Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Mol Cell ; 63(4): 674-685, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27499292

ABSTRACT

CENP-A is a histone variant, which replaces histone H3 at centromeres and confers unique properties to centromeric chromatin. The crystal structure of CENP-A nucleosome suggests flexible nucleosomal DNA ends, but their dynamics in solution remains elusive and their implication in centromere function is unknown. Using electron cryo-microscopy, we determined the dynamic solution properties of the CENP-A nucleosome. Our biochemical, proteomic, and genetic data reveal that higher flexibility of DNA ends impairs histone H1 binding to the CENP-A nucleosome. Substituting the 2-turn αN-helix of CENP-A with the 3-turn αN-helix of H3 results in compact particles with rigidified DNA ends, able to bind histone H1. In vivo replacement of CENP-A with H3-CENP-A hybrid nucleosomes leads to H1 recruitment, delocalization of kinetochore proteins, and significant mitotic and cytokinesis defects. Our data reveal that the evolutionarily conserved flexible ends of the CENP-A nucleosomes are essential to ensure the fidelity of the mitotic pathway.


Subject(s)
Autoantigens/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA/metabolism , Histones/metabolism , Kinetochores/metabolism , Mitosis/physiology , Nucleosomes/metabolism , Animals , Autoantigens/genetics , Autoantigens/ultrastructure , Binding Sites , Centromere Protein A , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/ultrastructure , Cryoelectron Microscopy , Cytokinesis , DNA/chemistry , Genotype , HeLa Cells , Humans , Kinetochores/ultrastructure , Mice , Mice, Knockout , Models, Molecular , Mutation , Nucleic Acid Conformation , Nucleosomes/ultrastructure , Phenotype , Protein Binding , Protein Conformation, alpha-Helical , Structure-Activity Relationship , Transfection
2.
Nucleic Acids Res ; 50(13): 7350-7366, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35766398

ABSTRACT

The histone variant H3.3 is encoded by two distinct genes, H3f3a and H3f3b, exhibiting identical amino-acid sequence. H3.3 is required for spermatogenesis, but the molecular mechanism of its spermatogenic function remains obscure. Here, we have studied the role of each one of H3.3A and H3.3B proteins in spermatogenesis. We have generated transgenic conditional knock-out/knock-in (cKO/KI) epitope-tagged FLAG-FLAG-HA-H3.3B (H3.3BHA) and FLAG-FLAG-HA-H3.3A (H3.3AHA) mouse lines. We show that H3.3B, but not H3.3A, is required for spermatogenesis and male fertility. Analysis of the molecular mechanism unveils that the absence of H3.3B led to alterations in the meiotic/post-meiotic transition. Genome-wide RNA-seq reveals that the depletion of H3.3B in meiotic cells is associated with increased expression of the whole sex X and Y chromosomes as well as of both RLTR10B and RLTR10B2 retrotransposons. In contrast, the absence of H3.3B resulted in down-regulation of the expression of piRNA clusters. ChIP-seq experiments uncover that RLTR10B and RLTR10B2 retrotransposons, the whole sex chromosomes and the piRNA clusters are markedly enriched of H3.3. Taken together, our data dissect the molecular mechanism of H3.3B functions during spermatogenesis and demonstrate that H3.3B, depending on its chromatin localization, is involved in either up-regulation or down-regulation of expression of defined large chromatin regions.


Subject(s)
Histones , RNA, Small Interfering/metabolism , Retroelements , Spermatogenesis , Animals , Chromatin/genetics , Histones/genetics , Histones/metabolism , Male , Mice , Sex Chromosomes/metabolism
3.
Ecotoxicol Environ Saf ; 272: 116070, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38340603

ABSTRACT

Perfluorooctanesulfonic acid (PFOS) is a neurotoxic widespread organic contaminant which affects several brain functions including memory, motor coordination and social activity. PFOS has the ability to traverse the placenta and the blood brain barrier (BBB) and cause weight gain in female mice. It's also known that obesity and consumption of a high fat diet have negative effects on the brain, impairs cognition and increases the risk for the development of dementia. The combination effect of developmental exposure to PFOS and the intake of a high-fat diet (HFD) has not been explored. This study investigates the effect of PFOS and /or HFD on weight gain, behavior and transcriptomic and proteomic analysis of adult brain mice. We found that female mice exposed to PFOS alone showed an increase in weight, while HFD expectedly increased body weight. The combination of HFD and PFOS exacerbated generalized behavior such as time spent in the center and rearing, while PFOS alone impacted the distance travelled. These results suggest that PFOS exposure may promote hyperactivity. The combination of PFOS and HFD alter social behavior such as rearing and withdrawal. Although HFD interfered with memory retrieval, biomarkers of dementia did not change except for total Tau and phosphorylated Tau. Tau was impacted by either or both PFOS exposure and HFD. Consistent with behavioral observations, global cerebral transcriptomic analysis showed that PFOS exposure affects calcium signaling, MAPK pathways, ion transmembrane transport, and developmental processes. The combination of HFD with PFOS enhances the effect of PFOS in the brain and affects pathways related to ER stress, axon guidance and extension, and neural migration. Proteomic analysis showed that HFD enhances the impact of PFOS on inflammatory pathways, regulation of cell migration and proliferation, and MAPK signaling pathways. Overall, these data show that PFOS combined with HFD may reprogram the genome and modulate neuromotor development and may promote symptoms linked to attention deficit-hyperactivity disorders (ADHD) and autism spectrum disorders (ASD). Future work will be needed to confirm these connections.


Subject(s)
Alkanesulfonic Acids , Dementia , Fluorocarbons , Neurodevelopmental Disorders , Pregnancy , Mice , Animals , Female , Diet, High-Fat/adverse effects , Proteomics , Weight Gain , Mice, Inbred C57BL
4.
Neurobiol Dis ; 182: 106147, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37178811

ABSTRACT

Coronavirus disease of 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has sparked a global pandemic with severe complications and high morbidity rate. Neurological symptoms in COVID-19 patients, and neurological sequelae post COVID-19 recovery have been extensively reported. Yet, neurological molecular signature and signaling pathways that are affected in the central nervous system (CNS) of COVID-19 severe patients remain still unknown and need to be identified. Plasma samples from 49 severe COVID-19 patients, 50 mild COVID-19 patients, and 40 healthy controls were subjected to Olink proteomics analysis of 184 CNS-enriched proteins. By using a multi-approach bioinformatics analysis, we identified a 34-neurological protein signature for COVID-19 severity and unveiled dysregulated neurological pathways in severe cases. Here, we identified a new neurological protein signature for severe COVID-19 that was validated in different independent cohorts using blood and postmortem brain samples and shown to correlate with neurological diseases and pharmacological drugs. This protein signature could potentially aid the development of prognostic and diagnostic tools for neurological complications in post-COVID-19 convalescent patients with long term neurological sequelae.


Subject(s)
COVID-19 , Nervous System Diseases , Humans , COVID-19/complications , SARS-CoV-2 , Nervous System Diseases/etiology , Central Nervous System , Brain
5.
Int J Mol Sci ; 24(14)2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37511368

ABSTRACT

Excess hepatic lipid accumulation is the hallmark of non-alcoholic fatty liver disease (NAFLD), for which no medication is currently approved. However, glucagon-like peptide-1 receptor agonists (GLP-1RAs), already approved for treating type 2 diabetes, have lately emerged as possible treatments. Herein we aim to investigate how the GLP-1RA exendin-4 (Ex-4) affects the microRNA (miRNAs) expression profile using an in vitro model of steatosis. Total RNA, including miRNAs, was isolated from control, steatotic, and Ex-4-treated steatotic cells and used for probing a panel of 799 highly curated miRNAs using NanoString technology. Enrichment pathway analysis was used to find the signaling pathways and cellular functions associated with the differentially expressed miRNAs. Our data shows that Ex-4 reversed the expression of a set of miRNAs. Functional enrichment analysis highlighted many relevant signaling pathways and cellular functions enriched in the differentially expressed miRNAs, including hepatic fibrosis, insulin receptor, PPAR, Wnt/ß-Catenin, VEGF, and mTOR receptor signaling pathways, fibrosis of the liver, cirrhosis of the liver, proliferation of hepatic stellate cells, diabetes mellitus, glucose metabolism disorder and proliferation of liver cells. Our findings suggest that miRNAs may play essential roles in the processes driving steatosis reduction in response to GLP-1R agonists, which warrants further functional investigation.


Subject(s)
Diabetes Mellitus, Type 2 , MicroRNAs , Non-alcoholic Fatty Liver Disease , Humans , Exenatide/pharmacology , MicroRNAs/genetics , MicroRNAs/therapeutic use , Hep G2 Cells , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Glucagon-Like Peptide 1/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Liver Cirrhosis , Glucagon-Like Peptide-1 Receptor/genetics
6.
Int J Mol Sci ; 24(20)2023 Oct 16.
Article in English | MEDLINE | ID: mdl-37894896

ABSTRACT

The field of Alzheimer's disease (AD) has witnessed recent breakthroughs in the development of disease-modifying biologics and diagnostic markers. While immunotherapeutic interventions have provided much-awaited solutions, nucleic acid-based tools represent other avenues of intervention; however, these approaches are costly and invasive, and they have serious side effects. Previously, we have shown in AD animal models that tolfenamic acid (TA) can lower the expression of AD-related genes and their products and subsequently reduce pathological burden and improve cognition. Using TA as a scaffold and the zinc finger domain of SP1 as a pharmacophore, we developed safer and more potent brain-penetrating analogs that interfere with sequence-specific DNA binding at transcription start sites and predominantly modulate the expression of SP1 target genes. More importantly, the proteome of treated cells displayed ~75% of the downregulated products as SP1 targets. Specific levels of SP1-driven genes and AD biomarkers such as amyloid precursor protein (APP) and Tau proteins were also decreased as part of this targeted systemic response. These small molecules, therefore, offer a viable alternative to achieving desired therapeutic outcomes by interfering with both amyloid and Tau pathways with limited off-target systemic changes.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Mice, Transgenic , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , ortho-Aminobenzoates/pharmacology , ortho-Aminobenzoates/therapeutic use , tau Proteins/genetics , tau Proteins/metabolism , Amyloid beta-Peptides/metabolism
7.
J Immunol ; 204(1): 199-211, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31801817

ABSTRACT

Programmed cell death 1 (PD-1) is critical for T regulatory cells (Tregs) to maintain peripheral tolerance to self-antigens. In the tumor microenvironment, interaction between PD-1 and its ligands supports tumor immune evasion. Pembrolizumab blocks interactions of PD-1 with its ligands, enhancing antitumor and clinical responses. We and others have reported that pembrolizumab does not affect function or phenotype of thymic-derived Tregs; however, little is known about its effect on extrathymic differentiation of peripheral Tregs. In this study, we investigated the effect of pembrolizumab on in vitro-induced Tregs (iTregs). Our work showed that PD-1 blockade interferes with iTreg differentiation and has no potential effect on the stability of FOXP3 after differentiation. Additionally, we found that both nontreated and pembrolizumab-treated iTregs were suppressive. However, pembrolizumab-treated iTregs were relatively less suppressive in higher Treg ratios and failed to produce IL-10 compared with their nontreated counterparts. Different methods including transcriptomic analyses confirmed that the downregulation of FOXP3 was mediated by activating mTOR and STAT1 and inhibiting MAPK pathways, shifting the iTreg polarization in favor of Th1 and Th17 subsets. To confirm the role of mTOR activation, we found that rapamycin diminished the effect of pembrolizumab-mediated downregulation of FOXP3. Ingenuity pathway analysis revealed that pembrolizumab-treated iTregs showed upregulation of genes promoting DNA repair and immune cell trafficking, in addition to downregulation of genes supporting cellular assembly and organization. To our knowledge, this is the first study to show that pembrolizumab interferes with differentiation of human FOXP3+ iTregs and to disclose some of the molecular pathways involved.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Forkhead Transcription Factors/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , TOR Serine-Threonine Kinases/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Demethylation/drug effects , Forkhead Transcription Factors/immunology , Healthy Volunteers , Humans , Protein Stability/drug effects , T-Lymphocytes, Regulatory/immunology , Up-Regulation/drug effects
8.
Nature ; 505(7485): 648-53, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24463511

ABSTRACT

H2A.Z is an essential histone variant implicated in the regulation of key nuclear events. However, the metazoan chaperones responsible for H2A.Z deposition and its removal from chromatin remain unknown. Here we report the identification and characterization of the human protein ANP32E as a specific H2A.Z chaperone. We show that ANP32E is a member of the presumed H2A.Z histone-exchange complex p400/TIP60. ANP32E interacts with a short region of the docking domain of H2A.Z through a new motif termed H2A.Z interacting domain (ZID). The 1.48 Å resolution crystal structure of the complex formed between the ANP32E-ZID and the H2A.Z/H2B dimer and biochemical data support an underlying molecular mechanism for H2A.Z/H2B eviction from the nucleosome and its stabilization by ANP32E through a specific extension of the H2A.Z carboxy-terminal α-helix. Finally, analysis of H2A.Z localization in ANP32E(-/-) cells by chromatin immunoprecipitation followed by sequencing shows genome-wide enrichment, redistribution and accumulation of H2A.Z at specific chromatin control regions, in particular at enhancers and insulators.


Subject(s)
Chromatin/chemistry , Chromatin/metabolism , Histones/metabolism , Molecular Chaperones/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Amino Acid Sequence , Cell Line , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin Immunoprecipitation , Crystallography, X-Ray , DNA/genetics , DNA/metabolism , Genome, Human/genetics , Histones/chemistry , Histones/isolation & purification , Humans , Models, Molecular , Molecular Chaperones/chemistry , Molecular Sequence Data , Nuclear Proteins/chemistry , Nucleosomes/chemistry , Nucleosomes/metabolism , Phosphoproteins/chemistry , Protein Binding , Protein Conformation , Substrate Specificity
9.
Bioinformatics ; 34(17): 3004-3012, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29635310

ABSTRACT

Motivation: Single-molecule localization microscopy (SMLM) can play an important role in integrated structural biology approaches to identify, localize and determine the 3D structure of cellular structures. While many tools exist for the 3D analysis and visualization of crystal or cryo-EM structures little exists for 3D SMLM data, which can provide unique insights but are particularly challenging to analyze in three dimensions especially in a dense cellular context. Results: We developed 3DClusterViSu, a method based on 3D Voronoi tessellations that allows local density estimation, segmentation and quantification of 3D SMLM data and visualization of protein clusters within a 3D tool. We show its robust performance on microtubules and histone proteins H2B and CENP-A with distinct spatial distributions. 3DClusterViSu will favor multi-scale and multi-resolution synergies to allow integrating molecular and cellular levels in the analysis of macromolecular complexes. Availability and impementation: 3DClusterViSu is available under http://cbi-dev.igbmc.fr/cbi/voronoi3D. Supplementary information: Supplementary figures are available at Bioinformatics online.


Subject(s)
Cluster Analysis , Single Molecule Imaging , Centromere Protein A/analysis , Histones/analysis , Humans , Imaging, Three-Dimensional , Software
10.
PLoS Genet ; 12(7): e1006221, 2016 07.
Article in English | MEDLINE | ID: mdl-27467129

ABSTRACT

FACT, in addition to its role in transcription, is likely implicated in both transcription-coupled nucleotide excision repair and DNA double strand break repair. Here, we present evidence that FACT could be directly involved in Base Excision Repair and elucidate the chromatin remodeling mechanisms of FACT during BER. We found that, upon oxidative stress, FACT is released from transcription related protein complexes to get associated with repair proteins and chromatin remodelers from the SWI/SNF family. We also showed the rapid recruitment of FACT to the site of damage, coincident with the glycosylase OGG1, upon the local generation of oxidized DNA. Interestingly, FACT facilitates uracil-DNA glycosylase in the removal of uracil from nucleosomal DNA thanks to an enhancement in the remodeling activity of RSC. This discloses a novel property of FACT wherein it has a co-remodeling activity and strongly enhances the remodeling capacity of the chromatin remodelers. Altogether, our data suggest that FACT may acts in concert with RSC to facilitate excision of DNA lesions during the initial step of BER.


Subject(s)
DNA Repair/genetics , DNA-Binding Proteins/genetics , High Mobility Group Proteins/genetics , Histones/genetics , Transcription, Genetic , Transcriptional Elongation Factors/genetics , Animals , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA Damage/genetics , DNA-Binding Proteins/biosynthesis , HeLa Cells , High Mobility Group Proteins/biosynthesis , Histones/metabolism , Humans , Nucleosomes/genetics , Oxidative Stress/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Transcriptional Elongation Factors/biosynthesis , Uracil/metabolism , Xenopus laevis
11.
Genes Dev ; 24(12): 1253-65, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20504901

ABSTRACT

The histone variant H3.3 marks active chromatin by replacing the conventional histone H3.1. In this study, we investigate the detailed mechanism of H3.3 replication-independent deposition. We found that the death domain-associated protein DAXX and the chromatin remodeling factor ATRX (alpha-thalassemia/mental retardation syndrome protein) are specifically associated with the H3.3 deposition machinery. Bacterially expressed DAXX has a marked binding preference for H3.3 and assists the deposition of (H3.3-H4)(2) tetramers on naked DNA, thus showing that DAXX is a H3.3 histone chaperone. In DAXX-depleted cells, a fraction of H3.3 was found associated with the replication-dependent machinery of deposition, suggesting that cells adapt to the depletion. The reintroduced DAXX in these cells colocalizes with H3.3 into the promyelocytic leukemia protein (PML) bodies. Moreover, DAXX associates with pericentric DNA repeats, and modulates the transcription from these repeats through assembly of H3.3 nucleosomes. These findings establish a new link between the PML bodies and the regulation of pericentric DNA repeat chromatin structure. Taken together, our data demonstrate that DAXX functions as a bona fide histone chaperone involved in the replication-independent deposition of H3.3.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins/metabolism , Cell Line , Co-Repressor Proteins , DNA Helicases/metabolism , HeLa Cells , Heterochromatin/metabolism , Humans , Mice , Molecular Chaperones , Multiprotein Complexes , Nuclear Proteins/genetics , Nucleosomes/metabolism , Protein Stability , Transcription Factors/metabolism , X-linked Nuclear Protein
12.
Proc Natl Acad Sci U S A ; 110(21): 8579-84, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23657009

ABSTRACT

The role of the mitotic phosphorylation of the amino (NH2) terminus of Centromere Protein A (CENP-A), the histone variant epigenetic centromeric marker, remains elusive. Here, we show that the NH2 terminus of human CENP-A is essential for mitotic progression and that localization of CENP-C, another key centromeric protein, requires only phosphorylation of the CENP-A NH2 terminus, and is independent of the CENP-A NH2 terminus length and amino acid sequence. Mitotic CENP-A nucleosomal complexes contain CENP-C and phosphobinding 14-3-3 proteins. In contrast, mitotic nucleosomal complexes carrying nonphosphorylatable CENP-A-S7A contained only low levels of CENP-C and no detectable 14-3-3 proteins. Direct interactions between the phosphorylated form of CENP-A and 14-3-3 proteins as well as between 14-3-3 proteins and CENP-C were demonstrated. Taken together, our results reveal that 14-3-3 proteins could act as specific mitotic "bridges," linking phosphorylated CENP-A and CENP-C, which are necessary for the platform function of CENP-A centromeric chromatin in the assembly and maintenance of active kinetochores.


Subject(s)
Autoantigens/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Kinetochores/metabolism , Mitosis/physiology , Nucleosomes/metabolism , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Amino Acid Substitution , Autoantigens/genetics , Centromere Protein A , Chromosomal Proteins, Non-Histone/genetics , HeLa Cells , Humans , Mutation, Missense , Nucleosomes/genetics , Phosphorylation , Protein Structure, Tertiary
13.
Proc Natl Acad Sci U S A ; 108(20): 8212-7, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21531907

ABSTRACT

TRIM24 (TIF1α), TRIM28 (TIF1ß), and TRIM33 (TIF1γ) are three related cofactors belonging to the tripartite motif superfamily that interact with distinct transcription factors. TRIM24 interacts with the liganded retinoic acid (RA) receptor to repress its transcriptional activity. Germ line inactivation of TRIM24 in mice deregulates RA-signaling in hepatocytes leading to the development of hepatocellular carcinoma (HCC). Here we show that TRIM24 can be purified as at least two macromolecular complexes comprising either TRIM33 or TRIM33 and TRIM28. Somatic hepatocyte-specific inactivation of TRIM24, TRIM28, or TRIM33 all promote HCC in a cell-autonomous manner in mice. Moreover, HCC formation upon TRIM24 inactivation is strongly potentiated by further loss of TRIM33. These results demonstrate that the TIF1-related subfamily of TRIM proteins interact both physically and functionally to modulate HCC formation in mice.


Subject(s)
Carcinoma, Hepatocellular/etiology , Liver Neoplasms/etiology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/pathology , Hepatocytes/pathology , Liver Neoplasms/pathology , Mice , Multiprotein Complexes/isolation & purification , Multiprotein Complexes/physiology , Protein Binding , Receptors, Retinoic Acid , Tripartite Motif-Containing Protein 28
14.
Heliyon ; 10(9): e30267, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38711666

ABSTRACT

Wastewater-based epidemiology (WBE) has been proven effective for the monitoring of infectious disease outbreaks during mass gathering events and for timely public health interventions. As part of Qatar's efforts to monitor and combat the spread of infectious diseases during the FIFA World Cup Qatar 2022™ (FWC'22), wastewater surveillance was used to monitor the spread of SARS-CoV-2, human enterovirus, and poliovirus. The screening covered five major wastewater treatment plants servicing the event locations between October 2022 and January 2023. Viruses were concentrated from the wastewater samples by PEG precipitation, followed by qRT-PCR to measure the viral load in the wastewater. As expected, SARS-CoV-2 and enterovirus RNA were detected in all samples, while poliovirus was not detected. The concentration of SARS-CoV-2 was correlated with population density, such as areas surrounding the World Cup venues, and with the number of reported clinical cases. Additionally, we observed temporal fluctuations in viral RNA concentrations, with peak levels coinciding with the group stage matches of the FWC'22. This study has been useful in providing public health authorities with an efficient and cost-effective surveillance system for potential infectious disease outbreaks during mega-events.

15.
Proc Natl Acad Sci U S A ; 107(4): 1349-54, 2010 Jan 26.
Article in English | MEDLINE | ID: mdl-20080577

ABSTRACT

The human histone H3 variant, CENP-A, replaces the conventional histone H3 in centromeric chromatin and, together with centromere-specific DNA-binding factors, directs the assembly of the kinetochore. We purified the prenucelosomal e-CENP-A complex. We found that HJURP, a member of the complex, was required for cell cycle specific targeting of CENP-A to centromeres. HJURP facilitated efficient deposition of CENP-A/H4 tetramers to naked DNA in vitro. Bacterially expressed HJURP binds at a stoichiometric ratio to the CENP-A/H4 tetramer but not to the H3/H4 tetramer. The binding occurred through a conserved HJURP short N-terminal domain, termed CBD. The novel characteristic identified in vertebrates that we named TLTY box of CBD, was essential for formation of the HJURP-CENP-A/H4 complex. Our data identified HJURP as a vertebrate CENP-A chaperone and dissected its mode of interactions with CENP-A.


Subject(s)
Autoantigens/chemistry , Autoantigens/metabolism , Centromere/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Protein Interaction Domains and Motifs , Amino Acid Sequence , Animals , Autoantigens/genetics , Cell Cycle , Centromere Protein A , Chromosomal Proteins, Non-Histone/genetics , Conserved Sequence , DNA-Binding Proteins/genetics , HeLa Cells , Histones/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Nucleosomes/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , RNA, Small Interfering/genetics , Sequence Alignment
16.
Cell Death Dis ; 14(7): 415, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37438342

ABSTRACT

Previous studies have suggested that breast cancer (BC) from the Middle East and North Africa (MENA) is presented at younger age with advanced tumor stage, indicating underlying biological differences. Given the scant transcriptomic data on BC from the MENA region and to better understand the biology of this disease, we performed mRNA and microRNA (miRNA) transcriptomic profiling on a local cohort of BC (n = 96) from Qatar. Our data revealed the differentially expressed genes and miRNAs as function of BC molecular subtypes (HR+, HER2+, HER2+HR+, and TNBC), tumor grade (GIII vs GI-II), patients' age (young (≤40) vs old (>40)), and ethnicity (MENA vs non-MENA). Our profiling data revealed close similarity between TNBC and HER2+, while the transcriptome of HER2+HR+ tumor was resemblant of that from HR+ tumors. Network analysis identified complex miRNA-mRNA regulatory networks in each BC molecular subtype, in high vs low grade tumors, in tumors from young vs old patients, and in tumors from MENA vs non-MENA, thus implicating miRNA-mediated gene regulation as an essential mechanism in shaping the transcriptome of BC. Integration of our transcriptomic data with CRISPR-Cas9 functional screen data and the OncoKB database identified numerous dependencies and therapeutic vulnerabilities in each BC molecular subtype, while CDC123 was functionally validated as potential therapeutic target for TNBC. Cox regression survival analyses identified mRNA and miRNA-based signatures predicative of worse and better relapse free survival (RFS), which were validated in larger BC cohorts. Our data provides comprehensive transcriptomic profiling and unraveled the miRNA-mRNA regulatory networks in BC patients from the region and identified novel actionable gene targets, employing integrated approach. Findings from the current study have potential implications to improve the current standard-of-care for BC from the MENA as well as patients from other ethnicities.


Subject(s)
MicroRNAs , Triple Negative Breast Neoplasms , Humans , MicroRNAs/genetics , Gene Expression Profiling , Transcriptome/genetics , RNA, Messenger/genetics
17.
Nat Commun ; 13(1): 946, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35177642

ABSTRACT

COVID-19 complications still present a huge burden on healthcare systems and warrant predictive risk models to triage patients and inform early intervention. Here, we profile 893 plasma proteins from 50 severe and 50 mild-moderate COVID-19 patients, and 50 healthy controls, and show that 375 proteins are differentially expressed in the plasma of severe COVID-19 patients. These differentially expressed plasma proteins are implicated in the pathogenesis of COVID-19 and present targets for candidate drugs to prevent or treat severe complications. Based on the plasma proteomics and clinical lab tests, we also report a 12-plasma protein signature and a model of seven routine clinical tests that validate in an independent cohort as early risk predictors of COVID-19 severity and patient survival. The risk predictors and candidate drugs described in our study can be used and developed for personalized management of SARS-CoV-2 infected patients.


Subject(s)
Blood Proteins/analysis , COVID-19/mortality , COVID-19/pathology , Severity of Illness Index , Adult , Cytokines/blood , Female , Humans , Male , Middle Aged , Prognosis , Proteomics/methods , SARS-CoV-2/drug effects , Young Adult , COVID-19 Drug Treatment
18.
Viruses ; 13(5)2021 04 28.
Article in English | MEDLINE | ID: mdl-33925055

ABSTRACT

The COVID-19 pandemic has affected all individuals across the globe in some way. Despite large numbers of reported seroprevalence studies, there remains a limited understanding of how the magnitude and epitope utilization of the humoral immune response to SARS-CoV-2 viral anti-gens varies within populations following natural infection. Here, we designed a quantitative, multi-epitope protein microarray comprising various nucleocapsid protein structural motifs, including two structural domains and three intrinsically disordered regions. Quantitative data from the microarray provided complete differentiation between cases and pre-pandemic controls (100% sensitivity and specificity) in a case-control cohort (n = 100). We then assessed the influence of disease severity, age, and ethnicity on the strength and breadth of the humoral response in a multi-ethnic cohort (n = 138). As expected, patients with severe disease showed significantly higher antibody titers and interestingly also had significantly broader epitope coverage. A significant increase in antibody titer and epitope coverage was observed with increasing age, in both mild and severe disease, which is promising for vaccine efficacy in older individuals. Additionally, we observed significant differences in the breadth and strength of the humoral immune response in relation to ethnicity, which may reflect differences in genetic and lifestyle factors. Furthermore, our data enabled localization of the immuno-dominant epitope to the C-terminal structural domain of the viral nucleocapsid protein in two independent cohorts. Overall, we have designed, validated, and tested an advanced serological assay that enables accurate quantitation of the humoral response post natural infection and that has revealed unexpected differences in the magnitude and epitope utilization within a population.


Subject(s)
Antibodies, Viral/immunology , COVID-19/immunology , SARS-CoV-2/immunology , Adolescent , Adult , Antigens, Viral/immunology , COVID-19/epidemiology , COVID-19/virology , COVID-19 Serological Testing , Case-Control Studies , Cohort Studies , Epitopes , Ethnicity , Female , Humans , Immunity, Humoral , Immunoglobulin G/immunology , Male , Middle Aged , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/immunology , Pandemics , SARS-CoV-2/genetics , Sensitivity and Specificity , Seroepidemiologic Studies , Severity of Illness Index , Young Adult
19.
Vaccines (Basel) ; 8(1)2020 02 06.
Article in English | MEDLINE | ID: mdl-32041340

ABSTRACT

T cell immunoglobulin mucin-3 (TIM-3) is an immune checkpoint identified as one of the key players in regulating T-cell responses. Studies have shown that TIM-3 is upregulated in the tumor microenvironment (TME). However, the precise role of TIM-3 in colorectal cancer (CRC) TME is yet to be elucidated. We performed phenotypic and molecular characterization of TIM-3+ T cells in the TME and circulation of CRC patients by analyzing tumor tissues (TT, TILs), normal tissues (NT, NILs), and peripheral blood mononuclear cells (PBMC). TIM-3 was upregulated on both CD4+ and CD3+CD4- (CD8+) TILs. CD4+TIM-3+ TILs expressed higher levels of T regulatory cell (Tregs)-signature genes, including FoxP3 and Helios, compared with their TIM-3- counterparts. Transcriptomic and ingenuity pathway analyses showed that TIM-3 potentially activates inflammatory and tumor metastatic pathways. Moreover, NF-κB-mediated transcription factors were upregulated in CD4+TIM-3+ TILs, which could favor proliferation/invasion and induce inflammatory and T-cell exhaustion pathways. In addition, we found that CD4+TIM-3+ TILs potentially support tumor invasion and metastasis, compared with conventional CD4+CD25+ Tregs in the CRC TME. However, functional studies are warranted to support these findings. In conclusion, this study discloses some of the functional pathways of TIM-3+ TILs, which could improve their targeting in more specific therapeutic approaches in CRC patients.

20.
Front Oncol ; 9: 910, 2019.
Article in English | MEDLINE | ID: mdl-31620367

ABSTRACT

Breast cancer (BC) is the leading cause of cancer-related death in women. Therefore, a better understanding of BC biology and signaling pathways might lead to the development of novel biomarkers and targeted therapies. Although a number of transcriptomic studies have been performed on breast cancer patients from various geographic regions, there are almost no such comprehensive studies performed on breast cancer from patients in the gulf region. This study aimed to provide a better understanding of the altered molecular networks in BC from the gulf region. Herein, we compared the transcriptome of BC to adjacent normal tissue from six BC patients and identified 1,108 upregulated and 518 downregulated transcripts. A selected number of genes from the RNA-Seq analysis were subsequently validated using qRT-PCR. Differentially expressed (2.0-fold change, adj. p < 0.05) transcripts were subjected to ingenuity pathway analysis, which revealed a myriad of affected signaling pathways and functional categories. Activation of ERBB2, FOXM1, ESR1, and IGFBP2 mechanistic networks was most prominent in BC tissue. Additionally, BC tissue exhibited marked enrichment in genes promoting cellular proliferation, migration, survival, and DNA replication and repair. The presence of genes indicative of immune cell infiltration and activation was also observed in BC tissue. We observed high concordance [43.5% (upregulated) and 62.1% (downregulated)] between differentially expressed genes in our study group and those reported for the TCGA BC cohort. Our data provide novel insight on BC biology and suggest common altered molecular networks in BC in this geographic region. Our data suggest future development of therapeutic interventions targeting those common signaling pathways.

SELECTION OF CITATIONS
SEARCH DETAIL