Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
J Immunol ; 164(4): 1689-94, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657611

ABSTRACT

Apoptosis is one of the key regulatory mechanisms in tissue modeling and development. In the thymus, 95-98% of all thymocytes die by apoptosis because they failed to express a TCR with an optimal affinity for the selecting intrathymic peptide-MHC complexes. We studied the possible role of two prominent nerve growth factor (NGF-TNF) family member systems, Fas ligand (FasL)-Fas receptor (FasR) and TNF-alpha-TNFR, in apoptosis of murine CD8+4+ double-positive (DP) thymocytes induced via TCR-CD3- and cAMP-mediated signaling. TCR-CD3epsilon-mediated apoptosis of DP thymocytes was found not to be dependent on either of the two systems. The FasL-FasR system was also found to be dispensable for the cAMP-mediated apoptosis. By contrast, cAMP agonists (dibutyryl-cAMP and forskolin) induced apoptosis via TNF-alpha, as evidenced by 1) the ability of anti-TNF-alpha mAbs to abrogate cAMP analogue-induced DP apoptosis in a dose-dependent manner; and 2) increased resistance of DP thymocytes from TNF-alpha-/- and TNFR I-/-II-/- animals to cAMP agonist-mediated apoptosis. cAMP agonists induced DP thymocyte death by a combination of two mechanisms: first, they induced selective up-regulation of TNF-alpha production, and, second, they sensitized DP thymocytes to TNF-alpha. The latter effect may be due to the down-regulation of TNFR-associated factor 2 protein. These results identify TNF-alpha as the critical mediator of cAMP-induced apoptosis in thymocytes and provide a molecular explanation for how the cAMP stimulators, including the sex steroids, may modulate T cell production output, as observed under physiological and pharmacological conditions.


Subject(s)
Apoptosis/immunology , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cyclic AMP/physiology , T-Lymphocyte Subsets/immunology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis/genetics , Bucladesine/pharmacology , Colforsin/pharmacology , Cyclic AMP/agonists , Dose-Response Relationship, Immunologic , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Knockout , Receptor-CD3 Complex, Antigen, T-Cell/physiology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism
2.
J Immunol ; 164(4): 1695-8, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657612

ABSTRACT

The murine class I molecule H-2Kb and its natural gene conversion variant, H-2Kbm8, which differs from H-2Kb solely at 4 aa at the bottom of the peptide-binding B pocket, are expressed in coisogenic mouse strains C57BL/6 (B6) and B6.C-H-2bm8 (bm8). These two strains provide an excellent opportunity to study the effects of Mhc class I polymorphism on the T cell repertoire. We recently discovered a gain in the antiviral CTL repertoire in bm8 mice as a consequence of the emergence of the Mhc class I allele H-2Kbm8. In this report we sought to determine the mechanism behind the generation of this increased CTL diversity. Our results demonstrate that repertoire diversification occurred by a gain in intrathymic positive selection. As previously shown, the emergence of the same Mhc allele also caused a loss in positive selection of T cell repertoire specific for another Ag, OVA-8. This indicates that a reciprocal loss-and-gain pattern of intrathymic selection exists between H-2Kb and H-2Kbm8. Therefore, in the thymus of an individual, a new Mhc allele can select new T cell specificities, while abandoning some T cell specificities selected by the wild-type allele. A byproduct of this repertoire shift is a net gain of T cell repertoire of the species, which is likely to improve its survival fitness.


Subject(s)
Major Histocompatibility Complex/genetics , Polymorphism, Genetic/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Animals , DNA-Binding Proteins , H-2 Antigens/genetics , H-2 Antigens/immunology , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/genetics , Simplexvirus/immunology , Species Specificity , T-Lymphocytes, Cytotoxic/immunology , Thymus Gland/cytology , Viral Proteins/immunology
3.
Eur J Immunol ; 29(12): 3971-7, 1999 12.
Article in English | MEDLINE | ID: mdl-10602006

ABSTRACT

Cytotoxicity is a major effector function of CD8(+) T cells. Although mitogen-activated protein kinase (MAP kinase) / extracellular regulatory kinase (ERK) activity is indispensable for cytotoxic activity of most CD8(+) T cells a portion of CD8(+) T cells appears resistant to MEK inhibition as cytotoxicity of bulk cultures was partially preserved in the presence of a MEK inhibitor. We have also identified a long-term CD8(+) T cell line with unaltered cytolytic activity after prevention of ERK activation. Antigen-induced microtubule organizing center (MTOC) reorientation was not prevented in this CD8(+) cell line by MEK inhibition, in sharp contrast to the MTOC reorientation prevention in a CD8(+) T cell clone with MEK inhibition-sensitive cytolytic activity. These findings suggest that resistance of lysis to MEK inhibition may be due to a lack of ERK control over MTOC reorientation in some CD8(+) T cells. Thus, there appears to be a heterogeneity of ERK-regulated cytolytic activity in CD8(+) T cells, most likely resulting from a differential control of ERK over MTOC motility.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , MAP Kinase Signaling System/immunology , Mitogen-Activated Protein Kinases/immunology , Signal Transduction/immunology , Animals , CD8 Antigens/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
4.
J Immunol ; 163(8): 4262-8, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10510364

ABSTRACT

After puberty, the thymus undergoes a dramatic loss in volume, in weight and in the number of thymocytes, a phenomenon termed age-associated thymic involution. Recently, it was reported that age-associated thymic involution did not occur in mice expressing a rearranged transgenic (Tg) TCRalphabeta receptor. This finding implied that an age-associated defect in TCR rearrangement was the major, if not the only, cause for thymic involution. Here, we examined thymic involution in three other widely used MHC class I-restricted TCRalphabeta Tg mouse strains and compared it with that in non-Tg mice. In all three TCRalphabeta Tg strains, as in control mice, thymocyte numbers were reduced by approximately 90% between 2 and 24 mo of age. The presence or absence of the selecting MHC molecules did not alter this age-associated cell loss. Our results indicate that the expression of a rearranged TCR alone cannot, by itself, prevent thymic involution. Consequently, other presently unknown factors must also contribute to this phenomenon.


Subject(s)
Aging/immunology , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor/physiology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/physiology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Thymus Gland/physiology , Transgenes/immunology , Aging/genetics , Aging/physiology , Animals , Female , Immunophenotyping , Lymphocyte Count , Major Histocompatibility Complex/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism , Transgenes/physiology
5.
J Immunol ; 163(6): 3286-94, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10477598

ABSTRACT

The Ig superfamily members TCR and B cell receptor (BCR) share high structural and amino acid homology, yet interact with Ags in a distinct manner. The overall shape of the TCR ligand is rather constant, with the variation coming from the MHC polymorphism and the peptide heterogeneity. Consequently, the TCR alpha- and beta-chains form a relatively flat ligand-binding site that interacts with the peptide:MHC (pep:MHC) ligand in a fixed diagonal orientation relative to the MHC alpha-helices, with the alpha- and beta-chains of the TCR contacting the N and C termini of the pep:MHC complex, respectively. By contrast, the shape of BCR ligands varies dramatically, and the BCR exhibits much greater variability of the Ag-binding site. The mAbs 25-D1.16 (D1) and 22-C5.9 (C5), specific for the OVA-8:H-2Kb complex, allowed us to directly compare how TCR and BCR approach the same ligand. To that effect, we mapped D1 and C5 footprints over the OVA-8:H-2Kb complex. Using peptide variants and mutant MHC molecules, we show that the D1 and C5 contacts with the OVA-8:Kb complex C terminus overlap with the TCR beta-chain footprint, but that this footprint also extends to the regions of the molecule not contacted by the TCR. These studies suggest that D1 and C5 exhibit a hybrid mode of pep:MHC recognition, in part similar to that of the TCR beta-chain and in part similar to the conventional anti-MHC Ab.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibody Specificity , H-2 Antigens/immunology , H-2 Antigens/metabolism , Peptide Fragments/immunology , Peptide Fragments/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Antibodies, Monoclonal/chemistry , Arginine/chemistry , Cell Line , Conserved Sequence , Egg Proteins/chemistry , Egg Proteins/immunology , Egg Proteins/metabolism , Epitopes, T-Lymphocyte/chemistry , H-2 Antigens/chemistry , H-2 Antigens/genetics , Immunoglobulin Heavy Chains/chemistry , Ligands , Mice , Mice, Transgenic , Molecular Sequence Data , Ovalbumin/chemistry , Ovalbumin/immunology , Ovalbumin/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Conformation , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Solvents
6.
J Exp Med ; 190(6): 757-64, 1999 Sep 20.
Article in English | MEDLINE | ID: mdl-10499914

ABSTRACT

The majority ( approximately 70%) of postselection CD4(+) single-positive (SP) thymocytes are CD8(lo)CD4(hi). These cells express very low levels of CD8, undetectable by flow cytofluorimetric (FCM) analysis, but sufficiently high to allow purification by panning. Unlike the fully mature CD8(-)CD4(hi) thymocytes, which account for the remaining approximately 30% of the SP CD4(+) thymocytes, CD8(lo)CD4(hi) cells are functionally immature and short-lived unless they receive an unidentified maturation signal from the thymus. In this study, we tested the hypothesis that this signal is provided by a T cell receptor (TCR)-major histocompatibility complex (MHC) class II interaction. Using intrathymic transfer, we show that the immature CD8(lo)CD4(hi) cells could complete their intrathymic maturation and populate the peripheral lymphoid organs in the absence of MHC class II (and class I) molecules. Furthermore, in mice devoid of class II (and class I) molecules, the progeny of CD8(lo)CD4(hi) cells was long-lived and functionally reactive to allogeneic class II molecules, although their numbers in the spleen and the mesenteric lymph node were approximately 40-50% lower than those in class II(+) mice 5 mo after transfer. Control experiments demonstrated that the surviving cells did not originate from the contaminating mature thymocytes. These results demonstrate that the final maturation, proliferation, and peripheral survival (up to 5 mo) of at least some postselection CD4(+) SP cells do not require the TCR-MHC class II interaction. They also indicate that the TCR-MHC class II interaction(s) required for the intrathymic development of long-lived CD4(+) SP cells occurs before the CD4(hi) SP stage of development.


Subject(s)
Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/immunology , Animals , CD4 Antigens/immunology , CD8 Antigens/immunology , Cell Differentiation/immunology , Mice , Mice, Inbred C57BL
7.
J Immunol ; 162(11): 6384-91, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10352251

ABSTRACT

Previous studies from this laboratory have shown that >85% of old mice have stable B cell clonal populations detectable by Ig heavy chain complementary-determining region 3 mRNA size analysis and confirmed by sequence analysis. B cells from the same clone are frequently detected in several lymphoid compartments of the same mouse. We now report the phenotype of all ten stable B cell clonal populations detected in five 20-month-old C57BL/6 mice. These clonal B cells appear to develop in the periphery and nine of the ten B cell clonal populations expressed the CD5 cell surface marker. Stable B cell expansions may be dominated by cells at two stages of differentiation. Some B cell populations were detected with DNA as well as RNA and represent large clonal populations of B cells, detectable in several lymphoid compartments. These populations are found predominantly in B cell populations expressing CD45R/B220 and the mRNA coding for the membrane-bound form of the mu Ig heavy chain, which suggests a predominance of B lymphocytes in these populations. In other cases, smaller clonal populations were detected only in splenic RNA samples. These clonal populations were found predominantly among CD45R/B220- B cells and did not express the membrane-bound form of the micro Ig heavy chain. We offer the hypothesis that the B cell clonal populations present in old mice may be precursors of the two types of B cell neoplasms which are dominated by CD5+ B cells (B cell chronic lymphocytic leukemia) or plasma cells (multiple myeloma).


Subject(s)
Aging/immunology , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , Animals , B-Lymphocyte Subsets/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Lineage/immunology , Cellular Senescence/immunology , Clone Cells/cytology , Clone Cells/immunology , Clone Cells/metabolism , Female , Immunoglobulin mu-Chains/genetics , Leukocyte Common Antigens/biosynthesis , Mice , Mice, Inbred C57BL , Organ Specificity/immunology , Peritoneal Cavity/cytology , RNA, Messenger/biosynthesis , Receptors, Antigen, B-Cell/genetics , Stem Cells/cytology , Stem Cells/immunology
8.
Eur J Immunol ; 29(1): 30-7, 1999 01.
Article in English | MEDLINE | ID: mdl-9933083

ABSTRACT

Following subcutaneous implantation, the murine lymphoma E.G7 [a variant of EL-4, transfected with the chicken ovalbumin (OVA) gene] up-regulates the CD4 molecule. We previously showed that the administration of an anti-CD4 monoclonal antibody (mAb) to EG.7-bearing mice leads to a rapid and complete regression of large established tumors. This tumor regression was shown to require both CD8+ cells and functional Fcgamma receptors (FcgammaR), as it failed to occur in mice depleted of CD8 cells, or mice genetically deficient in FcgammaI/III (gamma-/-mice). Using adoptive transfer, we now show that the FcgammaR+ cells required for this regression are the CD11b+ (phagocytic) cells. Furthermore, experiments using peptide tolerization demonstrated that the critical CD8 CTL population in this model is tumor specific. Analysis of tumors at various stages of regression revealed a massive CD11b+FcgammaR+ and a marginal CD8 infiltration. In the presence of the CTL determinant OVA-8 on tumor cells and of the antitumor mAb, this CD8 infiltration became remarkable, and correlated with tumor regression. These results identify the specific cellular effectors essential for the mAb-mediated tumor regression, and suggest that FcgammaR-activated macrophages induced an expansion of tumor-eliminating CTL in situ.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lymphoma/immunology , Lymphoma/therapy , Animals , CD4 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Chickens , Hematopoietic Stem Cells/immunology , Kinetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Lymphoma/pathology , Macrophage-1 Antigen/metabolism , Mice , Mice, Knockout , Ovalbumin/genetics , Ovalbumin/immunology , Receptors, IgG/genetics , Receptors, IgG/metabolism , T-Lymphocytes, Cytotoxic/immunology , Transfection
9.
J Exp Med ; 188(9): 1553-61, 1998 Nov 02.
Article in English | MEDLINE | ID: mdl-9802967

ABSTRACT

In tumor transplantation models in mice, cytotoxic T lymphocytes (CTLs) are typically the primary effector cells. CTLs recognize major histocompatibility complex (MHC) class I-associated peptides expressed by tumors, leading to tumor rejection. Peptides presented by cancer cells can originate from viral proteins, normal self-proteins regulated during differentiation, or altered proteins derived from genetic alterations. However, many tumor peptides recognized by CTLs are poor immunogens, unable to induce activation and differentiation of effector CTLs. We used MHC binding motifs and the knowledge of class I:peptide:TCR structure to design heteroclitic CTL vaccines that exploit the expression of poorly immunogenic tumor peptides. The in vivo potency of this approach was demonstrated using viral and self-(differentiation) antigens as models. First, a synthetic variant of a viral antigen was expressed as a tumor antigen, and heteroclitic immunization with peptides and DNA was used to protect against tumor challenge and elicit regression of 3-d tumors. Second, a peptide from a relevant self-antigen of the tyrosinase family expressed by melanoma cells was used to design a heteroclitic peptide vaccine that successfully induced tumor protection. These results establish the in vivo applicability of heteroclitic immunization against tumors, including immunity to poorly immunogenic self-proteins.


Subject(s)
Cancer Vaccines/pharmacology , Immunization , Neoplasms, Experimental/immunology , Neoplasms, Experimental/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Antigens, Neoplasm/genetics , Antigens, Viral/genetics , Autoantigens , Base Sequence , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cross Reactions , DNA Primers/genetics , Female , Genetic Engineering , Histocompatibility Antigens Class I , Lymphoma/immunology , Lymphoma/prevention & control , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Polymerase Chain Reaction , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/pharmacology
10.
J Immunol ; 160(6): 2815-23, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9510184

ABSTRACT

The peptide-binding site of the murine MHC class I molecule H-2Kb contains a deep C pocket, that is critical for peptide binding, as it accepts the anchor phenylalanine or tyrosine residue located in the middle (position 5, P5F/Y) of H-2Kb binding peptides. H-2Kb predominantly binds octameric peptides. By both criteria, H-2Kb is unique among the known murine and human class I molecules, none of which have a deep C pocket or preferentially select octamers. We investigated the relative importance of the C pocket in peptide selection and binding by the MHC. An MHC class I H-2Kb variant, Kbw9, predicted to contain no C pocket, was engineered by replacing valine at MHC9 with tryptophan. This mutation drastically altered the selection of peptides bound to Kbw9. The Kbw9 molecule predominantly, if not exclusively, bound nonamers. New peptide anchor residues substituted for the loss of the P5F/Y:C pocket interaction. P3P/Y, which plays an auxiliary role in binding to Kb, assumed the role of a primary anchor, and P5R was selected as a new primary anchor, most likely contacting the E pocket. These experiments demonstrate that the presence of a deep C pocket is responsible for the selection of octameric peptides as the preferred ligands for Kb and provide insight into the adaptation of peptides to a rearranged MHC groove.


Subject(s)
H-2 Antigens/chemistry , Peptides/chemistry , Animals , Binding Sites , Cell Line , H-2 Antigens/genetics , Humans , Mice , Mutagenesis, Site-Directed , Protein Engineering , Structure-Activity Relationship
11.
Int Immunol ; 10(12): 1807-17, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9885901

ABSTRACT

We characterized kinetic and biochemical changes during glucocorticoid (GC)-induced apoptosis of immature CD8+CD4+ double-positive (DP) thymocytes. A GC analog dexamethasone (Dex) induced rapid apoptotic commitment and a transient up-regulation of the NF-kappaB/RelA-p50-binding activity in DP cells. This required an early activation of proteasome, as judged by the ability of a specific proteasomal inhibitor, lactacystine, to delay apoptosis and to suppress Dex-dependent NF-kappaB activation. Dex-induced apoptotic commitment was preceded by the rapid (3 h) cleavage of both a typical caspase substrate, poly(ADP-ribose) polymerase (PARP), and of nuclear transcription factors AP-1, NF-kappaB p50-p50 and NUR-77. By contrast, phorbol myristate acetate (PMA) and/or ionomycin-induced apoptosis had much slower kinetics, were preceded by an early increase of NF-kappaB/RelA-p50, AP-1 and NUR-77 activities, and were insensitive to proteasome inhibition. Both the transgenic Bcl-2 and zVAD-fmk, an inhibitor of caspases, affected all features of Dex-induced apoptosis in a similar fashion, by inhibiting cell death and PARP cleavage, and by stabilizing AP-1, NF-kappaB p50-p50 and NUR-77 levels. Furthermore, Bcl-2 prevented Dex-induced RelA-p50 activation. However, a higher gene dosage of the transgenic Bcl-2 was required for protection against Dex, compared to the PMA and/or ionomycin-induced apoptosis. These findings highlight the unique mechanistic features of GC-induced apoptosis.


Subject(s)
Apoptosis/drug effects , Apoptosis/immunology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Dexamethasone/pharmacology , Dexamethasone/pharmacokinetics , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Caspase Inhibitors , Cysteine Endopeptidases/drug effects , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Female , Ionomycin/pharmacology , Ionophores/pharmacology , Mice , Mice, Inbred C57BL , Multienzyme Complexes/drug effects , Multienzyme Complexes/metabolism , NF-kappa B/metabolism , Proteasome Endopeptidase Complex , Proto-Oncogene Proteins c-bcl-2/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor RelA , Transcription Factors/metabolism , Transgenes , Up-Regulation/drug effects
12.
J Immunol ; 159(8): 3866-74, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9378974

ABSTRACT

The effect of age on the diversity of the murine Ig heavy chain repertoire has been studied in unimmunized C57BL/6 mice. We examined the heterogeneity of complementarity-determining region 3 (CDR3) sizes of Ig mRNA of the IgM and IgG isotypes using two VH families, VHJ558 and VHQ52, which together account for approximately 65% of the Ab repertoire. The broad and bell-shaped profiles representing the diversity of the VHJ558 family in the spleen of 2- to 6-mo-old C57BL/6 mice becomes significantly less diverse after 12 mo of age and by 18 mo of age, single CDR3 sizes that dominate the profiles can be observed in the spleens of > 85% of the mice. Readable sequences have been obtained from 40 dominant mRNA CDR3 size species indicating that they represent clonal populations of B lineage. There are no significant homologies among these sequences. Clones of B lymphocytes that express a dominant CDR3 mRNA species can also be found in the bone marrow, the mesenteric lymph nodes, and the thymus of C57BL/6 mice > 18 mo of age. Some clones of B cells can be detected in only one lymphoid compartment; others are found in two or more compartments. The splenic B cell clones in C57BL/6 mice > 18 mo of age are stable for at least 2 mo. The CDR3 mRNA species that dominate the splenic repertoire of Ig mRNA-expressing cells in vivo do not dominate the repertoire of splenic B cells activated in vitro by bacterial LPS, suggesting that they represent a modest population of B cells expressing high levels of Ig mRNA.


Subject(s)
Aging/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/physiology , Aging/genetics , Animals , Antibody Diversity/genetics , B-Lymphocyte Subsets/metabolism , Bone Marrow Cells/immunology , Clone Cells , Immunoglobulin Variable Region/genetics , Lymph Nodes/cytology , Lymphocyte Activation/genetics , Mesentery , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , Spleen/cytology , Thymus Gland/cytology
13.
J Immunol ; 158(6): 2527-34, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9058783

ABSTRACT

Apoptosis plays an essential role in the development and homeostasis of the immune system. During lymphocyte development, potentially autoreactive cells are eliminated via the activation of a tightly regulated cell death program(s). Similar processes operate in mature lymphocytes, to control the magnitude of the normal immune response by eliminating activated lymphocytes. However, differences in susceptibility to signal-induced apoptosis between immature and mature lymphocytes are numerous. One well-characterized example occurs in response to Ca2+ elevation: peripheral T lymphocytes are resistant, while immature thymocytes are highly susceptible, to Ca2+-mediated cell death (CMCD). In this study, we show that the immunosuppressant cyclosporin A (CsA) primes splenic lymphocytes to undergo CMCD upon ionomycin stimulation. This CsA-induced CMCD affected both T and B lymphocytes. CsA-plug Ca2+-mediated apoptosis was dissected into a two-step process: first, CsA and Ca2+ synergized to induce TGF-beta 1 secretion by B cells; and then TGF-beta 1 and Ca2+ synergistically triggered T and B lymphocyte apoptosis. Together, our results suggest that lymphocyte apoptosis may play a role in CsA-induced immunosuppression via a TGF-beta-dependent mechanism.


Subject(s)
Apoptosis/drug effects , Apoptosis/immunology , Calcium/metabolism , Calcium/physiology , Cyclosporine/pharmacology , Lymphocyte Subsets/drug effects , Transforming Growth Factor beta/physiology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Differentiation/immunology , Drug Synergism , Female , Intracellular Fluid/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/cytology , Transforming Growth Factor beta/metabolism
14.
J Biol Chem ; 272(13): 8558-66, 1997 Mar 28.
Article in English | MEDLINE | ID: mdl-9079686

ABSTRACT

Many signals that cause apoptotic cell death operate by inducing transcription and translation of other (presumably death effector) mediators, and it is well established that stimulus-induced apoptosis can often be blocked by inhibiting transcription and translation. Transcriptional regulation of apoptosis, however, is incompletely understood. To gain insight into nuclear events associated with signal-induced apoptosis during T cell development, we studied signal-induced apoptosis of ex vivo isolated immature CD8(+)4(+) double-positive (DP) thymocytes. Stimuli utilizing the T cell receptor (TCR) signaling pathway or its parts (an alphaCD3/TCR monoclonal antibody, a Ca2+ ionophore, or a protein kinase C-activating phorbol ester) or a stimulus that antagonizes TCR signaling and apoptosis in T cell hybridoma (forskolin, a cyclic AMP-signaling activator) resulted in massive apoptosis of DP thymocytes. At the same time, these stimuli induced qualitatively similar but quantitatively unique patterns of inducible transcription factors (TFs) NF-kappaB/RelA-p50, AP-1 (Fos-Jun), and NUR-77. We focused our attention on the role of AP-1 (Fos-Jun) complex, which was strongly induced by all of the above stimuli and thus was a candidate for a proapoptotic TF. However, we found that AP-1/c-Fos induction was vital in prolonging DP thymocyte life, as judged by increased spontaneous and induced death of DP cells in Fos-/- mice. In direct support of this hypothesis, experiments with antisense oligonucleotides demonstrated that c-Fos plays an essential role in protecting normal DP thymocytes from Ca2+- and cAMP-induced apoptosis but not from TCR-mediated death. Together, these results demonstrate a physiological role for c-Fos in maintaining longevity of DP thymocytes.


Subject(s)
Apoptosis , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Transcription Factors/metabolism , Adaptor Protein Complex alpha Subunits , Adaptor Proteins, Vesicular Transport , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , Mice , NF-kappa B/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1 , Oligonucleotides, Antisense/pharmacology , Receptors, Antigen, T-Cell/metabolism , Receptors, Cytoplasmic and Nuclear , Receptors, Steroid/metabolism , Signal Transduction
15.
Eur J Immunol ; 27(2): 374-82, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9045907

ABSTRACT

We investigated the effector mechanisms operating during the rejection of a transplantable solid lymphoma E.G7 (H-2b) which expresses the gene encoding chicken ovalbumin (OVA). Anti-OVA cytotoxic T lymphocytes (CTL) completely and specifically protected animals from the onset of, but could not eradicate established, E.G7 tumors. The growth of the same lymphoma was also effectively prevented by the antibody GK1.5, whose target molecule, CD4, was expressed on E.G7 cells in vivo. Furthermore, GK1.5 was able to eradicate established solid E.G7 tumors. GK1.5-mediated tumor elimination was due to its antitumor activity, and not to the elimination of regulatory CD4+ cells, based on unimpaired tumor growth in the absence of GK1.5 in animals that genetically lack CD4 T cells. In vitro, GK1.5 did not kill tumor cells: complement activation or apoptosis induction were not evident. In vivo, GK1.5-mediated tumor regression did not depend on natural killer cells, but it absolutely required CD8+ cells and intact Fcgamma receptor. We conclude that, in the E.G7 model, the collaboration of antibody and CTL immunity was crucial for the successful immunotherapy of established tumors. The mechanism of this collaboration is discussed.


Subject(s)
Antibodies, Neoplasm/immunology , CD8 Antigens/analysis , Cytotoxicity, Immunologic/immunology , Graft Rejection/immunology , Lymphoma/immunology , Neoplasm Transplantation/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , CD4 Antigens/immunology , Female , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Ovalbumin/analysis , Ovalbumin/immunology , Tumor Cells, Cultured
16.
Clin Cancer Res ; 3(12 Pt 1): 2191-6, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9815614

ABSTRACT

DNA immunization has been shown to elicit both antibody and CTL responses against antigens expressed by infectious organisms. Because CTL responses have been implicated in rejection of cancer, we investigated whether DNA immunization by particle bombardment using a gene gun could induce CTL responses that were capable of rejecting tumors in mice. DNA immunization by particle bombardment using genes encoding beta-galactosidase and ovalbumin primed mice to generate CTLs in two genetic backgrounds (DBA/2 and C57BL/6 strains, respectively). DNA immunization was more potent in inducing CTLs than immunization with an optimized regimen of ovalbumin peptide plus immune adjuvant. Immunity induced by DNA immunization protected mice against s.c. challenge with tumors expressing the beta-galactosidase antigen. Tumors were rejected even when DNA immunization was started 3 or 7 days after tumor challenge as tumors were becoming established. Tumor rejection required CD8(+) T cells, confirming a role for CTLs in vivo. These studies show that DNA immunization by particle bombardment can efficiently induce CTL responses that are capable of rejecting even established tumors.


Subject(s)
Mast-Cell Sarcoma/therapy , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA , Animals , Biolistics , Cancer Vaccines/administration & dosage , Injections, Subcutaneous , Mast-Cell Sarcoma/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Ovalbumin/genetics , Ovalbumin/immunology , Recombinant Proteins/immunology , Spleen/immunology , Transfection , Tumor Cells, Cultured , Vaccines, DNA/administration & dosage , beta-Galactosidase/genetics , beta-Galactosidase/immunology
17.
Int Immunol ; 9(11): 1701-7, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9418131

ABSTRACT

The immunodominant ovalbumin257-264 (OVA-8, SIINFEKL) and herpes simplex virus gB496-503 (HSV-8, SSIEFARL) peptides share 50% amino acid identity (residues P1, P3, P5 and P8) and bind with comparable efficacy to the murine MHC-encoded class I molecule H-2Kb. However, these two peptides bind differently to H-2Kbm8, a natural H-2Kb variant with a substitution in four amino acids on the floor of the peptide-binding site; HSV-8 binds with high and OVA-8 with a relatively low efficacy. To investigate which of the non-homologous peptide residues were responsible for this differential binding, we used substituted peptide variants and the class I thermodynamic stabilization assay. Variation at the solvent-exposed peptide residues P6 and P7 did not appreciably influence binding. By contrast, variation at the buried P2 and, surprisingly, at the solvent-exposed P4 residue was found to be important. Transplantation of the HSV-8 P2 or P4 residues onto the OVA-8 backbone created variant peptides O2S (P2I-->S) and O4E (P4N-->E) that bound considerably better to H-2Kbm8 than OVA-8. Furthermore, the double-substituted peptide, O2S4E, bound even better, revealing a cooperative effect of the two residues. The reciprocally substituted peptides H2I and H4N, generated by grafting the OVA-8 P2 and P4 residues onto the HSV-8 backbone respectively, bound to H-2Kbm8 slightly worse than HSV-8 but the double-substituted peptide H2I4N bound as poorly as OVA-8. Effects exerted by the P4 residue, which is solvent accessible and therefore available for the TCR contact, demonstrated that exposed peptide residues can, in certain situations, influence not only the TCR contact but also MHC-peptide binding.


Subject(s)
Azides/metabolism , Egg Proteins/metabolism , Histocompatibility Antigens Class I/metabolism , Oligopeptides/metabolism , Ovalbumin/metabolism , Peptide Fragments/metabolism , Animals , Azides/chemistry , Binding Sites , Egg Proteins/chemistry , Glutamic Acid/metabolism , H-2 Antigens/chemistry , H-2 Antigens/metabolism , Histocompatibility Antigens Class I/chemistry , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/metabolism , Mice , Oligopeptides/chemistry , Ovalbumin/chemistry , Peptide Fragments/chemistry , Protein Binding , Protein Structure, Secondary , Receptors, Antigen, T-Cell/metabolism , Sequence Homology, Amino Acid , Solvents
18.
J Exp Med ; 184(1): 253-8, 1996 Jul 01.
Article in English | MEDLINE | ID: mdl-8691139

ABSTRACT

We investigated mechanistic differences in antigen presentation between murine MHC class I variants H-2K(b) and H-2K(bm)8. H-2K(bm)8 differs from H-2K(b) by four residues at the floor of the peptide-binding site, affecting its B pocket which interacts with the second (P2) residue of the peptide. The rest of the molecule, including the T cell receptor (TCR)-contacting residues, is identical to H-2K(b). Due to this variation, CTLs that recognize the ovalbumin 257-264 and HSV gB 498-505 peptides on H-2K(b) cannot recognize them on H-2K(bm)8. This could be due to impaired peptide binding or an altered peptide: K(bm)8 conformation. Peptide binding studies ruled out the first explanation. Molecular modeling indicated that the most obvious consequence of amino acid variation between peptide/H-2K(b) and peptide/H-2K(bm)8 complexes would be a loss of the conserved hydrogen bond network in the B pocket of the latter. This could cause conformational variation of bound peptides. Intermolecular second-site reversion was used to test this hypothesis: P2-substituted OVA and HSV peptides, engineered to restore the hydrogen bond network of the B pocket, were the only ones which restored CTL recognition. These results provide a molecular understanding of peptide/MHC conformational variation.


Subject(s)
Antigen-Presenting Cells/immunology , H-2 Antigens/immunology , Peptides/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Antigens, Viral/chemistry , Antigens, Viral/immunology , Binding Sites , H-2 Antigens/chemistry , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Ovalbumin/chemistry , Ovalbumin/immunology , Peptides/chemistry , Protein Binding , Protein Conformation , Receptors, Antigen, T-Cell/chemistry , Simplexvirus/chemistry , Simplexvirus/immunology , Structure-Activity Relationship
19.
J Immunol ; 156(1): 192-200, 1996 Jan 01.
Article in English | MEDLINE | ID: mdl-8598462

ABSTRACT

Fas-mediated apoptosis is a form of cell death that operates through a receptor-ligand interaction. The FasR has been implicated directly in peripheral T cell tolerance and activation-induced apoptosis of T cells in vitro, although to date its expression on murine peripheral T cells has been characterized incompletely. In this study, we document substantial expression of FasR on the vast majority of recent thymic emigrants and resting peripheral T lymphocytes. FasR ligation can induce death in a minor (approximately 5%) subset of these cells. By contrast to rather slow activation-mediated FasR up-regulation in vitro, we demonstrate that in vivo T cell activation by alpha CD3 mAb or superantigen results in rapid up-regulation of the FasR. This up-regulation is paralleled by the kinetics of activation-induced apoptosis in lymph node T cells. However, we demonstrate that the FasR is not necessary for activation-induced cell death. Lymph node T cells from young, healthy, FasR expression-deficient MRL-Ipr/Ipr and animals could be activated in vivo through the TCR-CD3 complex. Most importantly, MRL-Ipr/Ipr T cells underwent massive activation-induced apoptosis in response to high and intermediate doses of alpha CD3. At a low alpha CD3 dose, however, both MRL-Ipr/Ipr and MRL +/+ T cells were activated similarly, but only the latter underwent adequate apoptosis. Taken together, these findings suggest that in vivo, the Fas pathway may not be the only regulator of activation-induced T cell death, but that this pathway may be critical in regulating responses to weak stimuli.


Subject(s)
Apoptosis/immunology , Lymphocyte Activation , Membrane Glycoproteins/physiology , T-Lymphocytes/immunology , fas Receptor/physiology , Animals , CD3 Complex/immunology , Cell Death/immunology , Cell Movement/immunology , Fas Ligand Protein , Interphase/immunology , Lymph Nodes/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Mutant Strains , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Spleen/cytology , T-Lymphocyte Subsets/immunology , Up-Regulation/immunology
20.
Proc Natl Acad Sci U S A ; 92(24): 11225-9, 1995 Nov 21.
Article in English | MEDLINE | ID: mdl-7479970

ABSTRACT

The Fas/APO-1 cytotoxic pathway plays an important role in the regulation of peripheral immunity. Recent evidence indicates that this regulatory function operates through deletion of activated T and B lymphocytes by CD4+ T cells expressing the Fas ligand. Because macrophages play a key role in peripheral immunity, we asked whether Fas was involved in T-cell-macrophage interactions. Two-color flow cytometry revealed that Fas receptor (FasR) was expressed on resting murine peritoneal macrophages. FasR expression was upregulated after activation of macrophages with cytokines or lipopolysaccharide, although only tumor necrosis factor-alpha rendered macrophages sensitive to anti-FasR antibody-mediated death. To determine the consequence of antigen presentation by macrophages to CD4+ T cells, macrophages were pulsed with antigen and then incubated with either Th1 or Th2 cell lines or clones. Th1, but not Th2, T cells induced lysis of 60-80% of normal macrophages, whereas macrophages obtained from mice with mutations in the FasR were totally resistant to Th1-mediated cytotoxicity. Macrophage cytotoxicity depended upon specific antigen recognition by T cells and was major histocompatibility complex restricted. These findings indicate that, in addition to deletion of activated lymphocytes, Fas plays an important role in deletion of activated macrophages after antigen presentation to Th1 CD4+ T cells. Failure to delete macrophages that constitutively present self-antigens may contribute to the expression of autoimmunity in mice deficient in FasR (lpr) or Fas ligand (gld).


Subject(s)
Macrophages/cytology , Th1 Cells/immunology , fas Receptor/physiology , Animals , Antigen-Presenting Cells/cytology , Cell Death , Cytotoxicity, Immunologic , Immunity, Cellular , Macrophage Activation , Major Histocompatibility Complex , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred CBA , Mice, Mutant Strains , Receptors, Cell Surface/physiology , Th1 Cells/cytology , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...