Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Nature ; 623(7987): 633-642, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37938770

ABSTRACT

Trimethylation of histone H3 lysine 9 (H3K9me3) is crucial for the regulation of gene repression and heterochromatin formation, cell-fate determination and organismal development1. H3K9me3 also provides an essential mechanism for silencing transposable elements1-4. However, previous studies have shown that canonical H3K9me3 readers (for example, HP1 (refs. 5-9) and MPP8 (refs. 10-12)) have limited roles in silencing endogenous retroviruses (ERVs), one of the main transposable element classes in the mammalian genome13. Here we report that trinucleotide-repeat-containing 18 (TNRC18), a poorly understood chromatin regulator, recognizes H3K9me3 to mediate the silencing of ERV class I (ERV1) elements such as LTR12 (ref. 14). Biochemical, biophysical and structural studies identified the carboxy-terminal bromo-adjacent homology (BAH) domain of TNRC18 (TNRC18(BAH)) as an H3K9me3-specific reader. Moreover, the amino-terminal segment of TNRC18 is a platform for the direct recruitment of co-repressors such as HDAC-Sin3-NCoR complexes, thus enforcing optimal repression of the H3K9me3-demarcated ERVs. Point mutagenesis that disrupts the TNRC18(BAH)-mediated H3K9me3 engagement caused neonatal death in mice and, in multiple mammalian cell models, led to derepressed expression of ERVs, which affected the landscape of cis-regulatory elements and, therefore, gene-expression programmes. Collectively, we describe a new H3K9me3-sensing and regulatory pathway that operates to epigenetically silence evolutionarily young ERVs and exert substantial effects on host genome integrity, transcriptomic regulation, immunity and development.


Subject(s)
Endogenous Retroviruses , Gene Silencing , Histones , Intracellular Signaling Peptides and Proteins , Lysine , Retroelements , Animals , Humans , Mice , Chromatin/genetics , Chromatin/metabolism , Co-Repressor Proteins/metabolism , Endogenous Retroviruses/genetics , Epigenesis, Genetic , Gene Expression Profiling , Genome/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lysine/metabolism , Methylation , Protein Domains , Retroelements/genetics , Terminal Repeat Sequences/genetics , Animals, Newborn , Cell Line
2.
Nature ; 595(7868): 591-595, 2021 07.
Article in English | MEDLINE | ID: mdl-34163069

ABSTRACT

The development of cancer is intimately associated with genetic abnormalities that target proteins with intrinsically disordered regions (IDRs). In human haematological malignancies, recurrent chromosomal translocation of nucleoporin (NUP98 or NUP214) generates an aberrant chimera that invariably retains the nucleoporin IDR-tandemly dispersed repeats of phenylalanine and glycine residues1,2. However, how unstructured IDRs contribute to oncogenesis remains unclear. Here we show that IDRs contained within NUP98-HOXA9, a homeodomain-containing transcription factor chimera recurrently detected in leukaemias1,2, are essential for establishing liquid-liquid phase separation (LLPS) puncta of chimera and for inducing leukaemic transformation. Notably, LLPS of NUP98-HOXA9 not only promotes chromatin occupancy of chimera transcription factors, but also is required for the formation of a broad 'super-enhancer'-like binding pattern typically seen at leukaemogenic genes, which potentiates transcriptional activation. An artificial HOX chimera, created by replacing the phenylalanine and glycine repeats of NUP98 with an unrelated LLPS-forming IDR of the FUS protein3,4, had similar enhancing effects on the genome-wide binding and target gene activation of the chimera. Deeply sequenced Hi-C revealed that phase-separated NUP98-HOXA9 induces CTCF-independent chromatin loops that are enriched at proto-oncogenes. Together, this report describes a proof-of-principle example in which cancer acquires mutation to establish oncogenic transcription factor condensates via phase separation, which simultaneously enhances their genomic targeting and induces organization of aberrant three-dimensional chromatin structure during tumourous transformation. As LLPS-competent molecules are frequently implicated in diseases1,2,4-7, this mechanism can potentially be generalized to many malignant and pathological settings.


Subject(s)
Chromatin/genetics , Homeodomain Proteins/genetics , Intrinsically Disordered Proteins/genetics , Neoplasms/pathology , Nuclear Pore Complex Proteins/genetics , Translocation, Genetic , Animals , Carcinogenesis , Female , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Transcription Factors/genetics , Transcriptional Activation
3.
Nucleic Acids Res ; 49(8): 4441-4455, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33823544

ABSTRACT

Trimethylation of histone H3 lysine 27 (H3K27me3) is important for gene silencing and imprinting, (epi)genome organization and organismal development. In a prevalent model, the functional readout of H3K27me3 in mammalian cells is achieved through the H3K27me3-recognizing chromodomain harbored within the chromobox (CBX) component of canonical Polycomb repressive complex 1 (cPRC1), which induces chromatin compaction and gene repression. Here, we report that binding of H3K27me3 by a Bromo Adjacent Homology (BAH) domain harbored within BAH domain-containing protein 1 (BAHD1) is required for overall BAHD1 targeting to chromatin and for optimal repression of the H3K27me3-demarcated genes in mammalian cells. Disruption of direct interaction between BAHD1BAH and H3K27me3 by point mutagenesis leads to chromatin remodeling, notably, increased histone acetylation, at its Polycomb gene targets. Mice carrying an H3K27me3-interaction-defective mutation of Bahd1BAH causes marked embryonic lethality, showing a requirement of this pathway for normal development. Altogether, this work demonstrates an H3K27me3-initiated signaling cascade that operates through a conserved BAH 'reader' module within BAHD1 in mammals.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Histones/metabolism , Polycomb-Group Proteins/metabolism , Acetylation , Animals , Chromatin/genetics , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation Sequencing , Chromosomal Proteins, Non-Histone/genetics , Female , Gene Expression Profiling , Gene Ontology , HEK293 Cells , Humans , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Polycomb-Group Proteins/genetics , Protein Domains
4.
Nucleic Acids Res ; 49(9): 4971-4988, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33849067

ABSTRACT

Castration-resistant prostate cancer (CRPC) is a terminal disease and the molecular underpinnings of CRPC development need to be better understood in order to improve its treatment. Here, we report that a transcription factor Yin Yang 1 (YY1) is significantly overexpressed during prostate cancer progression. Functional and cistrome studies of YY1 uncover its roles in promoting prostate oncogenesis in vitro and in vivo, as well as sustaining tumor metabolism including the Warburg effect and mitochondria respiration. Additionally, our integrated genomics and interactome profiling in prostate tumor show that YY1 and bromodomain-containing proteins (BRD2/4) co-occupy a majority of gene-regulatory elements, coactivating downstream targets. Via gene loss-of-function and rescue studies and mutagenesis of YY1-bound cis-elements, we unveil an oncogenic pathway in which YY1 directly binds and activates PFKP, a gene encoding the rate-limiting enzyme for glycolysis, significantly contributing to the YY1-enforced Warburg effect and malignant growth. Altogether, this study supports a master regulator role for YY1 in prostate tumorigenesis and reveals a YY1:BRD2/4-PFKP axis operating in advanced prostate cancer with implications for therapy.


Subject(s)
Gene Expression Regulation, Neoplastic , Phosphofructokinase-1, Type C/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , YY1 Transcription Factor/metabolism , Animals , Carcinogenesis , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Glycolysis , HEK293 Cells , Humans , Male , Mice, SCID , Phosphofructokinase-1, Type C/physiology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Transcription Factors/metabolism , Transcriptional Activation , YY1 Transcription Factor/genetics , YY1 Transcription Factor/physiology
5.
Mol Cell Proteomics ; 19(4): 730-743, 2020 04.
Article in English | MEDLINE | ID: mdl-32071147

ABSTRACT

Dynamic tyrosine phosphorylation is fundamental to a myriad of cellular processes. However, the inherently low abundance of tyrosine phosphorylation in the proteome and the inefficient enrichment of phosphotyrosine(pTyr)-containing peptides has led to poor pTyr peptide identification and quantitation, critically hindering researchers' ability to elucidate signaling pathways regulated by tyrosine phosphorylation in systems where cellular material is limited. The most popular approaches to wide-scale characterization of the tyrosine phosphoproteome use pTyr enrichment with pan-specific, anti-pTyr antibodies from a large amount of starting material. Methods that decrease the amount of starting material and increase the characterization depth of the tyrosine phosphoproteome while maintaining quantitative accuracy and precision would enable the discovery of tyrosine phosphorylation networks in rarer cell populations. To achieve these goals, the BOOST (Broad-spectrum Optimization Of Selective Triggering) method leveraging the multiplexing capability of tandem mass tags (TMT) and the use of pervanadate (PV) boost channels (cells treated with the broad-spectrum tyrosine phosphatase inhibitor PV) selectively increased the relative abundance of pTyr-containing peptides. After PV boost channels facilitated selective fragmentation of pTyr-containing peptides, TMT reporter ions delivered accurate quantitation of each peptide for the experimental samples while the quantitation from PV boost channels was ignored. This method yielded up to 6.3-fold boost in pTyr quantification depth of statistically significant data derived from contrived ratios, compared with TMT without PV boost channels or intensity-based label-free (LF) quantitation while maintaining quantitative accuracy and precision, allowing quantitation of over 2300 unique pTyr peptides from only 1 mg of T cell receptor-stimulated Jurkat T cells. The BOOST strategy can potentially be applied in analyses of other post-translational modifications where treatments that broadly elevate the levels of those modifications across the proteome are available.


Subject(s)
Phosphoproteins/metabolism , Phosphotyrosine/metabolism , Proteome/metabolism , Proteomics , Tandem Mass Spectrometry , Vanadates/metabolism , Humans , Ions , Jurkat Cells , Phosphopeptides/metabolism
6.
Blood ; 134(14): 1176-1189, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31383640

ABSTRACT

Dysregulation of polycomb repressive complex 2 (PRC2) promotes oncogenesis partly through its enzymatic function for inducing trimethylation of histone H3 lysine 27 (H3K27me3). However, it remains to be determined how PRC2 activity is regulated in normal and diseased settings. We here report a PRC2-associated cofactor, PHD finger protein 19 (PHF19; also known as polycomb-like 3), as a crucial mediator of tumorigenicity in multiple myeloma (MM). Overexpression and/or genomic amplification of PHF19 is found associated with malignant progression of MM and plasma cell leukemia, correlating to worse treatment outcomes. Using various MM models, we demonstrated a critical requirement of PHF19 for tumor growth in vitro and in vivo. Mechanistically, PHF19-mediated oncogenic effect relies on its PRC2-interacting and chromatin-binding functions. Chromatin immunoprecipitation followed by sequencing profiling showed a critical role for PHF19 in maintaining the H3K27me3 landscape. PHF19 depletion led to loss of broad H3K27me3 domains, possibly due to impaired H3K27me3 spreading from cytosine guanine dinucleotide islands, which is reminiscent to the reported effect of an "onco"-histone mutation, H3K27 to methionine (H3K27M). RNA-sequencing-based transcriptome profiling in MM lines also demonstrated a requirement of PHF19 for optimal silencing of PRC2 targets, which include cell cycle inhibitors and interferon-JAK-STAT signaling genes critically involved in tumor suppression. Correlation studies using patient sample data sets further support a clinical relevance of the PHF19-regulated pathways. Lastly, we show that MM cells are generally sensitive to PRC2 inhibitors. Collectively, this study demonstrates that PHF19 promotes MM tumorigenesis through enhancing H3K27me3 deposition and PRC2's gene-regulatory functions, lending support for PRC2 blockade as a means for MM therapeutics.


Subject(s)
Carcinogenesis/metabolism , DNA-Binding Proteins/metabolism , Histones/metabolism , Multiple Myeloma/metabolism , Polycomb Repressive Complex 2/metabolism , Transcription Factors/metabolism , Animals , Carcinogenesis/pathology , Cell Line, Tumor , Humans , Methylation , Mice , Multiple Myeloma/pathology
7.
Kidney Int ; 97(3): 602-608, 2020 03.
Article in English | MEDLINE | ID: mdl-32001064

ABSTRACT

Membranous-like glomerulopathy with masked IgG kappa deposits (MGMID) is a recently described pattern of glomerulonephritis with a unique histopathology. The pattern is characterized by subepithelial and/or mesangial immune deposits that are "masked", to immunoglobulin staining by routine immunofluorescence but strongly stain for IgG and kappa light chain after protease digestion. Patients with this pattern of glomerulonephritis are most commonly young females presenting with proteinuria and a vague history of autoimmune disease such as low titer antinuclear antibodies. Here we compared the mass spectrometry profile of laser capture microdissected glomeruli from nine MGMID renal biopsies with eight biopsies showing other patterns of membranous glomerulopathy. The protein most significantly increased in MGMID was serum amyloid P. Immunostaining showed serum amyloid P colocalized with IgG in the glomeruli of MGMID but not with PLA2R-associated membranous glomerulopathy. Serum amyloid P was positive in the glomeruli of all 32 MGMID biopsies but negative in biopsies of other types of membranous glomerulopathies such as those associated with PLA2R and THSD7A. There were four biopsies with glomerular serum amyloid P staining among the 173 biopsies that did not fulfill criteria for MGMID or amyloidosis. All four of these biopsies with positive serum amyloid P staining had a membranous pattern of glomerulopathy with IgG kappa deposits that only differed from MGMID by the lack of "masking". Thus, positive staining within glomerular deposits for serum amyloid P identifies a unique form of glomerulonephritis likely sharing a common pathophysiologic mechanism of disease.


Subject(s)
Glomerulonephritis, Membranous , Glomerulonephritis , Kidney Diseases , Female , Glomerulonephritis, Membranous/diagnosis , Humans , Immunoglobulin G , Kidney Glomerulus
8.
J Proteome Res ; 17(10): 3384-3395, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30209945

ABSTRACT

The staphylococcal accessory regulator A ( sarA) impacts the extracellular accumulation of Staphylococcus aureus virulence factors at the level of intracellular production and extracellular protease-mediated degradation. We previously used a proteomics approach that measures protein abundance of all proteoforms to demonstrate that mutation of sarA results in increased levels of extracellular proteases and assesses the impact of this on the accumulation of S. aureus exoproteins. Our previous approach was limited as it did not take into account that large, stable proteolytic products from a given protein could result in false negatives when quantified by total proteoforms. Here, our goal was to use an expanded proteomics approach utilizing a dual quantitative method for measuring abundance at both the total proteoform and full-length exoprotein levels to alleviate these false negatives and thereby provide for characterization of protease-dependent and -independent effects of sarA mutation on the S. aureus exoproteome. Proteins present in conditioned medium from overnight, stationary phase cultures of the USA300 strain LAC, an isogenic sarA mutant, and a sarA mutant unable to produce any of the known extracellular proteases ( sarA/protease) were resolved using one-dimensional gel electrophoresis. Quantitative proteomic comparisons of sarA versus sarA/protease mutants identified proteins that were cleaved in a protease-dependent manner owing to mutation of sarA, and comparisons of sarA/protease mutant versus the LAC parent strain identified proteins in which abundance was altered in a sarA mutant in a protease-independent manner. Furthermore, the proteins uniquely identified by the full-length data analysis approach eliminated false negatives observed in the total proteoform analysis. This expanded approach provided for a more comprehensive analysis of the impact of mutating sarA on the S. aureus exoproteome.


Subject(s)
Bacterial Proteins/metabolism , Peptide Hydrolases/metabolism , Proteome/metabolism , Proteomics/methods , Staphylococcus aureus/metabolism , Bacterial Proteins/genetics , Biofilms , Gene Expression Regulation, Bacterial , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/metabolism , Mutation/genetics , Proteome/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/physiology , Tandem Mass Spectrometry , Virulence/genetics
9.
Nucleic Acids Res ; 43(18): 8830-8, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26250113

ABSTRACT

Defects during chromosome replication in eukaryotes activate a signaling pathway called the S-phase checkpoint, which produces a multifaceted response that preserves genome integrity at stalled DNA replication forks. Work with budding yeast showed that the 'alternative clamp loader' known as Ctf18-RFC acts by an unknown mechanism to activate the checkpoint kinase Rad53, which then mediates much of the checkpoint response. Here we show that budding yeast Ctf18-RFC associates with DNA polymerase epsilon, via an evolutionarily conserved 'Pol ϵ binding module' in Ctf18-RFC that is produced by interaction of the carboxyl terminus of Ctf18 with the Ctf8 and Dcc1 subunits. Mutations at the end of Ctf18 disrupt the integrity of the Pol ϵ binding module and block the S-phase checkpoint pathway, downstream of the Mec1 kinase that is the budding yeast orthologue of mammalian ATR. Similar defects in checkpoint activation are produced by mutations that displace Pol ϵ from the replisome. These findings indicate that the association of Ctf18-RFC with Pol ϵ at defective replication forks is a key step in activation of the S-phase checkpoint.


Subject(s)
DNA Polymerase II/metabolism , Replication Protein C/metabolism , S Phase Cell Cycle Checkpoints , Saccharomyces cerevisiae Proteins/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Multienzyme Complexes/metabolism , Mutation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
10.
Cancer Immunol Immunother ; 65(5): 563-73, 2016 May.
Article in English | MEDLINE | ID: mdl-26980480

ABSTRACT

In the dose-escalation phase of a Phase I clinical trial in which six subjects each were vaccinated with PepCan at the 50, 100, 250, and 500 µg per peptide dose, the 50 µg dose showed the best histological regression rate. Ten additional subjects were vaccinated at this dose in the final dose phase. As with the dose-escalation phase, no dose-limiting toxicities were observed. Overall, the histological regression rates were 50% at the 50 µg dose (7 of 14) and 100 µg dose (3 of 6), and 45 % overall (14 of 31). Of subjects in whom HPV type 16 (HPV 16) was detected at entry, it became undetectable in three subjects after vaccination, and the viral loads significantly decreased in nine subjects in whom HPV 16 infection was detected at entry and exit (p = 0.008). Immune profiling revealed increased T-helper type 1 cells after vaccinations (p = 0.02 and 0.0004 after 2 and 4 vaccinations, respectively). T-helper type 2 cells initially increased after two vaccinations (p = 0.01), but decreased below the baseline level after four vaccinations although not significantly. Pre-vaccination regulatory T cell levels were significantly lower in histological responders compared to non-responders (p = 0.03). Feasibility of testing plasma for multiplex cytokine/chemokine analysis and of performing proteomic analysis of PBMCs was examined for potentially identifying biomarkers in the future. While these analyses are feasible to perform, attention needs to be given to how soon the blood samples would be processed after phlebotomy. As sufficient safety of PepCan has been demonstrated, enrollment for the Phase II clinical trial has been opened.


Subject(s)
Human papillomavirus 16/immunology , Papillomavirus Infections/immunology , Uterine Cervical Neoplasms/immunology , Viral Load/immunology , Adult , Chromatography, Liquid , Cytokines/blood , Cytokines/immunology , Dose-Response Relationship, Drug , Female , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Human papillomavirus 16/drug effects , Human papillomavirus 16/physiology , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Middle Aged , Papillomavirus Infections/drug therapy , Papillomavirus Infections/virology , Proteome/immunology , Proteome/metabolism , Proteomics/methods , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tandem Mass Spectrometry , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/virology , Vaccination/methods , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use , Viral Load/drug effects , Young Adult
11.
PLoS Pathog ; 9(9): e1003583, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24068923

ABSTRACT

Lytic gammaherpesvirus (GHV) replication facilitates the establishment of lifelong latent infection, which places the infected host at risk for numerous cancers. As obligate intracellular parasites, GHVs must control and usurp cellular signaling pathways in order to successfully replicate, disseminate to stable latency reservoirs in the host, and prevent immune-mediated clearance. To facilitate a systems-level understanding of phosphorylation-dependent signaling events directed by GHVs during lytic replication, we utilized label-free quantitative mass spectrometry to interrogate the lytic replication cycle of murine gammaherpesvirus-68 (MHV68). Compared to controls, MHV68 infection regulated by 2-fold or greater ca. 86% of identified phosphopeptides - a regulatory scale not previously observed in phosphoproteomic evaluations of discrete signal-inducing stimuli. Network analyses demonstrated that the infection-associated induction or repression of specific cellular proteins globally altered the flow of information through the host phosphoprotein network, yielding major changes to functional protein clusters and ontologically associated proteins. A series of orthogonal bioinformatics analyses revealed that MAPK and CDK-related signaling events were overrepresented in the infection-associated phosphoproteome and identified 155 host proteins, such as the transcription factor c-Jun, as putative downstream targets. Importantly, functional tests of bioinformatics-based predictions confirmed ERK1/2 and CDK1/2 as kinases that facilitate MHV68 replication and also demonstrated the importance of c-Jun. Finally, a transposon-mutant virus screen identified the MHV68 cyclin D ortholog as a viral protein that contributes to the prominent MAPK/CDK signature of the infection-associated phosphoproteome. Together, these analyses enhance an understanding of how GHVs reorganize and usurp intracellular signaling networks to facilitate infection and replication.


Subject(s)
Gammaherpesvirinae/physiology , Host-Pathogen Interactions , Models, Biological , Phosphoproteins/metabolism , Signal Transduction , Viral Proteins/metabolism , Virus Replication , 3T3 Cells , Animals , Chromatography, High Pressure Liquid , Computational Biology , Cyclin D/chemistry , Cyclin D/genetics , Cyclin D/metabolism , Gammaherpesvirinae/genetics , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , MAP Kinase Signaling System , Mice , Mutation , Phosphoproteins/chemistry , Phosphoproteins/genetics , Proteome/chemistry , Proteome/metabolism , Proteomics/methods , Proto-Oncogene Proteins c-jun/chemistry , Proto-Oncogene Proteins c-jun/metabolism , Tandem Mass Spectrometry , Viral Proteins/chemistry , Viral Proteins/genetics
12.
EMBO J ; 28(19): 2992-3004, 2009 Oct 07.
Article in English | MEDLINE | ID: mdl-19661920

ABSTRACT

The eukaryotic replisome is a crucial determinant of genome stability, but its structure is still poorly understood. We found previously that many regulatory proteins assemble around the MCM2-7 helicase at yeast replication forks to form the replisome progression complex (RPC), which might link MCM2-7 to other replisome components. Here, we show that the RPC associates with DNA polymerase alpha that primes each Okazaki fragment during lagging strand synthesis. Our data indicate that a complex of the GINS and Ctf4 components of the RPC is crucial to couple MCM2-7 to DNA polymerase alpha. Others have found recently that the Mrc1 subunit of RPCs binds DNA polymerase epsilon, which synthesises the leading strand at DNA replication forks. We show that cells lacking both Ctf4 and Mrc1 experience chronic activation of the DNA damage checkpoint during chromosome replication and do not complete the cell cycle. These findings indicate that coupling MCM2-7 to replicative polymerases is an important feature of the regulation of chromosome replication in eukaryotes, and highlight a key role for Ctf4 in this process.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Polymerase I/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , Fungal Proteins/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cell Cycle , Chromosomal Proteins, Non-Histone , DNA/metabolism , Minichromosome Maintenance Complex Component 7 , Protein Binding , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics
13.
J Virol ; 86(24): 13253-62, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23015701

ABSTRACT

Several studies have previously defined host-derived signaling events capable of driving lytic gammaherpesvirus replication or enhancing immediate-early viral gene expression. Yet signaling pathways that regulate later stages of the productive gammaherpesvirus replication cycle are still poorly defined. In this study, we utilized a mass spectrometric approach to identify c-Jun as an abundant cellular phosphoprotein present in late stages of lytic murine gammaherpesvirus 68 (MHV68) infection. Kinetically, c-Jun phosphorylation was enhanced as infection progressed, and this correlated with enhanced phosphorylation of the c-Jun amino-terminal kinases JNK1 and JNK2 and activation of AP-1 transcription. These events were dependent on progression beyond viral immediate-early gene expression, but not dependent on viral DNA replication. Both pharmacologic and dominant-negative blockade of JNK1/2 activity inhibited viral replication, and this correlated with inhibition of viral DNA synthesis and reduced viral gene expression. These data suggest a model in which MHV68 by necessity amplifies and usurps JNK/c-Jun signaling as infection progresses in order to facilitate late stages of the MHV68 lytic infection cycle.


Subject(s)
Gammaherpesvirinae/physiology , MAP Kinase Kinase 4/metabolism , Signal Transduction , Virus Replication , Amino Acid Sequence , Animals , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Tandem Mass Spectrometry , Viral Proteins/chemistry , Viral Proteins/metabolism
14.
Nat Cell Biol ; 8(4): 358-66, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16531994

ABSTRACT

The components of the replisome that preserve genomic stability by controlling the progression of eukaryotic DNA replication forks are poorly understood. Here, we show that the GINS (go ichi ni san) complex allows the MCM (minichromosome maintenance) helicase to interact with key regulatory proteins in large replisome progression complexes (RPCs) that are assembled during initiation and disassembled at the end of S phase. RPC components include the essential initiation and elongation factor, Cdc45, the checkpoint mediator Mrc1, the Tof1-Csm3 complex that allows replication forks to pause at protein-DNA barriers, the histone chaperone FACT (facilitates chromatin transcription) and Ctf4, which helps to establish sister chromatid cohesion. RPCs also interact with Mcm10 and topoisomerase I. During initiation, GINS is essential for a specific subset of RPC proteins to interact with MCM. GINS is also important for the normal progression of DNA replication forks, and we show that it is required after initiation to maintain the association between MCM and Cdc45 within RPCs.


Subject(s)
DNA Replication , DNA, Fungal/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromatography, Liquid , DNA, Fungal/genetics , DNA-Binding Proteins/genetics , Immunoprecipitation , Mass Spectrometry , Minichromosome Maintenance 1 Protein , S Phase/physiology , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics
15.
Hum Genomics ; 5(6): 703-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22155608

ABSTRACT

Many primary biological databases are dedicated to providing annotation for a specific type of biological molecule such as a clone, transcript, gene or protein, but often with limited cross-references. Therefore, enhanced mapping is required between these databases to facilitate the correlation of independent experimental datasets. For example, molecular biology experiments conducted on samples (DNA, mRNA or protein) often yield more than one type of 'omics' dataset as an object for analysis (eg a sample can have a genomics as well as proteomics expression dataset available for analysis). Thus, in order to map the two datasets, the identifier type from one dataset is required to be linked to another dataset, so preventing loss of critical information in downstream analysis. This identifier mapping can be performed using identifier converter software relevant to the query and target identifier databases. This review presents the publicly available web-based biological database identifier converters, with comparison of their usage, input and output formats, and the types of available query and target database identifier types.


Subject(s)
Computational Biology , Database Management Systems , Genomics , Proteomics , Humans , Software
16.
Nat Commun ; 12(1): 1045, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33594072

ABSTRACT

Recurring chromosomal translocation t(10;17)(p15;q21) present in a subset of human acute myeloid leukemia (AML) patients creates an aberrant fusion gene termed ZMYND11-MBTD1 (ZM); however, its function remains undetermined. Here, we show that ZM confers primary murine hematopoietic stem/progenitor cells indefinite self-renewal capability ex vivo and causes AML in vivo. Genomics profilings reveal that ZM directly binds to and maintains high expression of pro-leukemic genes including Hoxa, Meis1, Myb, Myc and Sox4. Mechanistically, ZM recruits the NuA4/Tip60 histone acetyltransferase complex to cis-regulatory elements, sustaining an active chromatin state enriched in histone acetylation and devoid of repressive histone marks. Systematic mutagenesis of ZM demonstrates essential requirements of Tip60 interaction and an H3K36me3-binding PWWP (Pro-Trp-Trp-Pro) domain for oncogenesis. Inhibitor of histone acetylation-'reading' bromodomain proteins, which act downstream of ZM, is efficacious in treating ZM-induced AML. Collectively, this study demonstrates AML-causing effects of ZM, examines its gene-regulatory roles, and reports an attractive mechanism-guided therapeutic strategy.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Co-Repressor Proteins/chemistry , Co-Repressor Proteins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Leukemia, Myeloid, Acute/pathology , Lysine Acetyltransferase 5/metabolism , Acetylation , Animals , Carcinogenesis , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic , Disease Models, Animal , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Leukemic , Genome, Human , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Mice, Inbred BALB C , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oncogene Proteins, Fusion/metabolism , Protein Binding , Protein Domains , Transcription Factors/metabolism
17.
J Proteome Res ; 8(12): 5629-41, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19899799

ABSTRACT

The Rac-specific GEF (guanine-nucleotide exchange factor) Tiam1 has important functions in multiple cellular processes including proliferation, apoptosis and adherens junction maintenance. Here we describe a modified tandem affinity purification (TAP) technique that we have applied to specifically enrich Tiam1-containing protein complexes from mammalian cells. Using this technique in conjunction with LC-MS/MS mass spectrometry, we have identified additional Tiam1-interacting proteins not seen with the standard technique, and have identified multiple 14-3-3 family members as Tiam1 interactors. We confirm the Tiam1/14-3-3 protein interaction by GST-pulldown and coimmunoprecipitation experiments, show that it is phosphorylation-dependent, and that they colocalize in cells. The interaction is largely dependent on the N-terminal region of Tiam1; within this region, there are four putative phospho-serine-containing 14-3-3 binding motifs, and we confirm that two of them (Ser172 and Ser231) are phosphorylated in cells using mass spectrometry. Moreover, we show that phosphorylation at three of these motifs (containing Ser60, Ser172 and Ser231) is required for the binding of 14-3-3 proteins to this region of Tiam1. We show that phosphorylation of these sites does not affect Tiam1 activity; significantly however, we demonstrate that phosphorylation of the Ser60-containing motif is required for the degradation of Tiam1. Thus, we have established and proven methodology that allows the identification of additional protein-protein interactions in mammalian cells, resulting in the discovery of a novel mechanism of regulating Tiam1 stability.


Subject(s)
14-3-3 Proteins/chemistry , Chromatography, Affinity/methods , Guanine Nucleotide Exchange Factors/chemistry , 14-3-3 Proteins/metabolism , Animals , Binding Sites , Cell Line , Chromatography, Affinity/instrumentation , Guanine Nucleotide Exchange Factors/isolation & purification , Guanine Nucleotide Exchange Factors/metabolism , Humans , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/isolation & purification , Phosphorylation , Protein Binding , Protein Stability , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , Tandem Mass Spectrometry
18.
PLoS One ; 14(1): e0199274, 2019.
Article in English | MEDLINE | ID: mdl-30699108

ABSTRACT

BACKGROUND: Resistant starch is a prebiotic metabolized by the gut bacteria. It has been shown to attenuate chronic kidney disease (CKD) progression in rats. Previous studies employed taxonomic analysis using 16S rRNA sequencing and untargeted metabolomics profiling. Here we expand these studies by metaproteomics, gaining new insight into the host-microbiome interaction. METHODS: Differences between cecum contents in CKD rats fed a diet containing resistant starch with those fed a diet containing digestible starch were examined by comparative metaproteomics analysis. Taxonomic information was obtained using unique protein sequences. Our methodology results in quantitative data covering both host and bacterial proteins. RESULTS: 5,834 proteins were quantified, with 947 proteins originating from the host organism. Taxonomic information derived from metaproteomics data surpassed previous 16S RNA analysis, and reached species resolutions for moderately abundant taxonomic groups. In particular, the Ruminococcaceae family becomes well resolved-with butyrate producers and amylolytic species such as R. bromii clearly visible and significantly higher while fibrolytic species such as R. flavefaciens are significantly lower with resistant starch feeding. The observed changes in protein patterns are consistent with fiber-associated improvement in CKD phenotype. Several known host CKD-associated proteins and biomarkers of impaired kidney function were significantly reduced with resistant starch supplementation. Data are available via ProteomeXchange with identifier PXD008845. CONCLUSIONS: Metaproteomics analysis of cecum contents of CKD rats with and without resistant starch supplementation reveals changes within gut microbiota at unprecedented resolution, providing both functional and taxonomic information. Proteins and organisms differentially abundant with RS supplementation point toward a shift from mucin degraders to butyrate producers.


Subject(s)
Bacterial Proteins/analysis , Cecum/microbiology , Gastrointestinal Microbiome , Proteome/analysis , Proteomics , Renal Insufficiency, Chronic/chemically induced , Ruminococcus , Starch/adverse effects , Animals , Disease Progression , Male , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/microbiology , Ruminococcus/classification , Ruminococcus/growth & development , Starch/pharmacology
19.
J Bacteriol ; 190(15): 5265-78, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18539737

ABSTRACT

One-dimensional polyacrylamide gel electrophoresis followed by nanocapillary liquid chromatography coupled with mass spectrometry was used to analyze proteins isolated from Staphylococcus aureus UAMS-1 after 3, 6, 12, and 24 h of in vitro growth. Protein abundance was determined using a quantitative value termed normalized peptide number, and overall, proteins known to be associated with the cell wall were more abundant early on in growth, while proteins known to be secreted into the surrounding milieu were more abundant late in growth. In addition, proteins from spent media and cell lysates of strain UAMS-1 and its isogenic sarA, agr, and sarA agr regulatory mutant strains during exponential growth were identified, and their relative abundances were compared. Extracellular proteins known to be regulated by the global regulators sarA and agr displayed protein levels in accordance with what is known regarding the effects of these regulators. For example, cysteine protease (SspB), endopeptidase (SspA), staphopain (ScpA), and aureolysin (Aur) were higher in abundance in the sarA and sarA agr mutants than in strain UAMS-1. The immunoglobulin G (IgG)-binding protein (Sbi), immunodominant staphylococcal antigen A (IsaA), IgG-binding protein A (Spa), and the heme-iron-binding protein (IsdA) were most abundant in the agr mutant background. Proteins whose abundance was decreased in the sarA mutant included fibrinogen-binding protein (Fib [Efb]), IsaA, lipase 1 and 2, and two proteins identified as putative leukocidin F and S subunits of the two-component leukotoxin family. Collectively, this approach identified 1,263 proteins (matches of two peptides or more) and provided a convenient and reliable way of identifying proteins and comparing their relative abundances.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Mutation , Proteome/analysis , Staphylococcus aureus/physiology , Trans-Activators/genetics , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Bacterial Proteins/physiology , Chromatography, Liquid , DNA Transposable Elements , Electrophoresis, Polyacrylamide Gel , Mutagenesis, Insertional , Staphylococcus aureus/genetics , Tandem Mass Spectrometry , Trans-Activators/physiology
20.
J Neurochem ; 105(5): 1979-95, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18266931

ABSTRACT

The myelin proteolipid protein gene (Plp1) encodes the most abundant protein found in CNS myelin, accounting for nearly one-half of the total protein. Its expression in oligodendrocytes is developmentally regulated - peaking during the active myelination period of CNS development. Previously, we have identified a novel enhancer (designated ASE) in intron 1 DNA that appears to be important in mediating the surge of Plp1 gene activity during the active myelination period. Evidence suggests that the ASE participates in the formation of a specialized multi-protein/DNA complex called an enhanceosome. The current study describes an optimized, five-step, DNA affinity chromatography purification procedure to purify nuclear proteins from mouse brain that bind to the 85-bp ASE sequence, specifically. Electrophoretic mobility shift assay analysis demonstrated that specific DNA-binding activity was retained throughout the purification procedure, resulting in concomitant enrichment of nucleoprotein complexes. Identification of the purported regulatory factors was achieved through mass spectrometry analysis and included over 20 sequence-specific DNA-binding proteins. Supplementary western blot analyses to determine which of these sequence-specific factors are present in oligodendrocytes, and their developmental and regional expression in whole brain, suggest that Puralpha and Purbeta rank highest among the candidate factors as constituents of the multi-protein complex formed on the ASE.


Subject(s)
Enhancer Elements, Genetic/physiology , Introns/physiology , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Nuclear Proteins/metabolism , Proteomics/methods , Animals , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Protein Binding/physiology
SELECTION OF CITATIONS
SEARCH DETAIL