Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
J Virol ; 87(1): 282-95, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23077301

ABSTRACT

Herpesvirus saimiri is known to encode a homolog of human complement regulators named complement control protein homolog (CCPH). We have previously reported that this virally encoded inhibitor effectively inactivates complement by supporting factor I-mediated inactivation of complement proteins C3b and C4b (termed cofactor activity), as well as by accelerating the irreversible decay of the classical/lectin and alternative pathway C3 convertases (termed decay-accelerating activity). To fine map its functional sites, in the present study, we have generated a homology model of CCPH and performed substitution mutagenesis of its conserved residues. Functional analyses of 24 substitution mutants of CCPH indicated that (i) amino acids R118 and F144 play a critical role in imparting C3b and C4b cofactor activities, (ii) amino acids R35, K142, and K191 are required for efficient decay of the C3 convertases, (iii) positively charged amino acids of the linker regions, which are dubbed to be critical for functioning in other complement regulators, are not crucial for its function, and (iv) S100K and G110D mutations substantially enhance its decay-accelerating activities without affecting the cofactor activities. Overall, our data point out that ionic interactions form a major component of the binding interface between CCPH and its interacting partners.


Subject(s)
Complement Factor I/genetics , Complement Factor I/metabolism , Herpesvirus 2, Saimiriine/immunology , Viral Proteins/genetics , Viral Proteins/metabolism , Amino Acid Substitution , DNA Mutational Analysis , Herpesvirus 2, Saimiriine/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding
3.
J Immunol ; 187(9): 4392-402, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22013205

ABSTRACT

Herpesvirus Saimiri gene 13 (HVS13) exhibits 57% identity with the predicted sequence of a T cell-derived molecule termed CTLA8. Recombinant HVS13 and CTLA8 stimulate transcriptional factor NF-kappaB activity and Interleukin-6 (IL-6) secretion in fibroblasts, and costimulate T cell proliferation. An HVS13.Fc fusion protein was used to isolate a cDNA encoding a novel receptor that also binds CTLA8. This receptor is unrelated to previously identified cytokine receptor families. A recombinant soluble receptor inhibited T cell proliferation and IL-2 production induced by PHA, concanavalin A (conA), and anti-TCR MAb. These results define CTLA8 and HVS13 as novel cytokines that bind to a novel cytokine receptor. We propose to call these molecules IL-17, vIL-17, and IL-17R, respectively.


Subject(s)
Herpesvirus 2, Saimiriine/immunology , Interleukin-17/history , Receptors, Interleukin-17/history , Repressor Proteins/history , Trans-Activators/history , Amino Acid Sequence , Animals , Aotidae , Base Sequence , Cell Line, Tumor , History, 20th Century , Humans , Mice , Molecular Sequence Data , Protein Binding/immunology , Rats
4.
Hum Immunol ; 72(10): 783-90, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21741423

ABSTRACT

The pathogenic mechanisms responsible for depletion of CD4(+) T cells in aquired immune deficiency syndrome (AIDS) are not fully understood. Systemic immune activation mediated by persistent infection of human immunodeficiency virus (HIV) seems to be one of the predictors of disease progression. We predicted that certain lymphocytes responsible for CD4(+) T-cell depletion could be induced in patients during prolonged activation of lymphocytes. Therefore, we have established an in vitro long-term culture system for peripheral blood mononuclear cells with PHA-P stimulation and Herpesvirus saimiri infection, and examined what types of cells having strong cytotoxic activity to be emerged under the activated conditions. We observed that percentage of CD56(+) T cells was gradually increased in cultures from 30 days after stimulation and exhibited a cytotoxic activity against both autologous and allogeneic targets. Interestingly, HIV-1 infection enhanced the susceptibility of CD4(+) T cells to their cytotoxic effectors, and CD4(+) T cells from HIV-1-infected individuals showed decreased survival rate in the presence of autologous CD56(+) T cells. These findings raised the possibility that induction of autoreactive CD56(+) T cells in consequence of immune activation might be contributed to the depletion of CD4(+) T cells in HIV-1-infected patients.


Subject(s)
Acquired Immunodeficiency Syndrome , CD4-Positive T-Lymphocytes/immunology , CD56 Antigen/immunology , Cell Survival/drug effects , HIV-1/immunology , Lymphocyte Activation/immunology , Lymphocyte Depletion , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/pathology , Autoimmunity/drug effects , CD4-Positive T-Lymphocytes/pathology , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Flow Cytometry , Herpesviridae Infections/immunology , Herpesvirus 2, Saimiriine/immunology , Humans , Immunity, Cellular/drug effects , Lymphocyte Activation/drug effects , Phytohemagglutinins/pharmacology , Polymerase Chain Reaction , Tumor Virus Infections/immunology
5.
Gene Ther ; 17(5): 653-61, 2010 May.
Article in English | MEDLINE | ID: mdl-20164858

ABSTRACT

The rhadinovirus herpesvirus saimiri (HVS) as a gene delivery vector allows large DNA insertions and long-termed gene expression. In the case of T-cell transduction, such vectors use the viral transformation-associated genes of HVS C488 for T-cell amplification. In this report, we investigated whether the gene for the catalytic telomerase subunit human telomerase reverse transcriptase (hTERT) can substitute for the transformation-associated genes in rhadinoviral T-cell transduction and amplification. By using virus mutants generated by en passant mutagenesis from bacterial artificial chromosomes, we observed a very early and functional transgene expression even by virus mutants without transformation-associated genes. The markers of T-cell transformation by HVS, namely CD2 hyperreactivity, overexpression of interleukin-26, and of the tyrosine kinase Lyn could neither be induced nor enhanced by ectopic hTERT expression. When the viral transformation-associated genes were replaced by the hTERT gene, it was not sufficient for growth transformation, although hTERT was efficiently transduced and functionally expressed by the rhadinovirus vector. Thus, the transformation-associated proteins StpC and Tip are responsible for the T-cell phenotype after transduction by HVS and, additionally, modulate telomerase activity independently of hTERT expression.


Subject(s)
Cell Transformation, Viral/genetics , Genetic Vectors , Herpesvirus 2, Saimiriine/genetics , T-Lymphocytes/enzymology , Telomerase/genetics , Transduction, Genetic/methods , CD2 Antigens/immunology , Herpesvirus 2, Saimiriine/immunology , Humans , Interleukins/immunology , Phosphoproteins/genetics , Viral Proteins/genetics , src-Family Kinases/analysis , src-Family Kinases/immunology
6.
J Virol ; 83(1): 273-82, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18945762

ABSTRACT

Herpesvirus saimiri (HVS) establishes a persistent infection in squirrel monkeys by maintaining its episome within T lymphocytes. The product of open reading frame 73 (ORF73) plays a key role in episomal maintenance and is the functional homologue of Epstein-Barr virus EBNA1 and Kaposi's sarcoma-associated herpesvirus LANA1 proteins. There is little sequence homology among these proteins, although all contain a central domain of repeating amino acids. The repeat domains of EBNA1 and LANA1 enhance the stability of these proteins and cause a retardation in self-protein synthesis, leading to poor recognition by CD8(+) cytotoxic T lymphocytes (CTL). The HVS ORF73 repeat domain is composed of a glutamic acid and glycine repeat linked to a glutamic acid and alanine repeat (EG-EA repeat). Here we show that the EG-EA repeat similarly causes a reduction in the recognition of ORF73 by CD8(+) CTL. However, deletion of the EG-EA repeat from HVS ORF73 had no affect on the stability of the protein or its rate of translation. In contrast, the presence of the EG-EA repeat was found to decrease the steady-state levels of ORF73 mRNA. The inhibitory properties of the EG-EA repeat were maintained when transferred to a heterologous protein, and manipulation of the repeat revealed that the motif EEAEEAEEE was sufficient to cause a reduction in recognition of ORF73 by CD8(+) CTL. Thus, the EG-EA repeat of HVS ORF73 plays a role in immune evasion but utilizes a mechanism distinct from that of the EBNA1 and LANA1 repeats.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpesvirus 2, Saimiriine/immunology , Histocompatibility Antigens Class I/immunology , RNA, Messenger/biosynthesis , RNA, Viral/biosynthesis , Viral Proteins/biosynthesis , Viral Proteins/immunology , Cell Line , Humans , Repetitive Sequences, Amino Acid/genetics , Repetitive Sequences, Amino Acid/immunology , Viral Proteins/genetics
8.
Virology ; 326(1): 67-78, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15262496

ABSTRACT

Herpesvirus saimiri (HVS) is a gamma(2)-herpesvirus sharing genomic colinearity and a high degree of functional homology with HHV-8. To begin exploring the correlates of HVS infectivity and neutralization, we designed and implemented a new reporter assay. Using this assay, we could demonstrate that HVS neutralizing antibodies are present at high levels in naturally infected squirrel monkeys and are strongly induced after pathogenic, experimental infection of common marmosets. Further, we demonstrated that viral entry is influenced by cellular glycosaminoglycans and that, similar to HHV-8, soluble heparin is capable of blocking infectivity. We next cloned and characterized the positional homologue of HHV-8 K8.1, HVS Orf51. N-glycosidase F treatment indicates that like K8.1, Orf51 is a glycoprotein. Found in the viral particle, it localizes to the endoplasmic reticulum of expressing cells. Like K8.1, Orf51 could bind to agarose-conjugated heparin, implicating this molecule in viral attachment to cells. These studies provide the groundwork for additional experiments into the role that this protein may be playing in viral pathogenicity, persistence, and cell tropism.


Subject(s)
Antibodies, Viral/blood , Glycoproteins/genetics , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/immunology , Membrane Glycoproteins/genetics , Viral Envelope Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Callithrix , Disease Models, Animal , Glycoproteins/immunology , Glycosaminoglycans/genetics , Glycosaminoglycans/metabolism , Heparin/pharmacology , Herpesviridae Infections/blood , Herpesviridae Infections/immunology , Herpesviridae Infections/veterinary , Herpesvirus 2, Saimiriine/pathogenicity , Membrane Glycoproteins/immunology , Molecular Sequence Data , Neutralization Tests/methods , Open Reading Frames/physiology , Rabbits , Saimiri , Tumor Virus Infections/blood , Tumor Virus Infections/immunology , Tumor Virus Infections/veterinary , Viral Envelope Proteins/immunology , Virulence/drug effects , Virus Replication
9.
Curr Protoc Immunol ; Chapter 7: 7.21.1-7.21.11, 2004 Nov.
Article in English | MEDLINE | ID: mdl-18432932

ABSTRACT

Viral transformation of T cells is an effective method for obtaining large numbers of T cells that are easily maintained in the laboratory. This unit describes a method for generating antigen-independent, virally-transformed T cells using a T-lymphotropic primate gamma-2 herpesvirus, Herpesvirus saimiri (HVS; strain C488). Support protocols for preparing and titrating HSV C488 stocks and testing the functional status of transformed T cells are also included.


Subject(s)
Cell Transformation, Viral , Herpesvirus 2, Saimiriine , T-Lymphocytes , Cell Transformation, Viral/immunology , Herpesvirus 2, Saimiriine/immunology , Humans , Immunologic Techniques , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Virology/methods
10.
J Virol ; 77(23): 12494-506, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14610173

ABSTRACT

Kaposi's sarcoma (KS)-associated herpesvirus (KSHV), a human oncogenic gamma-2-herpesvirus, transforms human endothelial cells and establishes latent infection at a low efficiency in vitro. During latent infection, only a limited number of genes are expressed, and the circularized viral genome is maintained as a multicopy episome. Latency-associated nuclear antigen (LANA), exclusively expressed during latency, has been shown to have a multifunctional role in KS pathogenesis. LANA tethers the viral episome to the host chromosome, thus ensuring efficient persistence of the viral genome during successive rounds of cell division. Besides episome maintenance, LANA modulates the expression of genes of various cellular and viral pathways, including those of retinoblastoma protein and p53. Herpesvirus saimiri (HVS), another gamma-2-herpesvirus, primarily infects New World primates. Orf73, encoding the nuclear antigen of HVS, is the positional homolog of the LANA gene, and the ORF73 protein has some sequence homology to KSHV LANA. However, the function of ORF73 of HVS has not been thoroughly investigated. In this report, we show that HVS ORF73 may be important for episome persistence and colocalizes with the HVS genomic DNA on metaphase chromosomes. Furthermore, HVS terminal repeats (TRs) contain a cis-acting sequence similar to that in KSHV TRs, suggesting that the LANA binding sequence is conserved between these two viruses. This cis-acting element is sufficient to bind HVS ORF73 from strains C488 and A11, and plasmids containing the HVS C488 TR element are maintained and replicate in HVS C488 ORF73-expressing cells.


Subject(s)
Chromosomes , Genome, Viral , Herpesvirus 2, Saimiriine/genetics , Metaphase , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Base Sequence , Callithrix , Cell Line, Transformed , DNA Replication , DNA, Viral/biosynthesis , Fluorescent Antibody Technique , Herpesvirus 2, Saimiriine/immunology , Humans , Molecular Sequence Data , Open Reading Frames/immunology , Sequence Homology, Amino Acid
11.
Cell Immunol ; 223(1): 35-45, 2003 May.
Article in English | MEDLINE | ID: mdl-12914756

ABSTRACT

We previously reported that human interleukin (IL)-2 dependent T cell lines derived from very late antigen (VLA)-1(+) CD45RO(+) peripheral blood (PB) T-cells adhere constitutively to collagen type IV, whereas lines from VLA-1(-) PB lymphocytes (L) adhere weakly. Here we report that the latter are induced to adhere by phorbol 12-myristate 13-acetate (PMA). Both PMA dependent and constitutive adhesion, including that of a Herpes Virus Saimiri (HVS) infected CD4(+)VLA-1(+) clone (HVST) were inhibited by anti-VLA-1 monoclonal antibodies (mAb), by inhibitors of phospholipase C (PLC)gamma and by lovastatin but not by a MEK1 inhibitor, whereas only PMA induced adhesion was blocked by inhibition of protein-kinase (PK) C. Furthermore, lovastatin enhanced PLCgamma and anti VLA-1 mAb blockade, and its effect was not reversed by mevalonic acid (MVA). Lovastatin also inhibited interferon (IFN)gamma secretion by T cells triggered with anti-CD3 and in cells detaching from collagen IV. These results suggest new ways for functional modulation of activated T-cells interacting with collagen.


Subject(s)
Collagen Type IV/immunology , Integrin alpha1beta1/physiology , Lovastatin/pharmacology , Protein Kinase C/metabolism , T-Lymphocytes/immunology , Type C Phospholipases/antagonists & inhibitors , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Adhesion/physiology , Collagen Type IV/physiology , Flavonoids/pharmacology , Flow Cytometry , Herpesvirus 2, Saimiriine/immunology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Integrin alpha1beta1/antagonists & inhibitors , Integrin alpha1beta1/immunology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lovastatin/immunology , MAP Kinase Kinase 1 , Mevalonic Acid/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Phospholipase C gamma , Pravastatin/immunology , Pravastatin/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , Tetradecanoylphorbol Acetate/pharmacology , Type C Phospholipases/immunology , Type C Phospholipases/pharmacology
12.
J Neuroimmunol ; 139(1-2): 119-32, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12799029

ABSTRACT

Human gammadelta T cells are an integral part of the innate immune system and have been difficult to study owing primarily to their relatively low abundance and their fastidious culture properties associated with short in vitro lifespan. Their increased presence within multiple sclerosis (MS) white matter plaques compared to peripheral blood (PB) suggests a specific interaction with central nervous system (CNS) tissues. This fact, together with their innate ability to lyse human oligodendrocytes in culture implicate them possibly in the pathogenesis of MS. To further investigate their potential role in MS, we studied whether gammadelta T cells could be effectively immortalized using Herpesvirus saimiri (HVS), so that they could be studied in longer-term cultures. Effective culture conditions were established resulting in efficient HVS growth transformation of multiple PB and CSF gammadelta T cell lines and clones that could exist in IL-2-dependent culture for periods in excess of 2 years. Phenotypic and functional comparison studies with parental nontransformed gammadelta T cells were performed to characterize the changes that possibly induced by viral transformation. Using panels of transformed gammadelta T cell clones representing discrete gammadelta TcR subtypes, there was no apparent correlation between intracytoplasmic cytokine expression or tumor cell cytotoxicity with a specific TcR. All transformed gammadelta T cells analyzed, regardless of their compartment of origin, strongly expressed intracytoplasmic IFN-gamma and TNF-alpha, but little IL-2 or anti-inflammatory IL-4 or IL-10. These results indicate that HVS transformation of gammadelta T cells can be used to generate lines and clones from both the CSF and PB compartments for further study and elucidation of their potential role in MS pathogenesis.


Subject(s)
Cell Line, Transformed/virology , Genetic Vectors , Herpesvirus 2, Saimiriine/immunology , Multiple Sclerosis/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/virology , Cell Culture Techniques , Cell Line, Transformed/cytology , Cell Line, Transformed/immunology , Clone Cells/drug effects , Clone Cells/immunology , Cytotoxins/pharmacology , Genetic Vectors/immunology , Humans , Interferon-gamma/metabolism , Interleukin-2/pharmacology , Multiple Sclerosis/blood , Multiple Sclerosis/cerebrospinal fluid , Phenotype , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/metabolism
13.
J Gen Virol ; 83(Pt 9): 2269-2278, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12185282

ABSTRACT

The simian herpesvirus saimiri (HVS) induces malignant T cell lymphomas and is closely related to Kaposi's sarcoma-associated herpesvirus (KSHV or HHV-8). Both belong to the gamma-2 herpesvirus subgroup. The viral genome of HVS consists of a unique region (L-DNA) that contains all of the viral genes flanked by non-coding terminal repeats (H-DNA). Here we describe the cloning of a 113 kb restriction fragment containing the L-DNA of an oncogenic HVS strain in an F' replicon-based E. coli vector. Cloned DNA was infectious and the ends of the progeny viral genome consisted of amplified tandem alternating repeats of vector and a single H-DNA unit. T cells infected with these viruses contained the linear DNA typically found a few weeks after infection, but were unable to form episomal circular viral DNA, which is the latent form of the viral genome. Recombinant viruses with reconstructed H-DNA were generated and T cells infected with these rescued viruses contained high copy numbers of episomal DNA. Plasmids expressing the latency-associated nuclear antigen (LANA) and containing various numbers of H-DNA repeats stably replicated as episomes, but constructs containing three repeat units produced the highest copy numbers. These data show that intact and multiple terminal repeats are essential components for episomal replication in latently infected T cells. Moreover, LANA and terminal repeats are sufficient for stable plasmid persistence. Cloned HVS can also be utilized for mutagenesis of HVS and for the expression of foreign genes through efficient manipulation of plasmids in E. coli.


Subject(s)
DNA, Viral/genetics , Herpesvirus 2, Saimiriine/genetics , Nuclear Proteins/genetics , Animals , Antigens, Viral , Cloning, Molecular , DNA, Circular/biosynthesis , DNA, Viral/biosynthesis , DNA, Viral/chemistry , Escherichia coli/genetics , Genetic Vectors , Herpesvirus 2, Saimiriine/immunology , Herpesvirus 2, Saimiriine/physiology , Humans , Leukocytes, Mononuclear/virology , Plasmids , Recombination, Genetic , Terminal Repeat Sequences , Transformation, Genetic , Virus Latency
14.
J Am Vet Med Assoc ; 217(3): 384-7, 341, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10935045

ABSTRACT

A CD8+ T-cell leukemia was diagnosed in an aged female rhesus macaque. Although leukemia and lymphoma in nonhuman primates are commonly associated with simian T-lymphotropic virus, gibbon ape leukemia virus, oncogenic herpesviruses, and types C, D, and E retroviruses, this monkey was not infected with any of these viruses. However, the monkey did have antibodies against herpesvirus saimiri. This likely represents cross-reactivity of the herpesvirus saimiri assay with rhesus monkey rhadinovirus (RRV) antibodies; RRV was first described in rhesus macaques that were identified as having antibodies against herpesvirus saimiri. Rhesus rhadinovirus is a gamma herpesvirus, related antigenically to herpesvirus saimiri and Kaposi's sarcoma-associated herpesvirus (KSHV), which have been linked to lymphoproliferative disorders in primates and humans, respectively. Moreover, an oncogene has been recently identified in the RRV genome that is equivalent in position to the herpesvirus saimiri and KSHV oncogenes. Presently, the association of RRV infection with disease in nonhuman primates is unknown.


Subject(s)
Leukemia, T-Cell/veterinary , Lymphoproliferative Disorders/veterinary , Macaca mulatta , Monkey Diseases/diagnosis , Age Factors , Animals , Antibodies, Viral/blood , Cross Reactions , Diagnosis, Differential , Female , Herpesvirus 2, Saimiriine/immunology , Leukemia, T-Cell/diagnosis , Leukemia, T-Cell/virology , Lymphoproliferative Disorders/diagnosis , Lymphoproliferative Disorders/virology , Monkey Diseases/virology , Rhadinovirus/immunology
15.
J Neuroimmunol ; 103(1): 1-7, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10674983

ABSTRACT

The finite life span of human T lymphocytes and their requirement of regular restimulation frequently limit human T cell studies. Once infected with H. saimiri, however, human and monkey T cells are transformed to stable growth without the need for further restimulation. H. saimiri persists in human growth-transformed T cells episomally and only a few viral genes are expressed. The release of infectious virus from transformed human T cells has not been observed. H. saimiri-transformed T cells have the phenotype of mature activated CD4+ or CD8+ T cells. Transformed T cells retain a structurally and functionally intact T cell receptor and respond specifically to recognition of their antigen. They produce Th1-like cytokines, provide B cell help, can be triggered to become cytotoxic, and are sensitive to a variety of apoptosis-inducing treatments. While H. saimiri-transformed T cells resemble native T cells in numerous aspects, their reactivity to CD2 is strikingly different: Native T cells are activated via CD2 by certain pairs of mAbs, but not by the mere binding of CD2 to its ligand CD58. In contrast, H. saimiri-transformed T cells are activated by a single crosslinked anti-CD2 mAb and also by interaction with CD58-bearing cells.


Subject(s)
Herpesvirus 2, Saimiriine/immunology , Lymphocyte Activation , T-Lymphocytes/immunology , Apoptosis , CD2 Antigens/physiology , Cell Communication , Cells, Cultured , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Humans , Receptors, Antigen, T-Cell/physiology
16.
J Immunol Methods ; 230(1-2): 47-58, 1999 Nov 19.
Article in English | MEDLINE | ID: mdl-10594353

ABSTRACT

Herpesvirus saimiri (HVS), a nonhuman primate gamma herpes virus, was used to immortalize pig-tailed macaque CD4(+) T lymphocytes. The HVS-immortalized T cell lines were used to develop CD4(+) T cell clones from two animals. Three CD4(+) T cell clones were further characterized for the expression of cell surface markers. All expressed CD2, CD4, CD58, CD69 and CD80 and therefore resembled activated T cells. These clones required exogenous IL-2 for efficient growth and were found to be highly susceptible to infection by the challenge virus, Chimeric simian/human immunodeficiency virus (SHIV(KU-1)). They could also be productively infected not only by the quasispecies of the challenge virus (SHIV(KU-1/PDJ) and SHIV(KU-1/PNA), isolated from macaque PDj and PNa, respectively) but also by a different chimeric simian/human immunodeficiency virus (SHIV(89.6P)) and simian immunodeficiency virus (SIV(MAC239)). The virus-infected CD4(+) T cell clones were also used as stimulators for generation of CTL effectors. These effectors exhibited excellent virus-specific lysis in chromium-release assays when syngenic SHIV(KU-1) infected autologous CD4(+) T cell clones were used as targets. The target cell lysis was virus specific, as uninfected control cells showed no or minimal lysis.


Subject(s)
Acquired Immunodeficiency Syndrome/immunology , CD4-Positive T-Lymphocytes/immunology , Herpesvirus 2, Saimiriine/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, CD/metabolism , Antigens, Viral , Cell Transformation, Viral , Chimera/immunology , Clone Cells , Cytotoxicity Tests, Immunologic , Disease Models, Animal , HIV/immunology , HIV/pathogenicity , HIV Antigens , Humans , Interleukin-2/pharmacology , Macaca nemestrina , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity
17.
J Biol Chem ; 274(49): 35119-28, 1999 Dec 03.
Article in English | MEDLINE | ID: mdl-10574994

ABSTRACT

Mature CD4(+) and CD8(+) T lymphocytes are believed to build and express essentially identical surface alphabeta T-cell receptor-CD3 (TCR.CD3) complexes. However, TCR.CD3 expression has been shown to be more impaired in CD8(+) cells than in CD4(+) cells when CD3gamma is absent in humans or mice. We have addressed this paradox by performing a detailed phenotypical and biochemical analysis of the TCR.CD3 complex in human CD3gamma-deficient CD8(+) and CD4(+) T cells. The results indicated that the membrane TCR.CD3 complex of CD8(+) T lymphocytes was conformationally different from that of CD4(+) lymphocytes in the absence of CD3gamma. In addition, CD8(+), but not CD4(+), CD3gamma-deficient T lymphocytes were shown to contain abnormally glycosylated TCRbeta proteins, together with a smaller, abnormal TCR chain (probably incompletely processed TCRalpha). These results suggest the existence of hitherto unrecognized biochemical differences between mature CD4(+) and CD8(+) T lymphocytes in the intracellular control of alphabetaTCR. CD3 assembly, maturation, or transport that are revealed when CD3gamma is absent. Such lineage-specific differences may be important in receptor-coreceptor interactions during antigen recognition.


Subject(s)
CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/chemistry , Receptor-CD3 Complex, Antigen, T-Cell/chemistry , Blotting, Northern , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Calcium/metabolism , Flow Cytometry , Genes, T-Cell Receptor alpha/genetics , Herpesvirus 2, Saimiriine/immunology , Humans , Phenotype , Precipitin Tests , Protein Conformation , Receptor-CD3 Complex, Antigen, T-Cell/genetics , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic
18.
Tissue Antigens ; 52(5): 430-4, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9864032

ABSTRACT

Mucosal intestinal lymphocytes form the first immune-cell line of defense in the intestine. Several methodologies, most of them cumbersome and time consuming, have been used to obtain T-cell clones to unveil their physiological role. In the present work we take advantage of the recently described technique of transformation of T lymphocytes using Herpesvirus saimiri to show that it is possible to immortalize intestinal T-cell lines derived from healthy and diseased colonic samples and thence easily obtain in vitro intestinal T-cell lines as a model for physiopathological studies. Intestinal samples were obtained by colonoscopy and digested with dispase and collagenase. Mucosal lymphocytes (assessed by the expression of the CD3 and CD103 markers) were isolated using a Percoll gradient centrifugation and transformed with Herpesvirus saimiri. Sustained growth was observed 3 months later, showing that the cells were successfully transformed, a finding further confirmed by PCR. All cell lines were CD8+TcRalphabeta+ and HLA-DR+. CD25 was expressed on 1% of Crohn's disease-derived cells and on 25% of cells derived from patients with ulcerative colitis. CD80 expression was found on 80-90% of the cells. These immortal cell lines of intestinal origin may be useful in future experiments aimed at elucidating the role of mucosal lymphocytes in health and disease.


Subject(s)
Herpesvirus 2, Saimiriine/immunology , Intestinal Mucosa/immunology , T-Lymphocytes/immunology , Antigens, CD/immunology , B7-1 Antigen/immunology , B7-2 Antigen , Biomarkers , CD28 Antigens/immunology , Cell Lineage , HLA-DR Antigens/immunology , Inflammatory Bowel Diseases/pathology , Membrane Glycoproteins/immunology , Receptors, Interleukin-2/immunology
19.
J Virol ; 72(8): 6770-6, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9658125

ABSTRACT

The product of open reading frame 14 (orf14) of herpesvirus saimiri (HVS) exhibits significant homology with mouse mammary tumor virus superantigen. orf14 encodes a 50-kDa secreted glycoprotein, as shown previously (Z. Yao, E. Maraskovsky, M. K. Spriggs, J. I. Cohen, R. J. Armitage, and M. R. Alderson, J. Immunol. 156:3260-3266, 1996). orf14 expressed from recombinant baculovirus powerfully induces proliferation of CD4-positive cells originating from several different species. To study the role of orf14 in transformation, a mutant form of HVS (HVS Deltaorf14) was constructed with a deletion in the orf14 gene. The transforming potential of HVS Deltaorf14 was tested in cell culture and in common marmosets. Parental HVS subgroup C strain 488 immortalized common marmoset T lymphocytes in vitro to interleukin-2-independent growth, while the HVS Deltaorf14 mutant did not produce such a growth transformation. In addition, HVS Deltaorf14 was nononcogenic in common marmosets. In contrast to other nononcogenic HVS mutant viruses which were repeatedly isolated from peripheral blood mononuclear cells of infected marmosets for more than 5 months, HVS Deltaorf14 did not persist at a high level in vivo. These results demonstrate that orf14 of HVS is not required for replication but is required for transformation and for high-level persistence in vivo.


Subject(s)
Cell Transformation, Viral , Herpesviridae Infections/virology , Herpesvirus 2, Saimiriine/physiology , Lymphoma/virology , Oncogene Proteins, Viral/physiology , Tumor Virus Infections/virology , Virus Latency , Animals , Aotidae , Callithrix , Cell Division , Cell Line , Disease Models, Animal , Herpesviridae Infections/immunology , Herpesvirus 2, Saimiriine/genetics , Herpesvirus 2, Saimiriine/immunology , Humans , Lymphoma/immunology , Macaca mulatta , Mutagenesis , Oncogene Proteins, Viral/genetics , Open Reading Frames , Rabbits , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Saimiri , Spodoptera , T-Lymphocytes/cytology , Tumor Virus Infections/immunology
20.
J Immunol ; 161(1): 60-4, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9647207

ABSTRACT

Molecular mimicry has been suggested as a mode of autoreactive T cell stimulation in autoimmune diseases. Myelin basic protein (MBP) peptide 1-11 induces experimental autoimmune encephalomyelitis (EAE) in susceptible strains of mice. Here we show that a herpesvirus Saimiri (HVS) peptide, AAQRRPSRPFA, with a limited homology to MBP1-11 peptide, ASQKRPSQRHG (underlined letters showing homology), can stimulate a panel of MBP-11-specific T cell hybridomas and more importantly cause EAE in mice. We demonstrate that this is due to cross-recognition of these two peptides by TCRs. Results presented in this communication are the first demonstration that a viral peptide with homology at just 5 amino acids with a self peptide can induce clinical signs of EAE in mice. These findings have important implications in understanding the breakdown of T cell tolerance to self Ags in autoimmune diseases by means of cross-reactivity with unrelated peptides.


Subject(s)
Antigens, Viral/immunology , Autoantigens/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin Basic Protein/immunology , Peptide Fragments/immunology , Sequence Homology, Amino Acid , Animals , Antigens, Viral/administration & dosage , Antigens, Viral/chemistry , Autoantigens/chemistry , Cross Reactions , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Herpesvirus 2, Saimiriine/immunology , Histocompatibility Antigens Class II/genetics , Hybridomas/immunology , Injections, Subcutaneous , Lymphocyte Activation , Mice , Mice, Inbred Strains , Peptide Fragments/administration & dosage , Peptide Fragments/chemical synthesis , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...