972 resultados para Peripheral blood human lymphocytes
Resumo:
We recently identified the winged-helix transcription factor Trident and described its expression pattern in synchronized fibroblasts. We have now studied Trident expression in cell lines, differentiating thymocytes and in lymphocytes derived from peripheral blood. During T cell differentiation, expression peaked in the actively dividing immature single positive cells. In peripheral blood lymphocytes, expression of Trident mRNA was absent, but could be induced upon stimulation with mitogens in vitro. These observations imply a function for Trident in dividing lymphocytes.
Resumo:
Antibody-dependent lymphocyte cytotoxicity against human colon carcinoma cells grown in vitro was demonstrated with rabbit anti-carcinoembryonic antigen (CEA) antisera and normal human lymphocytes. The same antisera produced no tumor cell lysis in a complement-dependent cytotoxicity test. The specificity of the reaction was demonstrated by the inhibition of antibody-dependent lymphocyte cytotoxicity after the addition of increasing amounts of purified CEA to the antiserum and by the fact that only tumor cell lines expressing CEA on their surface were lysed. Antibody-dependent lymphocyte cytotoxicity was also observed against two colon carcinoma cell lines that expressed Blood Group A antigen, using a human serum containing anti-Blood Group A antibodies of the immunoglobulin G class. This reaction was specifically inhibited by absorption with Blood Group A red cells, whereas the anti-CEA-dependent cytotoxicity was not inhibited by absorption with red cells of different blood groups.
Resumo:
OBJECTIVE: To determine in chimpanzees if candidate HIV-1 subunit protein vaccines were capable of eliciting long-lasting T-cell memory responses in the absence of viral infection, and to determine the specific characteristics of these responses. DESIGN: A longitudinal study of cell-mediated immune responses induced in three chimpanzees following immunization with subunit envelope glycoproteins of either HIV-1 or herpes simplex virus (HSV)-2. Following these pre-clinical observations, four human volunteers who had been immunized 7 years previously with the same HIV-1 vaccine candidate donated blood for assessment of immune responses. METHODS: Responses were monitored by protein and peptide based ELISpot assays, lymphocyte proliferation, and intracellular cytokine staining. Humoral responses were assessed by enzyme-linked immunosorbent assay and virus neutralization assays. RESULTS: Although antigen (Ag)-specific CD4 T-cell responses persisted for at least 5 years in chimpanzees, CD8 T-cell responses were discordant and declined within 2 years. Detailed cellular analyses revealed that strong Th1 in addition to Th2 type responses were induced by AS2/gp120 and persisted, whereas CD8 T-cell memory declined in peripheral blood. The specificity of both Th and cytotoxic T-lymphocyte responses revealed that the majority of responses were directed to conserved epitopes. The remarkable persistence of Ag-specific CD4 T-cell memory was characterized as a population of the CD45RA-CD62L-CCR7- "effector phenotype" producing the cytokines IFNgamma, IL-2 and IL-4 upon epitope-specific recognition. Importantly, results in chimpanzees were confirmed in peripheral blood of one of four human volunteers studied more than 7 years after immunization. CONCLUSION: These studies demonstrate that epitope-specific Th1 and Th2 cytokine-dependent Th responses can be induced and maintained for longer than 5 years by immunization with subunit proteins of HIV-1.
Resumo:
In chronic viral infections, CD8⁺ T cells become functionally deficient and display multiple molecular alterations. In contrast, only little is known of self- and tumor-specific CD8⁺ T cells from mice and humans. Here we determined molecular profiles of tumor-specific CD8⁺ T cells from melanoma patients. In peripheral blood from patients vaccinated with CpG and the melanoma antigen Melan-A/MART-1 peptide, we found functional effector T cell populations, with only small but nevertheless significant differences in T cells specific for persistent herpesviruses (EBV and CMV). In contrast, Melan-A/MART-1-specific T cells isolated from metastases from patients with melanoma expressed a large variety of genes associated with T cell exhaustion. The identified exhaustion profile revealed extended molecular alterations. Our data demonstrate a remarkable coexistence of effector cells in circulation and exhausted cells in the tumor environment. Functional T cell impairment is mediated by inhibitory receptors and further molecular pathways, which represent potential targets for cancer therapy.
Resumo:
Carcinoembryonic antigen (CEACAM5) is commonly overexpressed in human colon cancer. Several antigenic peptides recognized by cytolytic CD8+ T-cells have been identified and used in colon cancer phase-I vaccination clinical trials. The HLA-A*0201-binding CEA(694-702) peptide was recently isolated from acid eluted MHC-I associated peptides from a human colon tumor cell line. However, the immunogenicity of this peptide in humans remains unknown. We found that the peptide CEA(694-702) binds weakly to HLA-A*0201 molecules and is ineffective at inducing specific CD8+ T-cell responses in healthy donors. Immunogenic-altered peptide ligands with increased affinity for HLA-A*0201 were identified. Importantly, the elicited cytolytic T lymphocyte (CTL) lines and clones cross-reacted with the wild-type CEA(694-702) peptide. Tumor cells expressing CEA were recognized in a peptide and HLA-A*0201 restricted fashion, but high-CEA expression levels appear to be required for CTL recognition. Finally, CEA-specific T-cell precursors could be readily expanded by in vitro stimulation of peripheral blood mononuclear cell (PBMC) from colon cancer patients with altered CEA peptide. However, the CEA-specific CD8+ T-cell clones derived from cancer patients revealed low-functional avidity and impaired tumor-cell recognition. Together, using T-cells to demonstrate the processing and presentation of the peptide CEA694-702, we were able to corroborate its presentation by tumor cells. However, the low avidity of the specific CTLs generated from cancer patients as well as the high-antigen expression levels required for CTL recognition pose serious concerns for the use of CEA694-702 in cancer immunotherapy.
Resumo:
Vitiligo, a skin disorder characterized by the spontaneous destruction of melanocytes, is believed to be of autoimmune origin. We investigated the presence and functionality of CD8(+) T-cells specific for the melanocyte-associated antigens Melan-A, gp100, tyrosinase, and TRP-2 in the blood of HLA-A2(+) vitiligo patients. We enumerated antigen-specific CD8(+) T cells by major histocompatibility complex multimer staining directly ex vivo, as well as after 9 days of in vitro stimulation and assessed IFN-gamma secretion by enzyme-linked immunospot (Elispot) assay. Tyrosinase-, gp100-, or TRP-2-specific CD8(+) T cells could not be identified in the peripheral blood of individuals with vitiligo. Although Melan-A-specific T cells were detectable at levels comparable to Flu-MP-specific T cells by multimer staining, these lymphocytes did not express the skin-homing receptor cutaneous lymphocyte antigen, were phenotypically naïve (CD45RA(+)), and were unresponsive in the IFN-gamma Elispot assay, suggesting that they are unlikely to be involved in the etiopathogenesis of vitiligo.
Resumo:
To determine the phenotype of peripheral blood lymphocytes during the time-course of adjuvant arthritis (AA) to detect alterations that could be involved in the pathogenesis of the arthritic process. METHODS--Phenotype analysis was performed on days 7, 14, 21, 28, 42, 56 and 70 after arthritis induction using monoclonal antibodies to CD5, CD4 and CD8 subsets, and flow cytometry. The proportion of activated lymphocytes and lymphocytes was also assessed with monoclonal antibodies to IL-2R (CD25), to Ia antigen and by polyclonal antibodies to rat Ig. RESULTS--Adjuvant arthritis produced leukocytosis with neutrophilia. Rats with AA showed a marked increase in the number of both CD4+ and CD8+ cells. The ratio CD4/CD8 decreased because the rise in CD8+ cells was more pronounced than the increase in CD4+ cells. Changes in lymphocyte counts showed two well-defined periods: the first, from day 14 to day 28, during which the inflammation of the joints reached a maximum and changes in lymphocyte subsets were more pronounced, that is, there was a threefold increase in CD8+ lymphocytes over normal counts, and the second, from day 42 to day 70, in which modified parameters improved considerably but remained different from controls. CONCLUSION--Alterations were detected in the phenotype of peripheral blood lymphocytes in AA, which provides an additional marker of disease activity.
Resumo:
Many new types of vaccines against infectious or malignant diseases are currently being proposed. Careful characterization of the induced immune response is required in assessing their efficiency. While in most studies human tumor antigen-specific T cells are analyzed after in vitro re-stimulation, we investigated these T cells directly ex vivo using fluorescent tetramers. In peripheral blood lymphocytes from untreated melanoma patients with advanced disease, a fraction of tumor antigen (Melan-A/MART-1)-specific T cells were non-naive, thus revealing tumor-driven immune activation. After immunotherapy with synthetic peptides plus adjuvant, we detected tumor antigen-specific T cells that proliferated and differentiated to memory cells in vivo in some melanoma patients. However, these cells did not present the features of effector cells as found in cytomegalovirus specific T cells analyzed in parallel. Thus, peptide plus adjuvant vaccines can lead to activation and expansion of antigen specific CD8(+) T cells in PBL. Differentiation to protective CD8(+) effector cells may, however, require additional vaccine components that stimulate T cells more efficiently, a major challenge for the development of future immunotherapy.
Resumo:
Pooled F(ab')2 fragments of three MAbs against distinct epitopes of carcinoembryonic antigen (CEA) were used for radioimmunotherapy of nude mice bearing a subcutaneous human colon carcinoma xenograft. 9-10 d after transplantation when tumor nodules were in exponential growth, 36 mice were treated by intravenous injection of different amounts of 131I-labeled MAb F(ab')2. All 14 mice injected with a single dose of 2,200 (n = 10) or 2,800 microCi (n = 4) showed complete tumor remission. 8 of the 10 mice treated with 2,200 microCi survived in good health for 1 yr when they were killed and shown to be tumor free. Four of nine other mice treated with four fractionated doses of 400 microCi showed no tumor relapse for more than 9 mo. In contrast, all 15 mice injected with 1,600-3,000 microCi 131I-control IgG F(ab')2 showed tumor growth retardation of only 1-4 wk, and 15 of 16 mice injected with unlabeled anti-CEA MAb F(ab')2 showed unmodified tumor progression as compared with untreated mice. From tissue radioactivity distributions it was calculated that by an injection of 2,200 microCi 131I-MAb F(ab')2 a mean dose of 8,335 rad was selectively delivered to the tumor, while the tissue-absorbed radiation doses for the normal organs were: peripheral blood, 2,093; stomach, 1,668; kidney, 1,289; lung, 1,185; liver, 617; spleen, 501; small intestine, 427; large intestine, 367; bone, 337; and muscle, 198. These treatments were well tolerated since out of 19 mice with complete tumor remission only 4 required bone marrow transplantation and 17 were in good health for 6-12 mo of observation. The results demonstrate the selective destruction of established human colon carcinoma transplants by intravenous injection of either single or fractionated doses of 131I-MAb F(ab')2.
Resumo:
SUMMARYAs a result of evolution, humans are equipped with an intricate but very effective immune system with multiple defense mechanisms primarily providing protection from infections. This system comprises various cell types, including T-lymphocytes, which are able to recognize and directly kill infected cells. T-cells are not only able to recognize cells carrying foreign antigens, such as virus-infected cells, but also autologous cells. In autoimmune diseases, e.g. multiple sclerosis, T- cells attack autologous cells and cause the destruction of healthy tissue. To prevent aberrant immune reactions, but also to prevent damage caused by an overreacting immune response against foreign targets, there are multiple systems in place that attenuate T-cell responses.By contrast, anti-self immune responses may be highly welcome in malignant diseases. It has been demonstrated that activated T-cells are able to recognize and lyse tumor cells, and may even lead to successful cure of cancer patients. Through vaccination, and especially with the help of powerful adjuvants, frequencies of tumor-reactive T-cells can be augmented drastically. However, the efficacy of anti-tumor responses is diminished by the same checks and balances preventing the human body from harm induced by overly activated T-cells in infections.In the context of my thesis, we studied spontaneous and vaccination induced T-cell responses in melanoma patients. The aim of my studies was to identify situations of T-cell suppression, and pinpoint immune suppressive mechanisms triggered by malignant diseases. We applied recently developed techniques such as multiparameter flow cytometry and gene arrays, allowing the characterization of tumor-reactive T-cells directly ex vivo. In our project, we determined functional capabilities, protein expression, and gene expression profiles of small numbers of T- cells from metastatic tissue and blood obtained from healthy donors and melanoma patients. We found evidence that tumor-specific T-cells were functionally efficient effector cells in peripheral blood, but severely exhausted in metastatic tissue. Our molecular screening revealed the upregulation of multiple inhibitory receptors on tumor-specific T-cells, likely implied in T-cell exhaustion. Functional attenuation of tumor-specific T-cells via inhibitory receptors depended on the anatomical location and immune suppressive mechanisms in the tumor microenvironment, which appeared more important than self-tolerance and anergy mechanisms. Our data reveal novel potential targets for cancer therapy, and contribute to the understanding of cancer biology.RÉSUMÉAu cours de l'évolution, les êtres humains se sont vus doter d'un système immunitaire complexe mais très efficace, avec de multiples mécanismes de défense, principalement contre les infections. Ce système comprend différents types de cellules, dont les lymphocytes Τ qui sont capables de reconnaître et de tuer directement des cellules infectées. Les cellules Τ reconnaissent non seulement des cellules infectées par des virus, mais également des cellules autologues. Dans le cas de maladies auto-immunes, comme par exemple la sclérose en plaques, les cellules Τ s'attaquent à des cellules autologues, ce qui engendre la destruction des tissus sains. Il existe plusieurs systèmes de contrôle des réponses Τ afin de minimiser les réactions immunitaires aberrantes et d'empêcher les dégâts causés par une réponse immunitaire trop importante contre une cible étrangère.Dans le cas de maladies malignes en revanche, une réponse auto-immune peut être avantageuse. Il a été démontré que les lymphocytes Τ étaient également capables de reconnaître et de tuer des cellules tumorales, pouvant même mener à la guérison d'un patient cancéreux. La vaccination peut augmenter fortement la fréquence des cellules Τ réagissant contre une tumeur, particulièrement si elle est combinée avec des adjuvants puissants. Cependant, l'efficacité d'une réponse antitumorale est atténuée par ces mêmes mécanismes de contrôle qui protègent le corps humain des dégâts causés par des cellules Τ activées trop fortement pendant une infection.Dans le cadre de ma recherche de thèse, nous avons étudié les réponses Τ spontanées et induites par la vaccination dans des patients atteints du mélanome. Le but était d'identifier des conditions dans lesquelles les réponses des cellules Τ seraient atténuées, voire inhibées, et d'élucider les mécanismes de suppression immunitaire engendrés par le cancer. Par le biais de techniques nouvelles comprenant la cryométrie de flux et l'analyse globale de l'expression génique à partir d'un nombre minimal de cellules, il nous fut possible de caractériser des cellules Τ réactives contre des tumeurs directement ex vivo. Nous avons examiné les profiles d'expression de gènes et de protéines, ainsi que les capacités fonctionnelles des cellules Τ isolées à partir de tissus métastatiques et à partir du sang de patients. Nos résultats indiquent que les cellules Τ spécifiques aux antigènes tumoraux sont fonctionnelles dans le sang, mais qu'elles sont épuisées dans les tissus métastatiques. Nous avons découvert dans les cellules Τ antitumorales une augmentation de l'expression des récepteurs inhibiteurs probablement impliqués dans l'épuisement de ces lymphocytes T. Cette expression particulière de récepteurs inhibiteurs dépendrait donc de leur localisation anatomique et des mécanismes de suppression existant dans l'environnement immédiat de la tumeur. Nos données révèlent ainsi de nouvelles cibles potentielles pour l'immunothérapie du cancer et contribuent à la compréhension biologique du cancer.
Resumo:
The feline immunodeficiency virus (FIV) targets activated CD4-positive helper T cells preferentially, inducing an AIDS-like immunodeficiency in its natural host species, the domestic cat. The primary receptor for FIV is CD134, a member of the tumor necrosis factor receptor superfamily, and all primary viral strains tested to date use CD134 for infection. We examined the expression of CD134 in the cat using a novel anti-feline CD134 monoclonal antibody (MAb), 7D6, and showed that as in rats and humans, CD134 expression is restricted tightly to CD4+, and not CD8+, T cells, consistent with the selective targeting of these cells by FIV. However, FIV is also macrophage tropic, and in chronic infection the viral tropism broadens to include B cells and CD8+ T cells. Using 7D6, we revealed CD134 expression on a B220-positive (B-cell) population and on cultured macrophages but not peripheral blood monocytes. Moreover, macrophage CD134 expression and FIV infection were enhanced by activation in response to bacterial lipopolysaccharide. Consistent with CD134 expression on human and murine T cells, feline CD134 was abundant on mitogen-stimulated CD4+ T cells, with weaker expression on CD8+ T cells, concordant with the expansion of FIV into CD8+ T cells with progression of the infection. The interaction between FIV and CD134 was probed using MAb 7D6 and soluble CD134 ligand (CD134L), revealing strain-specific differences in sensitivity to both 7D6 and CD134L. Infection with isolates such as PPR and B2542 was inhibited well by both 7D6 and CD134L, suggesting a lower affinity of interaction. In contrast, GL8, CPG, and NCSU were relatively refractory to inhibition by both 7D6 and CD134L and, accordingly, may have a higher-affinity interaction with CD134, permitting infection of cells where CD134 levels are limiting.
Resumo:
We describe herein some immunological properties of human fetal bone cells recently tested for bone tissue-engineering applications. Adult mesenchymal stem cells (MSCs) and osteoblasts were included in the study for comparison. Surface markers involved in bone metabolism and immune recognition were analyzed using flow cytometry before and after differentiation or treatment with cytokines. Immunomodulatory properties were studied on activated peripheral blood mononuclear cells (PBMCs). The immuno-profile of fetal bone cells was further investigated at the gene expression level. Fetal bone cells and adult MSCs were positive for Stro-1, alkaline phosphatase, CD10, CD44, CD54, and beta2-microglobulin, but human leukocyte antigen (HLA)-I and CD80 were less present than on adult osteoblasts. All cells were negative for HLA-II. Treatment with recombinant human interferon gamma increased the presence of HLA-I in adult cells much more than in fetal cells. In the presence of activated PBMCs, fetal cells had antiproliferative effects, although with patterns not always comparable with those of adult MSCs and osteoblasts. Because of the immunological profile, and with their more-differentiated phenotype than of stem cells, fetal bone cells present an interesting potential for allogeneic cell source in tissue-engineering applications.
Resumo:
Recent studies have shown that in humans the germinal center reactions produce three types of V(D)J mutated B cells in similar proportions, i.e. Ig-switched, IgD-IgM+ (IgM-only) and IgD+IgM+ cells, and that together they form the CD27+ compartment of recirculating B cells. We investigated the Ig isotype switch capacity of these cells. Peripheral blood B subsets were sorted and IgG subclass secretion in presence or absence of IL-4 was compared in B cell assays which lead to Ig secretion in all (coculture with EL-4 thymoma cells) or only in CD27+ (CD40L stimulation) B cells. Already switched IgG+ B cells showed no significant sequential switch and IgM-only cells also had a low switch capacity, but IgD+CD27+ switched as much as IgD+CD27- B cells to all IgG subclasses. Thus, in switched B cells some alterations compromising further switch options occur frequently; IgM-only cells may result from aborted switch. However, IgD+CD27+ human B cells, extensively V(D)J mutated and "naive" regarding switch, build up a repertoire of B cells combining (1) novel cross-reactive specificities, (2) increased differentiation capacity (including after T-independent stimulation by Staphylococcus aureus Cowan I) and (3) the capacity to produce appropriate isotypes when they respond to novel pathogens.
Resumo:
Direct identification as well as isolation of antigen-specific T cells became possible since the development of "tetramers" based on avidin-fluorochrome conjugates associated with mono-biotinylated class I MHC-peptide monomeric complexes. In principle, a series of distinct class I MHC-peptide tetramers, each labelled with a different fluorochrome, would allow to simultaneously enumerate as many unique antigen-specific CD8(+) T cells. Practically, however, only phycoerythrin and allophycocyanin conjugated tetramers have been generally available, imposing serious constraints for multiple labeling. To overcome this limitation, we have developed dextramers which are multimers based on a dextran backbone bearing multiple fluorescein and streptavidin moieties. Here we demonstrate the functionality and optimization of these new probes on human CD8(+) T cell clones with four independent antigen specificities. Their applications to the analysis of relatively low frequency antigen-specific T cells in peripheral blood, as well as their use in fluorescence microscopy, are demonstrated. The data show that dextramers produce a stronger signal than their fluoresceinated tetramer counterparts. Thus, these could become the reagents of choice as the antigen-specific T cell labeling transitions from basic research to clinical application.
Resumo:
Antibody-dependent cell-mediated cytotoxicity (ADCC) against human colon carcinoma cells grown in vitro was demonstrated with two specific rabbit anti-carcinoembryonic antigen (cea) antisera. The same antisera did not lyse the colon carcinoma cells in the presence of complement but without lymphocytes. The normal human lymphocytes in the absence of anti-CEA antiserum had a very low cytotoxic activity during the three hours 51Cr release assay used in this study. Two colon carcinoma cell lines, HT-29 and Co-115, expressing CEA on their surface as demonstrated by immunofluorescence, were significantly lysed in the ADCC test, whereas control tumor cell lines, not expressing CEA, were not affected by the anti-CEA sera and the lymphocytes. The specificity of the reaction was further demonstrated by the inhibition of antibody-dependent cell-mediated cytotoxicity after the addition of increasing amounts of purified CEA to the antiserum. The absorption of the anti-CEA antisera was controlled by radioimmunoassay. Absorption of the antisera by normal lung extracts and red cells of different blood groups did not decrease the cytotoxicity.