Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 24(11): 1921-1932, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37813964

ABSTRACT

The malate shuttle is traditionally understood to maintain NAD+/NADH balance between the cytosol and mitochondria. Whether the malate shuttle has additional functions is unclear. Here we show that chronic viral infections induce CD8+ T cell expression of GOT1, a central enzyme in the malate shuttle. Got1 deficiency decreased the NAD+/NADH ratio and limited antiviral CD8+ T cell responses to chronic infection; however, increasing the NAD+/NADH ratio did not restore T cell responses. Got1 deficiency reduced the production of the ammonia scavenger 2-ketoglutarate (2-KG) from glutaminolysis and led to a toxic accumulation of ammonia in CD8+ T cells. Supplementation with 2-KG assimilated and detoxified ammonia in Got1-deficient T cells and restored antiviral responses. These data indicate that the major function of the malate shuttle in CD8+ T cells is not to maintain the NAD+/NADH balance but rather to detoxify ammonia and enable sustainable ammonia-neutral glutamine catabolism in CD8+ T cells during chronic infection.


Subject(s)
Ketoglutaric Acids , NAD , Humans , Oxidation-Reduction , NAD/metabolism , Ketoglutaric Acids/metabolism , Ammonia , Malates/metabolism , CD8-Positive T-Lymphocytes/metabolism , Persistent Infection , Antiviral Agents
2.
Immunity ; 54(7): 1561-1577.e7, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34102100

ABSTRACT

A common metabolic alteration in the tumor microenvironment (TME) is lipid accumulation, a feature associated with immune dysfunction. Here, we examined how CD8+ tumor infiltrating lymphocytes (TILs) respond to lipids within the TME. We found elevated concentrations of several classes of lipids in the TME and accumulation of these in CD8+ TILs. Lipid accumulation was associated with increased expression of CD36, a scavenger receptor for oxidized lipids, on CD8+ TILs, which also correlated with progressive T cell dysfunction. Cd36-/- T cells retained effector functions in the TME, as compared to WT counterparts. Mechanistically, CD36 promoted uptake of oxidized low-density lipoproteins (OxLDL) into T cells, and this induced lipid peroxidation and downstream activation of p38 kinase. Inhibition of p38 restored effector T cell functions in vitro, and resolution of lipid peroxidation by overexpression of glutathione peroxidase 4 restored functionalities in CD8+ TILs in vivo. Thus, an oxidized lipid-CD36 axis promotes intratumoral CD8+ T cell dysfunction and serves as a therapeutic avenue for immunotherapies.


Subject(s)
CD36 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Lipid Peroxidation/physiology , Lipoproteins, LDL/metabolism , Neoplasms/metabolism , Receptors, Scavenger/metabolism , Animals , Biological Transport/physiology , Cell Line, Tumor , HEK293 Cells , Humans , Leukocytes, Mononuclear/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Microenvironment/physiology
3.
Cell ; 161(4): 750-61, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25957683

ABSTRACT

Memory T cells are critical for long-term immunity against reinfection and require interleukin-7 (IL-7), but the mechanisms by which IL-7 controls memory T cell survival, particularly metabolic fitness, remain elusive. We discover that IL-7 induces expression of the glycerol channel aquaporin 9 (AQP9) in virus-specific memory CD8+ T cells, but not naive cells, and that AQP9 is vitally required for their long-term survival. AQP9 deficiency impairs glycerol import into memory CD8+ T cells for fatty acid esterification and triglyceride (TAG) synthesis and storage. These defects can be rescued by ectopic expression of TAG synthases, which restores lipid stores and memory T cell survival. Finally, we find that TAG synthesis is a central component of IL-7-mediated survival of human and mouse memory CD8+T cells. This study uncovers the metabolic mechanisms by which IL-7 tailors the metabolism of memory T cells to promote their longevity and fast response to rechallenge.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Survival , Immunologic Memory , Triglycerides/metabolism , Animals , Aquaporins/metabolism , Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/metabolism , Glycerol/metabolism , Humans , Interleukin-7/metabolism , Lymphocytic choriomeningitis virus/physiology , Mice , Signal Transduction
4.
Cell ; 162(6): 1217-28, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26321681

ABSTRACT

Activated T cells engage aerobic glycolysis and anabolic metabolism for growth, proliferation, and effector functions. We propose that a glucose-poor tumor microenvironment limits aerobic glycolysis in tumor-infiltrating T cells, which suppresses tumoricidal effector functions. We discovered a new role for the glycolytic metabolite phosphoenolpyruvate (PEP) in sustaining T cell receptor-mediated Ca(2+)-NFAT signaling and effector functions by repressing sarco/ER Ca(2+)-ATPase (SERCA) activity. Tumor-specific CD4 and CD8 T cells could be metabolically reprogrammed by increasing PEP production through overexpression of phosphoenolpyruvate carboxykinase 1 (PCK1), which bolstered effector functions. Moreover, PCK1-overexpressing T cells restricted tumor growth and prolonged the survival of melanoma-bearing mice. This study uncovers new metabolic checkpoints for T cell activity and demonstrates that metabolic reprogramming of tumor-reactive T cells can enhance anti-tumor T cell responses, illuminating new forms of immunotherapy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Melanoma/therapy , Monitoring, Immunologic , Phosphoenolpyruvate/metabolism , Tumor Microenvironment , Animals , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Glycolysis , Hexokinase/metabolism , Immunotherapy , Mice , NFATC Transcription Factors/metabolism , Receptors, Antigen, T-Cell/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction , Transforming Growth Factor beta/immunology
5.
Immunity ; 50(5): 1218-1231.e5, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30952607

ABSTRACT

Patients with the neurological disorder HSAN-I suffer frequent infections, attributed to a lack of pain sensation and failure to seek care for minor injuries. Whether protective CD8+ T cells are affected in HSAN-I patients remains unknown. Here, we report that HSAN-I-associated mutations in serine palmitoyltransferase subunit SPTLC2 dampened human T cell responses. Antigen stimulation and inflammation induced SPTLC2 expression, and murine T-cell-specific ablation of Sptlc2 impaired antiviral-T-cell expansion and effector function. Sptlc2 deficiency reduced sphingolipid biosynthetic flux and led to prolonged activation of the mechanistic target of rapamycin complex 1 (mTORC1), endoplasmic reticulum (ER) stress, and CD8+ T cell death. Protective CD8+ T cell responses in HSAN-I patient PBMCs and Sptlc2-deficient mice were restored by supplementing with sphingolipids and pharmacologically inhibiting ER stress-induced cell death. Therefore, SPTLC2 underpins protective immunity by translating extracellular stimuli into intracellular anabolic signals and antagonizes ER stress to promote T cell metabolic fitness.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hereditary Sensory and Autonomic Neuropathies/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Serine C-Palmitoyltransferase/genetics , Animals , Cell Proliferation , Cells, Cultured , Cytokines/biosynthesis , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/immunology , Female , Humans , Lymphocytic Choriomeningitis/virology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Signal Transduction/immunology , Sphingolipids/biosynthesis
6.
Eur J Immunol ; 54(7): e2350825, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38650034

ABSTRACT

Cyclosporin A is a well-established immunosuppressive drug used to treat or prevent graft-versus-host disease, the rejection of organ transplants, autoimmune disorders, and leukemia. It exerts its immunosuppressive effects by inhibiting calcineurin-mediated dephosphorylation of the nuclear factor of activated T cells (NFAT), thus preventing its nuclear entry and suppressing T cell activation. Here we report an unexpected immunostimulatory effect of cyclosporin A in activating the mammalian target of rapamycin complex 1 (mTORC1), a crucial metabolic hub required for T cell activation. Through screening a panel of tool compounds known to regulate mTORC1 activation, we found that cyclosporin A activated mTORC1 in CD8+ T cells in a 3-phosphoinositide-dependent protein kinase 1 (PDK1) and protein kinase B (PKB/AKT)-dependent manner. Mechanistically, cyclosporin A inhibited the calcineurin-mediated AKT dephosphorylation, thereby stabilizing mTORC1 signaling. Cyclosporin A synergized with mTORC1 pathway inhibitors, leading to potent suppression of proliferation and cytokine production in CD8+ T cells and an increase in the killing of acute T cell leukemia cells. Consequently, relying solely on CsA is insufficient to achieve optimal therapeutic outcomes. It is necessary to simultaneously target both the calcineurin-NFAT pathway and the mTORC1 pathway to maximize therapeutic efficacy.


Subject(s)
CD8-Positive T-Lymphocytes , Cyclosporine , Immunosuppressive Agents , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 1 , Signal Transduction , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Cyclosporine/pharmacology , Mechanistic Target of Rapamycin Complex 1/metabolism , Animals , Immunosuppressive Agents/pharmacology , Mice , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , NFATC Transcription Factors/metabolism , Humans , TOR Serine-Threonine Kinases/metabolism , Calcineurin/metabolism , Mice, Inbred C57BL , Phosphorylation/drug effects , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Cell Proliferation/drug effects , Multiprotein Complexes/metabolism
7.
Eur J Immunol ; 53(1): e2149400, 2023 01.
Article in English | MEDLINE | ID: mdl-36263815

ABSTRACT

While the immunosuppressive function of regulatory T (Treg) cells has been extensively studied, their immune-supportive roles have been less well investigated. Using a lymphocytic choriomeningitis virus (LCMV) Armstrong infection mouse model, we found that Treg cell-derived interleukin (IL)-15 is required for long-term maintenance of the KLRG1+ IL-7Rα- CD62L- terminal effector memory CD8+ T (tTEM) cell subset, but dispensable for the suppressive function of Treg cells themselves. In contrast, deletion of Il15 from other sources, including myeloid cells and muscles, did not affect the composition of the memory CD8+ T cell pool. Our findings identify Treg cells as an essential IL-15 source maintaining tTEM cells and suggest that Treg cells promote the diversity of immunological memory.


Subject(s)
Lymphocytic Choriomeningitis , T-Lymphocytes, Regulatory , Mice , Animals , Lymphocytic choriomeningitis virus , Immunologic Memory , Interleukin-15 , CD8-Positive T-Lymphocytes , Mice, Inbred C57BL , Interleukin-2
8.
Immunity ; 41(5): 802-14, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25464856

ABSTRACT

Protein kinase B (also known as AKT) and the mechanistic target of rapamycin (mTOR) are central regulators of T cell differentiation, proliferation, metabolism, and survival. Here, we show that during chronic murine lymphocytic choriomeningitis virus infection, activation of AKT and mTOR are impaired in antiviral cytotoxic T lymphocytes (CTLs), resulting in enhanced activity of the transcription factor FoxO1. Blockade of inhibitory receptor programmed cell death protein 1 (PD-1) in vivo increased mTOR activity in virus-specific CTLs, and its therapeutic effects were abrogated by the mTOR inhibitor rapamycin. FoxO1 functioned as a transcriptional activator of PD-1 that promoted the differentiation of terminally exhausted CTLs. Importantly, FoxO1-null CTLs failed to persist and control chronic viral infection. Collectively, this study shows that CTLs adapt to persistent infection through a positive feedback pathway (PD-1?FoxO1?PD-1) that functions to both desensitize virus-specific CTLs to antigen and support their survival during chronic viral infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Programmed Cell Death 1 Receptor/biosynthesis , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , CD28 Antigens/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Chronic Disease , Forkhead Box Protein O1 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Granzymes/biosynthesis , Humans , Interferon-gamma/biosynthesis , Jurkat Cells , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Programmed Cell Death 1 Receptor/immunology , Proto-Oncogene Proteins c-akt/biosynthesis , Receptors, Antigen, T-Cell/immunology , Sirolimus/pharmacology , T-Lymphocytes, Cytotoxic/cytology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/biosynthesis
9.
Nano Lett ; 22(12): 4661-4668, 2022 Jun 22.
Article in English | MEDLINE | ID: mdl-35640103

ABSTRACT

Confined nanospaces provide a new platform to promote catalytic reactions. However, the mechanism of catalytic enhancement in the nanospace still requires insightful exploration due to the lack of direct visualization. Here, we report operando investigations on the etching and growth of graphene in a two-dimensional (2D) confined space between graphene and a Cu substrate. We observed that the graphene layer between the Cu and top graphene layer was surprisingly very active in etching (more than 10 times faster than the etching of the top graphene layer). More strikingly, at a relatively low temperature (∼530 °C), the etched carbon radicals dissociated from the bottom layer, in turn feeding the growth of the top graphene layer with a very high efficiency. Our findings reveal the in situ dynamics of the anomalous confined catalytic processes in 2D confined spaces and thus pave the way for the design of high-efficiency catalysts.

10.
Medicina (Kaunas) ; 59(4)2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37109742

ABSTRACT

Background and Objectives: As is well understood, peroxisome proliferator-activated receptor gamma cofactor-related 1 (PPRC1) plays a central role in the transcriptional control of the mitochondrial biogenesis and oxidative phosphorylation (OXPHOS) process, yet its critical role in pan-cancer remains unclear. Materials and Methods: In this paper, the expression levels of PPRC1 in different tumor tissues and corresponding adjacent normal tissues were analyzed based on four databases: The Genotype-Tissue Expression (GTEx), Cancer Cell Line Encyclopedia (CCLE), The Cancer Genome Atlas (TCGA), and Tumor Immune Estimation Resource (TIMER). Meanwhile, the prognostic value of PPRC1 was inferred using Kaplan-Meier plotter and forest-plot studies. In addition, the correlation between PPRC1 expression and tumor immune cell infiltration, immune checkpoints, and the tumor-stemness index was analyzed using TCGA and TIMER databases. Results: According to our findings, the expression level of PPRC1 was found to be different in different cancer types and there was a positive correlation between PPRC1 expression and prognosis in several tumor types. In addition, PPRC1 expression was found to be significantly correlated with immune cell infiltration, immune checkpoints, and the tumor-stemness index in both ovarian and hepatocellular carcinoma. Conclusions: PPRC1 demonstrated promising potential as a novel biomarker in pan-cancer due to its potential association with immune cell infiltration, expression of immune checkpoints, and the tumor-stemness index.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Ovarian Neoplasms , Female , Humans , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Ovarian Neoplasms/genetics , Prognosis
11.
Int J Cancer ; 151(5): 797-808, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35499751

ABSTRACT

Memory CD8+ T cells mature after antigen clearance and ultimately express CD8 protein at levels higher than those detected in effector CD8+ T cells. However, it is not clear whether engagement of CD8 in the absence of antigenic stimulation will result in the functional activation of T cells. Here, we found that CD8 antibody-mediated activation of memory CD8+ T cells triggered T cell receptor (TCR) downstream signaling, enhanced T cell-mediated cytotoxicity and promoted effector cytokine production in a glucose- and glutamine-dependent manner. Furthermore, pretreatment of memory CD8+ T cells with an agonistic anti-CD8 antibody enhanced their tumoricidal activity in vitro and in vivo. From these studies, we conclude that CD8 agonism activates glucose and glutamine metabolism in memory T cells and enhances the efficacy of memory T cell-based cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Glutamine , Glucose/metabolism , Glutamine/metabolism , Humans , Immunologic Memory , Lymphocyte Activation , Memory T Cells , Receptors, Antigen, T-Cell , Signal Transduction
12.
BMC Cancer ; 21(1): 29, 2021 Jan 06.
Article in English | MEDLINE | ID: mdl-33407221

ABSTRACT

BACKGROUND: Endometrial cancer (EC) is one of the most common gynecological malignancies worldwide. However, the molecular mechanisms and the prognostic prediction for EC patients remain unclear. METHODS: In the current study, we performed an in-depth analysis of over 500 patients which were obtained from the Cancer Genome Atlas (TCGA) database. The bioinformatics analysis included gene set enrichment analysis (GSEA) and Cox and lasso regression analyses to ensure overall survival (OS)-related genes, moreover, to construct a prognostic model and a nomogram for EC patients. RESULTS: GSEA identified 4 gene sets significantly associated with EC, which are DNA repair, unfolded protein response, reactive oxygen species pathway and UV response up. Twenty-five OS-related DNA repair genes were screened out, after that, a 9-mRNA signature was constructed to measure the risk scores of patients with different outcomes. In addition, a nomogram contained the 9-mRNA model and clinical parameters was also presented to assess the prognosis. Patients with low risk were more likely to have sensitivity to paclitaxel, vinblastine, rapamycin, metformin, imatinib, Akt inhibitor and lapatinib. CONCLUSIONS: The identified highly enriched gene sets may offer a novel insight into the tumorigenesis and treatment of EC. Additionally, the constructed 9-mRNA model and the nomogram have prominent clinical implications for prognosis evaluation and specific therapy guidance for EC patients.


Subject(s)
Biomarkers, Tumor/genetics , DNA Repair Enzymes/genetics , Endometrial Neoplasms/classification , Endometrial Neoplasms/pathology , RNA, Messenger/genetics , Case-Control Studies , Endometrial Neoplasms/genetics , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Middle Aged , Prognosis , Survival Rate
13.
Mol Cell Biochem ; 476(3): 1541-1554, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33394271

ABSTRACT

Protective effect of Tagetes erecta flowers essential oils was investigated on oxidative stress, immune response, inflammation, and apoptosis against N-methyl-N'nitro-N-nitroguanidine (MNNG) induced gastric cancer in rats. Essential oil were extracted from Tagetes erecta flowers and analyzed using gas chromatography-mass spectrometry (GC-MS). For observing a protective effect against MNNG induced gastric cancer, we divided rats into 4 groups (group A to D) having 10 rats in each group. Performed various experiments and measured a different parameters to investigate antioxidant activity, immune response, anti-inflammatory and anti-apoptotic activity. The levels of malondialdehyde were markedly increased in the presence of N-methyl-N'nitro-N-nitroguanidine, whereas, the antioxidant activities of superoxide dismutase, and catalase were lowered in the treated rats in contrast with the control. Intervention with TEEO to gastric cancer-induced rats upregulated the redox status and the activity of the immune system to decrease cancer risk. The proinflammatory cytokines (IL-6 and TNF-α) secretions that were induced by MNNG were markedly inhibited by TEEO. Administration of TEEO also significantly reduced terminal deoxynucleotidyl transferase dUTP nick end labeling positive gastric cancer cells, expression of mRNA of caspase-3, and Bax. Whereas, the expression of Bcl-2 was increased. Additionally, downregulation of nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) and IκBα degradation and the nuclear factor-κB (NF-κB) p65 expression in tissues of the stomach of MNNG-induced-rats were markedly elevated due to TEEO. This suggested possession of TEEO with a protective shield against MNNG induced gastric cancer by the exertion of antioxidative stress, anti-apoptotic response, the anti-inflammatory response through Nrf2/HO-1, and NF-κB signaling pathways.


Subject(s)
Flowers , Heme Oxygenase (Decyclizing) , NF-KappaB Inhibitor alpha , Neoplasm Proteins , Nucleocytoplasmic Transport Proteins , Stomach Neoplasms , Tagetes , Animals , Male , Mice , Rats , Antioxidants/metabolism , Apoptosis , Catalase/metabolism , Cell Line, Tumor , Flowers/metabolism , Gas Chromatography-Mass Spectrometry , Guanidines , Heme Oxygenase (Decyclizing)/metabolism , Immunoglobulin A/chemistry , Immunoglobulin G/chemistry , Immunoglobulin M/chemistry , Inflammation , Methylnitronitrosoguanidine/chemistry , Neoplasm Proteins/metabolism , NF-KappaB Inhibitor alpha/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Oils, Volatile/chemistry , Oxidative Stress/drug effects , Plant Extracts/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats, Wistar , Signal Transduction , Stomach Neoplasms/metabolism , Tagetes/metabolism , NF-E2-Related Factor 2
14.
Angew Chem Int Ed Engl ; 60(5): 2371-2378, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33398902

ABSTRACT

Tensile-strained Mxene/carbon nanotube (CNT) porous microspheres were developed as an electrocatalyst for the lithium polysulfide (LiPS) redox reaction. The internal stress on the surface results in lattice distortion with expanding Ti-Ti bonds, endowing the Mxene nanosheet with abundant active sites and regulating the d-band center of Ti atoms upshifted closer to the Fermi level, leading to strengthened LiPS adsorbability and accelerated catalytic conversion. The macroporous framework offers uniformed sulfur distribution, potent sulfur immobilization, and large surface area. The composite interwoven by CNT tentacle enhances conductivity and prevents the restacking of Mxene sheets. This combination of tensile strain effect and hierarchical architecture design results in smooth and favorable trapping-diffusion-conversion of LiPS on the interface. The Li-S battery exhibits an initial capacity of 1451 mAh g-1 at 0.2 C, rate capability up to 8 C, and prolonged cycle life.

15.
Int J Cancer ; 147(2): 307-316, 2020 07 15.
Article in English | MEDLINE | ID: mdl-31994718

ABSTRACT

In noncancerous tissues, neighboring cells coexist in metabolic harmony. This metabolic harmony is disrupted in cancerous tissues, often accompanied by genetic mutations. Tumor cells fundamentally change the metabolite profiles in the tumor microenvironment to favor their own growth. In this review, we will discuss several examples in which genetic mutations reprogram tumor cell metabolic pathways, leading to the consumption of essential nutrients in the tumor microenvironment, production of toxic byproducts, and suppression of antitumor immune cell metabolic fitness and tumor-killing function. Finally, we will briefly discuss how immune checkpoint blockade overcomes the metabolic suppression of tumor-infiltrating immune cells.


Subject(s)
Mutation , Neoplasms/metabolism , Oncogene Proteins/genetics , Cellular Reprogramming , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/metabolism , Metabolic Networks and Pathways , Neoplasms/genetics , Tumor Microenvironment
16.
J Immunol ; 201(9): 2731-2743, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30257886

ABSTRACT

HIV-1 Nef is a multifunctional protein that optimizes virus spread and promotes immune evasion of infected cells to accelerate disease progression in AIDS patients. As one of its activities, Nef reduces the motility of infected CD4+ T lymphocytes in confined space. In vivo, Nef restricts T lymphocyte homing to lymph nodes as it reduces the ability for extravasation at the diapedesis step. Effects of Nef on T lymphocyte motility are typically mediated by its ability to reduce actin remodeling. However, interference with diapedesis does not depend on residues in Nef required for inhibition of host cell actin dynamics. In search for an alternative mechanism by which Nef could alter T lymphocyte extravasation, we noted that the viral protein interferes with the polarization of primary human CD4+ T lymphocytes upon infection with HIV-1. Expression of Nef alone is sufficient to disrupt T cell polarization, and this effect is conserved among lentiviral Nef proteins. Nef acts by arresting the oscillation of CD4+ T cells between polarized and nonpolarized morphologies. Mapping studies identified the binding site for the Nef-associated kinase complex (NAKC) as critical determinant of this Nef activity and a NAKC-binding-deficient Nef variant fails to impair CD4+ T lymphocyte extravasation and homing to lymph nodes. These results thus imply the disruption of T lymphocyte polarity via its NAKC binding site as a novel mechanism by which lentiviral Nef proteins alter T lymphocyte migration in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cell Polarity/immunology , Chemotaxis, Leukocyte/immunology , Transendothelial and Transepithelial Migration/immunology , nef Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Binding Sites , CD4-Positive T-Lymphocytes/immunology , Humans , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL
17.
Cell Mol Immunol ; 21(5): 419-435, 2024 May.
Article in English | MEDLINE | ID: mdl-38565887

ABSTRACT

T cells are an important component of adaptive immunity and protect the host from infectious diseases and cancers. However, uncontrolled T cell immunity may cause autoimmune disorders. In both situations, antigen-specific T cells undergo clonal expansion upon the engagement and activation of antigens. Cellular metabolism is reprogrammed to meet the increase in bioenergetic and biosynthetic demands associated with effector T cell expansion. Metabolites not only serve as building blocks or energy sources to fuel cell growth and expansion but also regulate a broad spectrum of cellular signals that instruct the differentiation of multiple T cell subsets. The realm of immunometabolism research is undergoing swift advancements. Encapsulating all the recent progress within this concise review in not possible. Instead, our objective is to provide a succinct introduction to this swiftly progressing research, concentrating on the metabolic intricacies of three pivotal nutrient classes-lipids, glucose, and amino acids-in T cells. We shed light on recent investigations elucidating the roles of these three groups of metabolites in mediating the metabolic and immune functions of T cells. Moreover, we delve into the prospect of "editing" metabolic pathways within T cells using pharmacological or genetic approaches, with the aim of synergizing this approach with existing immunotherapies and enhancing the efficacy of antitumor and antiinfection immune responses.


Subject(s)
Cell Differentiation , T-Lymphocyte Subsets , Humans , Animals , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Energy Metabolism , Glucose/metabolism , Amino Acids/metabolism
18.
Heliyon ; 10(13): e33917, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39091937

ABSTRACT

Background: Natural products exhibit structural complexity, diversity, and historical therapeutic significance, boasting attractive functions and biological activities that have significantly influenced drug discovery endeavors. The identification of target proteins of active natural compounds is crucial for advancing novel drug innovation. Currently, methods for identifying targets of natural products can be categorized into labeling and label-free approaches based on whether the natural bioactive constituents are modified into active probes. In addition, there is a new avenue for rapidly exploring the targets of natural products based on their innate functions. Aim: This review aimed to summarize recent advancements in both labeling and label-free approaches to the identification of targets for natural products, as well as the novel target identification method based on the natural functions of natural products. Methods: We systematically collected relevant articles published in recent years from PubMed, Web of Science, and ScienceDirect, focusing on methods employed for identifying protein targets of bioactive natural products. Furthermore, we systematically summarized the principles, procedures, and successful cases, as well as the advantages and limitations of each method. Results: Labeling methods allow for the direct labeling of target proteins and the exclusion of indirectly targeted proteins. However, these methods are not suitable for studying post-modified compounds with abolished activity, chemically challenging synthesis, or trace amounts of natural active compounds. Label-free methods can be employed to identify targets of any natural active compounds, including trace amounts and multicomponent mixtures, but their reliability is not as high as labeling methods. The structural complementarity between natural products and their innate receptors significantly increase the opportunities for finding more promising structural analogues of the natural products, and natural products may interact with several structural analogues of receptors in humans. Conclusion: Each approach presents benefits and drawbacks. In practice, a combination of methods is employed to identify targets of natural products. And natural products' innate functions-based approach is a rapid and selective strategy for target identification. This review provides valuable references for future research in this field, offering insights into techniques and methodologies.

19.
Nat Commun ; 15(1): 6067, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025856

ABSTRACT

After recognizing its ligand lipopolysaccharide, Toll-like receptor 4 (TLR4) recruits adaptor proteins to the cell membrane, thereby initiating downstream signaling and triggering inflammation. Whether this recruitment of adaptor proteins is dependent solely on protein-protein interactions is unknown. Here, we report that the sphingolipid sphinganine physically interacts with the adaptor proteins MyD88 and TIRAP and promotes MyD88 recruitment in macrophages. Myeloid cell-specific deficiency in serine palmitoyltransferase long chain base subunit 2, which encodes the key enzyme catalyzing sphingolipid biosynthesis, decreases the membrane recruitment of MyD88 and inhibits inflammatory responses in in vitro bone marrow-derived macrophage and in vivo sepsis models. In a melanoma mouse model, serine palmitoyltransferase long chain base subunit 2 deficiency decreases anti-tumor myeloid cell responses and increases tumor growth. Therefore, sphinganine biosynthesis is required for the initiation of TLR4 signal transduction and serves as a checkpoint for macrophage pattern recognition in sepsis and melanoma mouse models.


Subject(s)
Macrophages , Melanoma , Myeloid Differentiation Factor 88 , Sepsis , Serine C-Palmitoyltransferase , Sphingosine , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/metabolism , Sepsis/metabolism , Macrophages/metabolism , Myeloid Differentiation Factor 88/metabolism , Mice , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Melanoma/metabolism , Melanoma/pathology , Melanoma/genetics , Serine C-Palmitoyltransferase/metabolism , Serine C-Palmitoyltransferase/genetics , Humans , Signal Transduction , Disease Models, Animal , Inflammation/metabolism , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , HEK293 Cells , Lipopolysaccharides
20.
Sci Immunol ; 9(94): eadg8817, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38640251

ABSTRACT

CD4+ regulatory T (Treg) cells accumulate in the tumor microenvironment (TME) and suppress the immune system. Whether and how metabolite availability in the TME influences Treg cell differentiation is not understood. Here, we measured 630 metabolites in the TME and found that serine and palmitic acid, substrates required for the synthesis of sphingolipids, were enriched. A serine-free diet or a deficiency in Sptlc2, the rate-limiting enzyme catalyzing sphingolipid synthesis, suppressed Treg cell accumulation and inhibited tumor growth. Sphinganine, an intermediate metabolite in sphingolipid synthesis, physically interacted with the transcription factor c-Fos. Sphinganine c-Fos interactions enhanced the genome-wide recruitment of c-Fos to regions near the transcription start sites of target genes including Pdcd1 (encoding PD-1), which promoted Pdcd1 transcription and increased inducible Treg cell differentiation in vitro in a PD-1-dependent manner. Thus, Sptlc2-mediated sphingolipid synthesis translates the extracellular information of metabolite availability into nuclear signals for Treg cell differentiation and limits antitumor immunity.


Subject(s)
Neoplasms , Sphingosine , T-Lymphocytes, Regulatory , Programmed Cell Death 1 Receptor/metabolism , Serine/metabolism , Sphingolipids/metabolism , Sphingosine/analogs & derivatives , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL