Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Exp Med ; 219(10)2022 10 03.
Article in English | MEDLINE | ID: mdl-36094518

ABSTRACT

Human cells homozygous for rare loss-of-expression (LOE) TYK2 alleles have impaired, but not abolished, cellular responses to IFN-α/ß (underlying viral diseases in the patients) and to IL-12 and IL-23 (underlying mycobacterial diseases). Cells homozygous for the common P1104A TYK2 allele have selectively impaired responses to IL-23 (underlying isolated mycobacterial disease). We report three new forms of TYK2 deficiency in six patients from five families homozygous for rare TYK2 alleles (R864C, G996R, G634E, or G1010D) or compound heterozygous for P1104A and a rare allele (A928V). All these missense alleles encode detectable proteins. The R864C and G1010D alleles are hypomorphic and loss-of-function (LOF), respectively, across signaling pathways. By contrast, hypomorphic G996R, G634E, and A928V mutations selectively impair responses to IL-23, like P1104A. Impairment of the IL-23-dependent induction of IFN-γ is the only mechanism of mycobacterial disease common to patients with complete TYK2 deficiency with or without TYK2 expression, partial TYK2 deficiency across signaling pathways, or rare or common partial TYK2 deficiency specific for IL-23 signaling.


Subject(s)
Job Syndrome , TYK2 Kinase , Humans , Interferon-gamma/metabolism , Interleukin-23 , Job Syndrome/genetics , TYK2 Kinase/deficiency , TYK2 Kinase/genetics , TYK2 Kinase/metabolism
2.
Sci Immunol ; 3(30)2018 12 21.
Article in English | MEDLINE | ID: mdl-30578352

ABSTRACT

Inherited IL-12Rß1 and TYK2 deficiencies impair both IL-12- and IL-23-dependent IFN-γ immunity and are rare monogenic causes of tuberculosis, each found in less than 1/600,000 individuals. We show that homozygosity for the common TYK2 P1104A allele, which is found in about 1/600 Europeans and between 1/1000 and 1/10,000 individuals in regions other than East Asia, is more frequent in a cohort of patients with tuberculosis from endemic areas than in ethnicity-adjusted controls (P = 8.37 × 10-8; odds ratio, 89.31; 95% CI, 14.7 to 1725). Moreover, the frequency of P1104A in Europeans has decreased, from about 9% to 4.2%, over the past 4000 years, consistent with purging of this variant by endemic tuberculosis. Surprisingly, we also show that TYK2 P1104A impairs cellular responses to IL-23, but not to IFN-α, IL-10, or even IL-12, which, like IL-23, induces IFN-γ via activation of TYK2 and JAK2. Moreover, TYK2 P1104A is properly docked on cytokine receptors and can be phosphorylated by the proximal JAK, but lacks catalytic activity. Last, we show that the catalytic activity of TYK2 is essential for IL-23, but not IL-12, responses in cells expressing wild-type JAK2. In contrast, the catalytic activity of JAK2 is redundant for both IL-12 and IL-23 responses, because the catalytically inactive P1057A JAK2, which is also docked and phosphorylated, rescues signaling in cells expressing wild-type TYK2. In conclusion, homozygosity for the catalytically inactive P1104A missense variant of TYK2 selectively disrupts the induction of IFN-γ by IL-23 and is a common monogenic etiology of tuberculosis.


Subject(s)
Interferon-gamma/immunology , Interleukin-23/immunology , Mutation, Missense/genetics , TYK2 Kinase/genetics , Tuberculosis/immunology , Cells, Cultured , Homozygote , Humans , Interleukin-23/deficiency , TYK2 Kinase/immunology
3.
Sci Immunol ; 3(24)2018 06 15.
Article in English | MEDLINE | ID: mdl-29907691

ABSTRACT

Heterozygosity for human signal transducer and activator of transcription 3 (STAT3) dominant-negative (DN) mutations underlies an autosomal dominant form of hyper-immunoglobulin E syndrome (HIES). We describe patients with an autosomal recessive form of HIES due to loss-of-function mutations of a previously uncharacterized gene, ZNF341 ZNF341 is a transcription factor that resides in the nucleus, where it binds a specific DNA motif present in various genes, including the STAT3 promoter. The patients' cells have low basal levels of STAT3 mRNA and protein. The autoinduction of STAT3 production, activation, and function by STAT3-activating cytokines is strongly impaired. Like patients with STAT3 DN mutations, ZNF341-deficient patients lack T helper 17 (TH17) cells, have an excess of TH2 cells, and have low memory B cells due to the tight dependence of STAT3 activity on ZNF341 in lymphocytes. Their milder extra-hematopoietic manifestations and stronger inflammatory responses reflect the lower ZNF341 dependence of STAT3 activity in other cell types. Human ZNF341 is essential for the STAT3 transcription-dependent autoinduction and sustained activity of STAT3.


Subject(s)
Gene Expression Regulation/immunology , Job Syndrome/genetics , STAT3 Transcription Factor/genetics , Transcription Factors/genetics , Transcription, Genetic/immunology , Adolescent , Adult , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Nucleus/metabolism , Consanguinity , Cytokines/immunology , Cytokines/metabolism , Exons/genetics , Female , Genes, Recessive/genetics , Genes, Recessive/immunology , Homozygote , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Job Syndrome/blood , Job Syndrome/immunology , Loss of Function Mutation , Lymphocyte Count , Male , Middle Aged , Pedigree , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism , Exome Sequencing , Young Adult , Zinc Fingers/genetics
4.
J Exp Med ; 213(11): 2413-2435, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27647349

ABSTRACT

Combined immunodeficiency (CID) refers to inborn errors of human T cells that also affect B cells because of the T cell deficit or an additional B cell-intrinsic deficit. In this study, we report six patients from three unrelated families with biallelic loss-of-function mutations in RLTPR, the mouse orthologue of which is essential for CD28 signaling. The patients have cutaneous and pulmonary allergy, as well as a variety of bacterial and fungal infectious diseases, including invasive tuberculosis and mucocutaneous candidiasis. Proportions of circulating regulatory T cells and memory CD4+ T cells are reduced. Their CD4+ T cells do not respond to CD28 stimulation. Their CD4+ T cells exhibit a "Th2" cell bias ex vivo and when cultured in vitro, contrasting with the paucity of "Th1," "Th17," and T follicular helper cells. The patients also display few memory B cells and poor antibody responses. This B cell phenotype does not result solely from the T cell deficiency, as the patients' B cells fail to activate NF-κB upon B cell receptor (BCR) stimulation. Human RLTPR deficiency is a CID affecting at least the CD28-responsive pathway in T cells and the BCR-responsive pathway in B cells.


Subject(s)
Alleles , B-Lymphocytes/immunology , Microfilament Proteins/genetics , Mutation/genetics , T-Lymphocytes/immunology , Adolescent , Adult , Base Sequence , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Child , Child, Preschool , Dimerization , Female , HEK293 Cells , Humans , Immunologic Memory , Immunophenotyping , Leukocytes/pathology , Male , NF-kappa B/metabolism , Pedigree , Phenotype , Receptors, Antigen, B-Cell , Signal Transduction , Th17 Cells/immunology , Th2 Cells/immunology , Young Adult
5.
J Exp Med ; 212(10): 1641-62, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26304966

ABSTRACT

Autosomal recessive, complete TYK2 deficiency was previously described in a patient (P1) with intracellular bacterial and viral infections and features of hyper-IgE syndrome (HIES), including atopic dermatitis, high serum IgE levels, and staphylococcal abscesses. We identified seven other TYK2-deficient patients from five families and four different ethnic groups. These patients were homozygous for one of five null mutations, different from that seen in P1. They displayed mycobacterial and/or viral infections, but no HIES. All eight TYK2-deficient patients displayed impaired but not abolished cellular responses to (a) IL-12 and IFN-α/ß, accounting for mycobacterial and viral infections, respectively; (b) IL-23, with normal proportions of circulating IL-17(+) T cells, accounting for their apparent lack of mucocutaneous candidiasis; and (c) IL-10, with no overt clinical consequences, including a lack of inflammatory bowel disease. Cellular responses to IL-21, IL-27, IFN-γ, IL-28/29 (IFN-λ), and leukemia inhibitory factor (LIF) were normal. The leukocytes and fibroblasts of all seven newly identified TYK2-deficient patients, unlike those of P1, responded normally to IL-6, possibly accounting for the lack of HIES in these patients. The expression of exogenous wild-type TYK2 or the silencing of endogenous TYK2 did not rescue IL-6 hyporesponsiveness, suggesting that this phenotype was not a consequence of the TYK2 genotype. The core clinical phenotype of TYK2 deficiency is mycobacterial and/or viral infections, caused by impaired responses to IL-12 and IFN-α/ß. Moreover, impaired IL-6 responses and HIES do not appear to be intrinsic features of TYK2 deficiency in humans.


Subject(s)
Job Syndrome/etiology , TYK2 Kinase/deficiency , Adolescent , Case-Control Studies , Child , Child, Preschool , Female , Humans , Infant , Interferon-gamma/metabolism , Interleukin-10/pharmacology , Interleukin-12/metabolism , Interleukin-12/pharmacology , Interleukin-23/pharmacology , Interleukin-6/pharmacology , Job Syndrome/complications , Job Syndrome/genetics , Leukocytes/drug effects , Leukocytes/metabolism , Male , Mutation , Mycobacterium Infections/etiology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , TYK2 Kinase/genetics , TYK2 Kinase/metabolism , Virus Diseases/etiology , Young Adult
6.
Virus Res ; 165(2): 179-89, 2012 May.
Article in English | MEDLINE | ID: mdl-22374337

ABSTRACT

The potential of Herpesvirus saimiri (HVS) subgroups A, B and C and Herpesvirus ateles (HVA) to transform primary T cells to permanent growth in vitro is restricted by the primate host species and by viral variability represented by distinct viral oncoproteins. We now addressed the relation between the transforming potential of the different viruses and the signaling pathways activated by transiently expressed oncoproteins. Marmoset lymphocytes were transformed by all HVS subgroups as well as HVA, while transformation of human cells was restricted to HVS-C and, unexpectedly, HVA. NF-κB and Src-family kinase (SFK) activity was required for survival of all transformed lymphocytes. Accordingly, NF-κB was induced by oncoproteins of all viruses. In contrast, SFK-related signaling was detectable only for oncoproteins of HVS-C and HVA. Thus, the restricted transformation of human lymphocytes likely correlates with the specific SFK targeting by these oncoproteins. These results will enable further studies into novel SFK effector mechanisms relevant for T-cell proliferation.


Subject(s)
Herpesvirus 2, Saimiriine/pathogenicity , Lymphocyte Activation , Oncogene Proteins/metabolism , Rhadinovirus/pathogenicity , Signal Transduction , Animals , Callithrix , Cells, Cultured , Humans , Primates
7.
J Virol ; 86(3): 1862-73, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22130528

ABSTRACT

Herpesviruses establish latency in suitable cells of the host organism after a primary lytic infection. Subgroup C strains of herpesvirus saimiri (HVS), a primate gamma-2 herpesvirus, are able to transform human and other primate T lymphocytes to stable growth in vitro. The viral genomes persist as nonintegrated, circular, and histone-associated episomes in the nuclei of those latently infected T cells. Epigenetic modifications of episomes are essential to restrict the transcription during latency to selected viral genes, such as the viral oncogenes stpC/tip and the orf73/LANA. In this study, we describe a genome-wide chromatin immunoprecipitation-on-chip (ChIP-on-chip) analysis to profile the occupancy of CTCF on the latent HVS genome. We then focused on two distinct, conserved CTCF binding sites (CBS) within the orf73/LANA promoter region. Analysis of recombinant viruses harboring deletions or mutations within the CBS indicated that the lytic replication of such viruses is not substantially influenced by CTCF. However, T cells latently infected with CBS mutants were impaired in their proliferation abilities and showed a significantly reduced episomal maintenance. We detected a reduced transcription of the orf73/LANA gene in the T cells, corresponding to the reduced viral genomes; this might contribute to the loss of HVS episomes, as LANA is central in the maintenance of viral episomes in the dividing T cell populations. These data demonstrate that the episomal stability of HVS genomes in latently infected human T cells is dependent on CTCF.


Subject(s)
Herpesvirus 2, Saimiriine/genetics , Plasmids/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism , T-Lymphocytes/virology , Viral Proteins/genetics , Virus Latency , Base Sequence , Binding Sites , CCCTC-Binding Factor , Cell Line , Chromatin Immunoprecipitation , Chromosomes, Artificial, Bacterial , DNA Primers , Herpesvirus 2, Saimiriine/physiology , Humans , Open Reading Frames , Polymerase Chain Reaction , Viral Proteins/metabolism
8.
J Biol Chem ; 285(22): 16495-503, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20353939

ABSTRACT

NF-kappaB transcription factors are key regulators of cellular proliferation and frequently contribute to oncogenesis. The herpesviral oncoprotein Tio, which promotes growth transformation of human T cells in a recombinant herpesvirus saimiri background, potently induces canonical NF-kappaB signaling through membrane recruitment of the ubiquitin ligase tumor necrosis factor receptor-associated factor 6 (TRAF6). Here, we show that, in addition to Tio-TRAF6 interaction, the Tio-induced canonical NF-kappaB signal requires the presence of the regulatory subunit of the inhibitor of kappaB kinase (IKK) complex, NF-kappaB essential modulator (NEMO), and the activity of its key kinase, IKKbeta, to up-regulate expression of endogenous cellular inhibitor of apoptosis 2 (cIAP2) and interleukin 8 (IL-8) proteins. Dependent on TRAF6 and NEMO, Tio enhances the expression of the noncanonical NF-kappaB proteins, p100 and RelB. Independent of TRAF6 and NEMO, Tio mediates stabilization of the noncanonical kinase, NF-kappaB-inducing kinase (NIK). Concomitantly, Tio induces efficient processing of the p100 precursor molecule to its active form, p52, as well as DNA binding of nuclear p52 and RelB. In human T cells transformed by infection with a Tio-recombinant virus, sustained expression of p100, RelB, and cIAP2 depends on IKKbeta activity, yet processing to p52 remains largely unaffected by IKKbeta inhibition. However, long term inhibition of IKKbeta disrupts the continuous growth of the transformed cells and induces cell death. Hence, the Tio oncoprotein triggers noncanonical NF-kappaB signaling through NEMO-dependent up-regulation of p100 precursor and RelB, as well as through NEMO-independent generation of p52 effector.


Subject(s)
NF-kappa B/metabolism , Oncogene Proteins, Viral/metabolism , Baculoviral IAP Repeat-Containing 3 Protein , Binding Sites , Cell Proliferation , Humans , I-kappa B Kinase/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Interleukin-8/metabolism , Jurkat Cells , Models, Biological , Oncogene Proteins, Viral/physiology , Signal Transduction , T-Lymphocytes/metabolism , TNF Receptor-Associated Factor 6/metabolism , Transcription Factor RelB/metabolism , Ubiquitin-Protein Ligases/metabolism
9.
Virology ; 388(1): 15-20, 2009 May 25.
Article in English | MEDLINE | ID: mdl-19375768

ABSTRACT

The routine transformation of human CD8(pos) T cells by Herpesvirus saimiri has so far not been achieved in the case of pre-expanded antigen-specific CTLs. Here we transformed 73% of polyclonal EBV-specific CD8(pos) T cell cultures using an optimized culture medium supplemented with IL-2, IL-7, IL-12, and TGF-beta(1). Still, antigen-specific cytotoxicity was frequently lost and analysis of the TCR Vbeta-chain repertoire revealed a variable outgrowth of several initially subdominant populations. Limiting dilution cloning of cells in the presence of high titers of HVS did not result in clonal transformation but in the rapid loss of the viral genome in outgrowing clones. In summary, our data suggest that transformation of CD8(pos) T cells out of bulk cultures can be routinely achieved, while viral transformation itself remains an infrequent event on a per cell basis. The practical use of the improved immortalization of antigen-expanded CD8(pos) T cell lines, however, is limited by the arbitrary outgrowth of subdominant populations of unpredictable specificity.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/virology , Herpesvirus 2, Saimiriine/physiology , Antigens, Viral , CD8-Positive T-Lymphocytes/drug effects , Cell Culture Techniques , Cell Line, Transformed , Cytokines/pharmacology , Cytotoxicity, Immunologic , Genome, Viral , Humans
10.
J Virol ; 80(20): 9934-42, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17005671

ABSTRACT

Lymphoma induction and T-cell transformation by herpesvirus saimiri strain C488 depends on two viral oncoproteins, StpC and Tip. The major interaction partner of Tip is the protein tyrosine kinase Lck, a key regulator of T-cell activation. The Lck binding domain (LBD) of Tip comprises two interaction motifs, a proline-rich SH3 domain-binding sequence (SH3B) and a region with homology to the C terminus of Src family kinase domains (CSKH). In addition, biophysical binding analyses with purified Lck-SH2 domain suggest the phosphorylated tyrosine residue 127 of Tip (pY127) as a potential third Lck interaction site. Here, we addressed the relevance of the individual binding motifs, SH3B, CSKH, and pY127, for Tip-Lck interaction and for human T-cell transformation. Both motifs within the LBD displayed Lck binding activities and cooperated to achieve a highly efficient interaction, while pY127, the major tyrosine phosphorylation site of Tip, did not enhance Lck binding in T cells. Herpesvirus saimiri strain C488 recombinants lacking one or both LBD motifs of Tip lost their transforming potential on human cord blood lymphocytes. Recombinant virus expressing Tip with a mutation at position Y127 was still able to transform human T lymphocytes but, in contrast to wild-type virus, was strictly dependent on exogenous interleukin-2. Thus, the strong Lck binding mediated by cooperation of both LBD motifs was essential for the transformation of human T cells by herpesvirus saimiri C488. The major tyrosine phosphorylation site Y127 of Tip was particularly required for transformation in the absence of exogenous interleukin-2, suggesting its involvement in cytokine signaling pathways.


Subject(s)
Cell Transformation, Viral , Herpesvirus 2, Saimiriine/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Phosphoproteins/metabolism , T-Lymphocytes/virology , Viral Proteins/metabolism , Amino Acid Motifs , Cell Line , Cells, Cultured , Herpesvirus 2, Saimiriine/genetics , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/chemistry , Mutation , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphorylation , Protein Binding , Recombination, Genetic , Sequence Deletion , T-Lymphocytes/cytology , Tyrosine/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics
11.
J Virol ; 79(16): 10507-13, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16051843

ABSTRACT

Human T cells are transformed to antigen-independent permanent growth in vitro upon infection with herpesvirus saimiri subgroup C strains. The viral oncoproteins required for this process, StpC and Tip, could be replaced by Tio, the oncoprotein of herpesvirus ateles. Here we demonstrate that proliferation of lymphocytes transformed with Tio-recombinant herpesvirus saimiri required the activity of Src family kinases. Src kinases had previously been identified as interaction partners of Tio. This interaction was now shown to be independent of any of the four tyrosine residues of Tio but to be dependent on an SH3-binding motif. Mutations within this motif abrogated the transforming capabilities of Tio-recombinant herpesvirus saimiri. Furthermore, kinase interaction resulted in the phosphorylation of Tio on a single tyrosine residue at position 136. Mutation of this residue in the viral context revealed that this phosphorylation site, but none of the other tyrosine residues, was required for T-cell transformation. These data indicate that the interaction of Tio with a Src kinase is essential for both the initiation and the maintenance of T-cell transformation by recombinant herpesvirus saimiri. The requirement for the tyrosine phosphorylation site at position 136 suggests a role for Tio beyond simple deregulation of the kinase.


Subject(s)
Cell Transformation, Viral , Herpesvirus 2, Saimiriine/genetics , Oncogene Proteins, Viral/metabolism , T-Lymphocytes/cytology , Tyrosine/metabolism , Amino Acid Sequence , DNA-Binding Proteins/metabolism , Humans , Molecular Sequence Data , Oncogene Proteins, Viral/chemistry , Phosphorylation , STAT3 Transcription Factor , Trans-Activators/metabolism , src Homology Domains , src-Family Kinases/physiology
12.
J Virol ; 79(9): 5713-20, 2005 May.
Article in English | MEDLINE | ID: mdl-15827186

ABSTRACT

Herpesvirus saimiri (saimirine herpesvirus 2) (HVS), a T-lymphotropic tumor virus, induces lymphoproliferative disease in several species of New World primates. In addition, strains of HVS subgroup C are able to transform T cells of Old World primates, including humans, to permanently growing T-cell lines. In concert with the Stp oncoprotein, the tyrosine kinase-interacting protein (Tip) of HVS C488 is required for T-cell transformation in vitro and lymphoma induction in vivo. Tip was previously shown to interact with the protein tyrosine kinase Lck. Constitutive activation of signal transducers and activators of transcription (STATs) has been associated with oncogenesis and has also been detected in HVS-transformed T-cell lines. Furthermore, Tip contains a putative consensus YXPQ binding motif for the SH2 (src homology 2) domains of STAT1 and STAT3. Tip tyrosine phosphorylation at this site was required for binding of STATs and induction of STAT-dependent transcription. Here we sought to address the relevance of STAT activation for transformation of human T cells by introducing a tyrosine-to-phenylalanine mutation in the YXPQ motif of Tip of HVS C488. Unexpectedly, the recombinant virus was still able to transform human T lymphocytes, but it had lost its capability to activate STAT3 as well as STAT1. This demonstrates that growth transformation by HVS is independent of STAT3 activation.


Subject(s)
Cell Transformation, Viral , DNA-Binding Proteins/metabolism , Herpesvirus 2, Saimiriine/physiology , T-Lymphocytes/virology , Trans-Activators/metabolism , Herpesvirus 2, Saimiriine/genetics , Humans , Mutation , Phosphoproteins/genetics , Phosphoproteins/metabolism , STAT3 Transcription Factor , T-Lymphocytes/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
13.
J Virol ; 78(18): 9814-9, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331715

ABSTRACT

Herpesvirus saimiri group C strains are capable of transforming human and simian T-lymphocyte populations to permanent antigen-independent growth. Two viral oncoproteins, StpC and Tip, that are encoded by a single bicistronic mRNA, act in concert to mediate this phenotype. A closely related New World monkey herpesvirus, herpesvirus ateles, transcribes a single spliced mRNA at an equivalent genome locus. The encoded protein, Tio, has sequence homologies to both StpC and Tip. We inserted the tio sequence of herpesvirus ateles strain 73 into a recombinant herpesvirus saimiri C488 lacking its own stpC/tip oncogene. Simian as well as human T lymphocytes were growth transformed by the chimeric Tio-expressing viruses. Thus, a single herpesvirus protein appears to be responsible for the oncogenic effects of herpesvirus ateles.


Subject(s)
Herpesvirus 2, Saimiriine/pathogenicity , Oncogene Proteins, Viral/physiology , Phosphoproteins/physiology , Rhadinovirus/pathogenicity , T-Lymphocytes/virology , Viral Proteins/physiology , Animals , Base Sequence , Cell Transformation, Viral , Cells, Cultured , DNA, Viral/genetics , Herpesvirus 2, Saimiriine/genetics , Humans , Oncogene Proteins, Viral/genetics , Phosphoproteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rhadinovirus/genetics , Saguinus , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...