Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Cell ; 186(22): 4898-4919.e25, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37827155

ABSTRACT

Expansions of repeat DNA tracts cause >70 diseases, and ongoing expansions in brains exacerbate disease. During expansion mutations, single-stranded DNAs (ssDNAs) form slipped-DNAs. We find the ssDNA-binding complexes canonical replication protein A (RPA1, RPA2, and RPA3) and Alternative-RPA (RPA1, RPA3, and primate-specific RPA4) are upregulated in Huntington disease and spinocerebellar ataxia type 1 (SCA1) patient brains. Protein interactomes of RPA and Alt-RPA reveal unique and shared partners, including modifiers of CAG instability and disease presentation. RPA enhances in vitro melting, FAN1 excision, and repair of slipped-CAGs and protects against CAG expansions in human cells. RPA overexpression in SCA1 mouse brains ablates expansions, coincident with decreased ATXN1 aggregation, reduced brain DNA damage, improved neuron morphology, and rescued motor phenotypes. In contrast, Alt-RPA inhibits melting, FAN1 excision, and repair of slipped-CAGs and promotes CAG expansions. These findings suggest a functional interplay between the two RPAs where Alt-RPA may antagonistically offset RPA's suppression of disease-associated repeat expansions, which may extend to other DNA processes.


Subject(s)
Replication Protein A , Trinucleotide Repeat Expansion , Animals , Humans , Mice , DNA/genetics , DNA Mismatch Repair , Huntington Disease/genetics , Proteins/genetics , Spinocerebellar Ataxias/genetics , Replication Protein A/metabolism
2.
Nucleic Acids Res ; 52(5): 2578-2589, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38261972

ABSTRACT

The loading of RecA onto ssDNA by RecBCD is an essential step of RecBCD-mediated homologous recombination. RecBCD facilitates RecA-loading onto ssDNA in a χ-dependent manner via its RecB nuclease domain (RecBn). Before recognition of χ, RecBn is sequestered through interactions with RecBCD. It was proposed that upon χ-recognition, RecBn undocks, allowing RecBn to swing out via a contiguous 70 amino acid linker to reveal the RecA-loading surface, and then recruit and load RecA onto ssDNA. We tested this hypothesis by examining the interactions between RecBn (RecB928-1180) and truncated RecBCD (RecB1-927CD) lacking the nuclease domain. The reconstituted complex of RecB1-927CD and RecBn is functional in vitro and in vivo. Our results indicate that despite being covalently severed from RecB1-927CD, RecBn can still load RecA onto ssDNA, establishing that RecBn does not function while only remaining tethered to the RecBCD complex via the linker. Instead, RecBCD undergoes a χ-induced intramolecular rearrangement to reveal the RecA-loading surface.


Subject(s)
Escherichia coli Proteins , Exodeoxyribonuclease V , Rec A Recombinases , DNA, Single-Stranded/genetics , Endonucleases/metabolism , Escherichia coli Proteins/metabolism , Exodeoxyribonuclease V/metabolism , Exodeoxyribonucleases/metabolism , Rec A Recombinases/metabolism
3.
Methods ; 224: 47-53, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38387709

ABSTRACT

Nucleotide excision repair (NER) promotes genomic integrity by removing bulky DNA adducts introduced by external factors such as ultraviolet light. Defects in NER enzymes are associated with pathological conditions such as Xeroderma Pigmentosum, trichothiodystrophy, and Cockayne syndrome. A critical step in NER is the binding of the Xeroderma Pigmentosum group A protein (XPA) to the ss/ds DNA junction. To better capture the dynamics of XPA interactions with DNA during NER we have utilized the fluorescence enhancement through non-canonical amino acids (FEncAA) approach. 4-azido-L-phenylalanine (4AZP or pAzF) was incorporated at Arg-158 in human XPA and conjugated to Cy3 using strain-promoted azide-alkyne cycloaddition. The resulting fluorescent XPA protein (XPACy3) shows no loss in DNA binding activity and generates a robust change in fluorescence upon binding to DNA. Here we describe methods to generate XPACy3 and detail in vitro experimental conditions required to stably maintain the protein during biochemical and biophysical studies.


Subject(s)
DNA Damage , DNA Repair , Humans , DNA Repair/genetics , DNA Damage/genetics , Excision Repair , Xeroderma Pigmentosum Group A Protein/genetics , Xeroderma Pigmentosum Group A Protein/chemistry , Xeroderma Pigmentosum Group A Protein/metabolism , DNA/chemistry , Ultraviolet Rays , Nucleotides , Protein Binding
4.
J Biol Chem ; 299(5): 104636, 2023 05.
Article in English | MEDLINE | ID: mdl-36963489

ABSTRACT

Base excision repair (BER) is carried out by a series of proteins that function in a step-by-step process to identify, remove, and replace DNA damage. During BER, the DNA transitions through various intermediate states as it is processed by each DNA repair enzyme. Left unrepaired, these BER intermediates can transition into double-stranded DNA breaks and promote genome instability. Previous studies have proposed a short-lived complex consisting of the BER intermediate, the incoming enzyme, and the outgoing enzyme at each step of the BER pathway to protect the BER intermediate. The transfer of BER intermediates between enzymes, known as BER coordination or substrate channeling, remains poorly understood. Here, we utilize single-molecule total internal reflection fluorescence microscopy to investigate the mechanism of BER coordination between apurinic/apyrimidinic endonuclease 1 (APE1) and DNA polymerase ß (Pol ß). When preformed complexes of APE1 and the incised abasic site product (APE1 product and Pol ß substrate) were subsequently bound by Pol ß, the Pol ß enzyme dissociated shortly after binding in most of the observations. In the events where Pol ß binding was followed by APE1 dissociation during substrate channeling, Pol ß remained bound for a longer period of time to allow disassociation of APE1. Our results indicate that transfer of the BER intermediate from APE1 to Pol ß during BER is dependent on the dissociation kinetics of APE1 and the duration of the ternary complex on the incised abasic site.


Subject(s)
DNA Polymerase beta , DNA Repair , DNA Damage , DNA Polymerase beta/genetics , DNA Polymerase beta/metabolism , DNA Repair/physiology , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Single Molecule Imaging , Microscopy, Fluorescence , Humans
5.
EMBO J ; 39(7): e104547, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32090346

ABSTRACT

In selecting ssDNA over dsDNA, the RAD51 DNA strand exchange protein has to overcome the entropy associated with straightening of single-strand DNA upon nucleoprotein filament formation. New work in The EMBO Journal (Paoletti et al, 2020), combined biophysical analysis of the RAD51-ssDNA interaction with mathematical modeling to show that the flexibility of DNA positively correlates with nucleation and extension of the RAD51 nucleoprotein filament and that the entropic penalty associated with restricting ssDNA flexibility is offset by a strong RAD51-RAD51 interaction within the nucleoprotein filament.


Subject(s)
DNA , Rad51 Recombinase , DNA, Single-Stranded , DNA-Binding Proteins/metabolism , Nucleoproteins , Rad51 Recombinase/metabolism
6.
Blood ; 139(7): 1039-1051, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34767620

ABSTRACT

Human telomere biology disorders (TBD)/short telomere syndromes (STS) are heterogeneous disorders caused by inherited loss-of-function mutations in telomere-associated genes. Here, we identify 3 germline heterozygous missense variants in the RPA1 gene in 4 unrelated probands presenting with short telomeres and varying clinical features of TBD/STS, including bone marrow failure, myelodysplastic syndrome, T- and B-cell lymphopenia, pulmonary fibrosis, or skin manifestations. All variants cluster to DNA-binding domain A of RPA1 protein. RPA1 is a single-strand DNA-binding protein required for DNA replication and repair and involved in telomere maintenance. We showed that RPA1E240K and RPA1V227A proteins exhibit increased binding to single-strand and telomeric DNA, implying a gain in DNA-binding function, whereas RPA1T270A has binding properties similar to wild-type protein. To study the mutational effect in a cellular system, CRISPR/Cas9 was used to knock-in the RPA1E240K mutation into healthy inducible pluripotent stem cells. This resulted in severe telomere shortening and impaired hematopoietic differentiation. Furthermore, in patients with RPA1E240K, we discovered somatic genetic rescue in hematopoietic cells due to an acquired truncating cis RPA1 mutation or a uniparental isodisomy 17p with loss of mutant allele, coinciding with stabilized blood counts. Using single-cell sequencing, the 2 somatic genetic rescue events were proven to be independently acquired in hematopoietic stem cells. In summary, we describe the first human disease caused by germline RPA1 variants in individuals with TBD/STS.


Subject(s)
Bone Marrow Failure Disorders/pathology , Gain of Function Mutation , Heterozygote , Myelodysplastic Syndromes/pathology , Replication Protein A/genetics , Telomere Shortening , Telomere/genetics , Adolescent , Adult , Bone Marrow Failure Disorders/etiology , Bone Marrow Failure Disorders/metabolism , Cell Differentiation , Child , Female , Humans , Infant, Newborn , Male , Middle Aged , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/metabolism , Young Adult
7.
Semin Cell Dev Biol ; 113: 27-37, 2021 05.
Article in English | MEDLINE | ID: mdl-33967572

ABSTRACT

DNA replication is laden with obstacles that slow, stall, collapse, and break DNA replication forks. At each obstacle, there is a decision to be made whether to bypass the lesion, repair or restart the damaged fork, or to protect stalled forks from further demise. Each "decision" draws upon multitude of proteins participating in various mechanisms that allow repair and restart of replication forks. Specific functions for many of these proteins have been described and an understanding of how they come together in supporting replication forks is starting to emerge. Many questions, however, remain regarding selection of the mechanisms that enable faithful genome duplication and how "normal" intermediates in these mechanisms are sometimes funneled into "rogue" processes that destabilize the genome and lead to cancer, cell death, and emergence of chemotherapeutic resistance. In this review we will discuss molecular mechanisms of DNA damage bypass and replication fork protection and repair. We will specifically focus on the key players that define which mechanism is employed including: PCNA and its control by posttranslational modifications, translesion synthesis DNA polymerases, molecular motors that catalyze reversal of stalled replication forks, proteins that antagonize fork reversal and protect reversed forks from nucleolytic degradation, and the machinery of homologous recombination that helps to reestablish broken forks. We will also discuss risks to genome integrity inherent in each of these mechanisms.


Subject(s)
DNA Damage/genetics , DNA Replication/genetics , Humans
8.
Nucleic Acids Res ; 49(4): 2005-2026, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33476370

ABSTRACT

Replication Protein A (RPA) is a critical complex that acts in replication and promotes homologous recombination by allowing recombinase recruitment to processed DSB ends. Most organisms possess three RPA subunits (RPA1, RPA2, RPA3) that form a trimeric complex critical for viability. The Caenorhabditis elegans genome encodes RPA-1, RPA-2 and an RPA-2 paralog RPA-4. In our analysis, we determined that RPA-2 is critical for germline replication and normal repair of meiotic DSBs. Interestingly, RPA-1 but not RPA-2 is essential for somatic replication, in contrast to other organisms that require both subunits. Six different hetero- and homodimeric complexes containing permutations of RPA-1, RPA-2 and RPA-4 can be detected in whole animal extracts. Our in vivo studies indicate that RPA-1/4 dimer is less abundant in the nucleus and its formation is inhibited by RPA-2. While RPA-4 does not participate in replication or recombination, we find that RPA-4 inhibits RAD-51 filament formation and promotes apoptosis of a subset of damaged nuclei. Altogether these findings point to sub-functionalization and antagonistic roles of RPA complexes in C. elegans.


Subject(s)
Apoptosis , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/genetics , DNA Replication , Meiosis/genetics , Recombination, Genetic , Replication Protein A/physiology , Animals , Caenorhabditis elegans Proteins/analysis , Caenorhabditis elegans Proteins/metabolism , DNA Breaks, Double-Stranded , Mitosis/genetics , Rad51 Recombinase/analysis , Replication Protein A/metabolism
9.
Nucleic Acids Res ; 49(9): e53, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33660771

ABSTRACT

Molecular machines within cells dynamically assemble, disassemble and reorganize. Molecular interactions between their components can be observed at the single-molecule level and quantified using colocalization single-molecule spectroscopy, in which individual labeled molecules are seen transiently associating with a surface-tethered partner, or other total internal reflection fluorescence microscopy approaches in which the interactions elicit changes in fluorescence in the labeled surface-tethered partner. When multiple interacting partners can form ternary, quaternary and higher order complexes, the types of spatial and temporal organization of these complexes can be deduced from the order of appearance and reorganization of the components. Time evolution of complex architectures can be followed by changes in the fluorescence behavior in multiple channels. Here, we describe the kinetic event resolving algorithm (KERA), a software tool for organizing and sorting the discretized fluorescent trajectories from a range of single-molecule experiments. KERA organizes the data in groups by transition patterns, and displays exhaustive dwell time data for each interaction sequence. Enumerating and quantifying sequences of molecular interactions provides important information regarding the underlying mechanism of the assembly, dynamics and architecture of the macromolecular complexes. We demonstrate KERA's utility by analyzing conformational dynamics of two DNA binding proteins: replication protein A and xeroderma pigmentosum complementation group D helicase.


Subject(s)
Software , Algorithms , DNA/chemistry , Fluorescence , Kinetics , Protein Conformation , Protein Domains , Replication Protein A/chemistry , Xeroderma Pigmentosum Group D Protein/chemistry
10.
Int J Mol Sci ; 24(12)2023 Jun 17.
Article in English | MEDLINE | ID: mdl-37373425

ABSTRACT

As many as 700,000 unique sequences in the human genome are predicted to fold into G-quadruplexes (G4s), non-canonical structures formed by Hoogsteen guanine-guanine pairing within G-rich nucleic acids. G4s play both physiological and pathological roles in many vital cellular processes including DNA replication, DNA repair and RNA transcription. Several reagents have been developed to visualize G4s in vitro and in cells. Recently, Zhen et al. synthesized a small protein G4P based on the G4 recognition motif from RHAU (DHX36) helicase (RHAU specific motif, RSM). G4P was reported to bind the G4 structures in cells and in vitro, and to display better selectivity toward G4s than the previously published BG4 antibody. To get insight into G4P- G4 interaction kinetics and selectivity, we purified G4P and its expanded variants, and analyzed their G4 binding using single-molecule total internal reflection fluorescence microscopy and mass photometry. We found that G4P binds to various G4s with affinities defined mostly by the association rate. Doubling the number of the RSM units in the G4P increases the protein's affinity for telomeric G4s and its ability to interact with sequences folding into multiple G4s.


Subject(s)
G-Quadruplexes , Humans , DEAD-box RNA Helicases/metabolism , RNA/metabolism , DNA Helicases/metabolism
11.
Crit Rev Biochem Mol Biol ; 55(5): 482-507, 2020 10.
Article in English | MEDLINE | ID: mdl-32856505

ABSTRACT

The heterotrimeric eukaryotic Replication protein A (RPA) is a master regulator of numerous DNA metabolic processes. For a long time, it has been viewed as an inert protector of ssDNA and a platform for assembly of various genome maintenance and signaling machines. Later, the modular organization of the RPA DNA binding domains suggested a possibility for dynamic interaction with ssDNA. This modular organization has inspired several models for the RPA-ssDNA interaction that aimed to explain how RPA, the high-affinity ssDNA binding protein, is replaced by the downstream players in DNA replication, recombination, and repair that bind ssDNA with much lower affinity. Recent studies, and in particular single-molecule observations of RPA-ssDNA interactions, led to the development of a new model for the ssDNA handoff from RPA to a specific downstream factor where not only stability and structural rearrangements but also RPA conformational dynamics guide the ssDNA handoff. Here we will review the current knowledge of the RPA structure, its dynamic interaction with ssDNA, and how RPA conformational dynamics may be influenced by posttranslational modification and proteins that interact with RPA, as well as how RPA dynamics may be harnessed in cellular decision making.


Subject(s)
DNA Repair , DNA Replication , Eukaryota/metabolism , Recombination, Genetic , Replication Protein A/metabolism , Animals , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Eukaryota/genetics , Humans , Protein Conformation , Replication Protein A/genetics
12.
Nucleic Acids Res ; 48(2): 694-708, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31799622

ABSTRACT

The proper repair of deleterious DNA lesions such as double strand breaks prevents genomic instability and carcinogenesis. In yeast, the Rad52 protein mediates DSB repair via homologous recombination. In mammalian cells, despite the presence of the RAD52 protein, the tumour suppressor protein BRCA2 acts as the predominant mediator during homologous recombination. For decades, it has been believed that the RAD52 protein played only a back-up role in the repair of DSBs performing an error-prone single strand annealing (SSA). Recent studies have identified several new functions of the RAD52 protein and have drawn attention to its important role in genome maintenance. Here, we show that RAD52 activities are enhanced by interacting with a small and highly acidic protein called DSS1. Binding of DSS1 to RAD52 changes the RAD52 oligomeric conformation, modulates its DNA binding properties, stimulates SSA activity and promotes strand invasion. Our work introduces for the first time RAD52 as another interacting partner of DSS1 and shows that both proteins are important players in the SSA and BIR pathways of DSB repair.


Subject(s)
Carcinogenesis/genetics , Homologous Recombination/genetics , Proteasome Endopeptidase Complex/genetics , Rad52 DNA Repair and Recombination Protein/genetics , BRCA2 Protein/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA-Binding Proteins/genetics , Genome, Human/genetics , Genomic Instability/genetics , Humans , Osteosarcoma/genetics , Osteosarcoma/pathology , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
13.
Nano Lett ; 20(1): 314-319, 2020 Jan 08.
Article in English | MEDLINE | ID: mdl-31851824

ABSTRACT

Quantum dots inserted in semiconducting nanowires are an interesting platform for the fabrication of single photon devices. To fully understand the physical properties of these objects, we need to correlate the optical, transport, and structural properties on the same nanostructure. In this work, we study the spectral tunability of the emission of a single quantum dot in a GaN nanowire by applying external bias. The nanowires are dispersed and contacted on electron beam transparent Si3N4 membranes, so that transmission electron microscopy observations, photocurrent, and micro-photoluminescence measurements under bias can be performed on the same specimen. The emission from a single dot blue or red shifts when the external electric field compensates or enhances the internal electric field generated by the spontaneous and piezoelectric polarization. A detailed study of two nanowire specimens emitting at 327.5 and 307.5 nm shows spectral shifts at rates of 20 and 12 meV/V, respectively. Theoretical calculations facilitated by the modeling of the exact heterostructure provide a good description of the experimental observations. When the bias-induced band bending is strong enough to favor tunneling of the electron in the dot toward the stem or the cap, the spectral shift saturates and additional transitions associated with charged excitons can be observed.

14.
Nanotechnology ; 31(47): 472001, 2020 Nov 20.
Article in English | MEDLINE | ID: mdl-32503014

ABSTRACT

Understanding the interplay between the structure, composition and opto-electronic properties of semiconductor nano-objects requires combining transmission electron microscopy (TEM) based techniques with electrical and optical measurements on the very same specimen. Recent developments in TEM technologies allow not only the identification and in-situ electrical characterization of a particular object, but also the direct visualization of its modification in-situ by techniques such as Joule heating. Over the past years, we have carried out a number of studies in these fields that are reviewed in this contribution. In particular, we discuss here i) correlated studies where the same unique object is characterized electro-optically and by TEM, ii) in-situ Joule heating studies where a solid-state metal-semiconductor reaction is monitored in the TEM, and iii) in-situ biasing studies to better understand the electrical properties of contacted single nanowires. In addition, we provide detailed fabrication steps for the silicon nitride membrane-chips crucial to these correlated and in-situ measurements.

15.
Proc Natl Acad Sci U S A ; 113(41): E6045-E6054, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27671650

ABSTRACT

The DNA strand exchange protein RAD51 facilitates the central step in homologous recombination, a process fundamentally important for accurate repair of damaged chromosomes, restart of collapsed replication forks, and telomere maintenance. The active form of RAD51 is a nucleoprotein filament that assembles on single-stranded DNA (ssDNA) at the sites of DNA damage. The c-Abl tyrosine kinase and its oncogenic counterpart BCR-ABL fusion kinase phosphorylate human RAD51 on tyrosine residues 54 and 315. We combined biochemical reconstitutions of the DNA strand exchange reactions with total internal reflection fluorescence microscopy to determine how the two phosphorylation events affect the biochemical activities of human RAD51 and properties of the RAD51 nucleoprotein filament. By mimicking RAD51 tyrosine phosphorylation with a nonnatural amino acid, p-carboxymethyl-l-phenylalanine (pCMF), we demonstrated that Y54 phosphorylation enhances the RAD51 recombinase activity by at least two different mechanisms, modifies the RAD51 nucleoprotein filament formation, and allows RAD51 to compete efficiently with ssDNA binding protein RPA. In contrast, Y315 phosphorylation has little effect on the RAD51 activities. Based on our work and previous cellular studies, we propose a mechanism underlying RAD51 activation by c-Abl/BCR-ABL kinases.


Subject(s)
Nucleoproteins/metabolism , Phosphotyrosine/metabolism , Rad51 Recombinase/metabolism , Biological Mimicry , DNA/genetics , DNA/metabolism , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Enzyme Activation , Homologous Recombination , Humans , Hydrolysis , Models, Molecular , Mutation , Nucleoproteins/chemistry , Phosphorylation , Phosphotyrosine/chemistry , Phosphotyrosine/genetics , Protein Conformation , Protein Multimerization , Proto-Oncogene Proteins c-abl/metabolism , Rad51 Recombinase/chemistry , Rad51 Recombinase/genetics , Recombinant Proteins
16.
J Biol Chem ; 292(26): 11136-11137, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28667173

ABSTRACT

The exchange of DNA strands between broken and intact molecules lies at the heart of fundamental cellular processes ranging from repairing DNA damage by homologous recombination to generation of genetic diversity during sexual reproduction. New work by Lee and colleagues utilizes the DNA curtain method, an elegant single-molecule technique, to demonstrate common and idiosyncratic features in the DNA strand exchange mechanisms of three RecA-family recombinases, bacterial RecA, and eukaryotic Rad51 and Dmc1 proteins.


Subject(s)
Bacteria/enzymology , Bacterial Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Rad51 Recombinase/metabolism , Rec A Recombinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology
17.
J Biol Chem ; 292(43): 17777-17793, 2017 10 27.
Article in English | MEDLINE | ID: mdl-28882897

ABSTRACT

T-cell lymphoma invasion and metastasis 1 (Tiam1) is a Dbl-family guanine nucleotide exchange factor (GEF) that specifically activates the Rho-family GTPase Rac1 in response to upstream signals, thereby regulating cellular processes including cell adhesion and migration. Tiam1 contains multiple domains, including an N-terminal pleckstrin homology coiled-coiled extension (PHn-CC-Ex) and catalytic Dbl homology and C-terminal pleckstrin homology (DH-PHc) domain. Previous studies indicate that larger fragments of Tiam1, such as the region encompassing the N-terminal to C-terminal pleckstrin homology domains (PHn-PHc), are auto-inhibited. However, the domains in this region responsible for inhibition remain unknown. Here, we show that the PHn-CC-Ex domain inhibits Tiam1 GEF activity by directly interacting with the catalytic DH-PHc domain, preventing Rac1 binding and activation. Enzyme kinetics experiments suggested that Tiam1 is auto-inhibited through occlusion of the catalytic site rather than by allostery. Small angle X-ray scattering and ensemble modeling yielded models of the PHn-PHc fragment that indicate it is in equilibrium between "open" and "closed" conformational states. Finally, single-molecule experiments support a model in which conformational sampling between the open and closed states of Tiam1 contributes to Rac1 dissociation. Our results highlight the role of the PHn-CC-Ex domain in Tiam1 GEF regulation and suggest a combinatorial model for GEF inhibition and activation of the Rac1 signaling pathway.


Subject(s)
Guanine Nucleotide Exchange Factors/chemistry , rac1 GTP-Binding Protein/chemistry , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Kinetics , Pleckstrin Homology Domains , Protein Binding , Signal Transduction/physiology , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , X-Ray Diffraction , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
18.
Nanotechnology ; 29(25): 255204, 2018 Jun 22.
Article in English | MEDLINE | ID: mdl-29558360

ABSTRACT

Nanowire photodetectors are investigated because of their compatibility with flexible electronics, or for the implementation of on-chip optical interconnects. Such devices are characterized by ultrahigh photocurrent gain, but their photoresponse scales sublinearly with the optical power. Here, we present a study of single-nanowire photodetectors displaying a linear response to ultraviolet illumination. Their structure consists of a GaN nanowire incorporating an AlN/GaN/AlN heterostructure, which generates an internal electric field. The activity of the heterostructure is confirmed by the rectifying behavior of the current-voltage characteristics in the dark, as well as by the asymmetry of the photoresponse in magnitude and linearity. Under reverse bias (negative bias on the GaN cap segment), the detectors behave linearly with the impinging optical power when the nanowire diameter is below a certain threshold (≈80 nm), which corresponds to the total depletion of the nanowire stem due to the Fermi level pinning at the sidewalls. In the case of nanowires that are only partially depleted, their nonlinearity is explained by a nonlinear variation of the diameter of their central conducting channel under illumination.

19.
Nucleic Acids Res ; 44(18): 8742-8753, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27342280

ABSTRACT

Guanine rich nucleic acid sequences can form G-quadruplex (G4) structures that interfere with DNA replication, repair and RNA transcription. The human FANCJ helicase contributes to maintaining genomic integrity by promoting DNA replication through G4-forming DNA regions. Here, we combined single-molecule and ensemble biochemical analysis to show that FANCJ possesses a G4-specific recognition site. Through this interaction, FANCJ targets G4-containing DNA where its helicase and G4-binding activities enable repeated rounds of stepwise G4-unfolding and refolding. In contrast to other G4-remodeling enzymes, FANCJ partially stabilizes the G-quadruplex. This would preserve the substrate for the REV1 translesion DNA synthesis polymerase to incorporate cytosine across from a replication-stalling G-quadruplex. The residues responsible for G-quadruplex recognition also participate in interaction with MLH1 mismatch-repair protein, suggesting that the FANCJ activity supporting replication and its participation in DNA interstrand crosslink repair and/or heteroduplex rejection are mutually exclusive. Our findings not only describe the mechanism by which FANCJ recognizes G-quadruplexes and mediates their stepwise unfolding, but also explain how FANCJ chooses between supporting DNA repair versus promoting DNA replication through G-rich sequences.


Subject(s)
Basic-Leucine Zipper Transcription Factors/chemistry , DNA/chemistry , Fanconi Anemia Complementation Group Proteins/chemistry , G-Quadruplexes , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Line , DNA/metabolism , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Humans , Models, Biological , Models, Molecular , Protein Binding , Protein Conformation , Recombinant Fusion Proteins
20.
Nucleic Acids Res ; 44(17): 8250-60, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27325737

ABSTRACT

Large multi-protein complexes play important roles in many biological processes, including DNA replication and repair, transcription, and signal transduction. One of the challenges in studying such complexes is to understand their mechanisms of assembly and disassembly and their architectures. Using single-molecule total internal reflection (TIRF) microscopy, we have examined the assembly and disassembly of the multi-protein complex that carries out translesion synthesis, the error-prone replication of damaged DNA. We show that the ternary complexes containing proliferating cell nuclear antigen (PCNA) and two non-classical DNA polymerases, Rev1 and DNA polymerase η, have two architectures: PCNA tool belts and Rev1 bridges. Moreover, these complexes are dynamic and their architectures can interconvert without dissociation. The formation of PCNA tool belts and Rev1 bridges and the ability of these complexes to change architectures are likely means of facilitating selection of the appropriate non-classical polymerase and polymerase-switching events.


Subject(s)
DNA-Directed DNA Polymerase/metabolism , DNA/biosynthesis , Nucleotidyltransferases/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Motifs , Amino Acid Substitution , Binding Sites , DNA-Directed DNA Polymerase/chemistry , Fluorescence , Kinetics , Multiprotein Complexes/metabolism , Mutant Proteins/metabolism , Nucleotidyltransferases/chemistry , Protein Binding , Protein Domains , Saccharomyces cerevisiae Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL