Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38999934

ABSTRACT

Biomolecular condensates (BMCs) exhibit physiological and pathological relevance in biological systems. Both liquid and solid condensates play significant roles in the spatiotemporal regulation and organization of macromolecules and their biological activities. Some pathological solid condensates, such as Lewy Bodies and other fibrillar aggregates, have been hypothesized to originate from liquid condensates. With the prevalence of BMCs having functional and dysfunctional roles, it is imperative to understand the mechanism of biomolecular condensate formation and initiation. Using the low-complexity domain (LCD) of heterogenous ribonuclear protein A1 (hnRNPA1) as our model, we monitored initial assembly events using dynamic light scattering (DLS) while modulating pH and salt conditions to perturb macromolecule and condensate properties. We observed the formation of nanometer-sized BMCs (nano-condensates) distinct from protein monomers and micron-sized condensates. We also observed that conditions that solubilize micron-sized protein condensates do not solubilize nano-condensates, indicating that the balance of forces that stabilize nano-condensates and micron-sized condensates are distinct. These findings provide insight into the forces that drive protein phase separation and potential nucleation structures of macromolecular condensation.


Subject(s)
Dynamic Light Scattering , Heterogeneous Nuclear Ribonucleoprotein A1 , Humans , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1/chemistry , Protein Domains , Biomolecular Condensates/chemistry , Biomolecular Condensates/metabolism , Hydrogen-Ion Concentration
2.
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
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.
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
5.
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
6.
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
7.
Langmuir ; 35(9): 3391-3403, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30712354

ABSTRACT

The synthesis of a novel poly(propyleneimine) (PPI) dendron in gram scale as well as its use in the formation of a highly stable, dendronized gold nanoparticle (AuNP)-based drug delivery platform is described herein. The AuNP-based platform is composed of three complementary parts: (i) a 15 nm AuNP core, (ii) a heterofunctional thioctic acid-terminated tetraethylene glycol spacer, and (iii) a third-generation PPI dendron with a unique protonation profile and diverse end-group functionalization that allows for further derivatization. The prepared dendronized AuNPs are able to withstand several rounds of lyophilization cycles with no sign of aggregation, are stable in phosphate-buffered saline and Hanks' buffer as well as in serum, and are resistant to degradation by glutathione exchange reactions. This nanocarrier platform displays a dense coating, with >1400 dendrons/AuNPs, which will enable very high payload. Furthermore, while amine-terminated AuNPs expectedly showed cytotoxicity against the MCF-7 breast cancer cell line from a NP concentration of 1 nM, the mixed monolayer AuNPs (coated with 40/60 amine/carboxylate dendrons) interestingly did not exhibit any sign of toxicity at concentrations as high as 15 nM, similar to the carboxylate-terminated AuNPs. The described dendronized AuNPs address the current practical need for a stable NP-based drug delivery platform which is scalable and easily conjugable, has long-term stability in solution, and can be conveniently formulated as a powder and redispersed in desired buffer or serum.


Subject(s)
Dendrimers/chemistry , Metal Nanoparticles/chemistry , Dendrimers/chemical synthesis , Dendrimers/toxicity , Gold/chemistry , Humans , MCF-7 Cells , Metal Nanoparticles/toxicity , Polypropylenes/chemical synthesis , Polypropylenes/chemistry , Polypropylenes/toxicity , Propionates/chemical synthesis , Propionates/chemistry , Propionates/toxicity , Thioctic Acid/analogs & derivatives , Thioctic Acid/chemical synthesis , Thioctic Acid/toxicity
8.
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
9.
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
10.
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
11.
Int J Mol Sci ; 19(5)2018 May 04.
Article in English | MEDLINE | ID: mdl-29734651

ABSTRACT

Neuropathological aggregates of the intrinsically disordered microtubule-associated protein Tau are hallmarks of Alzheimer’s disease, with decades of research devoted to studying the protein’s aggregation properties both in vitro and in vivo. Recent demonstrations that Tau is capable of undergoing liquid-liquid phase separation (LLPS) reveal the possibility that protein-enriched phase separated compartments could serve as initiation sites for Tau aggregation, as shown for other amyloidogenic proteins, such as the Fused in Sarcoma protein (FUS) and TAR DNA-binding protein-43 (TDP-43). Although truncation, mutation, and hyperphosphorylation have been shown to enhance Tau LLPS and aggregation, the effect of hyperacetylation on Tau aggregation remains unclear. Here, we investigate how the acetylation of Tau affects its potential to undergo phase separation and aggregation. Our data show that the hyperacetylation of Tau by p300 histone acetyltransferase (HAT) disfavors LLPS, inhibits heparin-induced aggregation, and impedes access to LLPS-initiated microtubule assembly. We propose that Tau acetylation prevents the toxic effects of LLPS-dependent aggregation but, nevertheless, contributes to Tau loss-of-function pathology by inhibiting Tau LLPS-mediated microtubule assembly.


Subject(s)
Alzheimer Disease/metabolism , Protein Aggregation, Pathological/metabolism , p300-CBP Transcription Factors/metabolism , tau Proteins/metabolism , Acetylation , Alzheimer Disease/genetics , Alzheimer Disease/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heparin/chemistry , Humans , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Liquid-Liquid Extraction , Microtubules/genetics , Microtubules/metabolism , Phosphorylation , Protein Aggregation, Pathological/genetics , p300-CBP Transcription Factors/genetics , tau Proteins/chemistry , tau Proteins/genetics
12.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...