Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Hepatol ; 77(5): 1359-1372, 2022 11.
Article in English | MEDLINE | ID: mdl-35738508

ABSTRACT

BACKGROUND & AIMS: The landscape and function of the immune infiltrate of intrahepatic cholangiocarcinoma (iCCA), a rare, yet aggressive tumor of the biliary tract, remains poorly characterized, limiting development of successful immunotherapies. Herein, we aimed to define the molecular characteristics of tumor-infiltrating leukocytes with a special focus on CD4+ regulatory T cells (Tregs). METHODS: We used high-dimensional single-cell technologies to characterize the T-cell and myeloid compartments of iCCA tissues, comparing these with their tumor-free peritumoral and circulating counterparts. We further used genomics and cellular assays to define the iCCA-specific role of a novel transcription factor, mesenchyme homeobox 1 (MEOX1), in Treg biology. RESULTS: We found poor infiltration of putative tumor-specific CD39+ CD8+ T cells accompanied by abundant infiltration of hyperactivated CD4+ Tregs. Single-cell RNA-sequencing identified an altered network of transcription factors in iCCA-infiltrating compared to peritumoral T cells, suggesting reduced effector functions by tumor-infiltrating CD8+ T cells and enhanced immunosuppression by CD4+ Tregs. Specifically, we found that expression of MEOX1 was highly enriched in tumor-infiltrating Tregs, and demonstrated that MEOX1 overexpression is sufficient to reprogram circulating Tregs to acquire the transcriptional and epigenetic landscape of tumor-infiltrating Tregs. Accordingly, enrichment of the MEOX1-dependent gene program in Tregs was strongly associated with poor prognosis in a large cohort of patients with iCCA. CONCLUSIONS: We observed abundant infiltration of hyperactivated CD4+ Tregs in iCCA tumors along with reduced CD8+ T-cell effector functions. Interfering with hyperactivated Tregs should be explored as an approach to enhance antitumor immunity in iCCA. LAY SUMMARY: Immune cells have the potential to slow or halt the progression of tumors. However, some tumors, such as intrahepatic cholangiocarcinoma, are associated with very limited immune responses (and infiltration of cancer-targeting immune cells). Herein, we show that a specific population of regulatory T cells (a type of immune cell that actually suppresses the immune response) are hyperactivated in intrahepatic cholangiocarcinoma. Targeting these cells could enable cancer-targeting immune cells to act more effectively and should be looked at as a potential therapeutic approach to this aggressive cancer type.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cholangiocarcinoma/pathology , RNA/metabolism , T-Lymphocytes, Regulatory , Transcription Factors/metabolism , Tumor Microenvironment , Single-Cell Analysis
3.
Commun Biol ; 4(1): 1117, 2021 09 22.
Article in English | MEDLINE | ID: mdl-34552178

ABSTRACT

Luminal-like breast cancer (BC) constitutes the majority of BC subtypes, but, differently from highly aggressive triple negative BC, is poorly infiltrated by the immune system. The quality of the immune infiltrate in luminal-like BCs has been poorly studied, thereby limiting further investigation of immunotherapeutic strategies. By using high-dimensional single-cell technologies, we identify heterogeneous behavior within the tissue-resident memory CD8+ T (Trm) cells infiltrating luminal-like tumors. A subset of CD127- CD39hi Trm cells, preferentially present in the tumor compared to the adjacent normal breast tissue or peripheral blood, retains enhanced degranulation capacity compared to the CD127+ CD39lo Trm counterpart ex vivo, and is specifically associated with positive prognosis. Nevertheless, such prognostic benefit is lost in the presence of highly-suppressive CCR8hi ICOShi IRF4+ effector Tregs. Thus, combinatorial strategies aiming at boosting Trm function and infiltration while relieving from Treg-mediated immunosuppression should be investigated to achieve proper tumor control in luminal-like BCs.


Subject(s)
Apyrase/metabolism , Breast Neoplasms/genetics , CD8-Positive T-Lymphocytes/metabolism , Breast Neoplasms/diagnosis , Humans , Prognosis , Single-Cell Analysis
4.
Semin Cancer Biol ; 72: 90-101, 2021 07.
Article in English | MEDLINE | ID: mdl-32492452

ABSTRACT

Cancer immunotherapy has witnessed a new renaissance with the advent of immune checkpoint inhibitors, which reactivate T cells and foster endogenous anti-tumor responses. The excellent results of immunotherapy in the field of melanoma, renal cancer, lung cancer, and other cancer types that have traditionally been known to be immunogenic, rekindled the interest of the oncology community in extending the benefits to all cancers including breast cancer (BC). In this review, we highlight the current state of using T cells as both markers for clinical practice and therapeutic options for BC.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/therapy , Immune System , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes/immunology , Animals , Female , Humans
5.
Nat Commun ; 11(1): 821, 2020 02 10.
Article in English | MEDLINE | ID: mdl-32041953

ABSTRACT

The diversity of the naïve T cell repertoire drives the replenishment potential and capacity of memory T cells to respond to immune challenges. Attrition of the immune system is associated with an increased prevalence of pathologies in aged individuals, but whether stem cell memory T lymphocytes (TSCM) contribute to such attrition is still unclear. Using single cells RNA sequencing and high-dimensional flow cytometry, we demonstrate that TSCM heterogeneity results from differential engagement of Wnt signaling. In humans, aging is associated with the coupled loss of Wnt/ß-catenin signature in CD4 TSCM and systemic increase in the levels of Dickkopf-related protein 1, a natural inhibitor of the Wnt/ß-catenin pathway. Functional assays support recent thymic emigrants as the precursors of CD4 TSCM. Our data thus hint that reversing TSCM defects by metabolic targeting of the Wnt/ß-catenin pathway may be a viable approach to restore and preserve immune homeostasis in the context of immunological history.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Precursor Cells, T-Lymphoid/immunology , Wnt Signaling Pathway/immunology , Aging/immunology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Gene Expression Profiling , HIV Infections/immunology , Humans , Immunologic Memory , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Thymus Gland/immunology , Wnt Signaling Pathway/genetics , beta Catenin/immunology
6.
Methods Enzymol ; 631: 137-158, 2020.
Article in English | MEDLINE | ID: mdl-31948545

ABSTRACT

Among the multiple factors that are responsible for the success of adoptive cell transfer (ACT) immunotherapy for cancer, the differentiation status of the in vitro expanded T cell product at the time of transfer seems to play a major role. In particular, less differentiated memory CD8+ T cells endowed with self-renewing capacity and multipotency exert the most potent antitumor activity. To this aim, expansion protocols that generate sufficient numbers of tumor-specific CD8+ T cells with superior capacity to persist in vivo following ACT are needed. We describe a procedure for the differentiation of TCF-1+ stem-like CD8+ memory T cells from peripheral blood naïve precursors that takes advantage of the use of antioxidants, in particular N-acetylcysteine (NAC), in combination with T cell receptor stimulation and proinflammatory cytokines. We additionally describe how to conduct in vitro assays to test the stem-like features of the generated cells at the phenotypic, functional and metabolic level. Balancing the oxidative metabolism by the addition of antioxidants during in vitro manipulation of CD8+ T cells results in the generation of cell products with potent antitumor characteristics following ACT.


Subject(s)
Acetylcysteine/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity Tests, Immunologic/methods , Immunologic Memory , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Antioxidants/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Humans , Stem Cells
7.
J Immunol ; 203(12): 3179-3189, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31740485

ABSTRACT

In mice, the ability of naive T (TN) cells to mount an effector response correlates with TCR sensitivity for self-derived Ags, which can be quantified indirectly by measuring surface expression levels of CD5. Equivalent findings have not been reported previously in humans. We identified two discrete subsets of human CD8+ TN cells, defined by the absence or presence of the chemokine receptor CXCR3. The more abundant CXCR3+ TN cell subset displayed an effector-like transcriptional profile and expressed TCRs with physicochemical characteristics indicative of enhanced interactions with peptide-HLA class I Ags. Moreover, CXCR3+ TN cells frequently produced IL-2 and TNF in response to nonspecific activation directly ex vivo and differentiated readily into Ag-specific effector cells in vitro. Comparative analyses further revealed that human CXCR3+ TN cells were transcriptionally equivalent to murine CXCR3+ TN cells, which expressed high levels of CD5. These findings provide support for the notion that effector differentiation is shaped by heterogeneity in the preimmune repertoire of human CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Receptors, CXCR3/metabolism , Adult , Age Factors , Aged , Animals , Biomarkers , Cells, Cultured , Female , Humans , Immunologic Memory , Immunophenotyping , Lymphocyte Activation/immunology , Male , Mice , Middle Aged , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Young Adult
8.
PLoS One ; 14(6): e0218354, 2019.
Article in English | MEDLINE | ID: mdl-31173608

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0091952.].

9.
JCI Insight ; 3(18)2018 09 20.
Article in English | MEDLINE | ID: mdl-30232291

ABSTRACT

Adoptive T cell transfer (ACT) immunotherapy benefits from early differentiated stem cell memory T (Tscm) cells capable of persisting in the long term and generating potent antitumor effectors. Due to their paucity ex vivo, Tscm cells can be derived from naive precursors, but the molecular signals at the basis of Tscm cell generation are ill-defined. We found that less differentiated human circulating CD8+ T cells display substantial antioxidant capacity ex vivo compared with more differentiated central and effector memory T cells. Limiting ROS metabolism with antioxidants during naive T cell activation hindered terminal differentiation, while allowing expansion and generation of Tscm cells. N-acetylcysteine (NAC), the most effective molecule in this regard, induced transcriptional and metabolic programs characteristic of self-renewing memory T cells. Upon ACT, NAC-generated Tscm cells established long-term memory in vivo and exerted more potent antitumor immunity in a xenogeneic model when redirected with CD19-specific CAR, highlighting the translational relevance of NAC as a simple and inexpensive method to improve ACT.


Subject(s)
Antineoplastic Agents/immunology , Antioxidants/metabolism , Antioxidants/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Stem Cells/drug effects , Animals , Antigens, CD19 , Cell Differentiation/drug effects , Female , Gene Expression Profiling , Humans , Immunity, Cellular , Immunologic Memory , Immunotherapy, Adoptive , Male , Mice , Mice, Inbred NOD
10.
Int Rev Cell Mol Biol ; 341: 63-124, 2018.
Article in English | MEDLINE | ID: mdl-30262035

ABSTRACT

On antigen encounter, naïve CD8+ and CD4+ T cells differentiate into a large number of effector cells that migrate to inflamed tissues to fight infections or tumors. Following elimination of the target, a few cells remain in the long-term, the so-called memory T cells that are capable to reexpand and respond more vigorously on a second encounter with the cognate antigen. While the naïve T cell compartment is fairly homogenous, effector and memory T cells are largely diverse, comprising dozens of subsets with diverse functions, molecular characteristics, and localization in the body. In addition, CD4+ and, to some extent, also CD8+ T cells can differentiate into several effector subsets according to their pattern of cytokine expression, including the T helper 1 (Th1), Th2, and Th17 cells. It has become clear that specific subsets of T cells dominate different types of infections and pathological conditions and have different capacities to infiltrate and reject tumors. Their correct phenotypic identification is therefore of foremost importance for their live purification by magnetic or fluorescence-activated cell sorting and subsequent molecular characterization. Here, we present a comprehensive list of the main T cell subpopulations with a major focus on human cells along with their surface phenotypic properties.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/classification , CD8-Positive T-Lymphocytes/immunology , Cytokines/genetics , Gene Expression Regulation , Humans , Receptors, Immunologic
11.
Eur J Immunol ; 48(8): 1281-1284, 2018 08.
Article in English | MEDLINE | ID: mdl-30133745

ABSTRACT

Interferon regulatory factor 4 (IRF4) regulates the clonal expansion and metabolic activity of activated T cells, but the precise context and mechanisms of its function in these processes are unclear. In this issue of the European Journal of Immunology, Miyakoda et al. [Eur. J. Immunol. 2018. 48: 1319-1328] show that IRF4 is required for activation and expansion of naïve and memory CD8+ T cells driven by T-cell receptor (TCR) signaling, but dispensable for memory CD8+ T-cell maintenance and homeostatic proliferation driven by homeostatic cytokines. The authors show that the function of IRF4 in CD8+ T-cell expansion is partially dependent upon activation of the PI3K/AKT pathway through direct or indirect attenuation of PTEN expression. These data shed light upon the differential intracellular pathways required for naïve and memory T cells to respond to self-antigens and/or homeostatic cytokines, and highlight the potential translational relevance of these findings in the context of immune reconstitution such as following allogeneic stem cell transplantation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interferon Regulatory Factors/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Cell Differentiation/immunology , Cell Proliferation , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Mice , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/immunology
12.
Eur J Immunol ; 47(9): 1468-1476, 2017 09.
Article in English | MEDLINE | ID: mdl-28671275

ABSTRACT

Human T memory stem (TSCM ) cells with superior persistence capacity and effector functions are emerging as important players in the maintenance of long-lived T-cell memory and are thus considered an attractive population to be used in adoptive transfer-based immunotherapy of cancer. However, the molecular signals regulating their generation remain poorly defined. Here we show that curtailed T-cell receptor stimulation curbs human effector CD8+ T-cell differentiation and allows the generation of CD45RO- CD45RA+ CCR7+ CD27+ CD95+ -phenotype cells from highly purified naïve T-cell precursors, resembling naturally-occurring human TSCM . These cells proliferate extensively in vitro and in vivo, express low amounts of effector-associated genes and transcription factors and undergo considerable self-renewal in response to IL-15 while retaining effector differentiation potential. Such a phenotype is associated with a lower number of mitochondria compared to highly-activated effector T cells committed to terminal differentiation. These results shed light on the molecular signals that are required to generate long-lived memory T cells with potential application in adoptive cell transfer immunotherapy.


Subject(s)
Adult Stem Cells/physiology , CD8-Positive T-Lymphocytes/physiology , Cancer Vaccines/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Animals , Antigens, CD/metabolism , Cell Differentiation , Cell Proliferation , Cell Self Renewal , Cells, Cultured , Humans , Immunologic Memory , Immunophenotyping , Interleukin-15/metabolism , Lymphocyte Activation , Mice , Mice, SCID , Neoplasms/immunology , Phenotype , Receptors, Antigen, T-Cell/metabolism
13.
Cancer Res ; 75(24): 5187-5193, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26627006

ABSTRACT

Preclinical models revealed that the immune system can mediate rejection of established tumors, but direct evidence in humans has been limited to largely immunogenic tumors, such as melanoma. The recent success of immune checkpoint inhibitors and adoptive T-cell transfer immunotherapy in clinical trials has instilled new hope for the use of T-cell immunotherapy in the treatment of cancer. IL15, a potent immunostimulatory cytokine, both potentiates host T-cells and natural killer (NK) cell immune responses and promotes the generation of long-lived memory T cells with superior functional capacity, with potential use in adoptive T-cell transfer protocols. IL15 has been recently tested in the clinic and showed dramatic effects at the level of responding NK and CD8(+) memory T cells. The recent advances in the knowledge of IL15-dependent regulation of T-cell responses, gene expression, and metabolic adaptation have important implications for the use of IL15 in T-cell-based immunotherapy of cancer.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-15/immunology , Neoplasms/therapy , T-Lymphocytes/transplantation , Animals , Humans , Neoplasms/immunology , Stem Cells/immunology
14.
PLoS One ; 9(3): e91952, 2014.
Article in English | MEDLINE | ID: mdl-24633152

ABSTRACT

Germline and somatic biallelic mutations of the Tuberous sclerosis complex (TSC) 1 and TSC2 gene products cause TSC, an autosomal dominant multifocal hamartomatosis with variable neurological manifestations. The consequences of TSC1 or TSC2 loss in cells of hematopoietic origin have recently started to be unveiled in mice and showed to hinder the development of proper T cell immunity. To date, the consequences of germline TSC1 mutations and/or its loss in mature human T cells remain to be determined. To address these issues, we analyzed subset representation, phenotype and responsiveness to mitogens in T cells from patients with inherited monoallelic TSC1 mutations, and induced shRNA-mediated TSC1 down-regulation in primary and transformed human T cells. We report that, the distribution of peripheral CD4 and CD8 T cell subsets, their cytokine-secretion profile, and responsiveness to in vitro stimulation were largely preserved in TSC subjects with monoallelic TSC1 germline mutations when compared to healthy controls. Sufficient levels of hamartin and tuberin and proper control of mTOR-dependent signaling in primary T cells from TSC subjects best explained this. In contrast, shRNA-induced down-regulation of TSC1, likely mimicking biallelic inactivation of TSC1, compromised hamartin and tuberin expression and mTORC2/AKT/FoxO1/3 signaling causing both primary and transformed T cells to die by apoptosis. Thus, our results indicate that, while one functional TSC1 allele preserves human T lymphocytes development and homeostasis, TSC1 acute down-regulation is detrimental to the survival of both primary and transformed T cells.


Subject(s)
Alleles , Germ-Line Mutation , Homeostasis , T-Lymphocyte Subsets/cytology , Tuberous Sclerosis/genetics , Tuberous Sclerosis/immunology , Tumor Suppressor Proteins/genetics , Adult , Animals , Apoptosis/genetics , Cell Proliferation , Cell Survival/genetics , Down-Regulation/genetics , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Silencing , Humans , Mice , Middle Aged , Mitochondria/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/deficiency
15.
Eur J Immunol ; 41(7): 2086-96, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21480212

ABSTRACT

The mammalian target of rapamycin (mTOR) controls T-cell differentiation in response to polarizing cytokines. We previously found that mTOR blockade by rapamycin (RAPA) delays the G1-S cell cycle transition and lymphocyte proliferation. Here, we report that both mTOR complex 1 and mTOR complex 2 are readily activated following TCR/CD28 engagement and are critical for early expression of Ifng, Il4 and Foxp3, and for effector T cell differentiation in the absence of polarizing cytokines. While inhibition of mTOR complex 1 and cell division were evident at low doses of RAPA, inhibition of mTOR complex 2, Ifng, Il4 and Foxp3 expression, and T-cell polarization required higher doses and more prolonged treatments. We found that while T-bet and GATA3 were readily induced following TCR/CD28 engagement, administration of RAPA delayed their expression, and interfered with the loss of DNA methylation within Ifng and Il4 promoter regions. In contrast, RAPA prevented activation-dependent DNA methylation of the Foxp3 promoter favoring Foxp3 expression. As a result, RAPA-cultured cells lacked immediate effector functions and instead were enriched for IL-2+ cells. We propose that mTOR-signaling, by timing the expression of critical transcription factors and DNA methylation of proximal promoter regions, regulates transcriptional competence at immunologically relevant sites and hence lymphocyte differentiation.


Subject(s)
CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/genetics , Interferon-gamma/genetics , Interleukin-4/genetics , Sirolimus/pharmacology , Transcription, Genetic , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , DNA Methylation , Forkhead Transcription Factors/metabolism , GATA3 Transcription Factor/biosynthesis , Interferon-gamma/metabolism , Interleukin-2/biosynthesis , Interleukin-4/metabolism , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred BALB C , Multiprotein Complexes , Polymerase Chain Reaction , Promoter Regions, Genetic , Proteins/metabolism , Signal Transduction , T-Box Domain Proteins/biosynthesis , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...