Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Immunol ; 25(1): 117-128, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38012417

ABSTRACT

In cancer and infections, self-renewing stem-like CD8+ T cells mediate the response of immunotherapies and replenish terminally exhausted T cells and effector-like T cells. However, the programs governing the lineage choice in chimeric antigen receptor (CAR) T cells are unclear. Here, by simultaneously profiling single-cell chromatin accessibility and transcriptome in the same CAR T cells, we identified heterogeneous chromatin states within CD8+ T cell subsets that foreshadowed transcriptional changes and were primed for regulation by distinct transcription factors. Transcription factors that controlled each CD8+ T cell subset were regulated by high numbers of enhancers and positioned as hubs of gene networks. FOXP1, a hub in the stem-like network, promoted expansion and stemness of CAR T cells and limited excessive effector differentiation. In the effector network, KLF2 enhanced effector CD8+ T cell differentiation and prevented terminal exhaustion. Thus, we identified gene networks and hub transcription factors that controlled the differentiation of stem-like CD8+ CAR T cells into effector or exhausted CD8+ CAR T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Transcription Factors , Transcription Factors/genetics , T-Lymphocyte Subsets , Cell Differentiation , Chromatin
2.
J Clin Invest ; 132(7)2022 04 01.
Article in English | MEDLINE | ID: mdl-35143421

ABSTRACT

The functional integrity of CD8+ T cells is tightly coupled to metabolic reprogramming, but how oxidative stress directs CD8+ T cell metabolic fitness in the tumor microenvironment (TME) remains elusive. Here, we report that SUMO-specific protease 7 (SENP7) senses oxidative stress to maintain the CD8+ T cell metabolic state and antitumor functions. SENP7-deficient CD8+ T cells exhibited decreased glycolysis and oxidative phosphorylation, resulting in attenuated proliferation in vitro and dampened antitumor functions in vivo. Mechanistically, CD8+ T cell-derived ROS triggered cytosolic SENP7-mediated PTEN deSUMOylation, thereby promoting PTEN degradation and preventing PTEN-dependent metabolic defects. Importantly, lowering T cell-intrinsic ROS restricted SENP7 cytosolic translocation and repressed CD8+ T cell metabolic and functional activity in human colorectal cancer samples. Our findings reveal that SENP7, as an oxidative stress sensor, sustains CD8+ T cell metabolic fitness and effector functions and unveil an oxidative stress-sensing machinery in tumor-infiltrating CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , CD8-Positive T-Lymphocytes/metabolism , Endopeptidases/metabolism , Humans , Neoplasms/metabolism , Oxidative Stress , Tumor Microenvironment
3.
J Clin Invest ; 131(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34403361

ABSTRACT

Proper metabolic activities facilitate T cell expansion and antitumor function; however, the mechanisms underlying disruption of the T cell metabolic program and function in the tumor microenvironment (TME) remain elusive. Here, we show a zinc finger protein 91-governed (ZFP91-governed) mechanism that disrupts the metabolic pathway and antitumor activity of tumor-infiltrating T cells. Single-cell RNA-Seq revealed that impairments in T cell proliferation and activation correlated with ZFP91 in tissue samples from patients with colorectal cancer. T cell-specific deletion of Zfp91 in mice led to enhanced T cell proliferation and potentiated T cell antitumor function. Loss of ZFP91 increased mammalian target of rapamycin complex 1 (mTORC1) activity to drive T cell glycolysis. Mechanistically, T cell antigen receptor-dependent (TCR-dependent) ZFP91 cytosolic translocation promoted protein phosphatase 2A (PP2A) complex assembly, thereby restricting mTORC1-mediated metabolic reprogramming. Our results demonstrate that ZFP91 perturbs T cell metabolic and functional states in the TME and suggest that targeting ZFP91 may improve the efficacy of cancer immunotherapy.


Subject(s)
Cytotoxicity, Immunologic , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes/immunology , Ubiquitin-Protein Ligases/physiology , Animals , Colorectal Neoplasms/immunology , Glycolysis , Humans , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 1/physiology , Mice , Mice, Inbred C57BL , Protein Phosphatase 2/metabolism , T-Lymphocytes/metabolism , Tumor Microenvironment
4.
Mol Cell ; 81(5): 940-952.e5, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33434504

ABSTRACT

STING-dependent cytosolic DNA sensing in dendritic cells (DCs) initiates antitumor immune responses, but how STING signaling is metabolically regulated in the tumor microenvironment remains unknown. Here, we show that oxidative stress is required for STING-induced DC antitumor function through a process that directs SUMO-specific protease 3 (SENP3) activity. DC-specific deletion of Senp3 drives tumor progression by blunting STING-dependent type-I interferon (IFN) signaling in DCs and dampening antitumor immune responses. DC-derived reactive oxygen species (ROS) trigger SENP3 accumulation and the SENP3-IFI204 interaction, thereby catalyzing IFI204 deSUMOylation and boosting STING signaling activation in mice. Consistently, SENP3 senses ROS to facilitate STING-dependent DC activity in tissue samples from colorectal cancer patients. Our results reveal that oxidative stress as a metabolic regulator promotes STING-mediated DC antitumor immune responses and highlights SENP3 as an overflow valve for STING signaling induction in the metabolically abnormal tumor microenvironment.


Subject(s)
Colorectal Neoplasms/genetics , Cysteine Endopeptidases/genetics , Dendritic Cells/immunology , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Allografts , Animals , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Cysteine Endopeptidases/immunology , Dendritic Cells/pathology , Female , HEK293 Cells , Humans , Interferon Type I/genetics , Interferon Type I/immunology , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/immunology , Oxidative Stress , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Signal Transduction , Survival Analysis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
5.
J Exp Med ; 218(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33355624

ABSTRACT

Autophagy programs the metabolic and functional fitness of regulatory T (T reg) cells to establish immune tolerance, yet the mechanisms governing autophagy initiation in T reg cells remain unclear. Here, we show that the E3 ubiquitin ligase ZFP91 facilitates autophagy activation to sustain T reg cell metabolic programming and functional integrity. T reg cell-specific deletion of Zfp91 caused T reg cell dysfunction and exacerbated colonic inflammation and inflammation-driven colon carcinogenesis. TCR-triggered autophagy induction largely relied on T reg cell-derived ZFP91 to restrict hyperglycolysis, which is required for the maintenance of T reg cell homeostasis. Mechanistically, ZFP91 rapidly translocated from the nucleus to the cytoplasm in response to TCR stimulation and then mediated BECN1 ubiquitination to promote BECN1-PIK3C3 complex formation. Therefore, our results highlight a ZFP91-dependent mechanism promoting TCR-initiated autophagosome maturation to maintain T reg cell homeostasis and function.


Subject(s)
Homeostasis/immunology , T-Lymphocytes, Regulatory/immunology , Ubiquitin-Protein Ligases/immunology , Animals , Autophagy/immunology , Beclin-1/immunology , Carcinogenesis/immunology , Colon/immunology , Disease Models, Animal , Female , Immune Tolerance/immunology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/immunology , Ubiquitination/immunology
6.
Cell Metab ; 30(2): 290-302.e5, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31204281

ABSTRACT

CD8+ T cell expansions and functions rely on glycolysis, but the mechanisms underlying CD8+ T cell glycolytic metabolism remain elusive. Here, we show that acylglycerol kinase (AGK) is required for the establishment and maintenance of CD8+ T cell metabolic and functional fitness. AGK deficiency dampens CD8+ T cell antitumor functions in vivo and perturbs CD8+ T cell proliferation in vitro. Activation of phosphatidylinositol-3-OH kinase (PI3K)-mammalian target of rapamycin (mTOR) signaling, which mediates elevated CD8+ T cell glycolysis, is tightly dependent on AGK kinase activity. Mechanistically, T cell antigen receptor (TCR)- and CD28-stimulated recruitment of PTEN to the plasma membrane facilitates AGK-PTEN interaction and AGK-triggered PTEN phosphorylation, thereby restricting PTEN phosphatase activity in CD8+ T cells. Collectively, these results demonstrate that AGK maintains CD8+ T cell metabolic and functional state by restraining PTEN activity and highlight a critical role for AGK in CD8+ T cell metabolic programming and effector function.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Phosphotransferases (Alcohol Group Acceptor)/immunology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Female , Male , Melanoma, Experimental/pathology , Mice , Mice, Transgenic
7.
J Exp Med ; 215(9): 2463-2476, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30115741

ABSTRACT

Metabolic programs are crucial for regulatory T (T reg) cell stability and function, but the underlying mechanisms that regulate T reg cell metabolism are elusive. Here, we report that lysosomal TRAF3IP3 acts as a pivotal regulator in the maintenance of T reg cell metabolic fitness. T reg-specific deletion of Traf3ip3 impairs T reg cell function, causing the development of inflammatory disorders and stronger antitumor T cell responses in mice. Excessive mechanistic target of rapamycin complex 1 (mTORC1)-mediated hyper-glycolytic metabolism is responsible for the instability of TRAF3IP3-deficient T reg cells. Mechanistically, TRAF3IP3 restricts mTORC1 signaling by recruiting the serine-threonine phosphatase catalytic subunit (PP2Ac) to the lysosome, thereby facilitating the interaction of PP2Ac with the mTORC1 component Raptor. Our results define TRAF3IP3 as a metabolic regulator in T reg cell stability and function and suggest a lysosome-specific mTORC1 signaling mechanism that regulates T reg cell metabolism.


Subject(s)
Carrier Proteins , Glycolysis , Lysosomes , Membrane Proteins , Signal Transduction , T-Lymphocytes, Regulatory , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Glycolysis/genetics , Glycolysis/immunology , Lysosomes/genetics , Lysosomes/immunology , Lysosomes/metabolism , Lysosomes/pathology , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Regulatory-Associated Protein of mTOR/genetics , Regulatory-Associated Protein of mTOR/immunology , Regulatory-Associated Protein of mTOR/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
8.
Nat Commun ; 9(1): 3157, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30089837

ABSTRACT

Regulatory T (Treg) cells are essential for maintaining immune homeostasis and tolerance, but the mechanisms regulating the stability and function of Treg cells have not been fully elucidated. Here we show SUMO-specific protease 3 (SENP3) is a pivotal regulator of Treg cells that functions by controlling the SUMOylation and nuclear localization of BACH2. Treg cell-specific deletion of Senp3 results in T cell activation, autoimmune symptoms and enhanced antitumor T cell responses. SENP3-mediated BACH2 deSUMOylation prevents the nuclear export of BACH2, thereby repressing the genes associated with CD4+ T effector cell differentiation and stabilizing Treg cell-specific gene signatures. Notably, SENP3 accumulation triggered by reactive oxygen species (ROS) is involved in Treg cell-mediated tumor immunosuppression. Our results not only establish the role of SENP3 in the maintenance of Treg cell stability and function via BACH2 deSUMOylation but also clarify the function of SENP3 in the regulation of ROS-induced immune tolerance.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Immune Tolerance/immunology , Peptide Hydrolases/metabolism , Sumoylation/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Active Transport, Cell Nucleus , Animals , Antineoplastic Agents/metabolism , Autoimmunity/immunology , Bone Marrow Cells , CD4-Positive T-Lymphocytes , Cell Differentiation/immunology , Cell Line, Tumor , Cell Nucleus/immunology , Cysteine Endopeptidases , Female , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , Homeostasis/immunology , Humans , Lymphocyte Activation/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Hydrolases/genetics , Reactive Oxygen Species , T-Lymphocytes, Regulatory/pathology
9.
Front Plant Sci ; 8: 1345, 2017.
Article in English | MEDLINE | ID: mdl-28824680

ABSTRACT

AcCATPO is a plant catalase-phenol oxidase recently identified from red amaranth. Its physiological function remains unexplored. As the starting step of functional analysis, here we report its subcellular localization and a non-canonical targeting signal. Commonly used bioinformatics programs predicted a peroxisomal localization for AcCATPO, but failed in identification of canonical peroxisomal targeting signals (PTS). The C-terminal GFP tagging led the fusion protein AcCATPO-GFP to the cytosol and the nucleus, but N-terminal tagging directed the GFP-AcCATPO to peroxisomes and nuclei, in transgenic tobacco. Deleting the tripeptide (PTM) at the extreme C-terminus almost ruled out the peroxisomal localization of GFP-AcCATPOΔ3, and removing the C-terminal decapeptide completely excluded peroxisomes as the residence of GFP-AcCATPOΔ10. Furthermore, this decapeptide as a targeting signal could import GFP-10aa to the peroxisome exclusively. Taken together, these results demonstrate that AcCATPO is localized to the peroxisome and the nucleus, and its peroxisomal localization is attributed to a non-canonical PTS1, the C-terminal decapeptide which contains an internal SRL motif and a conserved tripeptide P-S/T-I/M at the extreme of C-terminus. This work may further the study as to the physiological function of AcCATPO, especially clarify its involvement in betalain biosynthesis, and provide a clue to elucidate more non-canonic PTS.

10.
Front Plant Sci ; 6: 1228, 2015.
Article in English | MEDLINE | ID: mdl-26779247

ABSTRACT

Betalains are a group of nitrogen-containing pigments that color plants in most families of Caryophyllales. Their biosynthesis has long been proposed to begin with hydroxylation of L-tyrosine to L-DOPA through monophenolase activity of tyrosinase, but biochemical evidence in vivo remains lacking. Here we report that a Group 4 catalase, catalase-phenol oxidase (named as AcCATPO), was identified, purified and characterized from leaves of Amaranthus cruentus, a betalain plant. The purified enzyme appeared to be a homotrimeric protein composed of subunits of about 58 kDa, and demonstrated not only the catalase activity toward H2O2, but also the monophenolase activity toward L-tyrosine and diphenolase activity toward L-DOPA. Its catalase and phenol oxidase activities were inhibited by common classic catalase and tyrosinase inhibitors, respectively. All its peptide fragments identified by nano-LC-MS/MS were targeted to catalases, and matched with a cDNA-encoded polypeptide which contains both classic catalase and phenol oxidase active sites. These sites were also present in catalases of non-betalain plants analyzed. AcCATPO transcript abundance was positively correlated with the ratio of betaxanthin to betacyanin in both green and red leaf sectors of A. tricolor. These data shows that the fourth group catalase, catalase-phenol oxidase, is present in plant, and might be involved in betaxanthin biosynthesis.

SELECTION OF CITATIONS
SEARCH DETAIL
...