Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Biochemistry ; 63(9): 1067-1074, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38619104

ABSTRACT

NANOG protein levels correlate with stem cell pluripotency. NANOG concentrations fluctuate constantly with low NANOG levels leading to spontaneous cell differentiation. Previous literature implicated Pin1, a phosphorylation-dependent prolyl isomerase, as a key player in NANOG stabilization. Here, using NMR spectroscopy, we investigate the molecular interactions of Pin1 with the NANOG unstructured N-terminal domain that contains a PEST sequence with two phosphorylation sites. Phosphorylation of NANOG PEST peptides increases affinity to Pin1. By systematically increasing the amount of cis PEST conformers, we show that the peptides bind tighter to the prolyl isomerase domain (PPIase) of Pin1. Phosphorylation and cis Pro enhancement at both PEST sites lead to a 5-10-fold increase in NANOG binding to the Pin1 WW domain and PPIase domain, respectively. The cis-populated NANOG PEST peptides can be potential inhibitors for disrupting Pin1-dependent NANOG stabilization in cancer stem cells.


Subject(s)
NIMA-Interacting Peptidylprolyl Isomerase , Nanog Homeobox Protein , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/chemistry , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , Phosphorylation , Humans , Protein Stability , Protein Binding , Stereoisomerism
2.
Nat Chem Biol ; 20(4): 399-400, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38326412
3.
Int J Mol Sci ; 24(4)2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36834792

ABSTRACT

Cellular deposition of protein aggregates, one of the hallmarks of neurodegeneration, disrupts cellular functions and leads to neuronal death. Mutations, posttranslational modifications, and truncations are common molecular underpinnings in the formation of aberrant protein conformations that seed aggregation. The major proteins involved in neurodegeneration include amyloid beta (Aß) and tau in Alzheimer's disease, α-synuclein in Parkinson's disease, and TAR DNA-binding protein (TDP-43) in amyotrophic lateral sclerosis (ALS). These proteins are described as intrinsically disordered and possess enhanced ability to partition into biomolecular condensates. In this review, we discuss the role of protein misfolding and aggregation in neurodegenerative diseases, specifically highlighting implications of changes to the primary/secondary (mutations, posttranslational modifications, and truncations) and the quaternary/supramolecular (oligomerization and condensation) structural landscapes for the four aforementioned proteins. Understanding these aggregation mechanisms provides insights into neurodegenerative diseases and their common underlying molecular pathology.


Subject(s)
Alzheimer Disease , Amyotrophic Lateral Sclerosis , Intrinsically Disordered Proteins , Neurodegenerative Diseases , Parkinson Disease , Humans , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyotrophic Lateral Sclerosis/metabolism , Parkinson Disease/metabolism , tau Proteins
4.
Methods Mol Biol ; 2563: 135-148, 2023.
Article in English | MEDLINE | ID: mdl-36227471

ABSTRACT

Biomolecular condensates of ribonucleoproteins (RNPs) such as the transactivation response element (TAR) DNA-binding protein 43 (TDP-43) arise from liquid-liquid phase separation (LLPS) and play vital roles in various biological processes including the formation-dissolution of stress granules (SGs). These condensates are thought to be directly linked to neurodegenerative diseases, providing a depot of aggregation-prone proteins and serving as a cauldron of protein aggregation and fibrillation. Despite recent research efforts, biochemical processes and rearrangements within biomolecular condensates that trigger subsequent protein misfolding and aggregation remain to be elucidated. Fluorescence lifetime imaging microscopy (FLIM) provides a minimally intrusive high-sensitivity and high-resolution imaging method to monitor in-droplet spatiotemporal changes that initiate and lead to protein aggregation. In this chapter, we describe a FLIM application for characterizing chemical chaperone-assisted decoupling of TDP-43 liquid-liquid phase separation and aggregation/fibrillation, highlighting potential therapeutic strategies to combat pathological RNP-associated aggregates without compromising cellular stress responses.


Subject(s)
Biomolecular Condensates , Protein Aggregates , DNA-Binding Proteins/metabolism , Microscopy, Fluorescence , Ribonucleoproteins/metabolism
5.
Nat Cell Biol ; 24(5): 737-747, 2022 05.
Article in English | MEDLINE | ID: mdl-35484250

ABSTRACT

Human NANOG expression resets stem cells to ground-state pluripotency. Here we identify the unique features of human NANOG that relate to its dose-sensitive function as a master transcription factor. NANOG is largely disordered, with a C-terminal prion-like domain that phase-transitions to gel-like condensates. Full-length NANOG readily forms higher-order oligomers at low nanomolar concentrations, orders of magnitude lower than typical amyloids. Using single-molecule Förster resonance energy transfer and fluorescence cross-correlation techniques, we show that NANOG oligomerization is essential for bridging DNA elements in vitro. Using chromatin immunoprecipitation sequencing and Hi-C 3.0 in cells, we validate that NANOG prion-like domain assembly is essential for specific DNA recognition and distant chromatin interactions. Our results provide a physical basis for the indispensable role of NANOG in shaping the pluripotent genome. NANOG's unique ability to form prion-like assemblies could provide a cooperative and concerted DNA bridging mechanism that is essential for chromatin reorganization and dose-sensitive activation of ground-state pluripotency.


Subject(s)
Chromatin , Prions , Chromatin/genetics , DNA/genetics , Humans , Nanog Homeobox Protein/genetics , Prions/genetics
6.
Nat Commun ; 12(1): 5579, 2021 09 22.
Article in English | MEDLINE | ID: mdl-34552088

ABSTRACT

Expression of a few master transcription factors can reprogram the epigenetic landscape and three-dimensional chromatin topology of differentiated cells and achieve pluripotency. During reprogramming, thousands of long-range chromatin contacts are altered, and changes in promoter association with enhancers dramatically influence transcription. Molecular participants at these sites have been identified, but how this re-organization might be orchestrated is not known. Biomolecular condensation is implicated in subcellular organization, including the recruitment of RNA polymerase in transcriptional activation. Here, we show that reprogramming factor KLF4 undergoes biomolecular condensation even in the absence of its intrinsically disordered region. Liquid-liquid condensation of the isolated KLF4 DNA binding domain with a DNA fragment from the NANOG proximal promoter is enhanced by CpG methylation of a KLF4 cognate binding site. We propose KLF4-mediated condensation as one mechanism for selectively organizing and re-organizing the genome based on the local sequence and epigenetic state.


Subject(s)
Cellular Reprogramming , Chromatin/metabolism , DNA/metabolism , Kruppel-Like Transcription Factors/metabolism , Base Sequence , Cell Line , Cell Nucleus/metabolism , DNA/chemistry , DNA/genetics , DNA Methylation , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/genetics , Models, Molecular , Mutation , Nanog Homeobox Protein/genetics , Octamer Transcription Factor-3/genetics , Promoter Regions, Genetic , Protein Interaction Domains and Motifs , SOXB1 Transcription Factors/genetics , Zinc Fingers/genetics
7.
Protein Sci ; 30(7): 1408-1417, 2021 07.
Article in English | MEDLINE | ID: mdl-33982369

ABSTRACT

Membrane-less organelles and RNP granules are enriched in RNA and RNA-binding proteins containing disordered regions. Heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1), a key regulating protein in RNA metabolism, localizes to cytoplasmic RNP granules including stress granules. Dysfunctional nuclear-cytoplasmic transport and dynamic phase separation of hnRNPA1 leads to abnormal amyloid aggregation and neurodegeneration. The intrinsically disordered C-terminal domain (CTD) of hnRNPA1 mediates both dynamic liquid-liquid phase separation (LLPS) and aggregation. While cellular phase separation drives the formation of membrane-less organelles, aggregation within phase-separated compartments has been linked to neurodegenerative diseases. To understand some of the underlying mechanisms behind protein phase separation and LLPS-mediated aggregation, we studied LLPS of hnRNPA1 CTD in conditions that probe protein electrostatics, modulated specifically by varying pH conditions, and protein, salt and RNA concentrations. In the conditions investigated, we observed LLPS to be favored in acidic conditions, and by high protein, salt and RNA concentrations. We also observed that conditions that favor LLPS also enhance protein aggregation and fibrillation, which suggests an aggregation pathway that is LLPS-mediated. The results reported here also suggest that LLPS can play a direct role in facilitating protein aggregation, and that changes in cellular environment that affect protein electrostatics can contribute to the pathological aggregation exhibited in neurodegeneration.


Subject(s)
Heterogeneous Nuclear Ribonucleoprotein A1/chemistry , Intrinsically Disordered Proteins/chemistry , Protein Aggregates , Humans , Protein Domains , Static Electricity
8.
Semin Cell Dev Biol ; 99: 78-85, 2020 03.
Article in English | MEDLINE | ID: mdl-29753880

ABSTRACT

Intrinsically disordered proteins as computationally predicted account for ∼1/3 of eukaryotic proteomes, are involved in a plethora of biological functions, and have been linked to several human diseases as a result of their dysfunctions. Here, we present a picture wherein an energetic continuum describes protein structural and conformational propensities, ranging from the hyperstable folded proteins on one end to the hyperdestabilized and sometimes functionally disordered proteins on the other. We distinguish between proteins that are folding-competent but disordered because of marginal stability and those that are disordered due mainly to the absence of folding code-completing structure-determining interactions, and postulate that disordered proteins that are unstructured by way of partial population of protein denatured states represent a sizable proportion of the proteome.


Subject(s)
Intrinsically Disordered Proteins/metabolism , Humans , Intrinsically Disordered Proteins/chemistry , Ligands , Protein Conformation , Protein Folding , Proteome/chemistry , Proteome/metabolism
9.
Nucleic Acids Res ; 48(5): 2621-2642, 2020 03 18.
Article in English | MEDLINE | ID: mdl-31863590

ABSTRACT

Transposable elements (TEs) comprise a large proportion of long non-coding RNAs (lncRNAs). Here, we employed CRISPR to delete a short interspersed nuclear element (SINE) in Malat1, a cancer-associated lncRNA, to investigate its significance in cellular physiology. We show that Malat1 with a SINE deletion forms diffuse nuclear speckles and is frequently translocated to the cytoplasm. SINE-deleted cells exhibit an activated unfolded protein response and PKR and markedly increased DNA damage and apoptosis caused by dysregulation of TDP-43 localization and formation of cytotoxic inclusions. TDP-43 binds stronger to Malat1 without the SINE and is likely 'hijacked' by cytoplasmic Malat1 to the cytoplasm, resulting in the depletion of nuclear TDP-43 and redistribution of TDP-43 binding to repetitive element transcripts and mRNAs encoding mitotic and nuclear-cytoplasmic regulators. The SINE promotes Malat1 nuclear retention by facilitating Malat1 binding to HNRNPK, a protein that drives RNA nuclear retention, potentially through direct interactions of the SINE with KHDRBS1 and TRA2A, which bind to HNRNPK. Losing these RNA-protein interactions due to the SINE deletion likely creates more available TDP-43 binding sites on Malat1 and subsequent TDP-43 aggregation. These results highlight the significance of lncRNA TEs in TDP-43 proteostasis with potential implications in both cancer and neurodegenerative diseases.


Subject(s)
DNA-Binding Proteins/metabolism , Proteostasis/genetics , RNA, Long Noncoding/genetics , Short Interspersed Nucleotide Elements/genetics , Apoptosis , Cell Line , Cytoplasm/metabolism , DNA Damage , Endoplasmic Reticulum Stress , Enzyme Activation , Gene Dosage , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Humans , Mitosis , Models, Biological , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion/genetics , eIF-2 Kinase
10.
J Virol ; 93(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31375595

ABSTRACT

Influenza A virus (IAV) nonstructural protein 1 (NS1), a potent antagonist of the host immune response, is capable of interacting with RNA and a wide range of cellular proteins. NS1 consists of an RNA-binding domain (RBD) and an effector domain (ED) separated by a flexible linker region (LR). H5N1-NS1 has a characteristic 5-residue deletion in the LR, with either G (minor group) or E (major group) at the 71st position, and non-H5N1-NS1 contains E71 with an intact linker. Based on the orientation of the ED with respect to the RBD, previous crystallographic studies have shown that minor group H5N1-NS1(G71), a non-H5N1-NS1 [H6N6-NS1(E71)], and the LR deletion mutant H6N6-NS1(Δ80-84/E71) mimicking the major group H5N1-NS1 exhibit "open," "semiopen," and "closed" conformations, respectively, suggesting that NS1 exhibits a strain-dependent conformational preference. Here we report the first crystal structure of a naturally occurring H5N1-NS1(E71) and show that it adopts an open conformation similar to that of the minor group of H5N1-NS1 [H5N1-NS1(G71)]. We also show that H6N6-NS1(Δ80-84/E71) under a different crystallization condition and H6N6-NS1(Δ80-84/G71) also exhibit open conformations, suggesting that NS1 can adopt an open conformation irrespective of E or G at the 71st position. Our single-molecule fluorescence resonance energy transfer (FRET) analysis to investigate the conformational preference of NS1 in solution showed that all NS1 constructs predominantly exist in an open conformation. Further, our coimmunoprecipitation and binding studies showed that they all bind to cellular factors with similar affinities. Taken together, our studies suggest that NS1 exhibits strain-independent structural plasticity that allows it to interact with a wide variety of cellular ligands during viral infection.IMPORTANCE IAV is responsible for several pandemics over the last century and continues to infect millions annually. The frequent rise in drug-resistant strains necessitates exploring novel targets for developing antiviral drugs that can reduce the global burden of influenza infection. Because of its critical role in the replication and pathogenesis of IAV, nonstructural protein 1 (NS1) is a potential target for developing antivirals. Previous studies suggested that NS1 adopts strain-dependent "open," "semiopen," and "closed" conformations. Here we show, based on three crystal structures, that NS1 irrespective of strain differences can adopt an open conformation. We further show that NS1 from different strains primarily exists in an open conformation in solution and binds to cellular proteins with a similar affinity. Together, our findings suggest that conformational polymorphism facilitated by a flexible linker is intrinsic to NS1, and this may be the underlying factor allowing NS1 to bind several cellular factors during IAV replication.


Subject(s)
Influenza A virus/chemistry , Viral Nonstructural Proteins/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Fluorescence Resonance Energy Transfer , Influenza A virus/classification , Influenza A virus/genetics , Ligands , Mutation , Protein Binding , Protein Conformation , Structure-Activity Relationship , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
11.
Methods Mol Biol ; 1948: 221-233, 2019.
Article in English | MEDLINE | ID: mdl-30771181

ABSTRACT

Misfolding and aggregation of α-synuclein are linked to many neurodegenerative disorders, including Parkinson's and Alzheimer's disease. Despite intense research efforts, detailed structural characterization of early conformational transitions that initiate and drive α-synuclein aggregation remains elusive often due to the low sensitivity and ensemble averaging of commonly used techniques. Single-molecule Förster resonance energy transfer (smFRET) provides unique advantages in detecting minor conformations that initiate protein pathologic aggregation. In this chapter, we describe an smFRET-based method for characterizing early conformational conversions that are responsible for α-synuclein self-assembly and aggregation.


Subject(s)
Fluorescence Resonance Energy Transfer , Protein Aggregation, Pathological , Protein Conformation , alpha-Synuclein/chemistry , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Neurodegenerative Diseases/metabolism , Protein Aggregation, Pathological/metabolism , Protein Folding , Spectrum Analysis , alpha-Synuclein/metabolism
12.
Biochemistry ; 57(50): 6822-6826, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30520303

ABSTRACT

Ribonucleoprotein (RNP) condensations through liquid-liquid phase separation play vital roles in the dynamic formation-dissolution of stress granules (SGs). These condensations are, however, usually assumed to be linked to pathologic fibrillation. Here, we show that physiologic condensation and pathologic fibrillation of RNPs are independent processes that can be unlinked with the chemical chaperone trimethylamine N-oxide (TMAO). Using the low-complexity disordered domain of the archetypical SG-protein TDP-43 as a model system, we show that TMAO enhances RNP liquid condensation yet inhibits protein fibrillation. Our results demonstrate effective decoupling of physiologic condensation from pathologic aggregation and suggest that selective targeting of protein fibrillation (without altering condensation) can be employed as a therapeutic strategy for RNP aggregation-associated degenerative disorders.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Cytoplasmic Granules/chemistry , Cytoplasmic Granules/metabolism , DNA-Binding Proteins/genetics , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Liquid-Liquid Extraction , Methylamines/chemistry , Methylamines/metabolism , Microscopy, Fluorescence , Models, Biological , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Mutation , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/metabolism , Unfolded Protein Response
13.
Int J Mol Sci ; 19(12)2018 Dec 04.
Article in English | MEDLINE | ID: mdl-30518054

ABSTRACT

Sox2 is a pioneer transcription factor that initiates cell fate reprogramming through locus-specific differential regulation. Mechanistically, it was assumed that Sox2 achieves its regulatory diversity via heterodimerization with partner transcription factors. Here, utilizing single-molecule fluorescence spectroscopy, we show that Sox2 alone can modulate DNA structural landscape in a dosage-dependent manner. We propose that such stoichiometric tuning of regulatory DNAs is crucial to the diverse biological functions of Sox2, and represents a generic mechanism of conferring functional plasticity and multiplicity to transcription factors.


Subject(s)
DNA/chemistry , HMG-Box Domains , Nucleic Acid Conformation , SOXB1 Transcription Factors/chemistry , Single Molecule Imaging , Fluorescence Resonance Energy Transfer , Models, Molecular , Promoter Regions, Genetic/genetics , Protein Binding
14.
Nat Commun ; 9(1): 2631, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29980685

ABSTRACT

Rotaviruses (RVs) cause life-threatening diarrhea in infants and children worldwide. Recent biochemical and epidemiological studies underscore the importance of histo-blood group antigens (HBGA) as both cell attachment and susceptibility factors for the globally dominant P[4], P[6], and P[8] genotypes of human RVs. How these genotypes interact with HBGA is not known. Here, our crystal structures of P[4] and a neonate-specific P[6] VP8*s alone and in complex with H-type I HBGA reveal a unique glycan binding site that is conserved in the globally dominant genotypes and allows for the binding of ABH HBGAs, consistent with their prevalence. Remarkably, the VP8* of P[6] RVs isolated from neonates displays subtle structural changes in this binding site that may restrict its ability to bind branched glycans. This provides a structural basis for the age-restricted tropism of some P[6] RVs as developmentally regulated unbranched glycans are more abundant in the neonatal gut.


Subject(s)
Polysaccharides/metabolism , Rotavirus/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Blood Group Antigens/biosynthesis , Cell Line , Conserved Sequence , Crystallography, X-Ray , Fucose/metabolism , Humans , Infant, Newborn , Models, Molecular , Mutation/genetics , Polysaccharides/chemistry , RNA-Binding Proteins/chemistry , Rotavirus/pathogenicity , Rotavirus Infections/pathology , Rotavirus Infections/virology , Viral Nonstructural Proteins/chemistry
15.
Proc Natl Acad Sci U S A ; 115(23): 5962-5967, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29784777

ABSTRACT

The phosphoenolpyruvate-dependent phosphotransferase system (PTS) transports sugar into bacteria and phosphorylates the sugar for metabolic consumption. The PTS is important for the survival of bacteria and thus a potential target for antibiotics, but its mechanism of sugar uptake and phosphorylation remains unclear. The PTS is composed of multiple proteins, and the membrane-embedded Enzyme IIC (EIIC) component transports sugars across the membrane. Crystal structures of two members of the glucose superfamily of EIICs, bcChbC and bcMalT, were solved in the inward-facing and outward-facing conformations, and the structures suggest that sugar translocation could be achieved by movement of a structured domain that contains the sugar-binding site. However, different conformations have not been captured on the same transporter to allow precise description of the conformational changes. Here we present a crystal structure of bcMalT trapped in an inward-facing conformation by a mercury ion that bridges two strategically placed cysteine residues. The structure allows direct comparison of the outward- and inward-facing conformations and reveals a large rigid-body motion of the sugar-binding domain and other conformational changes that accompany the rigid-body motion. All-atom molecular dynamics simulations show that the inward-facing structure is stable with or without the cross-linking. The conformational changes were further validated by single-molecule Föster resonance energy transfer (smFRET). Combined, these results establish the elevator-type mechanism of transport in the glucose superfamily of EIIC transporters.


Subject(s)
Bacterial Proteins , Phosphoenolpyruvate Sugar Phosphotransferase System , Bacillus cereus/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Biological Transport , Cysteine/chemistry , Cysteine/metabolism , Fluorescence Resonance Energy Transfer , Molecular Dynamics Simulation , Phosphoenolpyruvate Sugar Phosphotransferase System/chemistry , Phosphoenolpyruvate Sugar Phosphotransferase System/metabolism , Phosphoenolpyruvate Sugar Phosphotransferase System/ultrastructure , Phosphorylation , Protein Conformation
16.
Eur Biophys J ; 47(1): 89-94, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29080139

ABSTRACT

Protein thermodynamic stability is intricately linked to cellular function, and altered stability can lead to dysfunction and disease. The linear extrapolation model (LEM) is commonly used to obtain protein unfolding free energies ([Formula: see text]) by extrapolation of solvent denaturation data to zero denaturant concentration. However, for some proteins, different denaturants result in non-coincident LEM-derived [Formula: see text] values, raising questions about the inherent assumption that the obtained [Formula: see text] values are intrinsic to the protein. Here, we used single-molecule FRET measurements to better understand such discrepancies by directly probing changes in the dimensions of the protein G B1 domain (GB1), a well-studied protein folding model, upon urea and guanidine hydrochloride denaturation. A comparison of the results for the two denaturants suggests denaturant-specific structural energetics in the GB1 denatured ensemble, revealing a role of the denatured state in the variable thermodynamic behavior of proteins.


Subject(s)
Bacterial Proteins/chemistry , Protein Denaturation/drug effects , Fluorescence Resonance Energy Transfer , Guanidine/pharmacology , Protein Domains , Thermodynamics , Urea/pharmacology
18.
Angew Chem Int Ed Engl ; 56(41): 12590-12593, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28833982

ABSTRACT

Transactivation response element (TAR) DNA-binding protein 43 (TDP-43) misfolding is implicated in several neurodegenerative diseases characterized by aggregated protein inclusions. Misfolding is believed to be mediated by both the N- and C-terminus of TDP-43; however, the mechanistic basis of the contribution of individual domains in the process remained elusive. Here, using single-molecule fluorescence and ensemble biophysical techniques, and a wide range of pH and temperature conditions, we show that TDP-43NTD is thermodynamically stable, well-folded and undergoes reversible oligomerization. We propose that, in full-length TDP-43, association between folded N-terminal domains enhances the propensity of the intrinsically unfolded C-terminal domains to drive pathological aggregation.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/chemistry , Protein Folding , Humans , Hydrogen-Ion Concentration , Models, Molecular , Protein Aggregation, Pathological/pathology , Protein Domains , Protein Multimerization , Thermodynamics
19.
Nat Commun ; 8: 16087, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28703127

ABSTRACT

Glutathione plays many important roles in biological processes; however, the dynamic changes of glutathione concentrations in living cells remain largely unknown. Here, we report a reversible reaction-based fluorescent probe-designated as RealThiol (RT)-that can quantitatively monitor the real-time glutathione dynamics in living cells. Using RT, we observe enhanced antioxidant capability of activated neurons and dynamic glutathione changes during ferroptosis. RT is thus a versatile tool that can be used for both confocal microscopy and flow cytometry based high-throughput quantification of glutathione levels in single cells. We envision that this new glutathione probe will enable opportunities to study glutathione dynamics and transportation and expand our understanding of the physiological and pathological roles of glutathione in living cells.


Subject(s)
Fluorescent Dyes , Fluorometry/methods , Glutathione/analysis , Glutathione/chemistry , HeLa Cells , Humans , Kinetics , Single-Cell Analysis
20.
Chemphyschem ; 16(1): 90-4, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25345588

ABSTRACT

Intrinsically disordered proteins (IDPs) are involved in diverse cellular functions. Many IDPs can interact with multiple binding partners, resulting in their folding into alternative ligand-specific functional structures. For such multi-structural IDPs, a key question is whether these multiple structures are fully encoded in the protein sequence, as is the case in many globular proteins. To answer this question, here we employed a combination of single-molecule and ensemble techniques to compare ligand-induced and osmolyte-forced folding of α-synuclein. Our results reveal context-dependent modulation of the protein's folding landscape, suggesting that the codes for the protein's native folds are partially encoded in its primary sequence, and are completed only upon interaction with binding partners. Our findings suggest a critical role for cellular interactions in expanding the repertoire of folds and functions available to disordered proteins.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Protein Folding , alpha-Synuclein/chemistry , Ligands
SELECTION OF CITATIONS
SEARCH DETAIL
...