Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Environ Pollut ; 360: 124677, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39127336

ABSTRACT

Mitochondria, as the powerhouse of the cell, play a vital role in maintaining cellular energy homeostasis and are known to be a primary target of cadmium (Cd) toxicity. The improper targeting of proteins to mitochondria can compromise the normal functions of the mitochondria. However, the precise mechanism by which protein localization contributes to the development of mitochondrial dysfunction induced by Cd is still not fully understood. For this research, Hy-Line white variety chicks (1-day-old) were used and equally distributed into 4 groups: the Control group (fed with a basic diet), the Cd35 group (basic diet with 35 mg/kg CdCl2), the Cd70 group (basic diet with 70 mg/kg CdCl2) and the Cd140 group (basic diet with 140 mg/kg CdCl2), respectively for 90 days. It was found that Cd caused the accumulation of heat shock factor 1 (HSF1) in the mitochondria, and the overexpression of HSF1 in the mitochondria led to mitochondrial dysfunction and neuronal damage. This process is due to the mitochondrial HSF1 (mtHSF1), causing mitochondrial fission through the upregulation of dynamin-related protein 1 (Drp1) content, while inhibiting oligomer formation of single-stranded DNA-binding protein 1 (SSBP1), resulting in the mitochondrial DNA (mtDNA) deletion. The findings unveil an unforeseen role of HSF1 in triggering mitochondrial dysfunction.

2.
Plant Sci ; 346: 112151, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38848768

ABSTRACT

Endosperm, the major storage organ in cereal grains, determines the grain yield and quality. Mitochondria provide the energy for dry matter accumulation, in the endosperm development. Although mitochondrial single-stranded DNA-binding proteins (mtSSBs) play a canonical role in the maintenance of single-stranded mitochondrial DNA, their molecular functions in RNA processing and endosperm development remain obscure. Here, we report a defective rice endosperm mutant, floury endosperm26 (flo26), which develops abnormal starch grains in the endosperm. Map-based cloning and complementation experiments showed that FLO26 allele encodes a mitochondrial single-stranded DNA-binding protein, named as mtSSB1.1. Loss of function of mtSSB1.1 affects the transcriptional level of many mitochondrially-encoded genes and RNA splicing of nad1, a core component of respiratory chain complex I in mitochondria. As a result, dysfunctional mature nad1 led to dramatically decreased complex I activity, thereby reducing ATP production. Our results reveal that mtSSB1.1 plays an important role in the maintenance of mitochondrial function and endosperm development by stabilizing the splicing of mitochondrial RNA in rice.


Subject(s)
Endosperm , Oryza , Plant Proteins , RNA Splicing , Oryza/genetics , Oryza/metabolism , Oryza/growth & development , Endosperm/genetics , Endosperm/metabolism , Endosperm/growth & development , Plant Proteins/genetics , Plant Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, Mitochondrial , Mitochondria/metabolism , Mitochondria/genetics , Gene Expression Regulation, Plant
3.
Brief Bioinform ; 25(4)2024 May 23.
Article in English | MEDLINE | ID: mdl-38856168

ABSTRACT

Nucleic acid-binding proteins (NABPs), including DNA-binding proteins (DBPs) and RNA-binding proteins (RBPs), play important roles in essential biological processes. To facilitate functional annotation and accurate prediction of different types of NABPs, many machine learning-based computational approaches have been developed. However, the datasets used for training and testing as well as the prediction scopes in these studies have limited their applications. In this paper, we developed new strategies to overcome these limitations by generating more accurate and robust datasets and developing deep learning-based methods including both hierarchical and multi-class approaches to predict the types of NABPs for any given protein. The deep learning models employ two layers of convolutional neural network and one layer of long short-term memory. Our approaches outperform existing DBP and RBP predictors with a balanced prediction between DBPs and RBPs, and are more practically useful in identifying novel NABPs. The multi-class approach greatly improves the prediction accuracy of DBPs and RBPs, especially for the DBPs with ~12% improvement. Moreover, we explored the prediction accuracy of single-stranded DNA binding proteins and their effect on the overall prediction accuracy of NABP predictions.


Subject(s)
Computational Biology , DNA-Binding Proteins , Deep Learning , RNA-Binding Proteins , RNA-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Computational Biology/methods , Neural Networks, Computer , Humans
4.
J Mol Biol ; 436(9): 168544, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38508303

ABSTRACT

Bacteriophage T4 gene 32 protein (gp32) is a single-stranded DNA (ssDNA) binding protein essential for DNA replication. gp32 forms stable protein filaments on ssDNA through cooperative interactions between its core and N-terminal domain. gp32's C-terminal domain (CTD) is believed to primarily help coordinate DNA replication via direct interactions with constituents of the replisome. However, the exact mechanisms of these interactions are not known, and it is unclear how tightly-bound gp32 filaments are readily displaced from ssDNA as required for genomic processing. Here, we utilized truncated gp32 variants to demonstrate a key role of the CTD in regulating gp32 dissociation. Using optical tweezers, we probed the binding and dissociation dynamics of CTD-truncated gp32, *I, to an 8.1 knt ssDNA molecule and compared these measurements with those for full-length gp32. The *I-ssDNA helical filament becomes progressively unwound with increased protein concentration but remains significantly more stable than that of full-length, wild-type gp32. Protein oversaturation, concomitant with filament unwinding, facilitates rapid dissociation of full-length gp32 from across the entire ssDNA segment. In contrast, *I primarily unbinds slowly from only the ends of the cooperative clusters, regardless of the protein density and degree of DNA unwinding. Our results suggest that the CTD may constrain the relative twist angle of proteins within the ssDNA filament such that upon critical unwinding the cooperative interprotein interactions largely vanish, facilitating prompt removal of gp32. We propose a model of CTD-mediated gp32 displacement via internal restructuring of its filament, providing a mechanism for rapid ssDNA clearing during genomic processing.


Subject(s)
Bacteriophage T4 , DNA, Single-Stranded , DNA-Binding Proteins , Protein Binding , Viral Proteins , Bacteriophage T4/genetics , Bacteriophage T4/metabolism , DNA Replication , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , DNA, Viral/genetics , DNA, Viral/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/chemistry , Optical Tweezers , Protein Domains , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/chemistry
5.
Protein Sci ; 33(4): e4959, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38511671

ABSTRACT

Single-stranded DNA binding proteins (SSBs) are ubiquitous across all domains of life and play essential roles via stabilizing and protecting single-stranded (ss) DNA as well as organizing multiprotein complexes during DNA replication, recombination, and repair. Two mammalian SSB paralogs (hSSB1 and hSSB2 in humans) were recently identified and shown to be involved in various genome maintenance processes. Following our recent discovery of the liquid-liquid phase separation (LLPS) propensity of Escherichia coli (Ec) SSB, here we show that hSSB2 also forms LLPS condensates under physiologically relevant ionic conditions. Similar to that seen for EcSSB, we demonstrate the essential contribution of hSSB2's C-terminal intrinsically disordered region (IDR) to condensate formation, and the selective enrichment of various genome metabolic proteins in hSSB2 condensates. However, in contrast to EcSSB-driven LLPS that is inhibited by ssDNA binding, hSSB2 phase separation requires single-stranded nucleic acid binding, and is especially facilitated by ssDNA. Our results reveal an evolutionarily conserved role for SSB-mediated LLPS in the spatiotemporal organization of genome maintenance complexes. At the same time, differential LLPS features of EcSSB and hSSB2 point to functional adaptations to prokaryotic versus eukaryotic genome metabolic contexts.


Subject(s)
DNA , Phase Separation , Animals , Humans , DNA-Binding Proteins/chemistry , DNA Repair , DNA Replication , DNA, Single-Stranded/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Mammals/genetics
6.
Int J Mol Sci ; 24(19)2023 Oct 04.
Article in English | MEDLINE | ID: mdl-37834349

ABSTRACT

Single-stranded DNA-binding proteins (SSBs) play a crucial role in DNA metabolism by binding and stabilizing single-stranded DNA (ssDNA) intermediates. Through their multifaceted roles in DNA replication, recombination, repair, replication restart, and other cellular processes, SSB emerges as a central player in maintaining genomic integrity. These attributes collectively position SSBs as essential guardians of genomic integrity, establishing interactions with an array of distinct proteins. Unlike Escherichia coli, which contains only one type of SSB, some bacteria have two paralogous SSBs, referred to as SsbA and SsbB. In this study, we identified Staphylococcus aureus SsbA (SaSsbA) as a fresh addition to the roster of the anticancer drug 5-fluorouracil (5-FU) binding proteins, thereby expanding the ambit of the 5-FU interactome to encompass this DNA replication protein. To investigate the binding mode, we solved the complexed crystal structure with 5-FU at 2.3 Å (PDB ID 7YM1). The structure of glycerol-bound SaSsbA was also determined at 1.8 Å (PDB ID 8GW5). The interaction between 5-FU and SaSsbA was found to involve R18, P21, V52, F54, Q78, R80, E94, and V96. Based on the collective results from mutational and structural analyses, it became evident that SaSsbA's mode of binding with 5-FU diverges from that of SaSsbB. This complexed structure also holds the potential to furnish valuable comprehension regarding how 5-FU might bind to and impede analogous proteins in humans, particularly within cancer-related signaling pathways. Leveraging the information furnished by the glycerol and 5-FU binding sites, the complexed structures of SaSsbA bring to the forefront the potential viability of several interactive residues as potential targets for therapeutic interventions aimed at curtailing SaSsbA activity. Acknowledging the capacity of microbiota to influence the host's response to 5-FU, there emerges a pressing need for further research to revisit the roles that bacterial and human SSBs play in the realm of anticancer therapy.


Subject(s)
Antineoplastic Agents , Bacterial Proteins , Humans , Bacterial Proteins/metabolism , Glycerol , DNA, Single-Stranded , Fluorouracil/pharmacology , Escherichia coli/metabolism , DNA Replication , Antineoplastic Agents/pharmacology , Protein Binding/genetics
7.
J Mol Biol ; 435(19): 168236, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37572935

ABSTRACT

RAD51 forms nucleoprotein filaments to promote homologous recombination, replication fork reversal, and fork protection. Numerous factors regulate the stability of these filaments and improper regulation leads to genomic instability and ultimately disease including cancer. RADX is a single stranded DNA binding protein that modulates RAD51 filament stability. Here, we utilize a CRISPR-dependent base editing screen to tile mutations across RADX to delineate motifs required for RADX function. We identified separation of function mutants of RADX that bind DNA and RAD51 but have a reduced ability to stimulate its ATP hydrolysis activity. Cells expressing these RADX mutants accumulate RAD51 on chromatin, exhibit replication defects, have reduced growth, accumulate DNA damage, and are hypersensitive to DNA damage and replication stress. These results indicate that RADX must promote RAD51 ATP turnover to regulate RAD51 and genome stability during DNA replication.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , RNA Editing , Rad51 Recombinase , Humans , Adenosine Triphosphate/metabolism , DNA Replication/genetics , DNA, Single-Stranded , Gene Editing , Genomic Instability/genetics , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
8.
Biomedicines ; 11(7)2023 Jun 25.
Article in English | MEDLINE | ID: mdl-37509458

ABSTRACT

Single-stranded DNA binding protein 2 (SSBP2) is a tumor suppressor candidate. In this study, the expression level and clinicopathological significance of SSBP2 in squamous cell carcinoma (SCC) and basal cell carcinoma (BCC) were evaluated. We also identified biological pathways associated with a set of genes potentially related to SSBP2. Immunohistochemistry (IHC) was performed on 70 SCC and 146 BCC cases to assess SSBP2 expression semi-quantitatively. In addition, the associations between SSBP2 expression and clinicopathological characteristics were analyzed. Gene ontology (GO) enrichment analysis was performed using publicly available data and web-based bioinformatics tools. Compared with BCC, SCC had a significantly low SSBP2 expression (p < 0.001). In total, 12 (17.1%) of the 70 SCC cases and 30 (20.5%) of the 146 BCC cases showed low SSBP2 expression. Among SCC cases, ulceration (p = 0.005) and a deep level of invasion (p = 0.012) showed an association with low SSBP2 expression. Local recurrence was slightly more common in the SCC subgroup with low SSBP2 expression, although the difference was not significant (p = 0.058). Using GO enrichment analysis, we identified several biological functions performed by a set of 36 genes in SCC. SSBP2 evaluation using IHC can be helpful in the differential diagnosis of SCC and BCC. SSBP2 expression was associated with tumor invasiveness in SCC.

9.
Int J Biol Macromol ; 248: 125885, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37473881

ABSTRACT

DR0041 ORF encodes an uncharacterized Deinococcus lineage protein. We earlier reported presence of DR0041 protein in DNA repair complexes of Ssb and RecA in Deinococcus radiodurans. Here, we systematically examined the role of DR0041 in DNA metabolism using various experimental methodologies including electrophoretic mobility assays, nuclease assays, strand exchange assays and transmission electron microscopy. Interaction between DR0041 and the C-terminal acidic tail of Ssb was assessed through co-expression and in vivo cross-linking studies. A knockout mutant was constructed to understand importance of DR0041 ORF for various physiological processes. Results highlight binding of DR0041 protein to single-stranded and double-stranded DNA, interaction with Ssb-coated single-stranded DNA without interference with RecA-mediated strand exchange, protection of DNA from exonucleases, and compaction of high molecular weight DNA molecules into tightly condensed forms. Bridging and compaction of sheared DNA by DR0041 protein might have implications in the preservation of damaged DNA templates to maintain genome integrity upon exposure to gamma irradiation. Our results suggest that DR0041 protein is dispensable for growth under standard growth conditions and following gamma irradiation but contributes to protection of DNA during transformation. We discuss the role of DR0041 protein from the perspective of protection of broken DNA templates and functional redundancy.


Subject(s)
Deinococcus , Deinococcus/genetics , Deinococcus/radiation effects , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , DNA/metabolism , DNA Repair , DNA, Single-Stranded/metabolism , Bacterial Proteins/chemistry
10.
J Biol Chem ; 299(6): 104773, 2023 06.
Article in English | MEDLINE | ID: mdl-37142225

ABSTRACT

The bacterial RadD enzyme is important for multiple genome maintenance pathways, including RecA DNA strand exchange and RecA-independent suppression of DNA crossover template switching. However, much remains unknown about the precise roles of RadD. One potential clue into RadD mechanisms is its direct interaction with the single-stranded DNA binding protein (SSB), which coats single-stranded DNA exposed during genome maintenance reactions in cells. Interaction with SSB stimulates the ATPase activity of RadD. To probe the mechanism and importance of RadD-SSB complex formation, we identified a pocket on RadD that is essential for binding SSB. In a mechanism shared with many other SSB-interacting proteins, RadD uses a hydrophobic pocket framed by basic residues to bind the C-terminal end of SSB. We found that RadD variants that substitute acidic residues for basic residues in the SSB binding site impair RadD:SSB complex formation and eliminate SSB stimulation of RadD ATPase activity in vitro. Additionally, mutant Escherichia coli strains carrying charge reversal radD changes display increased sensitivity to DNA damaging agents synergistically with deletions of radA and recG, although the phenotypes of the SSB-binding radD mutants are not as severe as a full radD deletion. This suggests that cellular RadD requires an intact interaction with SSB for full RadD function.


Subject(s)
DNA-Binding Proteins , Escherichia coli , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , DNA Repair/genetics , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Protein Binding , Mutation , Binding Sites , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Models, Molecular , Protein Structure, Quaternary
11.
Microorganisms ; 11(4)2023 Mar 27.
Article in English | MEDLINE | ID: mdl-37110272

ABSTRACT

Single-stranded DNA-binding proteins (SSBs) are essential for all living organisms. Whether SSBs can repair DNA double-strand breaks (DSBs) and improve the efficiency of CRISPR/Cas9-mediated genome editing has not been determined. Here, based on a pCas/pTargetF system, we constructed pCas-SSB and pCas-T4L by replacing the λ-Red recombinases with Escherichia coli SSB and phage T4 DNA ligase in pCas, respectively. Inactivation of the E. coli lacZ gene with homologous donor dsDNA increased the gene editing efficiency of pCas-SSB/pTargetF by 21.4% compared to pCas/pTargetF. Inactivation of the E. coli lacZ gene via NHEJ increased the gene editing efficiency of pCas-SSB/pTargetF by 33.2% compared to pCas-T4L/pTargetF. Furthermore, the gene-editing efficiency of pCas-SSB/pTargetF in E. coli (ΔrecA, ΔrecBCD, ΔSSB) with or without donor dsDNA did not differ. Additionally, pCas-SSB/pTargetF with donor dsDNA successfully deleted the wp116 gene in Pseudomonas sp. UW4. These results demonstrate that E. coli SSB repairs DSBs caused by CRISPR/Cas9 and effectively improves CRISPR/Cas9 genome editing in E. coli and Pseudomonas.

12.
Pharmaceutics ; 15(4)2023 Mar 23.
Article in English | MEDLINE | ID: mdl-37111518

ABSTRACT

Single-stranded DNA-binding protein (SSB) is a bacterial interaction hub and an appealing target for antimicrobial therapy. Understanding the structural adaptation of the disordered SSB C-terminus (SSB-Ct) to DNA metabolizing enzymes (e.g., ExoI and RecO) is essential for designing high-affinity SSB mimetic inhibitors. Molecular dynamics simulations revealed the transient interactions of SSB-Ct with two hot spots on ExoI and RecO. The residual flexibility of the peptide-protein complexes allows adaptive molecular recognition. Scanning with non-canonical amino acids revealed that modifications at both termini of SSB-Ct could increase the affinity, supporting the two-hot-spot binding model. Combining unnatural amino acid substitutions on both segments of the peptide resulted in enthalpy-enhanced affinity, accompanied by enthalpy-entropy compensation, as determined by isothermal calorimetry. NMR data and molecular modeling confirmed the reduced flexibility of the improved affinity complexes. Our results highlight that the SSB-Ct mimetics bind to the DNA metabolizing targets through the hot spots, interacting with both of segments of the ligands.

13.
Front Mol Biosci ; 10: 1113960, 2023.
Article in English | MEDLINE | ID: mdl-37006622

ABSTRACT

Mobile genetic elements can encode a wide variety of genes that support their own stability and mobility as well as genes that provide accessory functions to their hosts. Such genes can be adopted from host chromosomes and can be exchanged with other mobile elements. Due to their accessory nature, the evolutionary trajectories of these genes can differ from those of essential host genes. The mobilome therefore provides a rich source of genetic innovation. We previously described a new type of primase encoded by S. aureus SCCmec elements that is composed of an A-family polymerase catalytic domain in complex with a small second protein that confers single-stranded DNA binding. Here we use new structure prediction methods in conjunction with sequence database searches to show that related primases are widespread among putative mobile genetic elements in the Bacillota. Structure predictions show that the second protein adopts an OB fold (common among single-stranded DNA binding (SSB) proteins) and these predictions were far more powerful than simple sequence comparisons in identifying its homologs. The protein-protein interaction surface varies among these polymerase-SSB complexes appear to have arisen repeatedly by exploiting partial truncations of the polymerase's N-terminal accessory domains.

14.
Cell Host Microbe ; 31(3): 343-355.e5, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36893733

ABSTRACT

There is strong selection for the evolution of systems that protect bacterial populations from viral attack. We report a single phage defense protein, Hna, that provides protection against diverse phages in Sinorhizobium meliloti, a nitrogen-fixing alpha-proteobacterium. Homologs of Hna are distributed widely across bacterial lineages, and a homologous protein from Escherichia coli also confers phage defense. Hna contains superfamily II helicase motifs at its N terminus and a nuclease motif at its C terminus, with mutagenesis of these motifs inactivating viral defense. Hna variably impacts phage DNA replication but consistently triggers an abortive infection response in which infected cells carrying the system die but do not release phage progeny. A similar host cell response is triggered in cells containing Hna upon expression of a phage-encoded single-stranded DNA binding protein (SSB), independent of phage infection. Thus, we conclude that Hna limits phage spread by initiating abortive infection in response to a phage protein.


Subject(s)
Bacteriophages , Bacteriophages/genetics , DNA Replication
15.
J Hazard Mater ; 450: 131023, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36857823

ABSTRACT

Okadaic acid (OA), one of the most widely distributed marine toxins worldwide poses a severe threat to human health. Previous sensing methods for OA detection are usually based on antigen-antibody binding mechanism. However, the drawbacks of antibodies especially the enzyme-labeled antibodies, such as the harsh storage condition and high cost, lead to significant challenges to OA detection in biological samples. To overcome these limitations, a single-stranded DNA binding protein (SSB) coupled aptasensor was developed for OA detection. SSB was incubated on the microplate as a substitute for conventional OA-protein conjugations. Carbon-gold nanoparticles were synthesized and labeled with horseradish peroxidase and thiol-modified aptamers to obtain a capture probe (CGNs@HRP-Apt) instead of the enzyme-labeled antibody for signal amplification. OA and SSB competed to bind with limited aptamers on CGNs@HRP-Apt probes followed by colorimetric assay to obtain the optical signals correlated to OA concentration. To achieve on-site detection, a miniaturized and multichannel absorbance reader (Smart-plate reader) was self-designed with full automation for OA detection. Utilizing the SSB coupled aptasensor and the Smart-plate reader, our approach enables cost-effective and on-site OA sensing with a detection range of 2.5-80 ppb and an ultra-low limit of detection of 0.68 ppb. Moreover, novel OA detection kits based on the SSB coupled aptasensor were prepared which can effectively reduce the cost by 15 times lower than that of commercial ELISA kits. Therefore, the developed platform provides a favorable and promising avenue for marine toxin detection in aquaculture and food safety.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , Metal Nanoparticles , Humans , Gold , Marine Toxins , Carbon , Biosensing Techniques/methods , Okadaic Acid , DNA-Binding Proteins , Horseradish Peroxidase , Limit of Detection
16.
Microbiol Spectr ; 11(1): e0436922, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36602343

ABSTRACT

Epstein-Barr virus (EBV) replicates its genome in the nucleus and undergoes tegumentation and envelopment in the cytoplasm. We are interested in how the single-stranded DNA binding protein BALF2, which executes its function and distributes predominantly in the nucleus, is packaged into the tegument of virions. At the mid-stage of virus replication in epithelial TW01-EBV cells, a small pool of BALF2 colocalizes with tegument protein BBLF1, BGLF4 protein kinase, and the cis-Golgi marker GM130 at the perinuclear viral assembly compartment (AC). A possible nuclear localization signal (NLS) between amino acids 1100 and 1128 (C29), which contains positive charged amino acid 1113RRKRR1117, is able to promote yellow fluorescent protein (YFP)-LacZ into the nucleus. In addition, BALF2 interacts with the nucleocapsid-associated protein BVRF1, suggesting that BALF2 may be transported into the cytoplasm with nucleocapsids in a nuclear egress complex (NEC)-dependent manner. A group of proteins involved in intracellular transport were identified to interact with BALF2 in a proteomic analysis. Among them, the small GTPase Rab1A functioning in bi-directional trafficking at the ER-Golgi interface is also a tegument component. In reactivated TW01-EBV cells, BALF2 colocalizes with Rab1A in the cytoplasmic AC. Expression of dominant-negative GFP-Rab1A(N124I) diminished the accumulation of BALF2 in the AC, coupling with attenuation of gp350/220 glycosylation. Virion release was significantly downregulated by expressing dominant-negative GFP-Rab1A(N124I). Overall, the subcellular distribution of BALF2 is regulated through its complex interaction with various proteins. Rab1 activity is required for proper gp350/220 glycosylation and the maturation of EBV. IMPORTANCE Upon EBV lytic reactivation, the virus-encoded DNA replication machinery functions in the nucleus, while the newly synthesized DNA is encapsidated and transported to the cytoplasm for final virus assembly. The single-stranded DNA binding protein BALF2 executing functions within the nucleus was also identified in the tegument layer of mature virions. Here, we studied the functional domain of BALF2 that contributes to the nuclear targeting and used a proteomic approach to identify novel BALF2-interacting cellular proteins that may contribute to virion morphogenesis. The GTPase Rab1, a master regulator of anterograde and retrograde endoplasmic reticulum (ER)-Golgi trafficking, colocalizes with BALF2 in the juxtanuclear concave region at the midstage of EBV reactivation. Rab1 activity is required for BALF2 targeting to the cytoplasmic assembly compartment (AC) and for gp350/220 targeting to cis-Golgi for proper glycosylation and virion release. Our study hints that EBV hijacks the bi-directional ER-Golgi trafficking machinery to complete virus assembly.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Humans , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Herpesvirus 4, Human/genetics , Proteomics , Viral Proteins/genetics , Virion
17.
Biomolecules ; 12(9)2022 08 26.
Article in English | MEDLINE | ID: mdl-36139026

ABSTRACT

Single-stranded DNA (ssDNA) binding proteins (SSBs) are critical in maintaining genome stability by protecting the transient existence of ssDNA from damage during essential biological processes, such as DNA replication and gene transcription. The single-stranded region of telomeres also requires protection by ssDNA binding proteins from being attacked in case it is wrongly recognized as an anomaly. In addition to their critical roles in genome stability and integrity, it has been demonstrated that ssDNA and SSB-ssDNA interactions play critical roles in transcriptional regulation in all three domains of life and viruses. In this review, we present our current knowledge of the structure and function of SSBs and the structural features for SSB binding specificity. We then discuss the machine learning-based approaches that have been developed for the prediction of SSBs from double-stranded DNA (dsDNA) binding proteins (DSBs).


Subject(s)
DNA, Single-Stranded , DNA-Binding Proteins , DNA/chemistry , DNA-Binding Proteins/metabolism , Genomic Instability , Humans , Machine Learning , Protein Binding
18.
Biochem Biophys Res Commun ; 624: 68-74, 2022 10 08.
Article in English | MEDLINE | ID: mdl-35933928

ABSTRACT

African swine fever (ASF) is a lethal hemorrhagic disease that affects domestic pigs and wild boars. There is no medication available for ASF to date. The ability to mount antigen-specific responses to viral vectored CP312R makes it a crucial potential target for designing vaccines or drugs. This study determined the crystal structure of ASFV CP312R at 2.32 Å and found it to be a monomer with a single-stranded DNA binding core domain with a clear five-strands ß-barrel OB-fold architecture. Electrophoretic mobility shift assay and size-exclusion chromatography characterization assay further confirmed the single-stranded DNA (ssDNA)-binding property of ASFV CP312R. This study revealed the structure and preliminary ssDNA interaction mechanisms of ASFV CP312R, providing new clues for developing new antiviral strategies.


Subject(s)
African Swine Fever Virus , African Swine Fever , African Swine Fever Virus/genetics , Animals , DNA, Single-Stranded/metabolism , Sus scrofa/genetics , Swine , Viral Proteins/metabolism
19.
Int J Biol Macromol ; 216: 618-628, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35809674

ABSTRACT

The lysine (K) tRNA synthetase C-terminal (KTSC) domain containing proteins are widely spread in Bacteria, Archaea and Viruses, but the function of this short domain is unclear. The occurrence of the fusion of KTSC domain to a catalytic domain or domains related to DNA or RNA metabolisms suggests its potential role in DNA or RNA binding. Here, we report the characterization of Mvu8s from Methanolobus vulcani, which consists of a single KTSC domain. Mvu8s binds specifically to ssDNA with an affinity approximately 40- and 10-fold higher than those for dsDNA and ssRNA in vitro, respectively. It shows a slight preference to the G-rich DNA sequence but barely binds the A-stretch. Crystal structure of Mvu8s shows that it forms a homo-tetramer, with each monomer composed of a four-strand antiparallel ß-sheet and a helix-turn-helix in the order of ß1-ß2-ß3-α1-α2-ß4. Four basic residues (R3, R7, K54 and K58) were found to serve important roles in ssDNA-binding. And, the spiral arrangement of the DNA interfaces in Mvu8s homo-tetramer presumably results in ssDNA wrapping. Our results not only offer clues of the functions of the KTSC domain containing proteins but also expand our knowledge on the non-oligonucleotide-binding (OB) fold single-stranded DNA-binding proteins in Archaea.


Subject(s)
DNA, Single-Stranded , DNA-Binding Proteins , Catalytic Domain , DNA/metabolism , DNA-Binding Proteins/metabolism , Protein Binding/genetics , RNA/metabolism
20.
EMBO Mol Med ; 14(7): e15851, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35670111

ABSTRACT

Aberrant localization of proteins to mitochondria disturbs mitochondrial function and contributes to the pathogenesis of Huntington's disease (HD). However, the crucial factors and the molecular mechanisms remain elusive. Here, we found that heat shock transcription factor 1 (HSF1) accumulates in the mitochondria of HD cell models, a YAC128 mouse model, and human striatal organoids derived from HD induced pluripotent stem cells (iPSCs). Overexpression of mitochondria-targeting HSF1 (mtHSF1) in the striatum causes neurodegeneration and HD-like behavior in mice. Mechanistically, mtHSF1 facilitates mitochondrial fission by activating dynamin-related protein 1 (Drp1) phosphorylation at S616. Moreover, mtHSF1 suppresses single-stranded DNA-binding protein 1 (SSBP1) oligomer formation, which results in mitochondrial DNA (mtDNA) deletion. The suppression of HSF1 mitochondrial localization by DH1, a unique peptide inhibitor, abolishes HSF1-induced mitochondrial abnormalities and ameliorates deficits in an HD animal model and human striatal organoids. Altogether, our findings describe an unsuspected role of HSF1 in contributing to mitochondrial dysfunction, which may provide a promising therapeutic target for HD.


Subject(s)
Heat Shock Transcription Factors , Huntington Disease , Animals , Corpus Striatum/pathology , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Disease Models, Animal , Heat Shock Transcription Factors/metabolism , Huntington Disease/pathology , Mice , Mitochondria/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL