , Arrestins/metabolism , Allosteric Regulation , Signal Transduction/physiology , Receptors, G-Protein-Coupled/metabolism , Phosphorylation , beta-Arrestin 2/metabolism
15.
Proc Natl Acad Sci U S A ; 120(34): e2304071120, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37585458

ABSTRACT

Class IA phosphoinositide 3-kinase alpha (PI3Kα) is an important drug target because it is one of the most frequently mutated proteins in human cancers. However, small molecule inhibitors currently on the market or under development have safety concerns due to a lack of selectivity. Therefore, other chemical scaffolds or unique mechanisms of catalytic kinase inhibition are needed. Here, we report the cryo-electron microscopy structures of wild-type PI3Kα, the dimer of p110α and p85α, in complex with three Y-shaped ligands [cpd16 (compound 16), cpd17 (compound 17), and cpd18 (compound 18)] of different affinities and no inhibitory effect on the kinase activity. Unlike ATP-competitive inhibitors, cpd17 adopts a Y-shaped conformation with one arm inserted into a binding pocket formed by R770 and W780 and the other arm lodged in the ATP-binding pocket at an angle that is different from that of the ATP phosphate tail. Such a special interaction induces a conformation of PI3Kα resembling that of the unliganded protein. These observations were confirmed with two isomers (cpd16 and cpd18). Further analysis of these Y-shaped ligands revealed the structural basis of differential binding affinities caused by stereo- or regiochemical modifications. Our results may offer a different direction toward the design of therapeutic agents against PI3Kα.


Subject(s)
Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinases , Humans , Phosphatidylinositol 3-Kinases/metabolism , Ligands , Cryoelectron Microscopy , Adenosine Triphosphate/metabolism
16.
Trends Biochem Sci ; 46(5): 406-416, 2021 05.
Article in English | MEDLINE | ID: mdl-33446424

ABSTRACT

Bacterial microcompartments (MCPs) are extremely large (100-400 nm) and diverse proteinaceous organelles that compartmentalize multistep metabolic pathways, increasing their efficiency and sequestering toxic and/or volatile intermediates. This review highlights recent studies that have expanded our understanding of the diversity, structure, function, and potential biotechnological uses of MCPs. Several new types of MCPs have been identified and characterized revealing new functions and potential new associations with human disease. Recent structural studies of MCP proteins and recombinant MCP shells have provided new insights into MCP assembly and mechanisms and raised new questions about MCP structure. We also discuss recent work on biotechnology applications that use MCP principles to develop nanobioreactors, nanocontainers, and molecular scaffolds.


Subject(s)
Bacteria , Bacterial Proteins , Bacteria/metabolism , Bacterial Proteins/metabolism , Humans , Metabolic Networks and Pathways , Organelles/metabolism
17.
J Biol Chem ; 300(5): 107263, 2024 May.
Article in English | MEDLINE | ID: mdl-38582451

ABSTRACT

Synapse formation depends on the coordinated expression and regulation of scaffold proteins. The JNK family kinases play a role in scaffold protein regulation, but the nature of this functional interaction in dendritic spines requires further investigation. Here, using a combination of biochemical methods and live-cell imaging strategies, we show that the dynamics of the synaptic scaffold molecule SAP102 are negatively regulated by JNK inhibition, that SAP102 is a direct phosphorylation target of JNK3, and that SAP102 regulation by JNK is restricted to neurons that harbor mature synapses. We further demonstrate that SAP102 and JNK3 cooperate in the regulated trafficking of kainate receptors to the cell membrane. Specifically, we observe that SAP102, JNK3, and the kainate receptor subunit GluK2 exhibit overlapping expression at synaptic sites and that modulating JNK activity influences the surface expression of the kainate receptor subunit GluK2 in a neuronal context. We also show that SAP102 participates in this process in a JNK-dependent fashion. In summary, our data support a model in which JNK-mediated regulation of SAP102 influences the dynamic trafficking of kainate receptors to postsynaptic sites, and thus shed light on common pathophysiological mechanisms underlying the cognitive developmental defects associated with diverse mutations.


Subject(s)
Dendritic Spines , GluK2 Kainate Receptor , Receptors, Kainic Acid , Animals , Humans , Rats , Cell Membrane/metabolism , Dendritic Spines/metabolism , Hippocampus/metabolism , Hippocampus/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 10/genetics , Neurons/metabolism , Neuropeptides , Phosphorylation , Protein Transport , Receptors, Kainic Acid/metabolism , Receptors, Kainic Acid/genetics , Synapses/metabolism , Cells, Cultured
18.
Development ; 149(21)2022 11 01.
Article in English | MEDLINE | ID: mdl-36196585

ABSTRACT

The dentate gyrus, a gateway for input to the hippocampal formation, arises from progenitors in the medial telencephalic neuroepithelium adjacent to the cortical hem. Dentate progenitors navigate a complex migratory path guided by two cell populations that arise from the hem, the fimbrial glia and Cajal-Retzius (CR) cells. As the hem expresses multiple Wnt genes, we examined whether ß-catenin, which mediates canonical Wnt signaling and also participates in cell adhesion, is necessary for the development of hem-derived lineages. We report that, in mice, the fimbrial glial scaffold is disorganized and CR cells are mispositioned upon hem-specific disruption of ß-catenin. Consequently, the dentate migratory stream is severely affected, and the dentate gyrus fails to form. Using selective Cre drivers, we further determined that ß-catenin function is required in the fimbrial glial scaffold, but not in the CR cells, for guiding the dentate migration. Our findings highlight a primary requirement for ß-catenin for the organization of the fimbrial scaffold and a secondary role for this factor in dentate gyrus morphogenesis.


Subject(s)
Dentate Gyrus , Morphogenesis , beta Catenin , Animals , Mice , beta Catenin/metabolism , Dentate Gyrus/metabolism , Hippocampus/metabolism , Morphogenesis/genetics , Neuroglia/metabolism , Neurons/metabolism
19.
Brief Bioinform ; 24(2)2023 03 19.
Article in English | MEDLINE | ID: mdl-36754843

ABSTRACT

Scaffold proteins drive liquid-liquid phase separation (LLPS) to form biomolecular condensates and organize various biochemical reactions in cells. Dysregulation of scaffolds can lead to aberrant condensate assembly and various complex diseases. However, bioinformatics predictors dedicated to scaffolds are still lacking and their development suffers from an extreme imbalance between limited experimentally identified scaffolds and unlabeled candidates. Here, using the joint distribution of hybrid multimodal features, we implemented a positive unlabeled (PU) learning-based framework named PULPS that combined ProbTagging and penalty logistic regression (PLR) to profile the propensity of scaffolds. PULPS achieved the best AUC of 0.8353 and showed an area under the lift curve (AUL) of 0.8339 as an estimation of true performance. Upon reviewing recent experimentally verified scaffolds, we performed a partial recovery with 2.85% increase in AUL from 0.8339 to 0.8577. In comparison, PULPS showed a 45.7% improvement in AUL compared with PLR, whereas 8.2% superiority over other existing tools. Our study first proved that PU learning is more suitable for scaffold prediction and demonstrated the widespread existence of phase separation states. This profile also uncovered potential scaffolds that co-drive LLPS in the human proteome and generated candidates for further experiments. PULPS is free for academic research at http://pulps.zbiolab.cn.


Subject(s)
Cell Physiological Phenomena , Proteome , Humans
20.
Cell Mol Life Sci ; 81(1): 216, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740643

ABSTRACT

p50RhoGAP is a key protein that interacts with and downregulates the small GTPase RhoA. p50RhoGAP is a multifunctional protein containing the BNIP-2 and Cdc42GAP Homology (BCH) domain that facilitates protein-protein interactions and lipid binding and the GAP domain that regulates active RhoA population. We recently solved the structure of the BCH domain from yeast p50RhoGAP (YBCH) and showed that it maintains the adjacent GAP domain in an auto-inhibited state through the ß5 strand. Our previous WT YBCH structure shows that a unique kink at position 116 thought to be made by a proline residue between alpha helices α6 and α7 is essential for the formation of intertwined dimer from asymmetric monomers. Here we sought to establish the role and impact of this Pro116. However, the kink persists in the structure of P116A mutant YBCH domain, suggesting that the scaffold is not dictated by the proline residue at this position. We further identified Tyr124 (or Tyr188 in HBCH) as a conserved residue in the crucial ß5 strand. Extending to the human ortholog, when substituted to acidic residues, Tyr188D or Tyr188E, we observed an increase in RhoA binding and self-dimerization, indicative of a loss of inhibition of the GAP domain by the BCH domain. These results point to distinct roles and impact of the non-conserved and conserved amino acid positions in regulating the structural and functional complexity of the BCH domain.


Subject(s)
GTPase-Activating Proteins , Proline , Schizosaccharomyces pombe Proteins , Humans , Amino Acid Sequence , Conserved Sequence , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Models, Molecular , Proline/metabolism , Proline/chemistry , Proline/genetics , Protein Binding , Protein Domains , rhoA GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/chemistry , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Tyrosine/metabolism , Tyrosine/chemistry , Tyrosine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL