Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
Nature ; 552(7684): 278, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29168505

ABSTRACT

This corrects the article DOI: 10.1038/nature23884.

3.
Nature ; 549(7671): 219-226, 2017 09 13.
Article in English | MEDLINE | ID: mdl-28905911

ABSTRACT

The 4D Nucleome Network aims to develop and apply approaches to map the structure and dynamics of the human and mouse genomes in space and time with the goal of gaining deeper mechanistic insights into how the nucleus is organized and functions. The project will develop and benchmark experimental and computational approaches for measuring genome conformation and nuclear organization, and investigate how these contribute to gene regulation and other genome functions. Validated experimental technologies will be combined with biophysical approaches to generate quantitative models of spatial genome organization in different biological states, both in cell populations and in single cells.


Subject(s)
Cell Nucleus/genetics , Cell Nucleus/physiology , Genome , Models, Molecular , Molecular Imaging/methods , Spatio-Temporal Analysis , Animals , Cell Line , Chromatin/genetics , Chromatin/metabolism , Chromosomes/chemistry , Chromosomes/genetics , Chromosomes/metabolism , Genomics/methods , Genomics/organization & administration , Goals , Humans , Information Dissemination , Mice , Models, Biological , Reproducibility of Results , Single-Cell Analysis
4.
Science ; 357(6349)2017 07 28.
Article in English | MEDLINE | ID: mdl-28751582

ABSTRACT

The chromatin structure of DNA determines genome compaction and activity in the nucleus. On the basis of in vitro structures and electron microscopy (EM) studies, the hierarchical model is that 11-nanometer DNA-nucleosome polymers fold into 30- and subsequently into 120- and 300- to 700-nanometer fibers and mitotic chromosomes. To visualize chromatin in situ, we identified a fluorescent dye that stains DNA with an osmiophilic polymer and selectively enhances its contrast in EM. Using ChromEMT (ChromEM tomography), we reveal the ultrastructure and three-dimensional (3D) organization of individual chromatin polymers, megabase domains, and mitotic chromosomes. We show that chromatin is a disordered 5- to 24-nanometer-diameter curvilinear chain that is packed together at different 3D concentration distributions in interphase and mitosis. Chromatin chains have many different particle arrangements and bend at various lengths to achieve structural compaction and high packing densities.


Subject(s)
Chromatin/chemistry , Interphase , Mitosis , 3,3'-Diaminobenzidine/chemistry , Anthraquinones/chemistry , Cell Nucleus/ultrastructure , Chromatin/ultrastructure , DNA/chemistry , DNA/ultrastructure , Fluorescent Dyes/chemistry , Humans , Microscopy, Electron/methods , Nucleosomes/ultrastructure , Oxidation-Reduction , Staining and Labeling/methods
5.
Neuron ; 93(3): 574-586.e8, 2017 Feb 08.
Article in English | MEDLINE | ID: mdl-28111081

ABSTRACT

Microglia are the intrinsic immune sentinels of the central nervous system. Their activation restricts tissue injury and pathogen spread, but in some settings, including viral infection, this response can contribute to cell death and disease. Identifying mechanisms that control microglial responses is therefore an important objective. Using replication-incompetent adenovirus 5 (Ad5)-based vectors as a model, we investigated the mechanisms through which microglia recognize and respond to viral uptake. Transgenic, immunohistochemical, molecular-genetic, and fluorescence imaging approaches revealed that phosphatidylserine (PtdSer) exposure on the outer leaflet of transduced cells triggers their engulfment by microglia through TAM receptor-dependent mechanisms. We show that inhibition of phospholipid scramblase 1 (PLSCR1) activity reduces intracellular calcium dysregulation, prevents PtdSer externalization, and enables months-long protection of vector-transduced, transgene-expressing cells from microglial phagocytosis. Our study identifies PLSCR1 as a potent target through which the innate immune response to viral vectors, and potentially other stimuli, may be controlled.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/immunology , Genetic Vectors/immunology , Immunity, Innate/immunology , Microglia/immunology , Neurons/immunology , Phagocytosis/immunology , Phosphatidylserines/immunology , Phospholipid Transfer Proteins/immunology , Animals , Gene Knockdown Techniques , Immunohistochemistry , Mice, Transgenic , Neurons/virology , Optical Imaging , Phospholipid Transfer Proteins/genetics
7.
Cell ; 162(5): 987-1002, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26317467

ABSTRACT

In response to cellular genome breaks, MRE11/RAD50/NBS1 (MRN) activates a global ATM DNA damage response (DDR) that prevents cellular replication. Here, we show that MRN-ATM also has critical functions in defending the cell against DNA viruses. We reveal temporally distinct responses to adenovirus genomes: a critical MRN-ATM DDR that must be inactivated by E1B-55K/E4-ORF3 viral oncoproteins and a global MRN-independent ATM DDR to viral nuclear domains that does not impact viral replication. We show that MRN binds to adenovirus genomes and activates a localized ATM response that specifically prevents viral DNA replication. In contrast to chromosomal breaks, ATM activation is not amplified by H2AX across megabases of chromatin to induce global signaling and replicative arrest. Thus, γH2AX foci discriminate "self" and "non-self" genomes and determine whether a localized anti-viral or global ATM response is appropriate. This provides an elegant mechanism to neutralize viral genomes without jeopardizing cellular viability.


Subject(s)
Adenoviridae Infections/immunology , DNA Repair , Genome, Viral , Adenoviridae/genetics , Adenoviridae/physiology , Adenovirus E1B Proteins/metabolism , Adenovirus E4 Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins/metabolism , Genome, Human , Histones/metabolism , Humans , Phosphorylation , Virus Replication
8.
J Virol ; 89(20): 10260-72, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26223632

ABSTRACT

UNLABELLED: Adenovirus E4-ORF3 and E1B-55K converge in subverting critical overlapping cellular pathways to facilitate virus replication. Here, we show that E1B-55K and E4-ORF3 induce sumoylation and the assembly of SUMO2/3 viral genome replication domains. Using a conjugation-deficient SUMO2 construct, we demonstrate that SUMO2/3 is recruited to E2A viral genome replication domains through noncovalent interactions. E1B-55K and E4-ORF3 have critical functions in inactivating MRN and ATM to facilitate viral genome replication. We show that ATM kinase inhibitors rescue ΔE1B-55K/ΔE4-ORF3 viral genome replication and that the assembly of E2A domains recruits SUMO2/3 independently of E1B-55K and E4-ORF3. However, the morphology and organization of SUMO2/3-associated E2A domains is strikingly different from that in wild-type Ad5-infected cells. These data reveal that E1B-55K and E4-ORF3 specify the nuclear compartmentalization and structure of SUMO2/3-associated E2A domains, which could have important functions in viral replication. We show that E4-ORF3 specifically targets and sequesters the cellular E3 SUMO ligase PIAS3 but not PIAS1, PIAS2, or PIAS4. The assembly of E4-ORF3 into a multivalent nuclear matrix is required to target PIAS3. In contrast to MRN, PIAS3 is targeted by E4-ORF3 proteins from disparate adenovirus subgroups. Our studies reveal that PIAS3 is a novel and evolutionarily conserved target of E4-ORF3 in human adenovirus infections. Furthermore, we reveal that viral proteins not only disrupt but also usurp SUMO2/3 to transform the nucleus and assemble novel genomic domains that could facilitate pathological viral replication. IMPORTANCE: SUMO is a key posttranslational modification that modulates the function, localization, and assembly of protein complexes. In the ever-escalating host-pathogen arms race, viruses have evolved strategies to subvert sumoylation. Adenovirus is a small DNA tumor virus that is a global human pathogen and key biomedical agent in basic research and therapy. We show that adenovirus infection induces global changes in SUMO localization and conjugation. Using virus and SUMO mutants, we demonstrate that E1B-55K and E4-ORF3 disrupt and usurp SUMO2/3 interactions to transform the nucleus and assemble highly structured and compartmentalized viral genome domains. We reveal that the cellular E3 SUMO ligase PIAS3 is a novel and conserved target of E4-ORF3 proteins from disparate adenovirus subgroups. The induction of sumoylation and SUMO2/3 viral replication domains by early viral proteins could play an important role in determining the outcome of viral infection.


Subject(s)
Adenovirus E1B Proteins/metabolism , Adenovirus E4 Proteins/metabolism , Adenoviruses, Human/metabolism , Genome, Viral , Molecular Chaperones/metabolism , Protein Inhibitors of Activated STAT/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitins/metabolism , Adenovirus E1B Proteins/genetics , Adenovirus E4 Proteins/genetics , Adenoviruses, Human/genetics , Amino Acid Sequence , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/virology , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Models, Molecular , Molecular Chaperones/genetics , Molecular Sequence Data , Open Reading Frames , Osteoblasts/metabolism , Osteoblasts/virology , Protein Inhibitors of Activated STAT/genetics , Sequence Alignment , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation , Ubiquitins/genetics , Virus Replication
9.
Methods ; 90: 39-48, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26066760

ABSTRACT

Structural studies of viral proteins most often use high-resolution techniques such as X-ray crystallography, nuclear magnetic resonance, single particle negative stain, or cryo-electron microscopy (EM) to reveal atomic interactions of soluble, homogeneous viral proteins or viral protein complexes. Once viral proteins or complexes are separated from their host's cellular environment, their natural in situ structure and details of how they interact with other cellular components may be lost. EM has been an invaluable tool in virology since its introduction in the late 1940's and subsequent application to cells in the 1950's. EM studies have expanded our knowledge of viral entry, viral replication, alteration of cellular components, and viral lysis. Most of these early studies were focused on conspicuous morphological cellular changes, because classic EM metal stains were designed to highlight classes of cellular structures rather than specific molecular structures. Much later, to identify viral proteins inducing specific structural configurations at the cellular level, immunostaining with a primary antibody followed by colloidal gold secondary antibody was employed to mark the location of specific viral proteins. This technique can suffer from artifacts in cellular ultrastructure due to compromises required to provide access to the immuno-reagents. Immunolocalization methods also require the generation of highly specific antibodies, which may not be available for every viral protein. Here we discuss new methods to visualize viral proteins and structures at high resolutions in situ using correlated light and electron microscopy (CLEM). We discuss the use of genetically encoded protein fusions that oxidize diaminobenzidine (DAB) into an osmiophilic polymer that can be visualized by EM. Detailed protocols for applying the genetically encoded photo-oxidizing protein MiniSOG to a viral protein, photo-oxidation of the fusion protein to yield DAB polymer staining, and preparation of photo-oxidized samples for TEM and serial block-face scanning EM (SBEM) for large-scale volume EM data acquisition are also presented. As an example, we discuss the recent multi-scale analysis of Adenoviral protein E4-ORF3 that reveals a new type of multi-functional polymer that disrupts multiple cellular proteins. This new capability to visualize unambiguously specific viral protein structures at high resolutions in the native cellular environment is revealing new insights into how they usurp host proteins and functions to drive pathological viral replication.


Subject(s)
Microscopy, Electron/methods , Viral Proteins/chemistry , Adenoviridae , Cell Line , Host-Pathogen Interactions , Humans , Models, Chemical , Oxidation-Reduction , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry
10.
Cell Metab ; 19(4): 549-50, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24703688

ABSTRACT

Viral and cellular oncogenes converge in targeting critical protein interaction networks to reprogram the cellular DNA and protein replication machinery for pathological replication. In this issue, Thai et al. (2014) show that adenovirus E4ORF1 activates MYC glycolytic targets to induce a Warburg-like effect that converts glucose into nucleotides for viral replication.


Subject(s)
Adenovirus E4 Proteins/metabolism , Epithelial Cells/metabolism , Glucose/metabolism , Metabolic Networks and Pathways/physiology , Models, Biological , Proto-Oncogene Proteins c-myc/metabolism , Virus Replication/physiology , Humans
11.
Cell ; 151(2): 304-19, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23063122

ABSTRACT

Evolution of minimal DNA tumor virus' genomes has selected for small viral oncoproteins that hijack critical cellular protein interaction networks. The structural basis for the multiple and dominant functions of adenovirus oncoproteins has remained elusive. E4-ORF3 forms a nuclear polymer and simultaneously inactivates p53, PML, TRIM24, and MRE11/RAD50/NBS1 (MRN) tumor suppressors. We identify oligomerization mutants and solve the crystal structure of E4-ORF3. E4-ORF3 forms a dimer with a central ß core, and its structure is unrelated to known polymers or oncogenes. E4-ORF3 dimer units coassemble through reciprocal and nonreciprocal exchanges of their C-terminal tails. This results in linear and branched oligomer chains that further assemble in variable arrangements to form a polymer network that partitions the nuclear volume. E4-ORF3 assembly creates avidity-driven interactions with PML and an emergent MRN binding interface. This reveals an elegant structural solution whereby a small protein forms a multivalent matrix that traps disparate tumor suppressors.


Subject(s)
Adenovirus E4 Proteins/chemistry , Adenovirus E4 Proteins/metabolism , Adenoviruses, Human/metabolism , Tumor Suppressor Proteins/metabolism , Adenovirus Infections, Human/virology , Cell Line , Cells, Cultured , Crystallography, X-Ray , Humans , Plant Cells/virology , Protein Folding , Nicotiana/virology
12.
Article in English | MEDLINE | ID: mdl-21061422

ABSTRACT

One of the greatest challenges in biomedicine is to define the critical targets and network interactions that are subverted to elicit growth deregulation in human cells. Understanding and developing rational treatments for cancer requires a definition of the key molecular targets and how they interact to elicit the complex growth deregulation phenotype. Viral proteins provide discerning and powerful probes to understand both how cells work and how they can be manipulated using a minimal number of components. The small DNA viruses have evolved to target inherent weaknesses in cellular protein interaction networks to hijack the cellular DNA and protein replication machinery. In the battle to escape the inevitability of senescence and programmed cell death, cancers have converged on similar mechanisms, through the acquisition and selection of somatic mutations that drive unchecked cellular replication in tumors. Understanding the dynamic mechanisms through which a minimal number of viral proteins promote host cells to undergo unscheduled and pathological replication is a powerful strategy to identify critical targets that are also disrupted in cancer. Viruses can therefore be used as tools to probe the system-wide protein-protein interactions and structures that drive growth deregulation in human cells. Ultimately this can provide a path for developing system context-dependent therapeutics. This review will describe ongoing experimental approaches using viruses to study pathways deregulated in cancer, with a particular focus on viral cellular protein-protein interactions and structures.


Subject(s)
Neoplasms/metabolism , Viral Proteins/chemistry , DNA/chemistry , DNA/metabolism , Humans , Neoplasms/genetics , Neoplasms/virology , Protein Binding , Viral Proteins/metabolism , Virus Replication
13.
Mol Cell ; 39(6): 835-7, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20864031

ABSTRACT

The MAGE proteins are best known as curious tumor-specific antigens. However, Doyle et al. (2010) reveal that MAGE proteins interact with RING proteins to promote ubiquitylation which provides important new insights into the physiological and pathological functions of this enigmatic family of proteins.

14.
Nature ; 466(7310): 1076-81, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20740008

ABSTRACT

The transcription factor p53 (also known as TP53) guards against tumour and virus replication and is inactivated in almost all cancers. p53-activated transcription of target genes is thought to be synonymous with the stabilization of p53 in response to oncogenes and DNA damage. During adenovirus replication, the degradation of p53 by E1B-55k is considered essential for p53 inactivation, and is the basis for p53-selective viral cancer therapies. Here we reveal a dominant epigenetic mechanism that silences p53-activated transcription, irrespective of p53 phosphorylation and stabilization. We show that another adenoviral protein, E4-ORF3, inactivates p53 independently of E1B-55k by forming a nuclear structure that induces de novo H3K9me3 heterochromatin formation at p53 target promoters, preventing p53-DNA binding. This suppressive nuclear web is highly selective in silencing p53 promoters and operates in the backdrop of global transcriptional changes that drive oncogenic replication. These findings are important for understanding how high levels of wild-type p53 might also be inactivated in cancer as well as the mechanisms that induce aberrant epigenetic silencing of tumour-suppressor loci. Our study changes the longstanding definition of how p53 is inactivated in adenovirus infection and provides key insights that could enable the development of true p53-selective oncolytic viral therapies.


Subject(s)
Adenoviridae/metabolism , Gene Silencing , Heterochromatin/metabolism , Oncogene Proteins, Viral/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Cell Proliferation , Cells, Cultured , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HCT116 Cells , Histones/metabolism , Humans , Methylation , Neoplasms/metabolism , Neoplasms/virology , Protein Binding , Virus Replication
15.
J Virol ; 81(23): 13209-17, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17686851

ABSTRACT

During the late stages of adenovirus infection, the 100K protein (100K) inhibits the translation of cellular messages in the cytoplasm and regulates hexon trimerization and assembly in the nucleus. However, it is not known how it switches between these two functions. Here we show that 100K is methylated on arginine residues at its C terminus during infection and that this region is necessary for binding PRMT1 methylase. Methylated 100K is exclusively nuclear. Mutation of the third RGG motif (amino acids 741 to 743) prevents localization to the nucleus during infection, suggesting that methylation of that sequence is important for 100K shuttling. Treatment of infected cells with methylation inhibitors inhibits expression of late structural proteins. These data suggest that arginine methylation of 100K is necessary for its localization to the nucleus and is a critical cellular function necessary for productive adenovirus infection.


Subject(s)
Adenoviridae/metabolism , Arginine/metabolism , Viral Nonstructural Proteins/metabolism , Cell Line , Cell Nucleus/chemistry , Humans , Methylation , Protein Binding , Protein Interaction Mapping , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Viral Nonstructural Proteins/chemistry , Viral Structural Proteins/biosynthesis
16.
Cancer Cell ; 10(6): 501-14, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17157790

ABSTRACT

There is currently much interest in the idea of restoring p53 activity in tumor cells by inhibiting Hdm2/Mdm2. However, it has remained unclear whether this would also activate p53 in normal cells. Using a switchable endogenous p53 mouse model, which allows rapid and reversible toggling of p53 status between wild-type and null states, we show that p53 is spontaneously active in all tested tissues of mdm2-deficient mice, triggering fatal pathologies that include ablation of classically radiosensitive tissues. In apoptosis-resistant tissues, spontaneous unbuffered p53 activity triggers profound inhibition of cell proliferation. Such acute spontaneous p53 activity occurs in the absence of any detectable p53 posttranslational modification, DNA damage, or p19ARF signaling and triggers rapid p53 degradation.


Subject(s)
Proto-Oncogene Proteins c-mdm2/physiology , Tumor Suppressor Protein p53/physiology , ADP-Ribosylation Factor 1/physiology , Animals , DNA Damage , Imidazoles , Mice , Phosphorylation , Piperazines , Proto-Oncogene Proteins c-mdm2/deficiency , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Transcription, Genetic
18.
Oncogene ; 24(52): 7640-55, 2005 Nov 21.
Article in English | MEDLINE | ID: mdl-16299526

ABSTRACT

DNA viruses have enormous utility in cancer research, both as tools for tumor target discovery as well as agents for lytic cancer therapies. This is because there is a profound functional overlap between the DNA viral and tumor cell programs. DNA viruses encode proteins that elicit growth deregulation in infected cells similar to that engendered by mutations in tumor cells. Evolution has refined viral proteins to target the critical cellular hubs that regulate growth. Thus, viral proteins are discriminating biochemical probes that can be used to identify and characterize novel tumor targets. Moreover, the overlap between the DNA viral and tumor programs can also be exploited for the development of lytic cancer therapies. Discovering whether tumor cells selectively complement the replication of viral mutants can reveal novel oncolytic viral therapies, as well as unexpected tumor properties. For example, altered RNA export was recently uncovered as a novel tumor cell property that underlies ONYX-015 replication, a promising oncolytic adenoviral therapy. A perspective is provided on how adenovirus could be systematically exploited to map the requisite role, or indeed the redundancy, of cellular pathways that act in an integrated program to elicit pathological replication. This knowledge has important applications for the rational design of the next generation of oncolytic viruses, as well as the discovery of efficacious combination cancer therapies.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/pathogenicity , Viral Proteins/genetics , Viral Proteins/physiology , Adenoviridae , Aging , Cell Proliferation , DNA Viruses/pathogenicity , Humans , Neoplasms/virology , RNA/metabolism
19.
Cancer Cell ; 8(1): 61-74, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16023599

ABSTRACT

ONYX-015 is an E1B-55K-deleted adenovirus that has promising clinical activity as a cancer therapy. However, many tumor cells fail to support ONYX-015 oncolytic replication. E1B-55K functions include p53 degradation, RNA export, and host protein shutoff. Here, we show that resistant tumor cell lines fail to provide the RNA export functions of E1B-55K necessary for ONYX-015 replication; viral 100K mRNA export is necessary for host protein shutoff. However, heat shock rescues late viral RNA export and renders refractory tumor cells permissive to ONYX-015. These data indicate that heat shock and late adenoviral RNAs may converge upon a common mechanism for their export. Moreover, these data suggest that the concomitant induction of a heat shock response could significantly improve ONYX-015 cancer therapy.


Subject(s)
Adenoviridae/physiology , Drug Resistance, Neoplasm , Neoplasms/therapy , RNA Transport , Adenoviridae/genetics , Adenoviridae/metabolism , Adenovirus E1B Proteins/genetics , Adenovirus E1B Proteins/metabolism , Antineoplastic Agents/therapeutic use , Cells, Cultured , Cytopathogenic Effect, Viral/genetics , Hot Temperature/therapeutic use , Humans , Neoplasms/metabolism , Neoplasms/virology , Phenotype , RNA, Viral/metabolism , Shock/therapy , Viral Nonstructural Proteins/metabolism , Viral Vaccines , Virus Replication/genetics
20.
Cell Cycle ; 4(7): 883-8, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15970698

ABSTRACT

mTOR is a critical regulator of protein translation, and plays an important role in controlling cellular replication. Recent studies indicate that nutrient and growth factor mediated activation of mTOR is deregulated in human cancer, and therefore represents an attractive tumor target. However, activation of mTOR is a complex process that is not yet fully understood. DNA viruses and tumor cells often perturb similar cellular pathways to facilitate their replication. In a recent study, we used adenovirus as a novel tool to probe the mechanisms underlying the inappropriate activation of mTOR upon virus infection of quiescent primary cells. These studies revealed that adenovirus encodes two viral proteins, E4-ORF1 and E4-ORF4, which activate mTOR, even in the absence of nutrient/growth factor signals, and which play a role in promoting viral replication. E4-ORF1 mimics growth factor signaling to mTOR by activating PI3-kinase, whereas E4-ORF4, which binds and relocalizes PP2A, can substitute for glucose mediated activation of mTOR. We discuss insights from this study, together with the similarities that may exist between viruses and tumor cells with respect to the mechanistic and functional requirements for mTOR activation in driving their aberrant DNA replication.


Subject(s)
Adenoviridae/metabolism , Cell Cycle , Protein Biosynthesis , Gene Expression Regulation, Viral , Growth Substances , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...