Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Nat Cell Biol ; 25(10): 1535-1545, 2023 10.
Article in English | MEDLINE | ID: mdl-37735597

ABSTRACT

Specificity within the ubiquitin-proteasome system is primarily achieved through E3 ubiquitin ligases, but for many E3s their substrates-and in particular the molecular features (degrons) that they recognize-remain largely unknown. Current approaches for assigning E3s to their cognate substrates are tedious and low throughput. Here we developed a multiplex CRISPR screening platform to assign E3 ligases to their cognate substrates at scale. A proof-of-principle multiplex screen successfully performed ~100 CRISPR screens in a single experiment, refining known C-degron pathways and identifying an additional pathway through which Cul2FEM1B targets C-terminal proline. Further, by identifying substrates for Cul1FBXO38, Cul2APPBP2, Cul3GAN, Cul3KLHL8, Cul3KLHL9/13 and Cul3KLHL15, we demonstrate that the approach is compatible with pools of full-length protein substrates of varying stabilities and, when combined with site-saturation mutagenesis, can assign E3 ligases to their cognate degron motifs. Thus, multiplex CRISPR screening will accelerate our understanding of how specificity is achieved within the ubiquitin-proteasome system.


Subject(s)
Proteasome Endopeptidase Complex , Ubiquitin-Protein Ligases , Ubiquitin-Protein Ligases/metabolism , Proteasome Endopeptidase Complex/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Ubiquitin/genetics , Ubiquitin/metabolism
3.
Mol Cell ; 82(2): 479-491.e7, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34963054

ABSTRACT

Genetically encoded biosensors are powerful tools to monitor cellular behavior, but the difficulty in generating appropriate reporters for chromatin factors hampers our ability to dissect epigenetic pathways. Here, we present TRACE (transgene reporters across chromatin environments), a high-throughput, genome-wide technique to generate fluorescent human reporter cell lines responsive to manipulation of epigenetic factors. By profiling GFP expression from a large pool of individually barcoded lentiviral integrants in the presence and absence of a perturbation, we identify reporters responsive to pharmacological inhibition of the histone lysine demethylase LSD1 and genetic ablation of the PRC2 subunit SUZ12. Furthermore, by manipulating the HIV-1 host factor LEDGF through targeted deletion or fusion to chromatin reader domains, we alter lentiviral integration site preferences, thus broadening the types of chromatin examined by TRACE. The phenotypic reporters generated through TRACE will allow the genetic interrogation of a broad range of epigenetic pathways, furthering our mechanistic understanding of chromatin biology.


Subject(s)
Biosensing Techniques , Epigenesis, Genetic , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/genetics , Lentivirus/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Chromatin Assembly and Disassembly , Epigenome , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Lentivirus/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , THP-1 Cells , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Nat Commun ; 11(1): 4940, 2020 10 02.
Article in English | MEDLINE | ID: mdl-33009411

ABSTRACT

The HUSH complex represses retroviruses, transposons and genes to maintain the integrity of vertebrate genomes. HUSH regulates deposition of the epigenetic mark H3K9me3, but how its three core subunits - TASOR, MPP8 and Periphilin - contribute to assembly and targeting of the complex remains unknown. Here, we define the biochemical basis of HUSH assembly and find that its modular architecture resembles the yeast RNA-induced transcriptional silencing complex. TASOR, the central HUSH subunit, associates with RNA processing components. TASOR is required for H3K9me3 deposition over LINE-1 repeats and repetitive exons in transcribed genes. In the context of previous studies, this suggests that an RNA intermediate is important for HUSH activity. We dissect the TASOR and MPP8 domains necessary for transgene repression. Structure-function analyses reveal TASOR bears a catalytically-inactive PARP domain necessary for targeted H3K9me3 deposition. We conclude that TASOR is a multifunctional pseudo-PARP that directs HUSH assembly and epigenetic regulation of repetitive genomic targets.


Subject(s)
DNA Transposable Elements/genetics , Epigenesis, Genetic , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Amino Acid Sequence , Antigens, Neoplasm/metabolism , Binding Sites , Exons/genetics , Genome , HEK293 Cells , HeLa Cells , Histones/metabolism , Humans , Lysine/metabolism , Magnetic Resonance Spectroscopy , Methylation , NAD/metabolism , Nuclear Proteins/chemistry , Phosphoproteins/metabolism , Protein Binding , Protein Domains , RNA/metabolism , RNA Processing, Post-Transcriptional , Transcription, Genetic
5.
Nucleic Acids Res ; 48(18): 10313-10328, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32976585

ABSTRACT

Transcription of integrated DNA from viruses or transposable elements is tightly regulated to prevent pathogenesis. The Human Silencing Hub (HUSH), composed of Periphilin, TASOR and MPP8, silences transcriptionally active viral and endogenous transgenes. HUSH recruits effectors that alter the epigenetic landscape and chromatin structure, but how HUSH recognizes target loci and represses their expression remains unclear. We identify the physicochemical properties of Periphilin necessary for HUSH assembly and silencing. A disordered N-terminal domain (NTD) and structured C-terminal domain are essential for silencing. A crystal structure of the Periphilin-TASOR minimal core complex shows Periphilin forms an α-helical homodimer, bound by a single TASOR molecule. The NTD forms insoluble aggregates through an arginine/tyrosine-rich sequence reminiscent of low-complexity regions from self-associating RNA-binding proteins. Residues required for TASOR binding and aggregation were required for HUSH-dependent silencing and genome-wide deposition of repressive mark H3K9me3. The NTD was functionally complemented by low-complexity regions from certain RNA-binding proteins and proteins that form condensates or fibrils. Our work suggests the associative properties of Periphilin promote HUSH aggregation at target loci.


Subject(s)
Antigens, Neoplasm/ultrastructure , Nuclear Proteins/ultrastructure , RNA-Binding Proteins/chemistry , Transcription, Genetic , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Crystallography, X-Ray , DNA Transposable Elements/genetics , Epigenesis, Genetic/genetics , Gene Silencing , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Aggregates/genetics , Protein Binding/genetics , Protein Conformation, alpha-Helical , Protein Domains/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/ultrastructure , Viruses/genetics
6.
Am J Hum Genet ; 107(2): 352-363, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32693025

ABSTRACT

MORC2 encodes an ATPase that plays a role in chromatin remodeling, DNA repair, and transcriptional regulation. Heterozygous variants in MORC2 have been reported in individuals with autosomal-dominant Charcot-Marie-Tooth disease type 2Z and spinal muscular atrophy, and the onset of symptoms ranges from infancy to the second decade of life. Here, we present a cohort of 20 individuals referred for exome sequencing who harbor pathogenic variants in the ATPase module of MORC2. Individuals presented with a similar phenotype consisting of developmental delay, intellectual disability, growth retardation, microcephaly, and variable craniofacial dysmorphism. Weakness, hyporeflexia, and electrophysiologic abnormalities suggestive of neuropathy were frequently observed but were not the predominant feature. Five of 18 individuals for whom brain imaging was available had lesions reminiscent of those observed in Leigh syndrome, and five of six individuals who had dilated eye exams had retinal pigmentary abnormalities. Functional assays revealed that these MORC2 variants result in hyperactivation of epigenetic silencing by the HUSH complex, supporting their pathogenicity. The described set of morphological, growth, developmental, and neurological findings and medical concerns expands the spectrum of genetic disorders resulting from pathogenic variants in MORC2.


Subject(s)
Adenosine Triphosphatases/genetics , Craniofacial Abnormalities/genetics , Growth Disorders/genetics , Mutation/genetics , Neurodevelopmental Disorders/genetics , Transcription Factors/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Genetic Diseases, Inborn/genetics , Heterozygote , Humans , Infant , Intellectual Disability/genetics , Male , Microcephaly/genetics , Middle Aged , Phenotype , Young Adult
7.
Nat Protoc ; 14(1): 153-170, 2019 01.
Article in English | MEDLINE | ID: mdl-30518911

ABSTRACT

Alterations in chromatin structure play a major role in the epigenetic regulation of gene expression. Here, we describe a step-by-step protocol for differential viral accessibility (DIVA), a method for identifying changes in chromatin accessibility genome-wide. Commonly used methods for mapping accessible genomic loci have strong preferences toward detecting 'open' chromatin found at regulatory regions but are not well suited to studying chromatin accessibility in gene bodies and intergenic regions. DIVA overcomes this limitation, enabling a broader range of sites to be interrogated. Conceptually, DIVA is similar to ATAC-seq in that it relies on the integration of exogenous DNA into the genome to map accessible chromatin, except that chromatin architecture is probed through mapping integration sites of exogenous lentiviruses. An isogenic pair of cell lines are transduced with a lentiviral vector, followed by PCR amplification and Illumina sequencing of virus-genome junctions; the resulting sequences define a set of unique lentiviral integration sites, which are compared to determine whether genomic loci exhibit significantly altered accessibility between experimental and control cells. Experienced researchers will take 6 d to generate lentiviral stocks and transduce the target cells, a further 5 d to prepare the Illumina sequencing libraries and a few hours to perform the bioinformatic analysis.


Subject(s)
Chromatin/chemistry , Chromosome Mapping/methods , DNA, Viral/genetics , Genome, Human , Lentivirus/genetics , Virus Integration , Chromatin/virology , Chromosome Mapping/statistics & numerical data , Genetic Loci , Genomic Library , High-Throughput Nucleotide Sequencing , Humans , Polymerase Chain Reaction , Transduction, Genetic
8.
Genome Res ; 28(6): 836-845, 2018 06.
Article in English | MEDLINE | ID: mdl-29728366

ABSTRACT

Retrotransposons encompass half of the human genome and contribute to the formation of heterochromatin, which provides nuclear structure and regulates gene expression. Here, we asked if the human silencing hub (HUSH) complex is necessary to silence retrotransposons and whether it collaborates with TRIM28 and the chromatin remodeler ATRX at specific genomic loci. We show that the HUSH complex contributes to de novo repression and DNA methylation of an SVA retrotransposon reporter. By using naïve versus primed mouse pluripotent stem cells, we reveal a critical role for the HUSH complex in naïve cells, implicating it in programming epigenetic marks in development. Although the HUSH component FAM208A binds to endogenous retroviruses (ERVs) and long interspersed element-1s (LINE-1s or L1s), it is mainly required to repress evolutionarily young L1s (mouse-specific lineages <5 million years old). TRIM28, in contrast, is necessary to repress both ERVs and young L1s. Genes co-repressed by TRIM28 and FAM208A are evolutionarily young, or exhibit tissue-specific expression, are enriched in young L1s, and display evidence for regulation through LTR promoters. Finally, we demonstrate that the HUSH complex is also required to repress L1 elements in human cells. Overall, these data indicate that the HUSH complex and TRIM28 co-repress young retrotransposons and new genes rewired by retrotransposon noncoding DNA.


Subject(s)
Genome, Human , Nuclear Proteins/genetics , Retroelements/genetics , Tripartite Motif-Containing Protein 28/genetics , Animals , DNA Methylation/genetics , Endogenous Retroviruses/genetics , Heterochromatin/genetics , Humans , Long Interspersed Nucleotide Elements/genetics , Mice , Promoter Regions, Genetic
9.
Nat Commun ; 9(1): 651, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29440755

ABSTRACT

Missense mutations in MORC2 cause neuropathies including spinal muscular atrophy and Charcot-Marie-Tooth disease. We recently identified MORC2 as an effector of epigenetic silencing by the human silencing hub (HUSH). Here we report the biochemical and cellular activities of MORC2 variants, alongside crystal structures of wild-type and neuropathic forms of a human MORC2 fragment comprising the GHKL-type ATPase module and CW-type zinc finger. This fragment dimerizes upon binding ATP and contains a hinged, functionally critical coiled-coil insertion absent in other GHKL ATPases. We find that dimerization and DNA binding of the MORC2 ATPase module transduce HUSH-dependent silencing. Disease mutations change the dynamics of dimerization by distinct structural mechanisms: destabilizing the ATPase-CW module, trapping the ATP lid, or perturbing the dimer interface. These defects lead to the modulation of HUSH function, thus providing a molecular basis for understanding MORC2-associated neuropathies.


Subject(s)
Adenosine Triphosphatases/metabolism , Epigenesis, Genetic , Gene Silencing , Mutation, Missense , Nervous System Diseases/genetics , Transcription Factors/genetics , Adenosine Triphosphate/metabolism , Animals , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Crystallography, X-Ray , DNA/metabolism , HEK293 Cells , HeLa Cells , Humans , Muscular Atrophy, Spinal , Nervous System Diseases/pathology , Protein Binding , Protein Conformation , Protein Multimerization , Sf9 Cells , Transcription Factors/chemistry , Transcription Factors/metabolism , Zinc Fingers
10.
Mol Cell ; 69(1): 5-7, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29304334

ABSTRACT

Post-translational modifications of histones are widely used to discriminate between different types of chromatin. In a recent issue of Molecular Cell, Becker et al. (2017) delineate chromatin states based on physical properties, thereby expanding our understanding of chromatin function.


Subject(s)
Chromatin , Histone Code , Genomics , Histones/genetics , Protein Processing, Post-Translational , Proteomics
11.
Nat Genet ; 49(7): 1035-1044, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28581500

ABSTRACT

Dominant mutations in the MORC2 gene have recently been shown to cause axonal Charcot-Marie-Tooth (CMT) disease, but the cellular function of MORC2 is poorly understood. Here, through a genome-wide CRISPR-Cas9-mediated forward genetic screen, we identified MORC2 as an essential gene required for epigenetic silencing by the HUSH complex. HUSH recruits MORC2 to target sites in heterochromatin. We exploited a new method, differential viral accessibility (DIVA), to show that loss of MORC2 results in chromatin decompaction at these target loci, which is concomitant with a loss of H3K9me3 deposition and transcriptional derepression. The ATPase activity of MORC2 is critical for HUSH-mediated silencing, and the most common alteration affecting the ATPase domain in CMT patients (p.Arg252Trp) hyperactivates HUSH-mediated repression in neuronal cells. These data define a critical role for MORC2 in epigenetic silencing by the HUSH complex and provide a mechanistic basis underpinning the role of MORC2 mutations in CMT disease.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Chromatin Assembly and Disassembly/genetics , Epigenetic Repression/genetics , Gene Silencing , Heterochromatin/genetics , Transcription Factors/genetics , Adenosine Triphosphatases/metabolism , CRISPR-Cas Systems , Charcot-Marie-Tooth Disease/metabolism , HeLa Cells , Heterochromatin/metabolism , Histone Code , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Lysine/chemistry , Methylation , Multiprotein Complexes , Mutation, Missense , Neurons/metabolism , Protein Domains , Protein Interaction Mapping , Protein Methyltransferases/metabolism , Protein Processing, Post-Translational , Transcription Factors/physiology , Transgenes
12.
Cell Rep ; 17(3): 653-659, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27732843

ABSTRACT

The histone methyltransferase SETDB1 plays a central role in repressive chromatin processes, but the functional requirement for its binding partner ATF7IP has remained enigmatic. Here, we show that ATF7IP is essential for SETDB1 stability: nuclear SETDB1 protein is degraded by the proteasome upon ablation of ATF7IP. As a result, ATF7IP is critical for repression that requires H3K9 trimethylation by SETDB1, including transgene silencing by the HUSH complex. Furthermore, we show that loss of ATF7IP phenocopies loss of SETDB1 in genome-wide assays. ATF7IP and SETDB1 knockout cells exhibit near-identical defects in the global deposition of H3K9me3, which results in similar dysregulation of the transcriptome. Overall, these data identify a critical functional role for ATF7IP in heterochromatin formation by regulating SETDB1 abundance in the nucleus.


Subject(s)
Heterochromatin/metabolism , Multiprotein Complexes/metabolism , Protein Methyltransferases/metabolism , Transcription Factors/metabolism , Cell Nucleus/metabolism , Enzyme Stability , Epigenesis, Genetic , Gene Deletion , Gene Silencing , HeLa Cells , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Repressor Proteins , Transcriptome/genetics
13.
Nat Commun ; 7: 11786, 2016 06 10.
Article in English | MEDLINE | ID: mdl-27283361

ABSTRACT

The application of forward genetic screens to cultured human cells represents a powerful method to study gene function. The repurposing of the bacterial CRISPR/Cas9 system provides an effective method to disrupt gene function in mammalian cells, and has been applied to genome-wide screens. Here, we compare the efficacy of genome-wide CRISPR/Cas9-mediated forward genetic screens versus gene-trap mutagenesis screens in haploid human cells, which represent the existing 'gold standard' method. This head-to-head comparison aimed to identify genes required for the endoplasmic reticulum-associated degradation (ERAD) of MHC class I molecules. The two approaches show high concordance (>70%), successfully identifying the majority of the known components of the canonical glycoprotein ERAD pathway. Both screens also identify a role for the uncharacterized gene TXNDC11, which we show encodes an EDEM2/3-associated disulphide reductase. Genome-wide CRISPR/Cas9-mediated screens together with haploid genetic screens provide a powerful addition to the forward genetic toolbox.


Subject(s)
CRISPR-Cas Systems/genetics , Endoplasmic Reticulum-Associated Degradation/genetics , Genetic Testing , Haploidy , Mammals/genetics , Animals , Fluorescent Dyes/metabolism , Genes, Reporter , Glycoproteins/metabolism , HEK293 Cells , HeLa Cells , Histocompatibility Antigens Class I/metabolism , Humans , Oxidation-Reduction , Protein Binding , Protein Domains , Thioredoxins/chemistry , Thioredoxins/metabolism , alpha-Glucosidases/metabolism
14.
Bioessays ; 38(4): 333-43, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26853531

ABSTRACT

Much of what we understand about heterochromatin formation in mammals has been extrapolated from forward genetic screens for modifiers of position-effect variegation (PEV) in the fruit fly Drosophila melanogaster. The recent identification of the HUSH (Human Silencing Hub) complex suggests that more recent evolutionary developments contribute to the mechanisms underlying PEV in human cells. Although HUSH-mediated repression also involves heterochromatin spreading through the reading and writing of the repressive H3K9me3 histone modification, clear orthologues of HUSH subunits are not found in Drosophila but are conserved in vertebrates. Here we compare the insights into the mechanisms of PEV derived from genetic screens in the fly, the mouse and in human cells, review what is currently known about the HUSH complex and discuss the implications of HUSH-mediated silencing for viral latency. Future studies will provide mechanistic insight into HUSH complex function and reveal the relationship between HUSH and other epigenetic silencing complexes.


Subject(s)
Antigens, Neoplasm/genetics , Chromosomal Position Effects , Gene Silencing , Heterochromatin/metabolism , Histones/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Animals , Antigens, Neoplasm/metabolism , Biological Evolution , Cell Line, Tumor , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Genetic Testing , HIV-1/genetics , HIV-1/metabolism , Haploidy , Heterochromatin/chemistry , Histones/metabolism , Humans , Mice , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Virus Latency
15.
Science ; 348(6242): 1481-1485, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-26022416

ABSTRACT

Forward genetic screens in Drosophila melanogaster for modifiers of position-effect variegation have revealed the basis of much of our understanding of heterochromatin. We took an analogous approach to identify genes required for epigenetic repression in human cells. A nonlethal forward genetic screen in near-haploid KBM7 cells identified the HUSH (human silencing hub) complex, comprising three poorly characterized proteins, TASOR, MPP8, and periphilin; this complex is absent from Drosophila but is conserved from fish to humans. Loss of HUSH components resulted in decreased H3K9me3 both at endogenous genomic loci and at retroviruses integrated into heterochromatin. Our results suggest that the HUSH complex is recruited to genomic loci rich in H3K9me3, where subsequent recruitment of the methyltransferase SETDB1 is required for further H3K9me3 deposition to maintain transcriptional silencing.


Subject(s)
Antigens, Neoplasm/metabolism , Chromosomal Position Effects , Gene Silencing , Histones/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Animals , Antigens, Neoplasm/genetics , Conserved Sequence , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Evolution, Molecular , Genes, Reporter , Genetic Loci , Green Fluorescent Proteins/genetics , HeLa Cells , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase , Humans , Immunoprecipitation , Multiprotein Complexes/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Protein Methyltransferases/metabolism
16.
PLoS Pathog ; 9(11): e1003772, 2013.
Article in English | MEDLINE | ID: mdl-24278019

ABSTRACT

The Kaposi's sarcoma-associated herpesvirus gene products K3 and K5 are viral ubiquitin E3 ligases which downregulate MHC-I and additional cell surface immunoreceptors. To identify novel cellular genes required for K5 function we performed a forward genetic screen in near-haploid human KBM7 cells. The screen identified proteolipid protein 2 (PLP2), a MARVEL domain protein of unknown function, as essential for K5 activity. Genetic loss of PLP2 traps the viral ligase in the endoplasmic reticulum, where it is unable to ubiquitinate and degrade its substrates. Subsequent analysis of the plasma membrane proteome of K5-expressing KBM7 cells in the presence and absence of PLP2 revealed a wide range of novel K5 targets, all of which required PLP2 for their K5-mediated downregulation. This work ascribes a critical function to PLP2 for viral ligase activity and underlines the power of non-lethal haploid genetic screens in human cells to identify the genes involved in pathogen manipulation of the host immune system.


Subject(s)
Cell Membrane/metabolism , Down-Regulation , Herpesvirus 8, Human/enzymology , Immediate-Early Proteins/biosynthesis , MARVEL Domain-Containing Proteins/biosynthesis , Proteolipids/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Viral Proteins/biosynthesis , Cell Membrane/genetics , Cell Membrane/immunology , Genetic Testing , HeLa Cells , Hep G2 Cells , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/immunology , Humans , Immediate-Early Proteins/genetics , MARVEL Domain-Containing Proteins/genetics , MARVEL Domain-Containing Proteins/immunology , Proteolipids/genetics , Proteolipids/immunology , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/immunology , Sarcoma, Kaposi/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology , Viral Proteins/genetics , Viral Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...