Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Science ; 382(6674): 1073-1079, 2023 12.
Article in English | MEDLINE | ID: mdl-38033053

ABSTRACT

Skin-resident CD8+ T cells include distinct interferon-γ-producing [tissue-resident memory T type 1 (TRM1)] and interleukin-17 (IL-17)-producing (TRM17) subsets that differentially contribute to immune responses. However, whether these populations use common mechanisms to establish tissue residence is unknown. In this work, we show that TRM1 and TRM17 cells navigate divergent trajectories to acquire tissue residency in the skin. TRM1 cells depend on a T-bet-Hobit-IL-15 axis, whereas TRM17 cells develop independently of these factors. Instead, c-Maf commands a tissue-resident program in TRM17 cells parallel to that induced by Hobit in TRM1 cells, with an ICOS-c-Maf-IL-7 axis pivotal to TRM17 cell commitment. Accordingly, by targeting this pathway, skin TRM17 cells can be ablated without compromising their TRM1 counterparts. Thus, skin-resident T cells rely on distinct molecular circuitries, which can be exploited to strategically modulate local immunity.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Memory T Cells , Skin , CD8-Positive T-Lymphocytes/immunology , Memory T Cells/immunology , Skin/immunology , Humans , Th17 Cells/immunology , Inducible T-Cell Co-Stimulator Ligand/metabolism , Proto-Oncogene Proteins c-maf/metabolism , Interleukin-7/metabolism
2.
Cell ; 186(22): 4851-4867.e20, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37848036

ABSTRACT

Post-acute sequelae of COVID-19 (PASC, "Long COVID") pose a significant global health challenge. The pathophysiology is unknown, and no effective treatments have been found to date. Several hypotheses have been formulated to explain the etiology of PASC, including viral persistence, chronic inflammation, hypercoagulability, and autonomic dysfunction. Here, we propose a mechanism that links all four hypotheses in a single pathway and provides actionable insights for therapeutic interventions. We find that PASC are associated with serotonin reduction. Viral infection and type I interferon-driven inflammation reduce serotonin through three mechanisms: diminished intestinal absorption of the serotonin precursor tryptophan; platelet hyperactivation and thrombocytopenia, which impacts serotonin storage; and enhanced MAO-mediated serotonin turnover. Peripheral serotonin reduction, in turn, impedes the activity of the vagus nerve and thereby impairs hippocampal responses and memory. These findings provide a possible explanation for neurocognitive symptoms associated with viral persistence in Long COVID, which may extend to other post-viral syndromes.


Subject(s)
Post-Acute COVID-19 Syndrome , Serotonin , Humans , COVID-19/complications , Disease Progression , Inflammation , Post-Acute COVID-19 Syndrome/blood , Post-Acute COVID-19 Syndrome/pathology , Serotonin/blood , Virus Diseases
3.
Nat Rev Cancer ; 23(11): 780-798, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37821656

ABSTRACT

T cells can acquire a broad spectrum of differentiation states following activation. At the extreme ends of this continuum are short-lived cells equipped with effector machinery and more quiescent, long-lived cells with heightened proliferative potential and stem cell-like developmental plasticity. The latter encompass stem-like exhausted T cells and memory T cells, both of which have recently emerged as key determinants of cancer immunity and response to immunotherapy. Here, we discuss key similarities and differences in the regulation and function of stem-like exhausted CD8+ T cells and memory CD8+ T cells, and consider their context-specific contributions to protective immunity in diverse outcomes of cancer, including tumour escape, long-term control and eradication. Finally, we emphasize how recent advances in the understanding of the molecular regulation of stem-like exhausted T cells and memory T cells are being explored for clinical benefit in cancer immunotherapies such as checkpoint inhibition, adoptive cell therapy and vaccination.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , Memory T Cells , Neoplasms/therapy , Cell Differentiation , Stem Cells
4.
Immunity ; 56(7): 1664-1680.e9, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37392736

ABSTRACT

Memory CD8+ T cells can be broadly divided into circulating (TCIRCM) and tissue-resident memory T (TRM) populations. Despite well-defined migratory and transcriptional differences, the phenotypic and functional delineation of TCIRCM and TRM cells, particularly across tissues, remains elusive. Here, we utilized an antibody screening platform and machine learning prediction pipeline (InfinityFlow) to profile >200 proteins in TCIRCM and TRM cells in solid organs and barrier locations. High-dimensional analyses revealed unappreciated heterogeneity within TCIRCM and TRM cell lineages across nine different organs after either local or systemic murine infection models. Additionally, we demonstrated the relative effectiveness of strategies allowing for the selective ablation of TCIRCM or TRM populations across organs and identified CD55, KLRG1, CXCR6, and CD38 as stable markers for characterizing memory T cell function during inflammation. Together, these data and analytical framework provide an in-depth resource for memory T cell classification in both steady-state and inflammatory conditions.


Subject(s)
CD8-Positive T-Lymphocytes , Memory T Cells , Mice , Animals , Cell Lineage , Immunologic Memory
5.
Immunity ; 56(6): 1320-1340.e10, 2023 06 13.
Article in English | MEDLINE | ID: mdl-37315535

ABSTRACT

CD8+ T cell exhaustion (Tex) limits disease control during chronic viral infections and cancer. Here, we investigated the epigenetic factors mediating major chromatin-remodeling events in Tex-cell development. A protein-domain-focused in vivo CRISPR screen identified distinct functions for two versions of the SWI/SNF chromatin-remodeling complex in Tex-cell differentiation. Depletion of the canonical SWI/SNF form, BAF, impaired initial CD8+ T cell responses in acute and chronic infection. In contrast, disruption of PBAF enhanced Tex-cell proliferation and survival. Mechanistically, PBAF regulated the epigenetic and transcriptional transition from TCF-1+ progenitor Tex cells to more differentiated TCF-1- Tex subsets. Whereas PBAF acted to preserve Tex progenitor biology, BAF was required to generate effector-like Tex cells, suggesting that the balance of these factors coordinates Tex-cell subset differentiation. Targeting PBAF improved tumor control both alone and in combination with anti-PD-L1 immunotherapy. Thus, PBAF may present a therapeutic target in cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Chromatin Assembly and Disassembly , Chromatin , Cell Differentiation , Epigenesis, Genetic
6.
Eur J Immunol ; 53(11): e2250060, 2023 11.
Article in English | MEDLINE | ID: mdl-36597841

ABSTRACT

Tissue-resident memory T (TRM ) cells play a vital role in local immune protection against infection and cancer. The location of TRM cells within peripheral tissues at sites of pathogen invasion allows for the rapid detection and elimination of microbes, making their generation an attractive goal for the development of next-generation vaccines. Here, we discuss differential requirements for CD8+ TRM cell development across tissues with implications for establishing local prophylactic immunity, emphasizing the role of tissue-derived factors, local antigen, and adjuvants on TRM cell generation in the context of vaccination.


Subject(s)
CD8-Positive T-Lymphocytes , Vaccines , Humans , Immunologic Memory , Immune System , Vaccination
7.
Nat Immunol ; 23(8): 1236-1245, 2022 08.
Article in English | MEDLINE | ID: mdl-35882933

ABSTRACT

Tissue-resident memory T cells (TRM cells) provide rapid and superior control of localized infections. While the transcription factor Runx3 is a critical regulator of CD8+ T cell tissue residency, its expression is repressed in CD4+ T cells. Here, we show that, as a direct consequence of this Runx3-deficiency, CD4+ TRM cells lacked the transforming growth factor (TGF)-ß-responsive transcriptional network that underpins the tissue residency of epithelial CD8+ TRM cells. While CD4+ TRM cell formation required Runx1, this, along with the modest expression of Runx3 in CD4+ TRM cells, was insufficient to engage the TGF-ß-driven residency program. Ectopic expression of Runx3 in CD4+ T cells incited this TGF-ß-transcriptional network to promote prolonged survival, decreased tissue egress, a microanatomical redistribution towards epithelial layers and enhanced effector functionality. Thus, our results reveal distinct programming of tissue residency in CD8+ and CD4+ TRM cell subsets that is attributable to divergent Runx3 activity.


Subject(s)
Immunologic Memory , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Transforming Growth Factor beta/metabolism
8.
Trends Immunol ; 43(4): 280-282, 2022 04.
Article in English | MEDLINE | ID: mdl-35272933

ABSTRACT

Efficient generation of tissue-resident memory T (TRM) cells is essential for long-lived immune protection in barrier tissues. Peng et al. now show that the costimulatory molecule ICOS enhances CD8+ TRM cell lodgment by promoting early tissue retention.


Subject(s)
Internship and Residency , Bandages , CD8-Positive T-Lymphocytes/immunology , Humans , Immunologic Memory/immunology , Inducible T-Cell Co-Stimulator Protein , Transcription Factors
9.
J Exp Med ; 219(1)2022 01 03.
Article in English | MEDLINE | ID: mdl-34677611

ABSTRACT

Tissue-resident memory T (TRM) cells provide long-lasting immune protection. One of the key events controlling TRM cell development is the local retention of TRM cell precursors coupled to downregulation of molecules necessary for tissue exit. Sphingosine-1-phosphate receptor 5 (S1PR5) is a migratory receptor with an uncharted function in T cells. Here, we show that S1PR5 plays a critical role in T cell infiltration and emigration from peripheral organs, as well as being specifically downregulated in TRM cells. Consequentially, TRM cell development was selectively impaired upon ectopic expression of S1pr5, whereas loss of S1pr5 enhanced skin TRM cell formation by promoting peripheral T cell sequestration. Importantly, we found that T-bet and ZEB2 were required for S1pr5 induction and that local TGF-ß signaling was necessary to promote coordinated Tbx21, Zeb2, and S1pr5 downregulation. Moreover, S1PR5-mediated control of tissue residency was conserved across innate and adaptive immune compartments. Together, these results identify the T-bet-ZEB2-S1PR5 axis as a previously unappreciated mechanism modulating the generation of tissue-resident lymphocytes.


Subject(s)
Cell Differentiation/genetics , Lymphoid Tissue/metabolism , Memory T Cells/metabolism , Sphingosine-1-Phosphate Receptors/genetics , T-Lymphocytes/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Movement/genetics , Cells, Cultured , Gene Expression Profiling/methods , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA-Seq/methods , Sphingosine-1-Phosphate Receptors/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Zinc Finger E-box Binding Homeobox 2/genetics , Zinc Finger E-box Binding Homeobox 2/metabolism
10.
Nat Immunol ; 22(9): 1140-1151, 2021 09.
Article in English | MEDLINE | ID: mdl-34426691

ABSTRACT

Tissue-resident memory T (TRM) cells are non-recirculating cells that exist throughout the body. Although TRM cells in various organs rely on common transcriptional networks to establish tissue residency, location-specific factors adapt these cells to their tissue of lodgment. Here we analyze TRM cell heterogeneity between organs and find that the different environments in which these cells differentiate dictate TRM cell function, durability and malleability. We find that unequal responsiveness to TGFß is a major driver of this diversity. Notably, dampened TGFß signaling results in CD103- TRM cells with increased proliferative potential, enhanced function and reduced longevity compared with their TGFß-responsive CD103+ TRM counterparts. Furthermore, whereas CD103- TRM cells readily modified their phenotype upon relocation, CD103+ TRM cells were comparatively resistant to transdifferentiation. Thus, despite common requirements for TRM cell development, tissue adaptation of these cells confers discrete functional properties such that TRM cells exist along a spectrum of differentiation potential that is governed by their local tissue microenvironment.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Plasticity/immunology , Cellular Microenvironment/immunology , Immunologic Memory/immunology , Animals , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/cytology , Female , Integrin alpha Chains/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology , Transforming Growth Factor beta1/metabolism
11.
J Exp Med ; 218(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-33914023

ABSTRACT

Tissue-resident memory T cells (TRM cells) are key elements of tissue immunity. Here, we investigated the role of the regulator of T cell receptor and cytokine signaling, Ptpn2, in the formation and function of TRM cells in skin. Ptpn2-deficient CD8+ T cells displayed a marked defect in generating CD69+ CD103+ TRM cells in response to herpes simplex virus type 1 (HSV-1) skin infection. This was accompanied by a reduction in the proportion of KLRG1- memory precursor cells and a transcriptional bias toward terminal differentiation. Of note, forced expression of KLRG1 was sufficient to impede TRM cell formation. Normalizing memory precursor frequencies by transferring equal numbers of KLRG1- cells restored TRM generation, demonstrating that Ptpn2 impacted skin seeding with precursors rather than downstream TRM cell differentiation. Importantly, Ptpn2-deficient TRM cells augmented skin autoimmunity but also afforded superior protection from HSV-1 infection. Our results emphasize that KLRG1 repression is required for optimal TRM cell formation in skin and reveal an important role of Ptpn2 in regulating TRM cell functionality.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Lectins, C-Type/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/immunology , Receptors, Immunologic/immunology , Animals , Autoimmunity/immunology , Female , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Mice , Mice, Inbred C57BL , Skin/immunology
12.
Article in English | MEDLINE | ID: mdl-33753406

ABSTRACT

Memory T-cell responses are partitioned between the blood, secondary lymphoid organs, and nonlymphoid tissues. Tissue-resident memory T (Trm) cells are a population of immune cells that remain permanently in tissues without recirculating in blood. These nonrecirculating cells serve as a principal node in the anamnestic response to invading pathogens and developing malignancies. Here, we contemplate how T-cell tissue residency is defined and shapes protective immunity in the steady state and in the context of disease. We review the properties and heterogeneity of Trm cells, highlight the critical roles these cells play in maintaining tissue homeostasis and eliciting immune pathology, and explore how they might be exploited to treat disease.


Subject(s)
Immunologic Memory , Memory T Cells/physiology , Animals , Humans , Immunotherapy
13.
Cell Rep ; 33(13): 108567, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33378682

ABSTRACT

Concurrent infection with multiple pathogens occurs frequently in individuals and can result in exacerbated infections and altered immunity. However, the impact of such coinfections on immune responses remains poorly understood. Here, we reveal that systemic infection results in an inflammation-induced suppression of local immunity. During localized infection or vaccination in barrier tissues including the skin or respiratory tract, concurrent systemic infection induces a type I interferon-dependent lymphopenia that impairs lymphocyte recruitment to the draining lymph node (dLN) and induces sequestration of lymphocytes in non-draining LN. This contributes to suppressed fibroblastic reticular cell and endothelial cell expansion and dLN remodeling and impairs induction of B cell responses and antibody production. Our data suggest that contemporaneous systemic inflammation constrains the induction of regional immunity.


Subject(s)
Coinfection/immunology , Herpes Simplex/immunology , Inflammation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphoid Tissue/immunology , Simplexvirus/immunology , Animals , Antibody Formation , Fibroblasts/immunology , Herpes Simplex/virology , Interferon Type I/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytic Choriomeningitis/virology , Lymphoid Tissue/metabolism , Lymphopenia/immunology , Mice , Mice, Inbred C57BL , Stromal Cells/immunology , Stromal Cells/metabolism
14.
EMBO J ; 39(2): e103637, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31803974

ABSTRACT

Although adoptive T-cell therapy has shown remarkable clinical efficacy in haematological malignancies, its success in combating solid tumours has been limited. Here, we report that PTPN2 deletion in T cells enhances cancer immunosurveillance and the efficacy of adoptively transferred tumour-specific T cells. T-cell-specific PTPN2 deficiency prevented tumours forming in aged mice heterozygous for the tumour suppressor p53. Adoptive transfer of PTPN2-deficient CD8+ T cells markedly repressed tumour formation in mice bearing mammary tumours. Moreover, PTPN2 deletion in T cells expressing a chimeric antigen receptor (CAR) specific for the oncoprotein HER-2 increased the activation of the Src family kinase LCK and cytokine-induced STAT-5 signalling, thereby enhancing both CAR T-cell activation and homing to CXCL9/10-expressing tumours to eradicate HER-2+ mammary tumours in vivo. Our findings define PTPN2 as a target for bolstering T-cell-mediated anti-tumour immunity and CAR T-cell therapy against solid tumours.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Lymphocyte Activation/immunology , Neoplasms/therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Receptor, ErbB-2/physiology , Receptors, Antigen, T-Cell/immunology , Adoptive Transfer , Animals , Antigen Presentation/immunology , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , Signal Transduction
16.
Immunity ; 51(3): 418-420, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31533052

ABSTRACT

Factors regulating the differentiation of tissue-resident memory T (TRM) cells and tumor-infiltrating lymphocytes (TILs) are incompletely understood. In this issue of Immunity, Li et al. identify Bhlhe40 as a transcriptional regulator of CD8+ TRM cell and TIL persistence and activity by orchestrating metabolic and epigenetic programming.


Subject(s)
Immunologic Memory , Transcription Factors , CD8-Positive T-Lymphocytes , Lymphocytes, Tumor-Infiltrating , Social Control, Formal
17.
Trends Immunol ; 40(8): 735-747, 2019 08.
Article in English | MEDLINE | ID: mdl-31255505

ABSTRACT

Following their activation and expansion in response to foreign threats, many T cells are retained in peripheral tissues without recirculating in the blood. These tissue-resident CD8+ memory T (TRM) cells patrol barrier surfaces and nonlymphoid organs, where their critical role in protecting against viral and bacterial infections is well established. Recent evidence suggests that TRM cells also play a vital part in preventing the development and spread of solid tumors. Here, we discuss the emerging role of TRM cells in anticancer immunity. We highlight defining features of tumor-localizing TRM cells, examine the mechanisms through which they have recently been shown to suppress cancer growth, and explore their potential as future targets of cancer immunotherapy.


Subject(s)
Immunologic Memory , Immunologic Surveillance , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/etiology , Neoplasms/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Biomarkers , Disease Susceptibility , Humans , Immunotherapy , Lymphocytes, Tumor-Infiltrating/pathology , Neoplasms/pathology , Neoplasms/therapy , T-Lymphocyte Subsets/pathology , Tumor Microenvironment/immunology
18.
Cell Stress ; 3(5): 162-164, 2019 Apr 26.
Article in English | MEDLINE | ID: mdl-31225511

ABSTRACT

The immune system can prevent tumour development by engaging in a process termed cancer immunosurveillance, during which immune cells such as T cells restrict tumour growth either by completely eradicating cancer cells in a process of 'elimination' or by suppressing cancer cell outgrowth by establishing a state of tumour-immune 'equilibrium'. Most cancers develop within epithelial layers of tissues but circulating T cells are largely excluded from these epithelial tissue compartments in the absence of infection or overt inflammation. In contrast, CD8+ tissue-resident memory T (TRM) cells reside permanently within epithelial layers of peripheral tissues without recirculating in blood. Accumulating evidence suggests that TRM cells are found in diverse human solid cancers where they correlate with improved prognosis and can protect against tumour challenge in mice. However, the mechanisms through which these cells mediate cancer protection are poorly understood. In our recent study (Park SL et al, Nature 565(7739), 2019) we developed a melanoma model that allowed us to identify a critical role for TRM cells in the establishment and maintenance of tumour-immune equilibrium in skin. Our findings provide insight into the immune cell populations important for maintaining long-term tumour dormancy in peripheral tissues and imply that targeting TRM cells may serve as a novel cancer treatment strategy.

19.
J Exp Med ; 216(7): 1682-1699, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31142588

ABSTRACT

Interleukin (IL)-17-producing CD8+ T (Tc17) cells have emerged as key players in host-microbiota interactions, infection, and cancer. The factors that drive their development, in contrast to interferon (IFN)-γ-producing effector CD8+ T cells, are not clear. Here we demonstrate that the transcription factor TCF-1 (Tcf7) regulates CD8+ T cell fate decisions in double-positive (DP) thymocytes through the sequential suppression of MAF and RORγt, in parallel with TCF-1-driven modulation of chromatin state. Ablation of TCF-1 resulted in enhanced Tc17 cell development and exposed a gene set signature to drive tissue repair and lipid metabolism, which was distinct from other CD8+ T cell subsets. IL-17-producing CD8+ T cells isolated from healthy humans were also distinct from CD8+IL-17- T cells and enriched in pathways driven by MAF and RORγt Overall, our study reveals how TCF-1 exerts central control of T cell differentiation in the thymus by normally repressing Tc17 differentiation and promoting an effector fate outcome.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Hepatocyte Nuclear Factor 1-alpha/metabolism , Interleukin-17/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proto-Oncogene Proteins c-maf/metabolism , Animals , Chromatin/metabolism , Chromatin Immunoprecipitation Sequencing , Flow Cytometry , Hepatocyte Nuclear Factor 1-alpha/physiology , Humans , Lipid Metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocyte Subsets/physiology
20.
Nature ; 566(7745): E10, 2019 02.
Article in English | MEDLINE | ID: mdl-30742076

ABSTRACT

Panel j was inadvertently labelled as panel k in the caption to Fig. 4. Similarly, 'Fig. 4k' should have been 'Fig. 4j' in the sentence beginning 'TNF-α-deficient gBT-I cells were…'. In addition, the surname of author Umaimainthan Palendira was misspelled 'Palendria'. These errors have been corrected online.

SELECTION OF CITATIONS
SEARCH DETAIL
...