Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cell ; 168(3): 527-541.e29, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28111073

ABSTRACT

Advances in the synthesis and screening of small-molecule libraries have accelerated the discovery of chemical probes for studying biological processes. Still, only a small fraction of the human proteome has chemical ligands. Here, we describe a platform that marries fragment-based ligand discovery with quantitative chemical proteomics to map thousands of reversible small molecule-protein interactions directly in human cells, many of which can be site-specifically determined. We show that fragment hits can be advanced to furnish selective ligands that affect the activity of proteins heretofore lacking chemical probes. We further combine fragment-based chemical proteomics with phenotypic screening to identify small molecules that promote adipocyte differentiation by engaging the poorly characterized membrane protein PGRMC2. Fragment-based screening in human cells thus provides an extensive proteome-wide map of protein ligandability and facilitates the coordinated discovery of bioactive small molecules and their molecular targets.


Subject(s)
Drug Discovery/methods , Proteomics/methods , Adipocytes/cytology , Cell Differentiation , Crystallography, X-Ray , High-Throughput Screening Assays , Humans , Hydrolases/chemistry , Ligands , Membrane Proteins/antagonists & inhibitors , Oxidoreductases/chemistry , Protein Binding , Receptors, Progesterone/antagonists & inhibitors , Small Molecule Libraries
2.
Cell ; 163(1): 68-83, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26365491

ABSTRACT

cis-regulatory changes play a central role in morphological divergence, yet the regulatory principles underlying emergence of human traits remain poorly understood. Here, we use epigenomic profiling from human and chimpanzee cranial neural crest cells to systematically and quantitatively annotate divergence of craniofacial cis-regulatory landscapes. Epigenomic divergence is often attributable to genetic variation within TF motifs at orthologous enhancers, with a novel motif being most predictive of activity biases. We explore properties of this cis-regulatory change, revealing the role of particular retroelements, uncovering broad clusters of species-biased enhancers near genes associated with human facial variation, and demonstrating that cis-regulatory divergence is linked to quantitative expression differences of crucial neural crest regulators. Our work provides a wealth of candidates for future evolutionary studies and demonstrates the value of "cellular anthropology," a strategy of using in-vitro-derived embryonic cell types to elucidate both fundamental and evolving mechanisms underlying morphological variation in higher primates.


Subject(s)
Epigenomics/methods , Evolution, Molecular , Genetic Enhancement , Neural Crest/cytology , Pan troglodytes/genetics , Animals , Embryo, Mammalian/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Transgenic , Neural Crest/metabolism , Species Specificity
3.
Cell ; 163(3): 583-93, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26496605

ABSTRACT

LINE-1 retrotransposons are fast-evolving mobile genetic entities that play roles in gene regulation, pathological conditions, and evolution. Here, we show that the primate LINE-1 5'UTR contains a primate-specific open reading frame (ORF) in the antisense orientation that we named ORF0. The gene product of this ORF localizes to promyelocytic leukemia-adjacent nuclear bodies. ORF0 is present in more than 3,000 loci across human and chimpanzee genomes and has a promoter and a conserved strong Kozak sequence that supports translation. By virtue of containing two splice donor sites, ORF0 can also form fusion proteins with proximal exons. ORF0 transcripts are readily detected in induced pluripotent stem (iPS) cells from both primate species. Capped and polyadenylated ORF0 mRNAs are present in the cytoplasm, and endogenous ORF0 peptides are identified upon proteomic analysis. Finally, ORF0 enhances LINE-1 mobility. Taken together, these results suggest a role for ORF0 in retrotransposon-mediated diversity.


Subject(s)
Pan troglodytes/genetics , Retroelements , 5' Untranslated Regions , Amino Acid Sequence , Animals , Base Sequence , Cytoplasm/genetics , Humans , Long Interspersed Nucleotide Elements , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Open Reading Frames , RNA Processing, Post-Transcriptional , RNA, Antisense/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Ribosomes/metabolism , Sequence Alignment
4.
Genes Dev ; 30(10): 1155-71, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27198230

ABSTRACT

Nuclear pore complexes (NPCs) emerged as nuclear transport channels in eukaryotic cells ∼1.5 billion years ago. While the primary role of NPCs is to regulate nucleo-cytoplasmic transport, recent research suggests that certain NPC proteins have additionally acquired the role of affecting gene expression at the nuclear periphery and in the nucleoplasm in metazoans. Here we identify a widely expressed variant of the transmembrane nucleoporin (Nup) Pom121 (named sPom121, for "soluble Pom121") that arose by genomic rearrangement before the divergence of hominoids. sPom121 lacks the nuclear membrane-anchoring domain and thus does not localize to the NPC. Instead, sPom121 colocalizes and interacts with nucleoplasmic Nup98, a previously identified transcriptional regulator, at gene promoters to control transcription of its target genes in human cells. Interestingly, sPom121 transcripts appear independently in several mammalian species, suggesting convergent innovation of Nup-mediated transcription regulation during mammalian evolution. Our findings implicate alternate transcription initiation as a mechanism to increase the functional diversity of NPC components.


Subject(s)
Evolution, Molecular , Gene Expression Regulation , Membrane Glycoproteins/metabolism , Mutant Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , 5' Untranslated Regions/genetics , Cell Membrane/metabolism , Cell Nucleus/metabolism , Exons/genetics , HeLa Cells , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mutant Proteins/chemistry , Mutant Proteins/genetics , Nuclear Localization Signals , Nuclear Pore Complex Proteins/chemistry , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Domains , Protein Isoforms/genetics , Protein Isoforms/metabolism , Solubility , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Initiation Site
5.
Nature ; 536(7615): 205-9, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27487209

ABSTRACT

Genetic differences that specify unique aspects of human evolution have typically been identified by comparative analyses between the genomes of humans and closely related primates, including more recently the genomes of archaic hominins. Not all regions of the genome, however, are equally amenable to such study. Recurrent copy number variation (CNV) at chromosome 16p11.2 accounts for approximately 1% of cases of autism and is mediated by a complex set of segmental duplications, many of which arose recently during human evolution. Here we reconstruct the evolutionary history of the locus and identify bolA family member 2 (BOLA2) as a gene duplicated exclusively in Homo sapiens. We estimate that a 95-kilobase-pair segment containing BOLA2 duplicated across the critical region approximately 282 thousand years ago (ka), one of the latest among a series of genomic changes that dramatically restructured the locus during hominid evolution. All humans examined carried one or more copies of the duplication, which nearly fixed early in the human lineage--a pattern unlikely to have arisen so rapidly in the absence of selection (P < 0.0097). We show that the duplication of BOLA2 led to a novel, human-specific in-frame fusion transcript and that BOLA2 copy number correlates with both RNA expression (r = 0.36) and protein level (r = 0.65), with the greatest expression difference between human and chimpanzee in experimentally derived stem cells. Analyses of 152 patients carrying a chromosome 16p11. rearrangement show that more than 96% of breakpoints occur within the H. sapiens-specific duplication. In summary, the duplicative transposition of BOLA2 at the root of the H. sapiens lineage about 282 ka simultaneously increased copy number of a gene associated with iron homeostasis and predisposed our species to recurrent rearrangements associated with disease.


Subject(s)
Chromosomes, Human, Pair 16/genetics , DNA Copy Number Variations/genetics , Evolution, Molecular , Genetic Predisposition to Disease , Proteins/genetics , Animals , Autistic Disorder/genetics , Chromosome Breakage , Gene Duplication , Homeostasis/genetics , Humans , Iron/metabolism , Pan troglodytes/genetics , Pongo/genetics , Proteins/analysis , Recombination, Genetic , Species Specificity , Time Factors
7.
Nature ; 503(7477): 525-529, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-24153179

ABSTRACT

Identifying cellular and molecular differences between human and non-human primates (NHPs) is essential to the basic understanding of the evolution and diversity of our own species. Until now, preserved tissues have been the main source for most comparative studies between humans, chimpanzees (Pan troglodytes) and bonobos (Pan paniscus). However, these tissue samples do not fairly represent the distinctive traits of live cell behaviour and are not amenable to genetic manipulation. We propose that induced pluripotent stem (iPS) cells could be a unique biological resource to determine relevant phenotypical differences between human and NHPs, and that those differences could have potential adaptation and speciation value. Here we describe the generation and initial characterization of iPS cells from chimpanzees and bonobos as new tools to explore factors that may have contributed to great ape evolution. Comparative gene expression analysis of human and NHP iPS cells revealed differences in the regulation of long interspersed element-1 (L1, also known as LINE-1) transposons. A force of change in mammalian evolution, L1 elements are retrotransposons that have remained active during primate evolution. Decreased levels of L1-restricting factors APOBEC3B (also known as A3B) and PIWIL2 (ref. 7) in NHP iPS cells correlated with increased L1 mobility and endogenous L1 messenger RNA levels. Moreover, results from the manipulation of A3B and PIWIL2 levels in iPS cells supported a causal inverse relationship between levels of these proteins and L1 retrotransposition. Finally, we found increased copy numbers of species-specific L1 elements in the genome of chimpanzees compared to humans, supporting the idea that increased L1 mobility in NHPs is not limited to iPS cells in culture and may have also occurred in the germ line or embryonic cells developmentally upstream to germline specification during primate evolution. We propose that differences in L1 mobility may have differentially shaped the genomes of humans and NHPs and could have continuing adaptive significance.


Subject(s)
Long Interspersed Nucleotide Elements/genetics , Pan paniscus/genetics , Pan troglodytes/genetics , Pluripotent Stem Cells/metabolism , Animals , Argonaute Proteins/metabolism , Cell Line , Cell Shape , Cytidine Deaminase/metabolism , Evolution, Molecular , Genome, Human/genetics , High-Throughput Nucleotide Sequencing , Humans , Karyotyping , Mice, Nude , Minor Histocompatibility Antigens , Pan paniscus/metabolism , Pan troglodytes/metabolism , Pluripotent Stem Cells/cytology , RNA, Messenger/analysis , RNA, Messenger/genetics , Sequence Analysis, RNA , Species Specificity
8.
BMC Biol ; 15(1): 68, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28800766

ABSTRACT

Researchers have long sought to understand the genetic basis of the cognitive differences between primates, with particular focus on the human brain. Although all mutational types have worked in concert with evolutionary forces to generate the current human brain, in this review we will explore the impact of mobile elements, specifically non-LTR retrotransposons. Non-LTR retrotransposons have contributed coding and regulatory sequences to the genome throughout evolution. During primate evolution there have been multiple waves of LINE retrotransposition as well as the birth of new mobile elements such as the SINEs Alu and SVA and we will explore what kinds of impacts these may have had on the evolving human brain.


Subject(s)
Biological Evolution , Brain/physiology , Genome , Primates/physiology , Retroelements/genetics , Animals , Evolution, Molecular , Primates/genetics
9.
EMBO Rep ; 12(5): 444-50, 2011 May.
Article in English | MEDLINE | ID: mdl-21460793

ABSTRACT

Human apolipoprotein-B mRNA-editing catalytic polypeptide-like 3 (APOBEC3) proteins constitute a family of cytidine deaminases that mediate restriction of retroviruses, endogenous retro-elements and DNA viruses. It is well established that these enzymes are potent mutators of viral DNA, but it is unclear whether their editing activity is a threat to the integrity of the cellular genome. We show that expression of APOBEC3A can lead to induction of DNA breaks and activation of damage responses in a deaminase-dependent manner. Consistent with these observations, APOBEC3A expression induces cell-cycle arrest. These results indicate that cellular DNA is vulnerable to APOBEC3 activity and deregulated expression of APOBEC3A could threaten genomic integrity.


Subject(s)
Cell Cycle/physiology , Cytidine Deaminase/metabolism , DNA Damage/genetics , Proteins/metabolism , Blotting, Western , Bromodeoxyuridine , Cell Line , Fluorescent Antibody Technique , Histones/metabolism , Humans , In Situ Nick-End Labeling , Nucleotide Deaminases/metabolism , Phosphorylation , Uracil-DNA Glycosidase/metabolism
10.
J Virol ; 85(4): 1765-76, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21123384

ABSTRACT

Members of the human APOBEC3 family of editing enzymes can inhibit various mobile genetic elements. APOBEC3A (A3A) can block the retrotransposon LINE-1 and the parvovirus adeno-associated virus type 2 (AAV-2) but does not inhibit retroviruses. In contrast, APOBEC3G (A3G) can block retroviruses but has only limited effects on AAV-2 or LINE-1. What dictates this differential target specificity remains largely undefined. Here, we modeled the structure of A3A based on its homology with the C-terminal domain of A3G and further compared the sequence of human A3A to those of 11 nonhuman primate orthologues. We then used these data to perform a mutational analysis of A3A, examining its ability to restrict LINE-1, AAV-2, and foreign plasmid DNA and to edit a single-stranded DNA substrate. The results revealed an essential functional role for the predicted single-stranded DNA-docking groove located around the A3A catalytic site. Within this region, amino acid differences between A3A and A3G are predicted to affect the shape of the polynucleotide-binding groove. Correspondingly, transferring some of these A3A residues to A3G endows the latter protein with the ability to block LINE-1 and AAV-2. These results suggest that the target specificity of APOBEC3 family members is partly defined by structural features influencing their interaction with polynucleotide substrates.


Subject(s)
Cytidine Deaminase/chemistry , Cytidine Deaminase/metabolism , DNA, Single-Stranded/physiology , Dependovirus/physiology , Long Interspersed Nucleotide Elements/physiology , Polynucleotides/metabolism , Proteins/chemistry , Proteins/metabolism , APOBEC-3G Deaminase , Animals , Base Sequence , Cytidine Deaminase/genetics , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Dependovirus/genetics , Dependovirus/metabolism , HEK293 Cells , Haplorhini , HeLa Cells , Humans , Long Interspersed Nucleotide Elements/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Plasmids , Polynucleotides/chemistry , Proteins/genetics , Retroelements/genetics , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Structure-Activity Relationship , Substrate Specificity
11.
Curr Biol ; 32(22): 4797-4807.e5, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36228612

ABSTRACT

Unique aspects of human behavior are often attributed to differences in the relative size and organization of the human brain: these structural aspects originate during early development. Recent studies indicate that human neurodevelopment is considerably slower than that in other nonhuman primates, a finding that is termed neoteny. One aspect of neoteny is the slow onset of action potentials. However, which molecular mechanisms play a role in this process remain unclear. To examine the evolutionary constraints on the rate of neuronal maturation, we have generated transcriptional data tracking five time points, from the neural progenitor state to 8-week-old neurons, in primates spanning the catarrhine lineage, including Macaca mulatta, Gorilla gorilla, Pan paniscus, Pan troglodytes, and Homo sapiens. Despite finding an overall similarity of many transcriptional signatures, species-specific and clade-specific distinctions were observed. Among the genes that exhibited human-specific regulation, we identified a key pioneer transcription factor, GATA3, that was uniquely upregulated in humans during the neuronal maturation process. We further examined the regulatory nature of GATA3 in human cells and observed that downregulation quickened the speed of developing spontaneous action potentials, thereby modulating the human neotenic phenotype. These results provide evidence for the divergence of gene regulation as a key molecular mechanism underlying human neoteny.


Subject(s)
Hominidae , Transcriptome , Animals , Humans , Primates/genetics , Hominidae/genetics , Gorilla gorilla/genetics , Pan troglodytes/genetics , Pan paniscus , Macaca mulatta
12.
PLoS Pathog ; 5(5): e1000439, 2009 May.
Article in English | MEDLINE | ID: mdl-19461882

ABSTRACT

The APOBEC3 proteins form a multigene family of cytidine deaminases with inhibitory activity against viruses and retrotransposons. In contrast to APOBEC3G (A3G), APOBEC3A (A3A) has no effect on lentiviruses but dramatically inhibits replication of the parvovirus adeno-associated virus (AAV). To study the contribution of deaminase activity to the antiviral activity of A3A, we performed a comprehensive mutational analysis of A3A. By mutation of non-conserved residues, we found that regions outside of the catalytic active site contribute to both deaminase and antiviral activities. Using A3A point mutants and A3A/A3G chimeras, we show that deaminase activity is not required for inhibition of recombinant AAV production. We also found that deaminase-deficient A3A mutants block replication of both wild-type AAV and the autonomous parvovirus minute virus of mice (MVM). In addition, we identify specific residues of A3A that confer activity against AAV when substituted into A3G. In summary, our results demonstrate that deaminase activity is not necessary for the antiviral activity of A3A against parvoviruses.


Subject(s)
Cytidine Deaminase/physiology , Dependovirus/physiology , Minute Virus of Mice/physiology , Proteins/physiology , Virus Replication , APOBEC-3G Deaminase , Amino Acid Sequence , Cell Line, Transformed , Cell Line, Tumor , Cytidine Deaminase/chemistry , Cytidine Deaminase/genetics , DNA, Recombinant/genetics , DNA, Recombinant/metabolism , Fluorescence Resonance Energy Transfer , Fluorescent Antibody Technique , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Proteins/chemistry , Proteins/genetics
13.
Curr Biol ; 16(5): 480-5, 2006 Mar 07.
Article in English | MEDLINE | ID: mdl-16527742

ABSTRACT

APOBEC3 proteins constitute a family of cytidine deaminases that provide intracellular resistance to retrovirus replication and transposition of endogenous retroelements. One family member, APOBEC3A (hA3A), is an orphan, without any known antiviral activity. We show that hA3A is catalytically active and that it, but none of the other family members, potently inhibits replication of the parvovirus adeno-associated virus (AAV). hA3A was also a potent inhibitor of the endogenous LTR retroelements, MusD, IAP, and the non-LTR retroelement, LINE-1. Its function was dependent on the conserved amino acids of the hA3A active site, consistent with a role for cytidine deamination, although mutations in retroelement sequences were not found. These findings demonstrate the potent activity of hA3A, an APOBEC3 family member with no previously identified function. They also highlight the functional differences between APOBEC3 proteins. The APOBEC3 family members have distinct functions and may have evolved to resist various classes of genetic elements.


Subject(s)
Cytidine Deaminase/physiology , Dependovirus/physiology , Nuclear Proteins/physiology , Proteins/physiology , Retroelements/physiology , Cell Line, Tumor , Cell Nucleus/enzymology , Dependovirus/pathogenicity , Humans , Macrophages/enzymology , Monocytes/enzymology , RNA, Messenger/metabolism , Virus Replication/physiology
14.
Elife ; 82019 02 07.
Article in English | MEDLINE | ID: mdl-30730291

ABSTRACT

Comparative analyses of neuronal phenotypes in closely related species can shed light on neuronal changes occurring during evolution. The study of post-mortem brains of nonhuman primates (NHPs) has been limited and often does not recapitulate important species-specific developmental hallmarks. We utilize induced pluripotent stem cell (iPSC) technology to investigate the development of cortical pyramidal neurons following migration and maturation of cells grafted in the developing mouse cortex. Our results show differential migration patterns in human neural progenitor cells compared to those of chimpanzees and bonobos both in vitro and in vivo, suggesting heterochronic changes in human neurons. The strategy proposed here lays the groundwork for further comparative analyses between humans and NHPs and opens new avenues for understanding the differences in the neural underpinnings of cognition and neurological disease susceptibility between species.


Subject(s)
Neurons/cytology , Pan paniscus/physiology , Pan troglodytes/physiology , Animals , Cell Differentiation , Cell Line , Cell Movement/genetics , Dendrites/metabolism , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Species Specificity
15.
Biochem Biophys Res Commun ; 364(1): 92-9, 2007 Dec 07.
Article in English | MEDLINE | ID: mdl-17931602

ABSTRACT

The use of stem cells as a vehicle of therapeutic genes is an attractive approach for the development of new antitumoral strategies based on gene therapy. The aim of our study was to assess the potential of bone marrow-derived Multipotent Adult Progenitor Cells (rMAPCs) to differentiate in vitro and in vivo into endothelial cells and to be recruited to areas of tumor vasculogenesis. In vitro, rMAPCs obtained from Buffalo rats differentiated into cells expressing endothelial markers and demonstrated functional endothelial capacity. Intravenous injection of undifferentiated rMAPC transduced with a lentivirus expressing GFP in an orthotopic rat model of hepatocellular carcinoma, resulted in tumor recruitment of the injected cells and in vivo differentiation into endothelial cells in the tumor area with contribution to vasculogenesis. In summary, our results suggest that rMAPCs can be efficiently recruited by vascularized tumors and differentiate to endothelium and thus may represent a useful vehicle for delivery of therapeutic genes to sites of active tumor neovascularization.


Subject(s)
Carcinoma, Hepatocellular/blood supply , Liver Neoplasms, Experimental/blood supply , Multipotent Stem Cells/physiology , Neovascularization, Pathologic/physiopathology , Animals , Bone Marrow Cells/physiology , Endothelium, Vascular/cytology , Genetic Therapy/methods , Male , Rats
16.
Mol Imaging Biol ; 4(6): 415-24, 2002 Nov.
Article in English | MEDLINE | ID: mdl-14537106

ABSTRACT

PURPOSE: To develop a new fully-automated method for the synthesis of 9-(4-[18F]fluoro-3-hydroxymethylbutyl)guanine ([18F]FHBG) amenable for its routine use in gene therapy monitoring studies. PROCEDURES: A nuclear interface commercial synthesizer was substantially modified and adapted to the synthesis of the referred compound. After the fluorination reaction of the tosylate precursor, the intermediate product was purified by Solid Phase Extraction (SPE) before the hydrolysis. The final product was purified by semi-preparative high performance liquid chromatography (HPLC). RESULTS: [18F]FHBG was obtained in 10-15% yield in 65 minutes including HPLC purification. The radiotracer was > 99% chemically and radiochemically pure, sterile and free from pyrogens. The synthesized compound was shown to accumulate in thymidine kinase (tk) expressing cells both in cell culture, and in laboratory animals infected with an adenoviral vector containing the herpes simplex virus type 1 thymidine kinase (HSV1-tk) gene. CONCLUSIONS: This new procedure facilitates the compliance with the applicable regulatory guidelines for positron emission tomography (PET) radiopharmaceuticals and will assist the clinical application of [18F]FHBG-PET as a noninvasive way to monitor gene therapy in humans.

17.
Elife ; 3: e02008, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24843014

ABSTRACT

Long INterspersed Element-1 (LINE-1 or L1) retrotransposition poses a mutagenic threat to human genomes. Human cells have therefore evolved strategies to regulate L1 retrotransposition. The APOBEC3 (A3) gene family consists of seven enzymes that catalyze deamination of cytidine nucleotides to uridine nucleotides (C-to-U) in single-strand DNA substrates. Among these enzymes, APOBEC3A (A3A) is the most potent inhibitor of L1 retrotransposition in cultured cell assays. However, previous characterization of L1 retrotransposition events generated in the presence of A3A did not yield evidence of deamination. Thus, the molecular mechanism by which A3A inhibits L1 retrotransposition has remained enigmatic. Here, we have used in vitro and in vivo assays to demonstrate that A3A can inhibit L1 retrotransposition by deaminating transiently exposed single-strand DNA that arises during the process of L1 integration. These data provide a mechanistic explanation of how the A3A cytidine deaminase protein can inhibit L1 retrotransposition.DOI: http://dx.doi.org/10.7554/eLife.02008.001.


Subject(s)
Cytidine Deaminase/metabolism , DNA, Single-Stranded/metabolism , Long Interspersed Nucleotide Elements/genetics , Proteins/metabolism , Retroelements , Deamination , Humans
18.
Cell Cycle ; 11(1): 33-8, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22157092

ABSTRACT

The human APOBEC3 family of cytidine deaminases constitutes a cellular intrinsic defense mechanism that is effective against a range of viruses and retro-elements. While it is well established that these enzymes are powerful mutators of viral DNA, the possibility that their activity could threaten the integrity of the host genome has only recently begun to be investigated. Here, we discuss the implications of new evidence suggesting that APOBEC3 proteins can mediate the deamination of cellular DNA. The maintenance of genomic integrity in the face of this potential off-target activity must require high fidelity DNA repair and strict regulation of APOBEC3 gene expression and enzyme activity. Conversely, the ability of specific members of the APOBEC3 family to activate DNA damage signaling pathways might also reflect another way that these proteins contribute to the host immune response.


Subject(s)
Cytosine Deaminase/metabolism , Genomic Instability , APOBEC Deaminases , Cytidine Deaminase , Cytosine Deaminase/genetics , DNA/metabolism , DNA Damage , DNA Repair , Deamination , Humans , Immunity, Innate , Signal Transduction , Uracil-DNA Glycosidase/metabolism
19.
Clin Vaccine Immunol ; 14(11): 1532-5, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17881509

ABSTRACT

Treatment with a low dose of combined aspirin and clopidogrel, two antiplatelet drugs widely used in humans, markedly reduced the homing of virus-specific cytotoxic T lymphocytes and virus-nonspecific inflammatory leukocytes to the liver of mice acutely infected with a hepatotropic, replication-deficient, lacZ-expressing adenovirus (RAd35). Consequently, aspirin/clopidogrel-induced platelet dysfunction greatly diminished liver disease severity and inhibited viral clearance. Along with the finding that aspirin/clopidogrel caused neither bleeding nor anemia, our results suggest that antiplatelet drugs may be considered to limit excessive liver immunopathology and/or to facilitate the persistence of hepatotropic viral vectors utilized in gene therapy.


Subject(s)
Adenoviridae Infections/drug therapy , Adenoviridae/drug effects , Aspirin/therapeutic use , Liver Diseases/drug therapy , Platelet Aggregation Inhibitors/therapeutic use , Ticlopidine/analogs & derivatives , Adenoviridae/physiology , Adenoviridae Infections/virology , Animals , Clopidogrel , Defective Viruses/genetics , Defective Viruses/immunology , Immune System Diseases/drug therapy , Immune System Diseases/immunology , Immune System Diseases/virology , Leukocytes/immunology , Liver/drug effects , Liver/immunology , Liver/virology , Liver Diseases/immunology , Liver Diseases/virology , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Ticlopidine/therapeutic use
20.
J Virol ; 80(3): 1376-84, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16415015

ABSTRACT

Posttranscriptional gene silencing allows sequence-specific control of gene expression. Specificity is guaranteed by small antisense RNAs such as microRNAs (miRNAs) or small interfering RNAs (siRNAs). Functional miRNAs derive from longer double-stranded RNA (dsRNA) molecules that are cleaved to pre-miRNAs in the nucleus and are transported by exportin 5 (Exp 5) to the cytoplasm. Adenovirus-infected cells express virus-associated (VA) RNAs, which are dsRNA molecules similar in structure to pre-miRNAs. VA RNAs are also transported by Exp 5 to the cytoplasm, where they accumulate. Here we show that small RNAs derived from VA RNAs (svaRNAs), similar to miRNAs, can be found in adenovirus-infected cells. VA RNA processing to svaRNAs requires neither viral replication nor viral protein expression, as evidenced by the fact that svaRNA accumulation can be detected in cells transfected with VA sequences. svaRNAs are efficiently bound by Argonaute 2, the endonuclease of the RNA-induced silencing complex, and behave as functional siRNAs, in that they inhibit the expression of reporter genes with complementary sequences. Blocking svaRNA-mediated inhibition affects efficient adenovirus production, indicating that svaRNAs are required for virus viability. Thus, svaRNA-mediated silencing could represent a novel mechanism used by adenoviruses to control cellular or viral gene expression.


Subject(s)
Adenoviridae/genetics , Adenoviridae/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , 3T3 Cells , Adenoviridae/physiology , Animals , Base Sequence , Cell Line , Gene Expression Regulation, Viral , Gene Silencing , HeLa Cells , Humans , Mice , Molecular Sequence Data , Nucleic Acid Conformation , RNA Processing, Post-Transcriptional , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Viral/chemistry , Viral Proteins/biosynthesis , Viral Proteins/genetics , Virus Replication/genetics , Virus Replication/physiology
SELECTION OF CITATIONS
SEARCH DETAIL