Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Immunity ; 55(1): 98-114.e5, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34932944

ABSTRACT

Elevated gene expression of the costimulatory receptor Icos is a hallmark of CD8+ tissue-resident memory (Trm) T cells. Here, we examined the contribution of ICOS in Trm cell differentiation. Upon transfer into WT mice, Icos-/- CD8+ T cells exhibited defective Trm generation but produced recirculating memory populations normally. ICOS deficiency or ICOS-L blockade compromised establishment of CD8+ Trm cells but not their maintenance. ICOS ligation during CD8+ T cell priming did not determine Trm induction; rather, effector CD8+ T cells showed reduced Trm differentiation after seeding into Icosl-/- mice. IcosYF/YF CD8+ T cells were compromised in Trm generation, indicating a critical role for PI3K signaling. Modest transcriptional changes in the few Icos-/- Trm cells suggest that ICOS-PI3K signaling primarily enhances the efficiency of CD8+ T cell tissue residency. Thus, local ICOS signaling promotes production of Trm cells, providing insight into the contribution of costimulatory signals in the generation of tissue-resident populations.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Inducible T-Cell Co-Stimulator Protein/metabolism , Memory T Cells/immunology , Adoptive Transfer , Animals , Antibodies, Blocking/metabolism , Cell Differentiation , Cells, Cultured , Inducible T-Cell Co-Stimulator Ligand/immunology , Inducible T-Cell Co-Stimulator Ligand/metabolism , Inducible T-Cell Co-Stimulator Protein/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
2.
Immunity ; 53(1): 158-171.e6, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32640257

ABSTRACT

Tissue-resident memory (Trm) CD8+ T cells mediate protective immunity in barrier tissues, but the cues promoting Trm cell generation are poorly understood. Sensing of extracellular adenosine triphosphate (eATP) by the purinergic receptor P2RX7 is needed for recirculating CD8+ T cell memory, but its role for Trm cells is unclear. Here we showed that P2RX7 supported Trm cell generation by enhancing CD8+ T cell sensing of TGF-ß, which was necessary for tissue residency. P2RX7-deficient Trm cells progressively decayed in non-lymphoid tissues and expressed dysregulated Trm-specific markers. P2RX7 was required for efficient re-expression of the receptor TGF-ßRII through calcineurin signaling. Forced Tgfbr2 expression rescued P2RX7-deficient Trm cell generation, and TGF-ß sensitivity was dictated by P2RX7 agonists and antagonists. Forced Tgfbr2 also rescued P2RX7-deficient Trm cell mitochondrial function. Sustained P2RX7 signaling was required for long-term Trm cell maintenance, indicating that P2RX7 signaling drives induction and CD8+ T cell durability in barrier sites.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptors, Purinergic P2X7/metabolism , Transforming Growth Factor beta/immunology , Adenosine Triphosphate/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , Calcineurin/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
Proc Natl Acad Sci U S A ; 119(43): e2209021119, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36260745

ABSTRACT

Interleukin-15 (IL-15) is often considered a central regulator of memory CD8+ T cells, based primarily on studies of recirculating subsets. However, recent work identified IL-15-independent CD8+ T cell memory populations, including tissue-resident memory CD8+ T cells (TRM) in some nonlymphoid tissues (NLTs). Whether this reflects the existence of IL-15-insensitive memory CD8+ T cells is unclear. We report that IL-15 complexes (IL-15c) stimulate rapid proliferation and expansion of both tissue-resident and circulating memory CD8+ T cell subsets across lymphoid and nonlymphoid tissues with varying magnitude by tissue and memory subset, in some sites correlating with differing levels of the IL-2Rß. This was conserved for memory CD8+ T cells recognizing distinct antigens and elicited by different pathogens. Following IL-15c-induced expansion, divided cells contracted to baseline numbers and only slowly returned to basal proliferation, suggesting a mechanism to transiently amplify memory populations. Through parabiosis, we showed that IL-15c drive local proliferation of TRM, with a degree of recruitment of circulating cells to some NLTs. Hence, irrespective of homeostatic IL-15 dependence, IL-15 sensitivity is a defining feature of memory CD8+ T cell populations, with therapeutic potential for expansion of TRM and other memory subsets in an antigen-agnostic and temporally controlled fashion.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-15 , Immunologic Memory , T-Lymphocyte Subsets
4.
J Immunol ; 208(7): 1686-1699, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35264459

ABSTRACT

Development of CD8+ central memory T (Tcm) and resident memory T (Trm) cells, which promote immunity in the circulation and in barrier tissues, respectively, is not completely understood. Tcm and Trm cells may arise from common precursors; however, their fate-inducing signals are elusive. We found that virus-specific effector CD8+ T cells display heterogeneous expression of the extracellular ATP sensor P2RX7. P2RX7-high expression is confined, at peak effector phase, to CD62L+ memory precursors, which preferentially form Tcm cells. Among early effector CD8+ T cells, asymmetrical P2RX7 distribution correlated with distinct transcriptional signatures, with P2RX7-high cells enriched for memory and tissue residency sets. P2RX7-high early effectors preferentially form both Tcm and Trm cells. Defective Tcm and Trm cell formation in P2RX7 deficiency is significantly reverted when the transcriptional repressor Zeb2 is ablated. Mechanistically, P2RX7 negatively regulates Zeb2 expression, at least partially through TGF-ß sensing in early effector CD8+ T cells. Our study indicates that unequal P2RX7 upregulation in effector CD8+ T cells is a foundational element of the early Tcm/Trm fate.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Animals , Mice , Mice, Inbred C57BL , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism , Transforming Growth Factor beta/metabolism
5.
Int Immunol ; 32(9): 583-587, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32620009

ABSTRACT

Independent studies over the last decade have characterized the properties of non-circulating CD8+ 'resident' memory T cells (TRM), which offer barrier protective immunity in non-lymphoid tissues and CD4+ follicular helper T cells (TFH), which mediate B-cell help in lymphoid sites. Despite their very different biological roles in the immune system, intriguing parallels have been noted between the trafficking properties and differentiation cues of these populations, parallels which have only sharpened with recent findings. In this review, we explore the features that underlie these similarities and discuss whether these indicate meaningful homologies in the development of CD8+ TRM and CD4+ TFH or reflect resemblances which are only 'skin-deep'.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Animals , Humans
6.
J Immunol ; 203(4): 946-955, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31243092

ABSTRACT

Recent studies have characterized populations of memory CD8+ T cells that do not recirculate through the blood but are, instead, retained in nonlymphoid tissues. Such CD8+ tissue resident memory T cells (TRM) are critical for pathogen control at barrier sites. Identifying TRM and defining the basis for their tissue residency is therefore of considerable importance for understanding protective immunity and improved vaccine design. Expression of the molecule CD69 is widely used as a definitive marker for TRM, yet it is unclear whether CD69 is universally required for producing or retaining TRM Using multiple mouse models of acute immunization, we found that the functional requirement for CD69 was highly variable, depending on the tissue examined, playing no detectable role in generation of TRM at some sites (such as the small intestine), whereas CD69 was critical for establishing resident cells in the kidney. Likewise, forced expression of CD69 (but not expression of a CD69 mutant unable to bind the egress factor S1PR1) promoted CD8+ TRM generation in the kidney but not in other tissues. Our findings indicate that the functional relevance of CD69 in generation and maintenance of CD8+ TRM varies considerably, chiefly dependent on the specific nonlymphoid tissue studied. Together with previous reports that suggest uncoupling of CD69 expression and tissue residency, these findings prompt caution in reliance on CD69 expression as a consistent marker of CD8+ TRM.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Lectins, C-Type/immunology , T-Lymphocyte Subsets/immunology , Animals , Mice
7.
bioRxiv ; 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38895229

ABSTRACT

Interleukin-7 (IL-7) is considered a critical regulator of memory CD8+ T cell homeostasis, but this is primarily based on analysis of circulating and not tissue-resident memory (TRM) subsets. Furthermore, the cell-intrinsic requirement for IL-7 signaling during memory homeostasis has not been directly tested. Using inducible deletion, we found that Il7ra loss had only a modest effect on persistence of circulating memory and TRM subsets and that IL-7Rα was primarily required for normal basal proliferation. Loss of IL-15 signaling imposed heightened IL-7Rα dependence on memory CD8+ T cells, including TRM populations previously described as IL-15-independent. In the absence of IL-15 signaling, IL-7Rα was upregulated, and loss of IL-7Rα signaling reduced proliferation in response to IL-15, suggesting cross-regulation in memory CD8+ T cells. Thus, across subsets and tissues, IL-7 and IL-15 act in concert to support memory CD8+ T cells, conferring resilience to altered availability of either cytokine.

8.
Sci Immunol ; 9(96): eadj8356, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38941479

ABSTRACT

KLRG1+ CD8 T cells persist for months after clearance of acute infections and maintain high levels of effector molecules, contributing protective immunity against systemic pathogens. Upon secondary infection, these long-lived effector cells (LLECs) are incapable of forming other circulating KLRG1- memory subsets such as central and effector memory T cells. Thus, KLRG1+ memory T cells are frequently referred to as a terminally differentiated population that is relatively short lived. Here, we show that after viral infection of mice, effector cells derived from LLECs rapidly enter nonlymphoid tissues and reduce pathogen burden but are largely dependent on receiving antigen cues from vascular endothelial cells. Single-cell RNA sequencing reveals that secondary memory cells in nonlymphoid tissues arising from either KLRG1+ or KLRG1- memory precursors develop a similar resident memory transcriptional signature. Thus, although LLECs cannot differentiate into other circulating memory populations, they still retain the flexibility to enter tissues and establish residency.


Subject(s)
Immunologic Memory , Lectins, C-Type , Memory T Cells , Receptors, Immunologic , Animals , Female , Mice , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Lectins, C-Type/immunology , Memory T Cells/immunology , Mice, Inbred C57BL , Mice, Knockout , Receptors, Immunologic/immunology
9.
Front Chem ; 11: 1093435, 2023.
Article in English | MEDLINE | ID: mdl-36874070

ABSTRACT

In view of the composition characteristics of lithium, calcium and bromine rich in Nanyishan oil and gas field brine of western Qaidam Basin, Qinghai Province, as well as based on the results reported in relevant literature, the phase equilibrium relationship of ternary system LiBr-CaBr2-H2O at 298.15 K was studied by isothermal dissolution equilibrium method. The equilibrium solid phase crystallization regions, as well as the compositions of invariant point, in phase diagram of this ternary system were clarified. On basis of the above ternary system research, the stable phase equilibria of quaternary systems (LiBr-NaBr-CaBr2-H2O, LiBr-KBr-CaBr2-H2O and LiBr-MgBr2-CaBr2-H2O), as well as quinary systems (LiBr-NaBr-KBr-CaBr2-H2O, LiBr-NaBr-MgBr2-CaBr2-H2O and LiBr-KBr-MgBr2-CaBr2-H2O) were further carried out at 298.15 K. According to the above experimental results, the corresponding phase diagrams at 298.15 K were drawn, which revealed the phase relationship of each component in solution and the law of crystallization and dissolution, and meanwhile summarized changing trends. The research results of this paper lay a foundation for further research on the multitemperature phase equilibria and thermodynamic properties of lithium and bromine containing high-component brine system in later stage, and also provide basic thermodynamic data for guiding the comprehensive development and utilization of this oil and gas field brine resource.

10.
bioRxiv ; 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37131831

ABSTRACT

Sensing of extracellular metabolites controls CD8+ T cell function. Their accumulation can occur through export by specialized molecules, such as the release channel Pannexin-1 (Panx1). Whether Panx1 controls CD8+ T cell immune responses to antigen, however, has not been previously addressed. Here, we report that T cell-specific Panx1 is needed for CD8+ T cell responses to viral infections and cancer. We found that CD8-specific Panx1 favors memory CD8+ T cell survival primarily through ATP export and induction of mitochondrial metabolism. CD8-specific Panx1 is also crucial for the effector expansion of CD8+ T cells, however this regulation occurs independently of eATP. Instead, our results suggest a connection between Panx1-induced extracellular lactate accumulation and the complete activation of effector CD8+ T cells. In summary, Panx1 regulates effector and memory CD8+ T cells through export of distinct metabolites and by engaging different metabolic and signaling pathways.

11.
Cancer Immunol Res ; 10(7): 871-884, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35588154

ABSTRACT

Expression of the purinergic receptor P2RX7 by CD8+ T cells promotes the generation of memory populations following acute infections. However, data suggest that P2RX7 may limit the efficacy of antitumor responses. Herein, we show that P2RX7 is beneficial for optimal melanoma control in a mouse CD8+ T-cell adoptive transfer model. Tumor-specific P2rx7-/- CD8+ T cells exhibited impaired mitochondrial maintenance and function but did not display signs of overt exhaustion early in the antitumor response. However, as the tumor burden increased, the relative frequency of P2RX7-deficient CD8+ T cells declined within the tumor; this correlated with reduced proliferation, increased apoptosis, and mitochondrial dysfunction. Extending these studies, we found that the transient in vitro stimulation of P2RX7 using the ATP analogue BzATP led to enhanced B16 melanoma control by CD8+ T cells. These findings are in keeping with the concept that extracellular ATP (eATP) sensing by P2RX7 on CD8+ T cells is required for their ability to efficiently eliminate tumors by promoting mitochondrial fitness and underscore the potential for P2RX7 stimulation as a novel therapeutic treatment to enhance tumor immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Melanoma, Experimental , Adenosine Triphosphate/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell- and Tissue-Based Therapy , Immunotherapy, Adoptive , Melanoma, Experimental/metabolism , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism
12.
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
13.
Nat Commun ; 12(1): 2308, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33863906

ABSTRACT

Conventional T cells are selected by peptide-MHC expressed by cortical epithelial cells in the thymus, and not by cortical thymocytes themselves that do not express MHC I or MHC II. Instead, cortical thymocytes express non-peptide presenting MHC molecules like CD1d and MR1, and promote the selection of PLZF+ iNKT and MAIT cells, respectively. Here, we report an inducible class-I transactivator mouse that enables the expression of peptide presenting MHC I molecules in different cell types. We show that MHC I expression in DP thymocytes leads to expansion of peptide specific PLZF+ innate-like (PIL) T cells. Akin to iNKT cells, PIL T cells differentiate into three functional effector subsets in the thymus, and are dependent on SAP signaling. We demonstrate that PIL and NKT cells compete for a narrow niche, suggesting that the absence of peptide-MHC on DP thymocytes facilitates selection of non-peptide specific lymphocytes.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Immunity, Innate , Thymocytes/immunology , Thymus Gland/immunology , Animals , Cell Differentiation/immunology , Histocompatibility Antigens Class I/immunology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Mucosal-Associated Invariant T Cells/immunology , Natural Killer T-Cells/immunology , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Thymocytes/metabolism , Thymus Gland/cytology
14.
J BUON ; 25(3): 1636-1642, 2020.
Article in English | MEDLINE | ID: mdl-32862616

ABSTRACT

PURPOSE: The study aimed to explore the regulatory mechanism of micro ribonucleic acid (miR)-130a in the autophagy of bladder cancer cells through cylindromatosis (CYLD). METHODS: Human bladder cancer T24 cell line was used as the objects of the study. After miR-130a was knocked down using small-interfering RNA (siRNA) in T24 cell line, the changes in expressions of miR-130a and CYLD in each group were detected via quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The cell proliferation in each group was detected using cell counting kit-8 (CCK8) assay and flow cytometry. The changes in mRNA and protein levels of microtubule-associated protein 1 light chain 3 (LC3) and Beclin1 were determined using qRT-PCR and Western blotting. The autolysosomes were detected through acridine orange (AO)/ethidium staining bromide (ER) staining. Moreover, CYLD was knocked down using siRNA, and then the changes in mRNA expressions of miR-130a, LC3 and Beclin1 in each group were detected through qRT-PCR. RESULTS: After interference in miR-130a with siRNA, miR-130a-siRNA group had a significantly lower mRNA expression of miR-130a compared with NC-siRNA group and a significantly higher mRNA expression of CYLD (p<0.05), obviously inhibited cell proliferation (p<0.05), and decreased significantly mRNA and protein expressions of LC3 showing Beclin1 (p<0.05), and an evidently smaller number of autolysosomes. After knockdown of CYLD using siRNA, the mRNA expression of miR-130a had no significant changes (p>0.05), while the mRNA expressions of LC3 and Beclin1 declined significantly in CYLD-siRNA group compared with those in NC-siRNA group (p<0.05). CONCLUSION: MiR-130 can promote the autophagy of bladder cancer cells through regulating CYLD, thus facilitating the proliferation of tumor cells.


Subject(s)
Autophagy/genetics , Deubiquitinating Enzyme CYLD/genetics , MicroRNAs/genetics , Urinary Bladder Neoplasms/genetics , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Urinary Bladder Neoplasms/pathology
15.
Science ; 367(6475)2020 01 17.
Article in English | MEDLINE | ID: mdl-31949051

ABSTRACT

Negative checkpoint regulators (NCRs) temper the T cell immune response to self-antigens and limit the development of autoimmunity. Unlike all other NCRs that are expressed on activated T lymphocytes, V-type immunoglobulin domain-containing suppressor of T cell activation (VISTA) is expressed on naïve T cells. We report an unexpected heterogeneity within the naïve T cell compartment in mice, where loss of VISTA disrupted the major quiescent naïve T cell subset and enhanced self-reactivity. Agonistic VISTA engagement increased T cell tolerance by promoting antigen-induced peripheral T cell deletion. Although a critical player in naïve T cell homeostasis, the ability of VISTA to restrain naïve T cell responses was lost under inflammatory conditions. VISTA is therefore a distinctive NCR of naïve T cells that is critical for steady-state maintenance of quiescence and peripheral tolerance.


Subject(s)
B7 Antigens/physiology , Membrane Proteins/physiology , Peripheral Tolerance/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal , B7 Antigens/genetics , Lymphocyte Activation , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Peripheral Tolerance/genetics , Receptors, Antigen, T-Cell/physiology
16.
Sci Rep ; 6: 32336, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27578425

ABSTRACT

Tripartite motif 14 (TRIM14) was reported to function as a mitochondrial signaling adaptor in mediating innate immune responses. However, the involvement of TRIM14 in host defense against viral infection and molecular mechanisms remain unclear. Here, we demonstrated that enforced expression of TRIM14 could potently inhibit the infection and replication of HCV in hepatocytes, whereas TRIM14 knockout cells became more susceptible to HCV infection. Interestingly, further experiments revealed that such anti-HCV activity was independent of activating the NF-κB or interferon pathways but required the C-terminal SPRY domain of no signaling capacity. In searching for mechanisms how TRIM14 exerts its antiviral function we found that TRIM14 interacted with HCV encoded non-structural protein NS5A and could strongly induce its degradation dependent on the NS5A1 subdomain. Interestingly extensive domain mapping analyses revealed that NS5A degradation was mediated by the highly conserved SPRY domain of TRIM14, which might involve the K48 ubiquitination pathway. Collectively, our work uncovered a new mechanism responsible for host defense against HCV infection, and could potentially aid the development of novel anti-HCV therapeutics.


Subject(s)
Carrier Proteins/genetics , Hepacivirus/genetics , Hepatitis C/genetics , Viral Nonstructural Proteins/genetics , B30.2-SPRY Domain/genetics , Gene Knockout Techniques , Hepacivirus/chemistry , Hepacivirus/pathogenicity , Hepatitis C/virology , Hepatocytes , Host-Pathogen Interactions/genetics , Humans , Intracellular Signaling Peptides and Proteins , Proteolysis , Tripartite Motif Proteins , Ubiquitination , Viral Nonstructural Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL