Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters

Country/Region as subject
Publication year range
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.
Brief Bioinform ; 25(Supplement_1)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39041914

ABSTRACT

This manuscript describes the development of a resource module that is part of a learning platform named 'NIGMS Sandbox for Cloud-based Learning' https://github.com/NIGMS/NIGMS-Sandbox. The overall genesis of the Sandbox is described in the editorial NIGMS Sandbox at the beginning of this Supplement. This module delivers learning materials on protein quantification in an interactive format that uses appropriate cloud resources for data access and analyses. Quantitative proteomics is a rapidly growing discipline due to the cutting-edge technologies of high resolution mass spectrometry. There are many data types to consider for proteome quantification including data dependent acquisition, data independent acquisition, multiplexing with Tandem Mass Tag reporter ions, spectral counts, and more. As part of the NIH NIGMS Sandbox effort, we developed a learning module to introduce students to mass spectrometry terminology, normalization methods, statistical designs, and basics of R programming. By utilizing the Google Cloud environment, the learning module is easily accessible without the need for complex installation procedures. The proteome quantification module demonstrates the analysis using a provided TMT10plex data set using MS3 reporter ion intensity quantitative values in a Jupyter notebook with an R kernel. The learning module begins with the raw intensities, performs normalization, and differential abundance analysis using limma models, and is designed for researchers with a basic understanding of mass spectrometry and R programming language. Learners walk away with a better understanding of how to navigate Google Cloud Platform for proteomic research, and with the basics of mass spectrometry data analysis at the command line. This manuscript describes the development of a resource module that is part of a learning platform named ``NIGMS Sandbox for Cloud-based Learning'' https://github.com/NIGMS/NIGMS-Sandbox. The overall genesis of the Sandbox is described in the editorial NIGMS Sandbox [1] at the beginning of this Supplement. This module delivers learning materials on the analysis of bulk and single-cell ATAC-seq data in an interactive format that uses appropriate cloud resources for data access and analyses.


Subject(s)
Cloud Computing , Proteome , Proteomics , Software , Proteome/metabolism , Proteomics/methods , Mass Spectrometry , Humans
4.
J Virol ; 98(7): e0079424, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38940558

ABSTRACT

Coronavirus disease 2019 (COVID-19) has claimed millions of lives since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and lung disease appears the primary cause of death in COVID-19 patients. However, the underlying mechanisms of COVID-19 pathogenesis remain elusive, and there is no existing model where human disease can be faithfully recapitulated and conditions for the infection process can be experimentally controlled. Herein we report the establishment of an ex vivo human precision-cut lung slice (hPCLS) platform for studying SARS-CoV-2 pathogenicity and innate immune responses, and for evaluating the efficacy of antiviral drugs against SARS-CoV-2. We show that while SARS-CoV-2 continued to replicate during the course of infection of hPCLS, infectious virus production peaked within 2 days, and rapidly declined thereafter. Although most proinflammatory cytokines examined were induced by SARS-CoV-2 infection, the degree of induction and types of cytokines varied significantly among hPCLS from individual donors. Two cytokines in particular, IP-10 and IL-8, were highly and consistently induced, suggesting a role in the pathogenesis of COVID-19. Histopathological examination revealed focal cytopathic effects late in the infection. Transcriptomic and proteomic analyses identified molecular signatures and cellular pathways that are largely consistent with the progression of COVID-19 in patients. Furthermore, we show that homoharringtonine, a natural plant alkaloid derived from Cephalotoxus fortunei, not only inhibited virus replication but also production of pro-inflammatory cytokines, and thus ameliorated the histopathological changes caused by SARS-CoV-2 infection, demonstrating the usefulness of the hPCLS platform for evaluating antiviral drugs. IMPORTANCE: Here, established an ex vivo human precision-cut lung slice platform for assessing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, viral replication kinetics, innate immune response, disease progression, and antiviral drugs. Using this platform, we identified early induction of specific cytokines, especially IP-10 and IL-8, as potential predictors for severe coronavirus disease 2019 (COVID-19), and uncovered a hitherto unrecognized phenomenon that while infectious virus disappears at late times of infection, viral RNA persists and lung histopathology commences. This finding may have important clinical implications for both acute and post-acute sequelae of COVID-19. This platform recapitulates some of the characteristics of lung disease observed in severe COVID-19 patients and is therefore a useful platform for understanding mechanisms of SARS-CoV-2 pathogenesis and for evaluating the efficacy of antiviral drugs.


Subject(s)
Antiviral Agents , COVID-19 , Cytokines , Lung , SARS-CoV-2 , Virus Replication , Humans , Lung/virology , Lung/pathology , Lung/drug effects , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Antiviral Agents/pharmacology , COVID-19/virology , COVID-19/pathology , Cytokines/metabolism , Virus Replication/drug effects , Immunity, Innate , COVID-19 Drug Treatment
5.
Nucleic Acids Res ; 51(14): 7288-7313, 2023 08 11.
Article in English | MEDLINE | ID: mdl-37378433

ABSTRACT

We have conducted a detailed transcriptomic, proteomic and phosphoproteomic analysis of CDK8 and its paralog CDK19, alternative enzymatic components of the kinase module associated with transcriptional Mediator complex and implicated in development and diseases. This analysis was performed using genetic modifications of CDK8 and CDK19, selective CDK8/19 small molecule kinase inhibitors and a potent CDK8/19 PROTAC degrader. CDK8/19 inhibition in cells exposed to serum or to agonists of NFκB or protein kinase C (PKC) reduced the induction of signal-responsive genes, indicating a pleiotropic role of Mediator kinases in signal-induced transcriptional reprogramming. CDK8/19 inhibition under basal conditions initially downregulated a small group of genes, most of which were inducible by serum or PKC stimulation. Prolonged CDK8/19 inhibition or mutagenesis upregulated a larger gene set, along with a post-transcriptional increase in the proteins comprising the core Mediator complex and its kinase module. Regulation of both RNA and protein expression required CDK8/19 kinase activities but both enzymes protected their binding partner cyclin C from proteolytic degradation in a kinase-independent manner. Analysis of isogenic cell populations expressing CDK8, CDK19 or their kinase-inactive mutants revealed that CDK8 and CDK19 have the same qualitative effects on protein phosphorylation and gene expression at the RNA and protein levels, whereas differential effects of CDK8 versus CDK19 knockouts were attributable to quantitative differences in their expression and activity rather than different functions.


Subject(s)
Cyclin-Dependent Kinases , Mediator Complex , Humans , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Mediator Complex/genetics , Mediator Complex/metabolism , Phosphorylation , Proteomics , RNA/metabolism
6.
Mol Cell ; 62(4): 558-71, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27132940

ABSTRACT

Histone H2B monoubiquitination (H2Bub1) is centrally involved in gene regulation. The deubiquitination module (DUBm) of the SAGA complex is a major regulator of global H2Bub1 levels, and components of this DUBm are linked to both neurodegenerative diseases and cancer. Unexpectedly, we find that ablation of USP22, the enzymatic center of the DUBm, leads to a reduction, rather than an increase, in global H2bub1 levels. In contrast, depletion of non-enzymatic components, ATXN7L3 or ENY2, results in increased H2Bub1. These observations led us to discover two H2Bub1 DUBs, USP27X and USP51, which function independently of SAGA and compete with USP22 for ATXN7L3 and ENY2 for activity. Like USP22, USP51 and USP27X are required for normal cell proliferation, and their depletion suppresses tumor growth. Our results reveal that ATXN7L3 and ENY2 orchestrate activities of multiple deubiquitinating enzymes and that imbalances in these activities likely potentiate human diseases including cancer.


Subject(s)
Breast Neoplasms/enzymology , Cell Proliferation , Deubiquitinating Enzymes/metabolism , Histones/metabolism , Transcription Factors/metabolism , Tumor Burden , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Deubiquitinating Enzymes/genetics , Endopeptidases/genetics , Endopeptidases/metabolism , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genotype , HEK293 Cells , Humans , MCF-7 Cells , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Phenotype , RNA Interference , Signal Transduction , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism , Time Factors , Transcription Factors/genetics , Transfection , Ubiquitin Thiolesterase , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitination
7.
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
8.
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
9.
Int J Mol Sci ; 24(17)2023 Aug 23.
Article in English | MEDLINE | ID: mdl-37685904

ABSTRACT

Infection with hepatitis B virus (HBV) is a main risk factor for hepatocellular carcinoma (HCC). Extracellular vesicles, such as exosomes, play an important role in tumor development and metastasis, including regulation of HBV-related HCC. In this study, we have characterized exosome microRNA and proteins released in vitro from hepatitis B virus (HBV)-related HCC cell lines SNU-423 and SNU-182 and immortalized normal hepatocyte cell lines (THLE2 and THLE3) using microRNA sequencing and mass spectrometry. Bioinformatics, including functional enrichment and network analysis, combined with survival analysis using data related to HCC in The Cancer Genome Atlas (TCGA) database, were applied to examine the prognostic significance of the results. More than 40 microRNAs and 200 proteins were significantly dysregulated (p < 0.05) in the exosomes released from HCC cells in comparison with the normal liver cells. The functional analysis of the differentially expressed exosomal miRNAs (i.e., mir-483, mir-133a, mir-34a, mir-155, mir-183, mir-182), their predicted targets, and exosomal differentially expressed proteins (i.e., POSTN, STAM, EXOC8, SNX9, COL1A2, IDH1, FN1) showed correlation with pathways associated with HBV, virus activity and invasion, exosome formation and adhesion, and exogenous protein binding. The results from this study may help in our understanding of the role of HBV infection in the development of HCC and in the development of new targets for treatment or non-invasive predictive biomarkers of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/genetics , Hepatitis B virus , Liver Neoplasms/genetics , Hepatocytes
10.
Nature ; 532(7599): 329-33, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27027282

ABSTRACT

It has been widely accepted that 5-methylcytosine is the only form of DNA methylation in mammalian genomes. Here we identify N(6)-methyladenine as another form of DNA modification in mouse embryonic stem cells. Alkbh1 encodes a demethylase for N(6)-methyladenine. An increase of N(6)-methyladenine levels in Alkbh1-deficient cells leads to transcriptional silencing. N(6)-methyladenine deposition is inversely correlated with the evolutionary age of LINE-1 transposons; its deposition is strongly enriched at young (<1.5 million years old) but not old (>6 million years old) L1 elements. The deposition of N(6)-methyladenine correlates with epigenetic silencing of such LINE-1 transposons, together with their neighbouring enhancers and genes, thereby resisting the gene activation signals during embryonic stem cell differentiation. As young full-length LINE-1 transposons are strongly enriched on the X chromosome, genes located on the X chromosome are also silenced. Thus, N(6)-methyladenine developed a new role in epigenetic silencing in mammalian evolution distinct from its role in gene activation in other organisms. Our results demonstrate that N(6)-methyladenine constitutes a crucial component of the epigenetic regulation repertoire in mammalian genomes.


Subject(s)
Adenine/analogs & derivatives , DNA Methylation , Epigenesis, Genetic/genetics , Mouse Embryonic Stem Cells/metabolism , Adenine/metabolism , AlkB Homolog 1, Histone H2a Dioxygenase , Animals , Cell Differentiation/genetics , DNA Transposable Elements/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/deficiency , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Enhancer Elements, Genetic/genetics , Evolution, Molecular , Gene Silencing , Long Interspersed Nucleotide Elements/genetics , Mammals/genetics , Mice , Mouse Embryonic Stem Cells/cytology , Up-Regulation/genetics , X Chromosome/genetics , X Chromosome/metabolism
11.
Int J Mol Sci ; 23(2)2022 Jan 08.
Article in English | MEDLINE | ID: mdl-35054859

ABSTRACT

The damaging effects of ionizing radiation (IR) on bone mass are well-documented in mice and humans and are most likely due to increased osteoclast number and function. However, the mechanisms leading to inappropriate increases in osteoclastic bone resorption are only partially understood. Here, we show that exposure to multiple fractions of low-doses (10 fractions of 0.4 Gy total body irradiation [TBI]/week, i.e., fractionated exposure) and/or a single exposure to the same total dose of 4 Gy TBI causes a decrease in trabecular, but not cortical, bone mass in young adult male mice. This damaging effect was associated with highly activated bone resorption. Both osteoclast differentiation and maturation increased in cultures of bone marrow-derived macrophages from mice exposed to either fractionated or singular TBI. IR also increased the expression and enzymatic activity of mitochondrial deacetylase Sirtuin-3 (Sirt3)-an essential protein for osteoclast mitochondrial activity and bone resorption in the development of osteoporosis. Osteoclast progenitors lacking Sirt3 exposed to IR exhibited impaired resorptive activity. Taken together, targeting impairment of osteoclast mitochondrial activity could be a novel therapeutic strategy for IR-induced bone loss, and Sirt3 is likely a major mediator of this effect.


Subject(s)
Bone Resorption/pathology , Mitochondria/metabolism , Mitochondria/radiation effects , Osteoclasts/metabolism , Osteoclasts/radiation effects , Radiation, Ionizing , Animals , Cancellous Bone/pathology , Cancellous Bone/radiation effects , Cell Respiration/radiation effects , Dose Fractionation, Radiation , Male , Mice, Inbred C57BL , Sirtuin 3/metabolism
12.
BMC Bioinformatics ; 22(1): 275, 2021 May 26.
Article in English | MEDLINE | ID: mdl-34039258

ABSTRACT

BACKGROUND: Histone post-translational modifications (PTMs) play an important role in our system by regulating the structure of chromatin and therefore contribute to the regulation of gene and protein expression. Irregularities in histone PTMs can lead to a variety of different diseases including various forms of cancer. Histone modifications are analyzed using high resolution mass spectrometry, which generate large amounts of data that requires sophisticated bioinformatics tools for analysis and visualization. PTMViz is designed for downstream differential abundance analysis and visualization of both protein and/or histone modifications. RESULTS: PTMViz provides users with data tables and visualization plots of significantly differentiated proteins and histone PTMs between two sample groups. All the data is packaged into interactive data tables and graphs using the Shiny platform to help the user explore the results in a fast and efficient manner to assess if changes in the system are due to protein abundance changes or epigenetic changes. In the example data provided, we identified several proteins differentially regulated in the dopaminergic pathway between mice treated with methamphetamine compared to a saline control. We also identified histone post-translational modifications including histone H3K9me, H3K27me3, H4K16ac, and that were regulated due to drug exposure. CONCLUSIONS: Histone modifications play an integral role in the regulation of gene expression. PTMViz provides an interactive platform for analyzing proteins and histone post-translational modifications from mass spectrometry data in order to quickly identify differentially expressed proteins and PTMs.


Subject(s)
Histones , Protein Processing, Post-Translational , Animals , Chromatin , Histone Code , Histones/metabolism , Methylation , Mice
13.
Angiogenesis ; 24(3): 597-611, 2021 08.
Article in English | MEDLINE | ID: mdl-33629198

ABSTRACT

Antibiotic-induced microbial imbalance, or dysbiosis, has systemic and long-lasting effects on the host and response to cancer therapies. However, the effects on tumor endothelial cells are largely unknown. Therefore, the goal of the current study was to generate matched B16-F10 melanoma associated endothelial cell lines isolated from mice with and without antibiotic-induced dysbiosis. After validating endothelial cell markers on a genomic and proteomic level, functional angiogenesis assays (i.e., migration and tube formation) also confirmed their vasculature origin. Subsequently, we found that tumor endothelial cells derived from dysbiotic mice (TEC-Dys) were more sensitive to ionizing radiotherapy in the range of clinically-relevant hypofractionated doses, as compared to tumor endothelial cells derived from orthobiotic mice (TEC-Ortho). In order to identify tumor vasculature-associated drug targets during dysbiosis, we used tandem mass tag mass spectroscopy and focused on the statistically significant cellular membrane proteins overexpressed in TEC-Dys. By these criteria c-Met was the most differentially expressed protein, which was validated histologically by comparing tumors with or without dysbiosis. Moreover, in vitro, c-Met inhibitors Foretinib, Crizotinib and Cabozantinib were significantly more effective against TEC-Dys than TEC-Ortho. In vivo, Foretinib inhibited tumor growth to a greater extent during dysbiosis as compared to orthobiotic conditions. Thus, we surmise that tumor response in dysbiotic patients may be greatly improved by targeting dysbiosis-induced pathways, such as c-Met, distinct from the many targets suppressed due to dysbiosis.


Subject(s)
Dysbiosis , Endothelial Cells/enzymology , Melanoma, Experimental , Neovascularization, Pathologic , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met , Animals , Dysbiosis/enzymology , Dysbiosis/microbiology , Melanoma, Experimental/blood supply , Melanoma, Experimental/enzymology , Melanoma, Experimental/microbiology , Melanoma, Experimental/therapy , Mice , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/microbiology , Neovascularization, Pathologic/therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Radiotherapy
14.
FASEB J ; 34(11): 15516-15530, 2020 11.
Article in English | MEDLINE | ID: mdl-32981077

ABSTRACT

Many factors contribute to the health risks encountered by astronauts on missions outside Earth's atmosphere. Spaceflight-induced potential adverse neurovascular damage and late neurodegeneration are a chief concern. The goal of the present study was to characterize the effects of spaceflight on oxidative damage in the mouse brain and its impact on blood-brain barrier (BBB) integrity. Ten-week-old male C57BL/6 mice were launched to the International Space Station (ISS) for 35 days as part of Space-X 12 mission. Ground control (GC) mice were maintained on Earth in flight hardware cages. Within 38 ± 4 hours after returning from the ISS, mice were euthanized and brain tissues were collected for analysis. Quantitative assessment of brain tissue demonstrated that spaceflight caused an up to 2.2-fold increase in apoptosis in the hippocampus compared to the control group. Immunohistochemical analysis of the mouse brain revealed an increased expression of aquaporin4 (AQP4) in the flight hippocampus compared to the controls. There was also a significant increase in the expression of platelet endothelial cell adhesion molecule-1 (PECAM-1) and a decrease in the expression of the BBB-related tight junction protein, Zonula occludens-1 (ZO-1). These results indicate a disturbance of BBB integrity. Quantitative proteomic analysis showed significant alterations in pathways responsible for neurovascular integrity, mitochondrial function, neuronal structure, protein/organelle transport, and metabolism in the brain after spaceflight. Changes in pathways associated with adhesion and molecular remodeling were also documented. These data indicate that long-term spaceflight may have pathological and functional consequences associated with neurovascular damage and late neurodegeneration.


Subject(s)
Blood-Brain Barrier/pathology , Brain/pathology , Disease Models, Animal , Mitochondria/pathology , Oxidative Stress/radiation effects , Proteome/analysis , Space Flight/methods , Animals , Apoptosis , Biological Transport , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/radiation effects , Brain/metabolism , Brain/radiation effects , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/radiation effects , Proteome/radiation effects , Weightlessness
15.
J Proteome Res ; 19(3): 1183-1195, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32027144

ABSTRACT

Cells respond to environmental perturbations and insults through modulating protein abundance and function. However, the majority of studies have focused on changes in RNA abundance because quantitative transcriptomics has historically been more facile than quantitative proteomics. Modern Orbitrap mass spectrometers now provide sensitive and deep proteome coverage, allowing direct, global quantification of not only protein abundance but also post-translational modifications (PTMs) that regulate protein activity. We implemented and validated using the well-characterized heat shock response of budding yeast, a tandem mass tagging (TMT), triple-stage mass spectrometry (MS3) strategy to measure global changes in the proteome during the yeast heat shock response over nine time points. We report that basic-pH, ultra-high performance liquid chromatography (UPLC) fractionation of tryptic peptides yields superfractions of minimal redundancy, a crucial requirement for deep coverage and quantification by subsequent LC-MS3. We quantified 2275 proteins across three biological replicates and found that differential expression peaked near 90 min following heat shock (with 868 differentially expressed proteins at 5% false discovery rate). The sensitivity of the approach also allowed us to detect changes in the relative abundance of ubiquitination and phosphorylation PTMs over time. Remarkably, relative quantification of post-translationally modified peptides revealed striking evidence of regulation of the heat shock response by protein PTMs. These data demonstrate that the high precision of TMT-MS3 enables peptide-level quantification of samples, which can reveal important regulation of protein abundance and regulatory PTMs under various experimental conditions.


Subject(s)
Proteome , Proteomics , Chromatography, Liquid , Heat-Shock Response , Mass Spectrometry
16.
J Biol Chem ; 294(28): 10969-10986, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31152063

ABSTRACT

The Musashi family of mRNA translational regulators controls both physiological and pathological stem cell self-renewal primarily by repressing target mRNAs that promote differentiation. In response to differentiation cues, Musashi can switch from a repressor to an activator of target mRNA translation. However, the molecular events that distinguish Musashi-mediated translational activation from repression are not understood. We have previously reported that Musashi function is required for the maturation of Xenopus oocytes and specifically for translational activation of specific dormant maternal mRNAs. Here, we employed MS to identify cellular factors necessary for Musashi-dependent mRNA translational activation. We report that Musashi1 needs to associate with the embryonic poly(A)-binding protein (ePABP) or the canonical somatic cell poly(A)-binding protein PABPC1 for activation of Musashi target mRNA translation. Co-immunoprecipitation studies demonstrated an increased Musashi1 interaction with ePABP during oocyte maturation. Attenuation of endogenous ePABP activity severely compromised Musashi function, preventing downstream signaling and blocking oocyte maturation. Ectopic expression of either ePABP or PABPC1 restored Musashi-dependent mRNA translational activation and maturation of ePABP-attenuated oocytes. Consistent with these Xenopus findings, PABPC1 remained associated with Musashi under conditions of Musashi target mRNA de-repression and translation during mammalian stem cell differentiation. Because association of Musashi1 with poly(A)-binding proteins has previously been implicated only in repression of Musashi target mRNAs, our findings reveal novel context-dependent roles for the interaction of Musashi with poly(A)-binding protein family members in response to extracellular cues that control cell fate.


Subject(s)
Nerve Tissue Proteins/metabolism , Poly(A)-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Xenopus Proteins/metabolism , Animals , Cell Cycle , Cell Differentiation , Nerve Tissue Proteins/physiology , Oocytes/metabolism , Oogenesis/physiology , Poly(A)-Binding Protein I/genetics , Poly(A)-Binding Proteins/genetics , Polyadenylation , Protein Biosynthesis , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Ribonucleoproteins/physiology , Signal Transduction , Xenopus Proteins/physiology , Xenopus laevis/metabolism
17.
Am J Physiol Lung Cell Mol Physiol ; 318(4): L592-L605, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32022592

ABSTRACT

Respiratory disease is a leading cause of mortality in patients with osteogenesis imperfecta (OI), a connective tissue disease that causes severely reduced bone mass and is most commonly caused by dominant mutations in type I collagen genes. Previous studies proposed that impaired respiratory function in OI patients was secondary to skeletal deformities; however, recent evidence suggests the existence of a primary lung defect. Here, we analyzed the lung phenotype of Crtap knockout (KO) mice, a mouse model of recessive OI. While we confirm changes in the lung parenchyma that are reminiscent of emphysema, we show that CrtapKO lung fibroblasts synthesize type I collagen with altered posttranslation modifications consistent with those observed in bone and skin. Unrestrained whole body plethysmography showed a significant decrease in expiratory time, resulting in an increased ratio of inspiratory time over expiratory time and a concomitant increase of the inspiratory duty cycle in CrtapKO compared with WT mice. Closed-chest measurements using the forced oscillation technique showed increased respiratory system elastance, decreased respiratory system compliance, and increased tissue damping and elasticity in CrtapKO mice compared with WT. Pressure-volume curves showed significant differences in lung volumes and in the shape of the curves between CrtapKO mice and WT mice, with and without adjustment for body weight. This is the first evidence that collagen defects in OI cause primary changes in lung parenchyma and several respiratory parameters and thus negatively impact lung function.


Subject(s)
Collagen Type I/genetics , Extracellular Matrix Proteins/genetics , Molecular Chaperones/genetics , Osteogenesis Imperfecta/genetics , Animals , Cells, Cultured , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Protein Processing, Post-Translational/genetics
18.
Int J Mol Sci ; 21(5)2020 Mar 09.
Article in English | MEDLINE | ID: mdl-32182883

ABSTRACT

BACKGROUND: Chemotherapy treatment for breast cancer can induce cognitive impairments often involving oxidative stress. The brain, as a whole, is susceptible to oxidative stress due to its high-energy requirements, limited anaerobic respiration capacities, and limited antioxidant defenses. The goal of the current study was to determine if the manganese porphyrin superoxide dismutase mimetic MnTnBuOE-2-PyP (MnBuOE) could ameliorate the effects of doxorubicin, cyclophosphamide, and paclitaxel (AC-T) on mature dendrite morphology and cognitive function. METHODS: Four-month-old female C57BL/6 mice received intraperitoneal injections of chemotherapy followed by subcutaneous injections of MnBuOE. Four weeks following chemotherapy treatment, mice were tested for hippocampus-dependent cognitive performance in the Morris water maze. After testing, brains were collected for Golgi staining and molecular analyses. RESULTS: MnBuOE treatment preserved spatial memory during the Morris water-maze. MnBuOE/AC-T showed spatial memory retention during all probe trials. AC-T treatment significantly impaired spatial memory retention in the first and third probe trial (no platform). AC-T treatment decreased dendritic length in the Cornu Ammonis 1 (CA1) and dentate gyrus (DG) areas of the hippocampus while AC-T/MnBuOE maintained dendritic length. Comparative proteomic analysis revealed affected protein networks associated with cell morphology and behavior functions in both the AC-T and AC-T/MnBuOE treatment groups.


Subject(s)
Cognition/drug effects , Cyclophosphamide/pharmacology , Doxorubicin/pharmacology , Hippocampus/drug effects , Metalloporphyrins/pharmacology , Oxidation-Reduction/drug effects , Paclitaxel/pharmacology , Animals , Antioxidants/pharmacology , CA1 Region, Hippocampal/drug effects , Dentate Gyrus/drug effects , Female , Mice , Mice, Inbred C57BL , Neurogenesis/drug effects , Oxidative Stress/drug effects , Proteomics/methods , Radiation-Protective Agents/pharmacology , Spatial Memory/drug effects
19.
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
20.
Mol Cell Proteomics ; 15(3): 765-75, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26621846

ABSTRACT

Normal cell growth is characterized by a regulated epigenetic program that drives cellular activities such as gene transcription, DNA replication, and DNA damage repair. Perturbation of this epigenetic program can lead to events such as mis-regulation of gene transcription and diseases such as cancer. To begin to understand the epigenetic program correlated to the development of melanoma, we performed a novel quantitative mass spectrometric analysis of histone post-translational modifications mis-regulated in melanoma cell culture as well as patient tumors. Aggressive melanoma cell lines as well as metastatic melanoma were found to have elevated histone H3 Lys(27) trimethylation (H3K27me3) accompanied by overexpressed methyltransferase EZH2 that adds the specific modification. The altered epigenetic program that led to elevated H3K27me3 in melanoma cell culture was found to directly silence transcription of the tumor suppressor genes RUNX3 and E-cadherin. The EZH2-mediated silencing of RUNX3 and E-cadherin transcription was also validated in advanced stage human melanoma tissues. This is the first study focusing on the detailed epigenetic mechanisms leading to EZH2-mediated silencing of RUNX3 and E-cadherin tumor suppressors in melanoma. This study underscores the utility of using high resolution mass spectrometry to identify mis-regulated epigenetic programs in diseases such as cancer, which could ultimately lead to the identification of biological markers for diagnostic and prognostic applications.


Subject(s)
Histones/metabolism , Lysine/metabolism , Mass Spectrometry/methods , Multiple Myeloma/metabolism , Up-Regulation , Cadherins/genetics , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Methylation , Multiple Myeloma/genetics , Neoplasm Metastasis , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL