Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.642
Filter
1.
Gene ; 926: 148635, 2024 Oct 30.
Article in English | MEDLINE | ID: mdl-38830518

ABSTRACT

T cells utilized in adoptive T cell immunotherapy are typically activated in vitro. Although these cells demonstrate proliferation and anti-tumor activity following activation, they often face difficulties in sustaining long-term survival post-reinfusion. This issue is attributed to the induction of T cells into a terminal differentiation state upon activation, whereas early-stage differentiated T cells exhibit enhanced proliferation potential and survival capabilities. In previous study, we delineated four T cell subsets at varying stages of differentiation: TN, TSCM, TCM, and TEM, and acquired their miRNA expression profiles via high-throughput sequencing. In the current study, we performed a differential analysis of miRNA across these subsets, identifying a distinct miRNA, hsa-miR-744-5p, characterized by progressively increasing expression levels upon T cell activation. This miRNA is not expressed in TSCM but is notably present in TEM. Target genes of miR-744-5p were predicted, followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses, revealing that these genes predominantly associate with pathways related to the 'Wnt signaling pathway'. We established that miR-744-5p directly targets STK11, influencing its expression. Further, we investigated the implications of miR-744-5p on T cell differentiation and functionality. Overexpression of miR-744-5p in T cells resulted in heightened apoptosis, reduced proliferation, an increased proportion of late-stage differentiated T cells, and elevated secretion of the cytokine TNF-α. Moreover, post-overexpression of miR-744-5p led to a marked decline in the expression of early-stage differentiation-associated genes in T cells (CCR7, CD62L, LEF1, BCL2) and a significant rise in late-stage differentiation-associated genes (KLRG1, PDCD1, GZMB). In conclusion, our findings affirm that miR-744-5p contributes to the progressive differentiation of T cells by downregulating the STK11 gene expression.


Subject(s)
Cell Differentiation , MicroRNAs , Protein Serine-Threonine Kinases , Humans , AMP-Activated Protein Kinase Kinases , Cell Proliferation , Lymphocyte Activation , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Wnt Signaling Pathway
2.
Annu Rev Immunol ; 42(1): 317-345, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38941605

ABSTRACT

Regionalized immune surveillance relies on the concerted efforts of diverse memory T cell populations. Of these, tissue-resident memory T (TRM) cells are strategically positioned in barrier tissues, where they enable efficient frontline defense against infections and cancer. However, the long-term persistence of these cells has been implicated in a variety of immune-mediated pathologies. Consequently, modulating TRM cell populations represents an attractive strategy for novel vaccination and therapeutic interventions against tissue-based diseases. Here, we provide an updated overview of TRM cell heterogeneity and function across tissues and disease states. We discuss mechanisms of TRM cell-mediated immune protection and their potential contributions to autoimmune disorders. Finally, we examine how TRM cell responses might be durably boosted or dampened for therapeutic gain.


Subject(s)
Immunologic Memory , Memory T Cells , Humans , Animals , Memory T Cells/immunology , Memory T Cells/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Organ Specificity/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Immunologic Surveillance
3.
Nat Commun ; 15(1): 5078, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38871720

ABSTRACT

T cell receptor (TCR) signaling regulates important developmental transitions, partly through induction of the E protein antagonist, Id3. Although normal γδ T cell development depends on Id3, Id3 deficiency produces different phenotypes in distinct γδ T cell subsets. Here, we show that Id3 deficiency impairs development of the Vγ3+ subset, while markedly enhancing development of NKγδT cells expressing the invariant Vγ1Vδ6.3 TCR. These effects result from Id3 regulating both the generation of the Vγ1Vδ6.3 TCR and its capacity to support development. Indeed, the Trav15 segment, which encodes the Vδ6.3 TCR subunit, is directly bound by E proteins that control its expression. Once expressed, the Vγ1Vδ6.3 TCR specifies the innate-like NKγδT cell fate, even in progenitors beyond the normally permissive perinatal window, and this is enhanced by Id3-deficiency. These data indicate that the paradoxical behavior of NKγδT cells in Id3-deficient mice is determined by its stereotypic Vγ1Vδ6.3 TCR complex.


Subject(s)
Inhibitor of Differentiation Proteins , Receptors, Antigen, T-Cell, gamma-delta , Animals , Inhibitor of Differentiation Proteins/metabolism , Inhibitor of Differentiation Proteins/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/genetics , Mice , Mice, Knockout , Mice, Inbred C57BL , Cell Differentiation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Signal Transduction
4.
Front Immunol ; 15: 1385380, 2024.
Article in English | MEDLINE | ID: mdl-38827744

ABSTRACT

Introduction: Depending on the microenvironment, γδ T cells may assume characteristics similar to those of Th1, Th2, Th17, regulatory T cells or antigen presenting cells. Despite the wide documentation of the effect of Th1/Th2 balance on pregnancy associated malaria and outcomes, there are no reports on the relationship between γδ T cell phenotype change and Placental Malaria (PM) with pregnancy outcomes. This study sought to investigate the involvement of γδ T cells and its subsets in placental Plasmodium falciparum malaria. Methods: In a case-control study conducted in Yaoundé, Cameroon from March 2022 to May 2023, peripheral, placental and cord blood samples were collected from 50 women at delivery (29 PM negative: PM- and 21 PM positive: PM+; as diagnosed by light microscopy). Hemoglobin levels were measured using hemoglobinometer. PBMCs, IVBMCs and CBMCs were isolated using histopaque-1077 and used to characterize total γδ T cell populations and subsets (Vδ1+, Vδ2+, Vδ1-Vδ2-) by flow cytometry. Results: Placental Plasmodium falciparum infection was associated with significant increase in the frequency of total γδ T cells in IVBMC and of the Vδ1+ subset in PBMC and IVBMC, but decreased frequency of the Vδ2+ subset in PBMC and IVBMC. The expression of the activation marker: HLA-DR, and the exhaustion markers (PD1 and TIM3) within total γδ T cells and subsets were significantly up-regulated in PM+ compared to PM- group. The frequency of total γδ T cells in IVBMC, TIM-3 expression within total γδ T cells and subsets in IVBMC, as well as HLA-DR expression within total γδ T cells and Vδ2+ subset in IVBMC were negatively associated with maternal hemoglobin levels. Furthermore, the frequency of total γδ T cells in PBMC and PD1 expression within the Vδ2+ subset in CBMC were negatively associated with birth weight contrary to the frequency of Vδ1-Vδ2- subset in PBMC and HLA-DR expression within the Vδ2+ subset in IVBMC which positively associated with maternal hemoglobin level and birth weight, respectively. Conclusion: The data indicate up-regulation of activated and exhausted γδ T cells in Plasmodium falciparum placental malaria, with effects on pregnancy outcomes including maternal hemoglobin level and birth weight.


Subject(s)
Malaria, Falciparum , Placenta , Plasmodium falciparum , Pregnancy Complications, Parasitic , Pregnancy Outcome , Receptors, Antigen, T-Cell, gamma-delta , Humans , Female , Pregnancy , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Malaria, Falciparum/blood , Cameroon , Adult , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Plasmodium falciparum/immunology , Pregnancy Complications, Parasitic/immunology , Case-Control Studies , Young Adult , Placenta/immunology , Placenta/parasitology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Phenotype
5.
Crit Rev Immunol ; 44(6): 1-12, 2024.
Article in English | MEDLINE | ID: mdl-38848289

ABSTRACT

Systemic immune-inflammation index (SII) and T cell subsets show involvement in mortality risk in septic patients, and we explored their predictive value in sepsis. Subjects were categorized into the Sepsis (SP)/Septic Shock (SSP)/Septic Shock (SPS) groups. T cell subsets [T-helper (Th)1, Th2, regulatory T cells (Treg), Th17]/platelets (PLT)/neutrophils (NEU)/lymphocytes (LYM)/C-reactive protein (CRP)/procalcitonin (PCT)/interleukin (IL)-4/IL-10/fibrinogen (FIB) were measured by an automatic blood biochemical analyzer/flow cytometry/Countess II FL automatic blood cell analyzer, with SII calculated. The correlations between SII/T cell subsets with Acute Physiology and Chronic Health Evaluation (APACH) II/Sequential Organ Failure Assessment (SOFA) scores and the predictive value of SII/Th1/Th2 for septic diagnosis/prognosis were analyzed using Spearman/ROC curve/Kaplan-Meier. The three groups varied in PLT/NEU/LYM/CRP/PCT/IL-4/IL-10/FIB levels and APACH II/SOFA scores. Compared with the SP group, the other two groups showed elevated APACH II/SOFA scores and SII/Th1/Th2/Th17/Treg levels. SII/Th1/Th2 levels significantly positively correlated with APACH II/SOFA scores. SII/Th1/Th2 levels had high predictive value for septic diagnosis/prognosis, with their combination exhibiting higher predictive value. Septic patients with high SII/Th1/Th2 levels exhibited lower survival rates. Altogether, SII, Th1, and Th2 had good predictive value for the diagnosis and prognosis of patients with varying severity of sepsis, with their high levels increasing mortality in septic patients.


Subject(s)
Sepsis , Severity of Illness Index , T-Lymphocyte Subsets , Humans , Sepsis/diagnosis , Sepsis/immunology , Sepsis/mortality , Sepsis/blood , Prognosis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Inflammation/immunology , Inflammation/diagnosis , Male , Female , Middle Aged , Aged , Biomarkers/blood
6.
Front Immunol ; 15: 1400550, 2024.
Article in English | MEDLINE | ID: mdl-38835756

ABSTRACT

The dog is an important companion animal and also serves as model species for human diseases. Given the central role of T cells in immune responses, a basic understanding of canine conventional T cell receptor (TCR)αß+ T cells, comprising CD4+ single-positive (sp) T helper (Th) and CD8α+ sp cytotoxic T cell subsets, is available. However, characterization of canine non-conventional TCRαß+ CD4+CD8α+ double-positive (dp) and TCRαß+ CD4-CD8α- double-negative (dn) T cells is limited. In this study, we performed a comprehensive analysis of canine dp and dn T cells in comparison with their conventional counterparts. TCRαß+ T cells from peripheral blood of healthy dogs were sorted according to their CD4/CD8α phenotype into four populations (i.e. CD4+ sp, CD8α+ sp, dp, and dn) and selected surface markers, transcription factors and effector molecules were analyzed ex vivo and after in vitro stimulation by RT-qPCR. Novel characteristics of canine dp T cells were identified, expanding the previously characterized Th1-like phenotype to Th17-like and Th2-like properties. Overall, mRNA expression of various Th cell-associated cytokines (i.e. IFNG, IL17A, IL4, IL13) in dp T cells upon stimulation highlights their versatile immunological potential. Furthermore, we demonstrated that the CD4-CD8α- dn phenotype is stable during in vitro stimulation. Strikingly, dn T cells were found to express highest mRNA levels of type 2 effector cytokines (IL4, IL5, and IL13) upon stimulation. Their strong ability to produce IL-4 was confirmed at the protein level. Upon stimulation, the percentage of IL-4-producing cells was even higher in the non-conventional dn than in the conventional CD4+ sp population. Constitutive transcription of IL1RL1 (encoding IL-33Rα) further supports Th2-like properties within the dn T cell population. These data point to a role of dn T cells in type 2 immunity. In addition, the high potential of dn T cells to transcribe the gene encoding the co-inhibitory receptor CTLA-4 and to produce the inhibitory cytokine IL-10 indicates putative immunosuppressive capacity of this population. In summary, this study reveals important novel aspects of canine non-conventional T cells providing the basis for further studies on their effector and/or regulatory functions to elucidate their role in health and disease.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta , Th2 Cells , Animals , Dogs , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Th2 Cells/immunology , CD8 Antigens/metabolism , CD8 Antigens/immunology , Cytokines/metabolism , CD4 Antigens/metabolism , CD4 Antigens/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Immunophenotyping , Male
7.
Front Immunol ; 15: 1325356, 2024.
Article in English | MEDLINE | ID: mdl-38835766

ABSTRACT

Introduction: Circulating T follicular helper (cTfh) cells and circulating T peripheral helper (cTph) cells (which share common characteristics with the cTfh population) are implicated in the pathogenesis of immune-mediated and autoimmune diseases such as psoriasis (Ps). Their close interplay with the interleukin 17 (IL-17) axis and the ex vivo effect of IL-17-targeting biologic agents used to treat Ps on them are elusive. This study aimed to investigate the effect of biologics targeting IL-17 on cTfh and cTph cell subpopulations isolated from the blood of patients with Ps. Methods: Peripheral blood mononuclear cells (PBMCs) were isolated from patients with Ps at treatment initiation and three months later. Samples were also collected from controls. Cells were stained using monoclonal antibodies. Flow cytometry assessed the fraction of cTfh (CD3+CD4+CXCR5+) and cTph (CD3+CD4+CXCR5-PD-1hi) cells.. Results: Flow cytometric analysis showed increased fractions of activated cTfh subsets including ICOS+ and ICOS+PD-1+ expressing cells, in patients compared to controls. Biologic blocking of IL-17A diminished the cTfh population. Furthermore, ICOS+ and ICOS+PD-1+ sub-populations were also inhibited. Finally, the cTph cell fraction significantly decreased after three months of successful treatment with biologics. Conclusion: Early anti-IL-17-mediated clinical remission in Ps is associated with decreased cTfh and cTph cell subpopulations.


Subject(s)
Biological Products , Interleukin-17 , Psoriasis , Humans , Psoriasis/immunology , Psoriasis/drug therapy , Male , Female , Interleukin-17/metabolism , Interleukin-17/antagonists & inhibitors , Adult , Middle Aged , Biological Products/therapeutic use , Biological Products/pharmacology , T Follicular Helper Cells/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/drug effects
8.
J Clin Immunol ; 44(6): 139, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38822857

ABSTRACT

We evaluated the impact of early recovery of mucosal-associated invariant T cells (MAIT) and gamma-delta (γδ) T cells, especially Vδ2+ T cells, on the clinical outcomes of 76 patients who underwent allogeneic hematopoietic cell transplantation (allo-HCT). MAIT cells were identified at day 20-30 post-transplant using flow cytometry and defined as CD3+ TCRVα7.2+CD161+. Two subsets of Vδ2+ T cells were analyzed according to the expression of CD26. The cytotoxicity profile of MAIT and Vδ2+ T cells was analyzed according to the intracellular expression of perforin and granzyme B, and intracellular IFN-γ was evaluated after in vitro activation. CD26+Vδ2+ T cells displayed higher intracellular levels of IFN-γ, whereas CD26- Vδ2+ T were found to be more cytotoxic. Moreover, MAIT cell frequency was correlated with the frequency of Vδ2+ T cells with a better correlation observed with Vδ2+CD26+ than with the Vδ2+CD26- T cell subset. By using the composite endpoint graft-versus-host disease (GvHD)-free, relapse-free survival (GRFS) as the primary endpoint, we found that patients with a higher MAIT cell frequency at day 20-30 after allo-HCT had a significantly increased GRFS and a better overall survival (OS) and disease-free survival (DFS). Moreover, patients with a low CD69 expression by MAIT cells had an increased cumulative incidence of grade 2-4 acute GvHD (aGvHD). These results suggest that MAIT cell reconstitution may provide mitigating effects early after allo-HCT depending on their activation markers and functional status. Patients with a high frequency of Vδ2+CD26+ T cells had a significantly higher GRFS, OS and DFS, but there was no impact on cumulative incidence of grade 2-4 aGVHD, non-relapse mortality and relapse. These results revealed that the impact of Vδ2+ T cells on the success of allo-HCT may vary according to the frequency of the CD26+ subset.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Mucosal-Associated Invariant T Cells , Transplantation, Homologous , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Male , Female , Adult , Middle Aged , Graft vs Host Disease/immunology , Graft vs Host Disease/etiology , Mucosal-Associated Invariant T Cells/immunology , Young Adult , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adolescent , Aged , Treatment Outcome , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Dipeptidyl Peptidase 4/metabolism , Cytotoxicity, Immunologic
9.
Oncoimmunology ; 13(1): 2355684, 2024.
Article in English | MEDLINE | ID: mdl-38798746

ABSTRACT

Identifying tumor-relevant T cell subsets in the peripheral blood (PB) has become a potential strategy for cancer treatment. However, the subset of PB that could be used to treat cancer remains poorly defined. Here, we found that the CX3CR1+ T cell subset in the blood of patients with lung cancer exhibited effector properties and had a higher TCR matching ratio with tumor-infiltrating lymphocytes (TILs) compared to CX3CR1- T cells, as determined by paired single-cell RNA and TCR sequencing. Meanwhile, the anti-tumor activities, effector cytokine production, and mitochondrial function were enhanced in CX3CR1+ T cells both in vitro and in vivo. However, in the co-culture system of H322 cells with T cells, the percentages of apoptotic cells and Fas were substantially higher in CX3CR1+ T cells than those in CX3CR1- T cells. Fas-mediated apoptosis was rescued by treatment with an anti-PD-1 antibody. Accordingly, the combination of adoptive transfer of CX3CR1+ T cells and anti-PD-1 treatment considerably decreased Fas expression and improved the survival of lung xenograft mice. Moreover, an increased frequency of CX3CR1+ T cells in the PB correlated with a better response and prolonged survival of patients with lung cancer who received anti-PD-1 therapy. These findings indicate the promising potential of adoptive transfer of peripheral CX3CR1+ T cells as an individual cancer immunotherapy.


Subject(s)
CX3C Chemokine Receptor 1 , Immune Checkpoint Inhibitors , Lung Neoplasms , Lymphocytes, Tumor-Infiltrating , Programmed Cell Death 1 Receptor , Animals , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , CX3C Chemokine Receptor 1/metabolism , Humans , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/metabolism , Xenograft Model Antitumor Assays , Cell Line, Tumor , Female , Apoptosis/drug effects , Male , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
10.
Front Immunol ; 15: 1369202, 2024.
Article in English | MEDLINE | ID: mdl-38774876

ABSTRACT

Atherosclerosis poses a significant threat to human health, impacting overall well-being and imposing substantial financial burdens. Current treatment strategies mainly focus on managing low-density lipids (LDL) and optimizing liver functions. However, it's crucial to recognize that Atherosclerosis involves more than just lipid accumulation; it entails a complex interplay of immune responses. Research highlights the pivotal role of lipid-laden macrophages in the formation of atherosclerotic plaques. These macrophages attract lymphocytes like CD4 and CD8 to the inflamed site, potentially intensifying the inflammatory response. γδ T lymphocytes, with their diverse functions in innate and adaptive immune responses, pathogen defense, antigen presentation, and inflammation regulation, have been implicated in the early stages of Atherosclerosis. However, our understanding of the roles of γδ T cells in Atherosclerosis remains limited. This mini-review aims to shed light on the characteristics and functions of γδ T cells in Atherosclerosis. By gaining insights into the roles of γδ T cells, we may uncover a promising strategy to mitigate plaque buildup and dampen the inflammatory response, thereby opening new avenues for effectively managing this condition.


Subject(s)
Atherosclerosis , Receptors, Antigen, T-Cell, gamma-delta , Humans , Atherosclerosis/immunology , Atherosclerosis/metabolism , Animals , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Macrophages/immunology , Macrophages/metabolism , Plaque, Atherosclerotic/immunology , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Immunity, Innate , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Inflammation/immunology , Adaptive Immunity
11.
Front Immunol ; 15: 1378040, 2024.
Article in English | MEDLINE | ID: mdl-38698866

ABSTRACT

Background: Interleukin-17-producing CD4 T cells contribute to the control of Mycobacterium tuberculosis (Mtb) infection in humans; whether infection with human immunodeficiency virus (HIV) disproportionately affects distinct Th17-cell subsets that respond to Mtb is incompletely defined. Methods: We performed high-definition characterization of circulating Mtb-specific Th17 cells by spectral flow cytometry in people with latent TB and treated HIV (HIV-ART). We also measured kynurenine pathway activity by liquid chromatography-mass spectrometry (LC/MS) on plasma and tested the hypothesis that tryptophan catabolism influences Th17-cell frequencies in this context. Results: We identified two subsets of Th17 cells: subset 1 defined as CD4+Vα7.2-CD161+CD26+and subset 2 defined as CD4+Vα7.2-CCR6+CXCR3-cells of which subset 1 was significantly reduced in latent tuberculosis infection (LTBI) with HIV-ART, yet Mtb-responsive IL-17-producing CD4 T cells were preserved; we found that IL-17-producing CD4 T cells dominate the response to Mtb antigen but not cytomegalovirus (CMV) antigen or staphylococcal enterotoxin B (SEB), and tryptophan catabolism negatively correlates with both subset 1 and subset 2 Th17-cell frequencies. Conclusions: We found differential effects of ART-suppressed HIV on distinct subsets of Th17 cells, that IL-17-producing CD4 T cells dominate responses to Mtb but not CMV antigen or SEB, and that kynurenine pathway activity is associated with decreases of circulating Th17 cells that may contribute to tuberculosis immunity.


Subject(s)
Antigens, Bacterial , HIV Infections , Interleukin-17 , Latent Tuberculosis , Mycobacterium tuberculosis , Th17 Cells , Adult , Female , Humans , Male , Middle Aged , Antigens, Bacterial/immunology , HIV Infections/immunology , HIV Infections/virology , Immunophenotyping , Interleukin-17/metabolism , Interleukin-17/immunology , Kynurenine/metabolism , Latent Tuberculosis/immunology , Latent Tuberculosis/microbiology , Mycobacterium tuberculosis/immunology , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Tryptophan/metabolism
12.
Asian Pac J Cancer Prev ; 25(5): 1777-1785, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38809650

ABSTRACT

BACKGROUND: Deregulation of immune checkpoint is an important point in cancer evolution as well as patients outcome. T-cells is an important arm in immunity against cancer. This study aimed to assess CTLA4/LAG3 expression on different T-cell subsets and its effect on disease outcome. METHODS: This study included 81 newly diagnosed Egyptian adult AML patients. For each one of the patients CTLA4/ LAG3 expression on T-cell subsets was identified by flowcytometry before start of induction chemotherapy. RESULTS: Total CD3 count in AML patients was lower than control. LAG3 expression were significantly higher in total CD3, T-cell subsets (CD4, CD8) as compared to healthy control. Moreover, co-expression of LAG3/CTLA4 on T-cell subsets were significantly higher in AML as compared to healthy control . NPM-/ FLT3+ was significantly associated with high LAG3 expression in T-cells subsets as compared to other molecular subtypes. Shorter OS, DFS were significantly associated with higher expression of LAG3 on T-cells subsets as compared to patients harbor low expression. COX regression analysis revealed that high expression of CD3/LAG3, CD4/LAG3, CD8/LAG4, CD3/CTLA4/LAG3 were considered a poor prognostic risk factor. CONCLUSION: High LAG3/CTLA4 expression could predict AML Patients' outcome Conclusion: Our findings indicated that high expression of LAG3/CTL4 on T cells subsets identify a subgroup of AML patients with poor prognosis.


Subject(s)
Antigens, CD , Biomarkers, Tumor , CTLA-4 Antigen , Leukemia, Myeloid, Acute , Lymphocyte Activation Gene 3 Protein , T-Lymphocyte Subsets , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/metabolism , Male , Female , Prognosis , CTLA-4 Antigen/metabolism , Adult , Antigens, CD/metabolism , Middle Aged , Case-Control Studies , Biomarkers, Tumor/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Follow-Up Studies , Young Adult , Survival Rate , Adolescent
14.
Commun Biol ; 7(1): 526, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702425

ABSTRACT

COVID-19, caused by SARS-CoV-2, can lead to a severe inflammatory disease characterized by significant lymphopenia. However, the underlying cause for the depletion of T-cells in COVID-19 patients remains incompletely understood. In this study, we assessed the presence of different T-cell subsets in the progression of COVID-19 from mild to severe disease, with a focus on TCF1 expressing progenitor T-cells that are needed to replenish peripheral T-cells during infection. Our results showed a preferential decline in TCF1+ progenitor CD4 and CD8+ T-cells with disease severity. This decline was seen in various TCF1+ subsets including naive, memory and effector-memory cells, and surprisingly, was accompanied by a loss in cell division as seen by a marked decline in Ki67 expression. In addition, TCF1+ T-cells showed a reduction in pro-survival regulator, BcL2, and the appearance of a new population of TCF1 negative caspase-3 expressing cells in peripheral blood from patients with severe disease. The decline in TCF1+ T-cells was also seen in a subgroup of severe patients with vitamin D deficiency. Lastly, we found that sera from severe patients inhibited TCF1 transcription ex vivo which was attenuated by a blocking antibody against the cytokine, interleukin-12 (IL12). Collectively, our findings underscore the potential significance of TCF1+ progenitor T-cells in accounting for the loss of immunity in severe COVID-19 and outline an array of markers that could be used to identify disease progression.


Subject(s)
COVID-19 , Hepatocyte Nuclear Factor 1-alpha , SARS-CoV-2 , Severity of Illness Index , Humans , COVID-19/immunology , COVID-19/pathology , Male , Female , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Middle Aged , CD8-Positive T-Lymphocytes/immunology , Adult , CD4-Positive T-Lymphocytes/immunology , Aged , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
15.
J Exp Med ; 221(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38753245

ABSTRACT

Preterm infants are at high risk of developing neonatal sepsis. γδ T cells are thought to be an important set of effector cells in neonates. Here, γδ T cells were investigated in a longitudinal cohort of preterm neonates using next-generation sequencing, flow cytometry, and functional assays. During the first year of life, the Vγ9Vδ2 T cell subset showed dynamic phenotypic changes and elevated levels of fetal-derived Vγ9Vδ2 T cells were evident in infants with sepsis. Single-cell transcriptomics identified HLA-DRhiCD83+ γδ T cells in neonatal sepsis, which expressed genes related to antigen presentation. In vitro assays showed that CD83 was expressed on activated Vγ9Vδ2 T cells in preterm and term neonates, but not in adults. In contrast, activation of adult Vγ9Vδ2 T cells enhanced CD86 expression, which was presumably the key receptor to induce CD4 T cell proliferation. Together, we provide a map of the maturation of γδ T cells after preterm birth and highlight their phenotypic diversity in infections.


Subject(s)
Antigens, CD , CD83 Antigen , Infant, Premature , Receptors, Antigen, T-Cell, gamma-delta , Humans , Infant, Newborn , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Infant, Premature/immunology , Antigens, CD/metabolism , Antigens, CD/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Female , Male , Sepsis/immunology , Cohort Studies , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adult , Lymphocyte Activation/immunology , Neonatal Sepsis/immunology , Infant
16.
Immunol Lett ; 267: 106861, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38697225

ABSTRACT

Hematopoietic precursors (HPCs) entering into the thymus undergo a sequential process leading to the generation of a variety of T cell subsets. This developmental odyssey unfolds in distinct stages within the thymic cortex and medulla, shaping the landscape of T cell receptor (TCR) expression and guiding thymocytes through positive and negative selection. Initially, early thymic progenitors (ETPs) take residence in the thymic cortex, where thymocytes begin to express their TCR and undergo positive selection. Subsequently, thymocytes transition to the thymic medulla, where they undergo negative selection. Both murine and human thymocyte development can be broadly classified into distinct stages based on the expression of CD4 and CD8 coreceptors, resulting in categorizations as double negative (DN), double positive (DP) or single positive (SP) cells. Thymocyte migration to the appropriate thymic microenvironment at the right differentiation stage is pivotal for the development and the proper functioning of T cells, which is critical for adaptive immune responses. The journey of lymphoid progenitor cells into the T cell developmental pathway hinges on an ongoing dialogue between the differentiating cell and the signals emanating from the thymus niche. Herein, we review the contribution of the key factors mentioned above for the localization, migration and emigration of thymocytes.


Subject(s)
Cell Differentiation , Cell Movement , Thymocytes , Thymus Gland , Thymocytes/immunology , Thymocytes/cytology , Thymocytes/metabolism , Animals , Humans , Thymus Gland/cytology , Thymus Gland/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Receptors, Antigen, T-Cell/metabolism
17.
Front Immunol ; 15: 1321126, 2024.
Article in English | MEDLINE | ID: mdl-38711501

ABSTRACT

Introduction: γδ T cells recognize and exert cytotoxicity against tumor cells. They are also considered potential immune cells for immunotherapy. Our previous study revealed that the altered expression of immune checkpoint T-cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) on γδ T cells may result in immunosuppression and is possibly associated with a poor overall survival in acute myeloid leukemia (AML). However, whether γδ T-cell memory subsets are predominantly involved and whether they have a relationship with clinical outcomes in patients with AML under the age of 65 remain unclear. Methods: In this study, we developed a multicolor flow cytometry-based assay to monitor the frequency and distribution of γδ T-cell subsets, including central memory γδ T cells (TCM γδ), effector memory γδ T cells (TEM γδ), and TEM expressing CD45RA (TEMRA γδ), in peripheral blood from 30 young (≤65 years old) patients with newly diagnosed non-acute promyelocytic leukemia (also known as M3) AML (AMLy-DN), 14 young patients with AML in complete remission (AMLy-CR), and 30 healthy individuals (HIs). Results: Compared with HIs, patients with AMLy-DN exhibited a significantly higher differentiation of γδ T cells, which was characterized by decreased TCM γδ cells and increased TEMRA γδ cells. A generally higher TIGIT expression was observed in γδ T cells and relative subsets in patients with AMLy-DN, which was partially recovered in patients with AMLy-CR. Furthermore, 17 paired bone marrow from patients with AMLy-DN contained higher percentages of γδ and TIGIT+ γδ T cells and a lower percentage of TCM γδ T cells. Multivariate logistic regression analyses revealed the association of high percentage of TIGIT+ TCM γδ T cells with an increased risk of poor induction chemotherapy response. Conclusions: In this study, we investigated the distribution of γδ T cells and their memory subsets in patients with non-M3 AML and suggested TIGIT+ TCM γδ T cells as potential predictive markers of induction chemotherapy response.


Subject(s)
Receptors, Antigen, T-Cell, gamma-delta , Receptors, Immunologic , Humans , Receptors, Immunologic/metabolism , Male , Female , Adult , Middle Aged , Prognosis , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Young Adult , Aged , Memory T Cells/immunology , Memory T Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/diagnosis , Immunologic Memory , Leukemia, Promyelocytic, Acute/immunology , Leukemia, Promyelocytic, Acute/diagnosis , Leukemia, Promyelocytic, Acute/mortality , Immunophenotyping
18.
Front Immunol ; 15: 1401738, 2024.
Article in English | MEDLINE | ID: mdl-38774869

ABSTRACT

A balance between pro-inflammatory decidual CD4+ T cells and FOXP3+ regulatory T cells (FOXP3+ Tregs) is important for maintaining fetomaternal tolerance. Using single-cell RNA-sequencing and T cell receptor repertoire analysis, we determined that diversity and clonality of decidual CD4+ T cell subsets depend on gestational age. Th1/Th2 intermediate and Th1 subsets of CD4+ T cells were clonally expanded in both early and late gestation, whereas FOXP3+ Tregs were clonally expanded in late gestation. Th1/Th2 intermediate and FOXP3+ Treg subsets showed altered gene expression in preeclampsia (PE) compared to healthy late gestation. The Th1/Th2 intermediate subset exhibited elevated levels of cytotoxicity-related gene expression in PE. Moreover, increased Treg exhaustion was observed in the PE group, and FOXP3+ Treg subcluster analysis revealed that the effector Treg like subset drove the Treg exhaustion signatures in PE. The Th1/Th2 intermediate and effector Treg like subsets are possible inflammation-driving subsets in PE.


Subject(s)
Forkhead Transcription Factors , Gestational Age , Pre-Eclampsia , Single-Cell Analysis , T-Lymphocytes, Regulatory , Humans , Female , Pre-Eclampsia/immunology , Pre-Eclampsia/genetics , Pregnancy , Single-Cell Analysis/methods , Adult , T-Lymphocytes, Regulatory/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , CD4-Positive T-Lymphocytes/immunology , Sequence Analysis, RNA , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/immunology , Decidua/immunology
19.
Cytometry B Clin Cytom ; 106(3): 171-180, 2024 05.
Article in English | MEDLINE | ID: mdl-38695297

ABSTRACT

CD20+ T cells constitute a small subset of T cells. These are found among CD4+, CD8+, CD4+CD8+, CD4-CD8- T, and TCRγδ+ T cells, and have been poorly characterized. The aim of this study was to characterize peripheral blood (PB) CD20+ T cells and compare them to their PB CD20- T cell counterparts. PB from 17 healthy individuals was collected. The distribution of CD20+ T cells among maturation-associated T cells compartments (naïve, central memory, transitional memory, effector memory, and effector T cells), their polarization, activation status, and expression of immune-regulatory proteins were evaluated by flow cytometry. Their function was also assessed, by measuring IFN-γ, TNF-α, and IL-17 production. Compared with CD20- T cells, CD20+ T cells represent a higher proportion of transitional memory cells. Furthermore, CD20+ T cells display a proinflammatory phenotype, characterized by the expansion of Th1, Th1/17, and Tc1 cell subsets , associated to a high expression of activation (CD25) and exhaustion (PD-1) markers. In addition, the simultaneous production of the proinflammatory cytokines IFN-γ, TNF-α, and IL-17 was also detected in CD4+CD20+ T cells. Our results show that CD20+ T cells are phenotypically and functionally different from CD20- T cells, suggesting that these cells are a distinct subset of T cells.


Subject(s)
Antigens, CD20 , Flow Cytometry , Humans , Antigens, CD20/immunology , Male , Female , Adult , Interferon-gamma , Tumor Necrosis Factor-alpha , Interleukin-17/blood , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Lymphocyte Activation/immunology , Middle Aged , Immunologic Memory/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Cytokines/metabolism , Memory T Cells/immunology , Interleukin-2 Receptor alpha Subunit/immunology
20.
J Immunother Cancer ; 12(5)2024 May 30.
Article in English | MEDLINE | ID: mdl-38816233

ABSTRACT

BACKGROUND: The incidence of papillary thyroid cancer (PTC) continues to rise all over the world, 10-15% of the patients have a poor prognosis. Although immunotherapy has been applied in clinical practice, its therapeutic efficacy remains far from satisfactory, necessitating further investigation of the mechanism of PTC immune remodeling and exploration of novel treatment targets. METHODS: This study conducted a single-cell RNA sequencing (scRNA-seq) analysis using 18 surgical tissue specimens procured from 14 patients diagnosed with adjacent tissues, non-progressive PTC or progressive PTC. Key findings were authenticated through spatial transcriptomics RNA sequencing, immunohistochemistry, multiplex immunohistochemistry, and an independent bulk RNA-seq data set containing 502 samples. RESULTS: A total of 151,238 individual cells derived from 18 adjacent tissues, non-progressive PTC and progressive PTC specimens underwent scRNA-seq analysis. We found that progressive PTC exhibits the following characteristics: a significant decrease in overall immune cells, enhanced immune evasion of tumor cells, and disrupted antigen presentation function. Moreover, we identified a subpopulation of lysosomal associated membrane protein 3 (LAMP3+) dendritic cells (DCs) exhibiting heightened infiltration in progressive PTC and associated with advanced T stage and poor prognosis of PTC. LAMP3+ DCs promote CD8+ T cells exhaustion (mediated by NECTIN2-TIGIT) and increase infiltration abundance of regulatory T cells (mediated by chemokine (C-C motif) ligand 17 (CCL17)-chemokine (C-C motif) receptor 4 (CCR4)) establishing an immune-suppressive microenvironment. Ultimately, we unveiled that progressive PTC tumor cells facilitate the retention of LAMP3+ DCs within the tumor microenvironment through NECTIN3-NECTIN2 interactions, thereby rendering tumor cells more susceptible to immune evasion. CONCLUSION: Our findings expound valuable insights into the role of the interaction between LAMP3+ DCs and T-cell subpopulations and offer new and effective ideas and strategies for immunotherapy in patients with progressive PTC.


Subject(s)
Dendritic Cells , Thyroid Cancer, Papillary , Humans , Dendritic Cells/immunology , Dendritic Cells/metabolism , Thyroid Cancer, Papillary/immunology , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/genetics , Lysosomal-Associated Membrane Protein 3/metabolism , Thyroid Neoplasms/immunology , Thyroid Neoplasms/pathology , Thyroid Neoplasms/genetics , Male , Female , Tumor Microenvironment/immunology , Middle Aged , Tumor Escape , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Neoplasm Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...