Oligoclonal Expansion of TCR V beta Subfamily T Cells in Patients with B-ALL.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3840-3840
Author(s):  
Yangqiu Li ◽  
Meijuan Huang ◽  
Yubing Zhou ◽  
Shaohua Chen ◽  
Lijian Yang

Abstract Clonal expansion of TCR V beta subfamily T cells were identified in peripheral blood and tumor infiltrated tissues lymphocytes from some solid tumors and leukemias, which were thought to be relative to tumor associated antigen and be benefited for design of the strategy of specific immunotherapy. The aim of this study is to investigate the situation of TCR V beta gene repertoire and clonal expansion in peripheral blood T cells from patients with B-ALL. The complementarity determining region 3 (CDR3) of TCR V beta 24 subfamily genes in peripheral blood mononuclear cell from 13 cases with untreated B-ALL were amplified using RT-PCR. The PCR products were further labelled by fluorescein and then analyzed by genescan technique for detection of the CDR3 size, to determine the clonality of T cells. The results showed that only 2–18 V beta subfamilies were identified in 13 B-ALL cases respectively. The frequency expression of V beta subfamilies was V beat 13 (84.6%) and V beta 21 (76.9%). No positive products were detected in V beta 4, 6, 8, 18, 20 and V beta 24 subfamilies. Clonal expansive T cells in one or more V beta subfamilies were found in all 13 cases. The display frequency of clonal expansive T cells in V beta 21 (46.1%) and V beta 23 (30.8%) subfamilies were higher than others. In conclusions, the results indicated that the dominant utilization of TCR V beta repertoire could be found in peripheral blood T cells from patients with B-ALL, meanwhile clonal expansive T cells were existed. It may be a feature of the host immune response for leukemia associated antigen. The clonal expansive tendency of T cells in V beta 21 and V beta 23 subfamilies was rather obvious, which maybe correlated with certain malignant B-cell clone. Isolation and amplification of the clonal expanded V beta T cells in vitro may be necessary for identification of the specific cytotoxicity for leukemia cells.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3874-3874
Author(s):  
Bo Li ◽  
Yangqiu Li ◽  
Shaohua Chen ◽  
Lijian Yang ◽  
Jiayu Chen ◽  
...  

Abstract Benzene is a potent human leukemogen, but its mechanism of hematotoxicity is uncertain. It is well know that benzene inhibit T cells proliferation. Several reports revealed that clonal expansion TCR Vβ T cells could be found in workers exposed to benzene. In this study we observe the distribution of TCR Vα gene repertoire and clonal expansion in peripheral blood mononuclear cells from 9 donors and 16 workers exposed to benzene. Complementarity determining region 3 (CDR3) of TCR Vα subfamily genes were amplified using RT-PCR. The PCR products were further analyzed by genescan to evaluating clonality of T cells. 29 Vα subfamily could be detected in 9 donors.1~11 Vα subfamilies were identified in all but one of the workers studied. The most frequently expressed Vα subfamily were Vα3, Vα12 and Vα19 (68.8%), Vα14(56.3%), with a lower expression rate found in Vα5,Vα15, Vα16, Vα 22, Vα 23 and Vα 24 (6.3%). Clonal expansion T cells in one or more Vα subfamily were found in 12 out of all workers studied, including oligoclonal, oligocolonal trend and bioclonal patterns. The frequency of clonal expansion T cells in Vα12, Vα14 and Vα19 subfamilies were higher than others. In conclusion, skewed distribution and clonal expansion of TCR Vα subfamily T cells could be found in workers exposed to benzene. Vα12, Vα14 and Vα19 subfamilies may be highly sensitive to benzene exposed. This is the first report of clonal expansion TCR Vα T cells in the benzene-exposed group. The bias pattern of TCR Vα T cells may be due to the immune cytotocicity from benzene. However, whether the oligoclonality in some Vα subfamilies reflect the phenomenon of clone absense or may be a response clone to benzene-related impairment during exposed to benzene, remains an open question.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3904-3904
Author(s):  
Yangqiu Li ◽  
Ji Tang ◽  
Lijian Yang ◽  
Shaohua Chen ◽  
Yubing Zhou

Abstract The analysis of T cell receptor (TCR) Vβ repertoire is one of the sensitive methods to identify the clonal expansion T cells which response to tumor associated antigens. Understanding the clonality and restricted usage of TCR Vβ repertoire of expanded T-cells induced by PML-RARα peptide may be useful in helping design the new immunotherapeutic strategy specifically for acute promyelocytic leukemia (APL).The aim of the present study was to investigate the specific cytotoxicity and clonality of TCR Vβ repertoire in cord blood T cells induced by PML-RARα peptide (LSSCITQGKAIETQSSSSEE) in vitro. Cord blood mononuclear cells were amplified by IL-2, anti-CD3 and anti-CD28 antibody with different concentration (16.7μg /ml, 33.3μg /ml or 50μg /ml respectively) of synthetic PML-RARα peptide. The induced T cells were collected at different time points after culture (3, 6, 9, 10, 12 or 15 days). The expression and clonality of TCR Vβ subfamilies within induced T cells were analyzed by using RT-PCR and genescan technique. The cytotoxicity of induced T cells was detected by LDH release assay. The results showed that the best condition for T cells induction and amplication was at a concentration with 16.7μg /ml of PML-RARα peptide and at a culture duration with 10 to 15 day. TCR Vβ repertoire analysis showed that restricted expression and cloanl expansion of TCR Vβ subfamily cord blood T cells could be identified after induction by PML-RARα peptide. Clonal expanded T cells were found in Vβ13, Vβ14 and Vβ16 subfamlies respectively. The induced T cells were showed to have the specific cytotoxicity for NB4 cell line (effector cells: tagerted cells=20:1), the cytotoxicity rates were 49.65±6.7% (p<0.05) at day 10th and 73.13±8.42% (p<0.01) at day 15th after culture, which show statistical significance in compare to the control group (without PML-RARα induction). In conclusions, the PML-RARα peptide could induce the clonal expansion T cells from cord blood in vitro, which may have specific cytotoxicity for PML-RARα+ cells.


2008 ◽  
Vol 2008 ◽  
pp. 1-5 ◽  
Author(s):  
E. W. Baars ◽  
H. F. J. Savelkoul

In two in vitro studies, we examined the immunological (pathways of the) effects of Citrus/Cydonia comp. from, respectively, a healthy and an allergic donor; peripheral blood mononuclear cells (PBMCs) were isolated out of peripheral blood and analyzed in vitro after polyclonal stimulation of T-cells. The differentiation capacity and the influence with regard to Th1 (IFN-γ) and Th2 (IL-5) cells were examined. Citrus/Cydonia comp. has a selective effect on the differentiation of T-cells by producing relatively more IL-10 than IL-12. By that, it also seems to have an effect on the induction of regulatory (IL-10 producing) T-cell subsets. It is in vitro capable of neutralizing (to some extent) the changes, characteristic to allergic rhinitis, with regard to the maturation, differentiation, and activity of the immune system. Thus, Citrus/Cydonia comp. can potentially restore the disturbed immune state of rhinitis patients, which essentially could be sufficient to make allergic symptoms disappear permanently.


2021 ◽  
Vol 17 (3) ◽  
pp. 460-470
Author(s):  
Mohd Wajid Ali Khan ◽  

Human Vγ9/Vδ2 T cells (γδ T cells) are immune surveillance cells both in innate and adaptive immunity and are a possible target for anticancer therapies, which can induce immune responses in a variety of cancers. Small non-peptide antigens such as zoledronate can do activation and expansion of T cells in vitro. It is evident that for adoptive cancer therapies, large numbers of functional cells are needed into cancer patients. Hence, optimization of methods needs to be carried out for the efficient expansion of these T cells. Standardization of peripheral blood mononuclear cells (PBMCs) isolation was devised. Cytokines (interleukin 2 (IL-2) and interleukin 15 (IL-15)) and zoledronate were also standardized for different concentrations. It was found that an increased number of PBMCs were recovered when washing was done at 1100 revolution per minute (rpm). Significantly high expansion fold was (2524 ± 787 expansion fold) achieved when stimulation of PBMCs was done with 1 μM of zoledronate and both cytokines IL-2 and IL-15 supported the expansion and survival of cells ISSN 0973-2063 (online) 0973-8894 (print) Bioinformation 17(3): 460-469 (2021) ©Biomedical Informatics (2021) 461 at the concentrations of 100 IU/ml and 10 ng/ml respectively. 14-day cultures showed highly pure (91.6 ± 5.1%) and live (96.5 ± 2.5%) expanded γδ T cells. This study aimed to standardize an easy to manipulate technique for the expansion of γδ T cells, giving a higher yield.


2019 ◽  
Vol 104 (9) ◽  
pp. 4067-4077 ◽  
Author(s):  
Guo Chen ◽  
Yungang Ding ◽  
Qian Li ◽  
Yanbing Li ◽  
Xiaofeng Wen ◽  
...  

Abstract Purpose To investigate the change in IL-10–producing regulatory B cells (Breg), which suppress peripheral immune responses, in patients with thyroid-associated ophthalmopathy (TAO). Methods Peripheral blood mononuclear cells (PBMCs) were isolated from healthy controls (n = 54), patients with Graves disease (n = 26), and patients with TAO (N=125), and stimulated with CpG/CD40L. The frequency of IL-10–producing Bregs and the expression of IL-10 in response to TSH stimulation were measured by flow cytometry. CD4+ T cells were cultured with Breg-depleted PBMCs to elucidate the function of Bregs in patients with TAO. The potential immunoregulatory mechanism was also investigated by Western blot and chromatin immunoprecipitation assays. Results Patients with active TAO had higher baseline levels of Bregs in their peripheral blood than both healthy controls and inactive patients. TSH promoted Bregs. Bregs from patients with TAO were defective in suppressing the activation of interferon (IFN)-γ+ and IL-17+ T cells in vitro. Conclusions Regulatory B cells in patients with TAO are functionally defective, suggesting that the defective Bregs might be responsible for the pathogenesis of TAO.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1659-1659 ◽  
Author(s):  
Sarah M. Larson ◽  
Andy Tu ◽  
Shanta Senadheera ◽  
Michelle Ho ◽  
Donald B. Kohn ◽  
...  

Abstract Background Although significant improvements have been made, patients with relapsed or refractory B-cell malignancies continue to have unfavorable clinical outcomes. We hypothesize that transduction of hematopoietic stem cells (HSCs) with an anti-CD19 Chimeric Antigen Receptor (CAR) will produce a multi-lineage, persistent immunotherapy that can be controlled by the HSVsr39TK suicide gene. Methods First generation anti-CD19 CAR lentiviral constructs containing the HSVsr39TK suicide gene were developed to compare vectors containing the human elongation factor alpha short (EFS) or myeloproliferative sarcoma virus U3 (MNDU3) promoters for transduction efficiency, antigen-specific cytotoxicity and ganciclovir (GCV)-induced cell death in primary human T-cells. The CD28 costimulatory domain was added to the selected construct, and high titer lentiviral vectors were generated to evaluate transduction of human umbilical cord blood (UCB) HSCs for in vitro and in vivo assays. In vitro assays were performed after culture under myeloid differentiation conditions, followed by assessment of phenotype, transduction efficiency, cytotoxic function and GCV-induced cell death. In vivo assays were conducted through transplantation of gene-modified human HSCs into irradiated NSG pups, compared to humanized NSG injected with non-modified human HSCs. Once engraftment was identified, mice from each cohort were further separated into GCV treated and untreated groups. Following GCV administration, mice were harvested to evaluate the presence of human and CAR-modified cells in the bone marrow, spleen and peripheral blood. Results In human primary T cells, the MNDU3 promoter resulted in higher percentage of CAR expressing cells and mean fluorescence intensity compared to the EFS promoter. Cytotoxicity by the transduced T cells against the huCD19+Raji cell line showed similar target cell specific lysis among the constructs. Treatment with GCV effectively decreased the in vitro survival of the cells containing the HSVsr39TK gene compared to the non-transduced and control vector. The construct with MNDU3 promoter was then used with a CD28-containing second-generation anti-CD19 CAR (CCL-MND-αCD19/z/28-sr39). Once transduction efficiency and CAR function were validated in primary human T cells, this vector was used to transduce human UCB CD34+ cells. Following transduction, these cells were evaluated in vitro and in vivo. The cells used for the in vitro studies were cultured under myeloid differentiation conditions. The average number of CAR expressing cells was 45% at the clinically relevant vector copy number of 0.5-1 copies/cell. The myeloid cells transduced with the CCL-MND-αCD19/z/28-sr39 vector demonstrated CD19-specific killing and were eliminated by GCV. In vivo studies demonstrated successful engraftment of transduced HSC with CAR-expressing cells in the different hematopoietic lineages (T, NK, myeloid) detected among human cells in the bone marrow (1.2-15.4%, mean 7.6%), spleen (0.3-15.4%, mean 5.6%), and peripheral blood (0.5-30%, mean 9.2%). Mice engrafted with anti-CD19 CAR-modified HSCs exhibited decreased huCD19+ populations, compared to the mice engrafted with non-modified HSCs. Treatment with GCV resulted in significant decrease in CAR-expressing cells only in the mice transplanted with CD34+ cells transduced with the HSVsr39TK-containing vector. Discussion Here we demonstrate that HSCs can be effectively transduced with an anti-CD19 CAR linked to the HSVsr39TK suicide gene. The CAR was detected in human cells in the bone marrow, spleen and peripheral blood and resulted in decreased B-lineage populations as an index of antigen-specific cytotoxicity; the HSVsr39TK gene conferred sensitivity to ganciclovir which eliminated transduced cells. These results provide pre-clinical support for the use of a CD19 targeted CAR in HSCs for the treatment of B-cell malignancies. Disclosures: Larson: Millenium: Speakers Bureau.


2010 ◽  
Vol 17 (5) ◽  
pp. 556-566 ◽  
Author(s):  
Giovanni Frisullo ◽  
Raffaele Iorio ◽  
Domenico Plantone ◽  
Alessandro Marti ◽  
Viviana Nociti ◽  
...  

Circulating T cells and monocytes expressing T-bet, pSTAT1 and pSTAT3 increase in relapsing–remitting multiple sclerosis (RRMS) during relapse. Natalizumab (NZB) is an effective drug in RRMS, but exacerbation of the disease after its discontinuation has been described in some patients. The aim of this research was to study the effect of NZB treatment on circulating lymphomonocyte subpopulations expressing T-bet, pSTAT1, pSTAT3 and CD4+CD25+Foxp3+ regulatory T cells. Flow cytometry was used to evaluate the percentages of circulating CD4+ and CD8+ T cells, CD14+ monocytes and B cells expressing T-bet, pSTAT1, and pSTAT3, and CD4+CD25+Foxp3+ regulatory T cells from RRMS patients before and after 6–12 NZB infusions. In NZB-treated RRMS patients, the percentages of CD4+pSTAT1+ and CD8+pSTAT1+ T cells, CD14+pSTAT1+ monocytes, CD4+T-bet+, CD8+T-bet+ and CD4+pSTAT3+ T cells and CD14+pSTAT3+ monocytes increased after 12 drug infusions and were similar to those observed in untreated relapsing RRMS patients. Otherwise in vitro NZB exposure of peripheral blood mononuclear cells from untreated RRMS patients and controls had no effect. It was concluded that NZB treatment determines an accumulation of CD4+pSTAT1+, CD8+pSTAT1+, CD4+T-bet+, CD8+T-bet+ and CD4+STAT3+ T cells in peripheral blood that may account for the exacerbation of the disease observed in some patients after the discontinuation of the drug.


2018 ◽  
Vol 10 (437) ◽  
pp. eaar6759 ◽  
Author(s):  
Mohamed Abdel-Mohsen ◽  
Leticia Kuri-Cervantes ◽  
Judith Grau-Exposito ◽  
Adam M. Spivak ◽  
Racheal A. Nell ◽  
...  

The persistence of HIV reservoirs, including latently infected, resting CD4+ T cells, is the major obstacle to cure HIV infection. CD32a expression was recently reported to mark CD4+ T cells harboring a replication-competent HIV reservoir during antiretroviral therapy (ART) suppression. We aimed to determine whether CD32 expression marks HIV latently or transcriptionally active infected CD4+ T cells. Using peripheral blood and lymphoid tissue of ART-treated HIV+ or SIV+ subjects, we found that most of the circulating memory CD32+ CD4+ T cells expressed markers of activation, including CD69, HLA-DR, CD25, CD38, and Ki67, and bore a TH2 phenotype as defined by CXCR3, CCR4, and CCR6. CD32 expression did not selectively enrich for HIV- or SIV-infected CD4+ T cells in peripheral blood or lymphoid tissue; isolated CD32+ resting CD4+ T cells accounted for less than 3% of the total HIV DNA in CD4+ T cells. Cell-associated HIV DNA and RNA loads in CD4+ T cells positively correlated with the frequency of CD32+ CD69+ CD4+ T cells but not with CD32 expression on resting CD4+ T cells. Using RNA fluorescence in situ hybridization, CD32 coexpression with HIV RNA or p24 was detected after in vitro HIV infection (peripheral blood mononuclear cell and tissue) and in vivo within lymph node tissue from HIV-infected individuals. Together, these results indicate that CD32 is not a marker of resting CD4+ T cells or of enriched HIV DNA–positive cells after ART; rather, CD32 is predominately expressed on a subset of activated CD4+ T cells enriched for transcriptionally active HIV after long-term ART.


Sign in / Sign up

Export Citation Format

Share Document