Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
Cell Discov ; 10(1): 53, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38763950

ABSTRACT

Peripheral CD8+ T cell number is tightly controlled but the precise molecular mechanism regulating this process is still not fully understood. In this study, we found that epilepsy patients with loss of function mutation of DEPDC5 had reduced peripheral CD8+ T cells, and DEPDC5 expression positively correlated with tumor-infiltrating CD8+ T cells as well as overall cancer patient survival, indicating that DEPDC5 may control peripheral CD8+ T cell homeostasis. Significantly, mice with T cell-specific Depdc5 deletion also had reduced peripheral CD8+ T cells and impaired anti-tumor immunity. Mechanistically, Depdc5-deficient CD8+ T cells produced high levels of xanthine oxidase and lipid ROS due to hyper-mTORC1-induced expression of ATF4, leading to spontaneous ferroptosis. Together, our study links DEPDC5-mediated mTORC1 signaling with CD8+ T cell protection from ferroptosis, thereby revealing a novel strategy for enhancing anti-tumor immunity via suppression of ferroptosis.

4.
Cell Mol Immunol ; 20(9): 1023-1039, 2023 09.
Article in English | MEDLINE | ID: mdl-37582972

ABSTRACT

CD8+ T cells are the key executioners of the adaptive immune arm, which mediates antitumor and antiviral immunity. Naïve CD8+ T cells develop in the thymus and are quickly activated in the periphery after encountering a cognate antigen, which induces these cells to proliferate and differentiate into effector cells that fight the initial infection. Simultaneously, a fraction of these cells become long-lived memory CD8+ T cells that combat future infections. Notably, the generation and maintenance of memory cells is profoundly affected by various in vivo conditions, such as the mode of primary activation (e.g., acute vs. chronic immunization) or fluctuations in host metabolic, inflammatory, or aging factors. Therefore, many T cells may be lost or become exhausted and no longer functional. Complicated intracellular signaling pathways, transcription factors, epigenetic modifications, and metabolic processes are involved in this process. Therefore, understanding the cellular and molecular basis for the generation and fate of memory and exhausted CD8+ cells is central for harnessing cellular immunity. In this review, we focus on mammalian target of rapamycin (mTOR), particularly signaling mediated by mTOR complex (mTORC) 2 in memory and exhausted CD8+ T cells at the molecular level.


Subject(s)
CD8-Positive T-Lymphocytes , TOR Serine-Threonine Kinases , Cell Differentiation , TOR Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism , Immunologic Memory
5.
Arterioscler Thromb Vasc Biol ; 43(10): 1818-1832, 2023 10.
Article in English | MEDLINE | ID: mdl-37381985

ABSTRACT

BACKGROUND: Anti-ß2GP1 (ß2-glycoprotein 1) antibodies are the primary pathogenic antibody to promote thrombosis in antiphospholipid syndrome (APS), yet the underlying mechanism remains obscure. We aimed to explore the intracellular pathway that mediated platelet activation. METHODS: Platelets were isolated from patients with APS and subjected to RNA sequencing. Platelet aggregation, the release of platelet granules, platelet spreading, and clot retraction were detected to evaluate platelet activation. We purified anti-ß2GP1 antibodies from patients with APS and the total IgG from healthy donors to stimulate platelets with/without FcγRIIA (Fcγ receptor IIA) blocking antibody or Akt (protein kinase B) inhibitor. Platelet-specific Sin1 (stress-activated protein kinase-interacting protein) deficiency mice were established. The thrombus model of inferior vena cava flow restriction, ferric chloride-induced carotid injury model, and laser-induced vessel wall injury in cremaster arterioles model were constructed after administration of anti-ß2GP1 antibodies. RESULTS: Combined RNA sequencing and bioinformatics analysis suggested that APS platelets exhibited increased levels of mRNA associated with platelet activation, which was in line with the hyperactivation of APS platelets in response to stimuli. Platelet activation in APS platelets was accompanied by upregulation of the mTORC2 (mammalian target of the rapamycin complex 2)/Akt pathway and increased levels of SIN1 phosphorylation at threonine 86. Anti-ß2GP1 antibody derived from patients with APS enhanced platelet activation and upregulated the mTORC2/Akt pathway. Moreover, the Akt inhibitor weakened the potentiating effect of the anti-ß2GP1 antibody on platelet activation. Notably, Sin1 deficiency suppresses anti-ß2GP1 antibody-enhanced platelet activation in vitro and thrombosis in all 3 models. CONCLUSIONS: This study elucidated the novel mechanism involving the mTORC2/Akt pathway, which mediates the promotion of platelet activation and induction of thrombosis by the anti-ß2GP1 antibody. The findings suggest that SIN1 may be a promising therapeutic target for the treatment of APS.


Subject(s)
Antiphospholipid Syndrome , Thrombosis , Humans , Animals , Mice , Antiphospholipid Syndrome/complications , beta 2-Glycoprotein I , Proto-Oncogene Proteins c-akt/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Antibodies/metabolism , Platelet Activation , Carrier Proteins , Thrombosis/etiology , Mammals/metabolism
6.
J Mol Cell Biol ; 14(12)2023 04 20.
Article in English | MEDLINE | ID: mdl-36542462

ABSTRACT

It is well documented that the neonatal thymus-derived (neonatal-TD) regulatory T cells (Treg) are essential to prevent lethal autoimmune diseases and allergies, and neonatal and adult thymus possesses distinct output potentials for naïve T cells, including Treg. However, the molecular features and detailed functional differences between neonatal-TD and adult thymus-derived (adult-TD) T cells in terms of their ability to maintain immune homeostasis during long-term environmental influences are still largely unknown, partially due to the lack of appropriate animal models to precisely trace these cells at specific time points. In this study, neonatal-TD and adult-TD CD4+ T cells from the spleen and Peyer's patches were traced for 9 weeks by a T cell origin-time tracing mouse model and analysed by single-cell RNA sequencing. More Treg but fewer naïve T cells were found in neonatal-TD CD4+ T cells from both tissues than those from adult-TD counterparts. Interestingly, the neonatal-TD Treg in both the spleen and Peyer's patches exhibited augmented expression of Foxp3, Gata3, Ctla4, Icos, Il2ra, Tgfb1, and Nrp1, as well as enriched Gene Ontology terms like T cell activation and tolerance induction, indicating an enhanced immunosuppressive function. These results were further confirmed by flow cytometry analysis and in vitro immune suppression assays. Flow cytometry also revealed a significantly higher proportion of neonatal-TD Treg in total Treg than that of adult-TD counterparts, suggesting the longer lifespan of neonatal-TD Treg. To investigate the intrinsic features of neonatal-TD and adult-TD CD4+ T cells, a shortened tracing time was performed. Surprisingly, the neonatal-TD and adult-TD CD4+ T cells had similar proportions of Treg and did not exhibit significant differences in Foxp3, Gata3, Ctla4, Icos, Il2ra, and Tgfb1 expression levels after tracing for 12 days. On the other hand, neonatal-TD Treg present an increased Nrp1 expression level compared with adult-TD counterparts, indicating the enhanced stability. Together, our work reveals that the neonatal-TD Treg are more immunosuppressive, which is likely shaped primarily by environmental factors.


Subject(s)
Single-Cell Gene Expression Analysis , T-Lymphocytes, Regulatory , Animals , Mice , CD4-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/metabolism
7.
Sci China Life Sci ; 64(3): 389-403, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32737854

ABSTRACT

T cell-mediated immunity in the intestine is stringently controlled to ensure proper immunity against pathogenic microbes and to prevent autoimmunity, a known cause of inflammatory bowel disease. However, precisely how T cells regulate intestine immunity remains to be fully understood. In this study, we found that mitogen-activated protein kinase kinase kinase 2 (MAP3K2) is required for the CD4+ T cell-mediated inflammation in the intestine. Using a T cell transfer colitis model, we found that MAP3K2-deficient naïve CD4 T cells had a dramatically reduced ability to induce colitis compared to wild type T cells. In addition, significantly fewer IFN-γ- but more IL-17A-producing CD4+ T cells in the intestines of mice receiving MAP3K2-deficient T cells than in those from mice receiving wild type T cells was observed. Interestingly, under well-defined in vitro differentiation conditions, MAP3K2-deficient naïve T cells were not impaired in their ability to differentiate into Th1, Th17 and Treg. Furthermore, the MAP3K2-regulated colitis severity was mediated by Th1 but not Th17 cells in the intestine. At the molecular level, we showed that MAP3K2-mediated Th1 cell differentiation in the intestine was regulated by IL-18 and required specific JNK activation. Together, our study reveals a novel regulatory role of MAP3K2 in intestinal T cell immunity via the IL-18-MAP3K2-JNK axis and may provide a novel target for intervention in T cell-mediated colitis.


Subject(s)
Cell Differentiation/physiology , Colitis/immunology , Interleukin-18/physiology , MAP Kinase Kinase Kinase 2/physiology , Th1 Cells/cytology , Animals , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase Kinase 2/genetics , MAP Kinase Signaling System , Mice , Mice, Knockout , Th1 Cells/immunology , Thymus Gland/cytology
8.
Cell Mol Immunol ; 16(9): 757-769, 2019 09.
Article in English | MEDLINE | ID: mdl-30705387

ABSTRACT

Proper control of B cell growth and metabolism is crucial for B-cell-mediated immunity, but the underlying molecular mechanisms remain incompletely understood. In this study, Sin1, a key component of mTOR complex 2 (mTORC2), specifically regulates B cell growth and metabolism. Genetic ablation of Sin1 in B cells reduces the cell size at either the transitional stage or upon antigen stimulation and severely impairs metabolism. Sin1 deficiency also severely impairs B-cell proliferation, antibody responses, and anti-viral immunity. At the molecular level, Sin1 controls the expression and stability of the c-Myc protein and maintains the activity of mTORC1 through the Akt-dependent inactivation of GSK3 and TSC1/2, respectively. Therefore, our study reveals a novel and specific role for Sin1 in coordinating the activation of mTORC2 and mTORC1 to control B cell growth and metabolism.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Carrier Proteins/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , B-Lymphocytes/immunology , Cell Proliferation , Cells, Cultured , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction
9.
Natl Sci Rev ; 6(6): 1149-1162, 2019 Nov.
Article in English | MEDLINE | ID: mdl-34691993

ABSTRACT

The mammalian target of rapamycin (mTOR) is an evolutionarily conserved Ser/Thr protein kinase with essential cellular function via processing various extracellular and intracellular inputs. Two distinct multi-protein mTOR complexes (mTORC), mTORC1 and mTORC2, have been identified and well characterized in eukaryotic cells from yeast to human. Sin1, which stands for Sty1/Spc1-interacting protein1, also known as mitogen-activated protein kinase (MAPK) associated protein (MAPKAP)1, is an evolutionarily conserved adaptor protein. Mammalian Sin1 interacts with many cellular proteins, but it has been widely studied as an essential component of mTORC2, and it is crucial not only for the assembly of mTORC2 but also for the regulation of its substrate specificity. In this review, we summarize our current knowledge of the structure and functions of Sin1, focusing specifically on its protein interaction network and its roles in the mTOR pathway that could account for various cellular functions of mTOR in growth, metabolism, immunity and cancer.

10.
J Mol Cell Biol ; 11(2): 93-106, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30428057

ABSTRACT

Glucose metabolism plays a key role in thymocyte development. The mammalian target of rapamycin complex 2 (mTORC2) is a critical regulator of cell growth and metabolism, but its role in early thymocyte development and metabolism has not been fully studied. We show here that genetic ablation of Sin1, an essential component of mTORC2, in T lineage cells results in severely impaired thymocyte development at the CD4-CD8- double negative (DN) stages but not at the CD4+CD8+ double positive (DP) or later stages. Notably, Sin1-deficient DN thymocytes show markedly reduced proliferation and glycolysis. Importantly, we discover that the M2 isoform of pyruvate kinase (PKM2) is a novel and crucial Sin1 effector in promoting DN thymocyte development and metabolism. At the molecular level, we show that Sin1-mTORC2 controls PKM2 expression through an AKT-dependent PPAR-γ nuclear translocation. Together, our study unravels a novel mTORC2-PPAR-γ-PKM2 pathway in immune-metabolic regulation of early thymocyte development.


Subject(s)
Carrier Proteins/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Membrane Proteins/metabolism , T-Lymphocytes/metabolism , Thyroid Hormones/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carrier Proteins/genetics , Cell Differentiation , Cell Proliferation , Glycolysis/physiology , Mediator Complex Subunit 1/metabolism , Mice , Mice, Transgenic , TOR Serine-Threonine Kinases/metabolism , Thyroid Hormone-Binding Proteins
11.
Arterioscler Thromb Vasc Biol ; 38(12): 2793-2805, 2018 12.
Article in English | MEDLINE | ID: mdl-30571167

ABSTRACT

Objective- Microthrombosis as a serious consequence of myocardial infarction, impairs the microvascular environment and increases the occurrences of heart failure, arrhythmia, and death. Sin1 (stress-activated protein kinase-interacting protein) as an essential component of mTORC2 (mammalian target of rapamycin complex 2) is required for cell proliferation and metabolism in response to nutrients, stress, and reactive oxygen species and activates Akt and PKC (protein kinase C). However, the activation and function of Sin1/mTORC2 in ischemia-induced microthrombosis remain poorly understood. Approach and Results- The phosphorylation of the mTORC2 target Akt at S473 (serine 473) was significantly elevated in platelets from the distal end of left anterior descending obstructions from patients who underwent off-pump coronary artery bypass grafting compared with platelets from healthy subjects. Consistent with this finding, phosphorylation of T86 in Sin1 was also dramatically increased. Importantly, the augmented levels of phosphorylated Sin1 and Akt in platelets from 61 preoperative patients with ST-segment-elevation myocardial infarction correlated well with the no-reflow phenomena observed after revascularization. Platelet-specific Sin1 deficiency mice and Sin1 T86 phosphorylation deficiency mice were established to explore the underlying mechanisms in platelet activation. Mechanistically, Sin1 T86 phosphorylation amplifies mTORC2-mediated downstream signals; it is also required for αIIbß3-mediated outside-in signaling and plays a role in generating hypoxia/reactive oxygen species through NAD+/Sirt3 (sirtuin 3)/SOD2 (superoxide dismutase 2) pathway. Importantly, Sin1 deletion in platelets protected mice from ischemia-induced microvascular embolization and subsequent heart dysfunction in a mouse model of myocardial infarction. Conclusions- Together, the results of our study reveal a novel role for Sin1 in platelet activation. Thus, Sin1 may be a valuable therapeutic target for interventions for ischemia-induced myocardial infarction deterioration.


Subject(s)
Adaptor Proteins, Signal Transducing/blood , Blood Platelets/enzymology , Carrier Proteins/blood , Myocardial Infarction/complications , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Signal Transduction , Thrombosis/enzymology , Adult , Aged , Aged, 80 and over , Animals , Carrier Proteins/genetics , Cell Hypoxia , Disease Models, Animal , Female , Humans , Male , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Phosphorylation , Proto-Oncogene Proteins c-akt/blood , Reactive Oxygen Species/blood , Sirtuin 3/blood , Sirtuin 3/genetics , Superoxide Dismutase/blood , Superoxide Dismutase/genetics , Thrombosis/blood , Thrombosis/genetics , Thrombosis/prevention & control
12.
J Immunol ; 201(3): 908-915, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29934471

ABSTRACT

Recirculation of naive T cells between secondary lymphoid organs to receive survival cues and scan for signs of infection or other pathologic conditions is important for immune homeostasis and effective immune responses. Although the mechanisms that specifically guide the entry of naive T cells into secondary lymphoid organs are well studied, the mechanisms that keep them from fluxing into inappropriate or undesirable compartments, such as healthy tissues or bone marrow, are less well understood. In this study, we report an unexpected finding that under steady state, bone marrow homing of naive T cells is actively suppressed by mTORC2 signaling. We found that in mice, T cell-specific deletion of an essential mTORC2 component Sin1 results in increased accumulation of naive T cells in the bone marrow. Mechanistically, we show that loss of mTORC2 signaling in naive T cells results in enhanced FOXO1 activity, which leads to increased CXCR4 expression and chemotactic response to CXCL12, a key chemokine that promotes bone marrow homing and retention of T cells. Together, the results of our study reveal a novel role of mTORC2 in T cell homeostasis via active suppression of naive T cell bone marrow homing by the mTORC2-FOXO1-CXCR4 axis.


Subject(s)
Bone Marrow/immunology , Bone Marrow/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Receptors, CXCR4/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Carrier Proteins/metabolism , Chemokine CXCL12/metabolism , Forkhead Box Protein O1/metabolism , Homeostasis/immunology , Mice , Mice, Inbred C57BL
13.
Viruses ; 6(12): 4800-10, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25431948

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causal agent of all forms of Kaposi's sarcoma (KS), including AIDS-KS, endemic KS, classic KS and iatrogenic KS. Based on Open reading frame (ORF) K1 sequence analysis, KSHV has been classified into seven major molecular subtypes (A, B, C, D, E, F and Z). The distribution of KSHV strains varies according to geography and ethnicity. Xinjiang is a unique region where the seroprevalence of KSHV is significantly higher than other parts of China. The genotyping of KSHV strains in this region has not been thoroughly studied. The present study aimed to evaluate the frequency of KSHV genotypes isolated from KS tissues in Classical KS and AIDS KS patients from Xinjiang, China. ORF-K1 of KSHV from tissue samples of 28 KS patients was amplified and sequenced. Two subtypes of KSHV were identified according to K1 genotyping. Twenty-three of them belonged to subtype A, while five of them were subtype C. More genotype A than genotype C strains were found in both Classical KS and AIDS KS. No significant difference was found in the prevalence of different genotype between Classical KS and AIDS KS.


Subject(s)
Herpesvirus 8, Human/genetics , Sarcoma, Kaposi/virology , Adult , Aged , China/epidemiology , China/ethnology , Female , Genotype , Herpesvirus 8, Human/classification , Herpesvirus 8, Human/isolation & purification , Humans , Male , Middle Aged , Open Reading Frames , Phylogeny , Sarcoma, Kaposi/epidemiology , Sarcoma, Kaposi/ethnology , Young Adult
14.
Jpn J Infect Dis ; 65(4): 350-3, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22814163

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causal agent of Kaposi's sarcoma (KS), a common cancer in patients with acquired immunodeficiency syndrome. The risk factors for KSHV infection have been extensively studied for Western countries but remain largely undefined for other parts of the world. Schistosomiasis, caused by Schistosoma japonicum infection, was recently identified as a cofactor for KSHV infection in rural Egypt. In this study, we examined the seroprevalence of KSHV in a population along the Yangtze River in China that has a high incidence of schistosomiasis. KSHV seroprevalence in subjects with schistosomiasis was slightly higher than that in subjects without schistosomiasis, but the difference was not statistically significant (8.4% versus 6.6%; P = 0.204). However, after adjusting for gender, KSHV seroprevalence in men with schistosomiasis was found to be significantly higher than that in men without schistosomiasis (8.4% versus 2.8%; odds ratio [OR], 3.170; 95% confidence interval [CI], 1.501-6.694; P = 0.002). Compared to men, women showed significantly higher seroprevalence of KSHV (5.9% versus 9.3%; OR, 1.621; 95% CI, 1.084-2.425; P = 0.019).


Subject(s)
Herpesviridae Infections/complications , Herpesviridae Infections/epidemiology , Herpesvirus 8, Human/immunology , Schistosomiasis/complications , Adolescent , Adult , Aged , China/epidemiology , Female , Herpesviridae Infections/diagnosis , Humans , Male , Middle Aged , Prevalence , Risk Factors , Schistosomiasis/diagnosis , Seroepidemiologic Studies , Sex Factors , Young Adult
15.
Virol Sin ; 25(3): 168-76, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20960290

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is causally related to Kaposi's sarcoma (KS), primary effusion lymphoma (PEL) and a proportion of cases of multicentric Castleman's disease (MCD). The ORF73 protein was cloned into pQE80L-orf73 and expressed in E.coli and purified. The expressed recombinant ORF73 was identified by sodium dodecyl sulfatepolyacrylamide gel electrophoresis (SDS-PAGE). A protein of about 27 kDa was expressed as expected. Western Blotting showed that the purified recombinant ORF73 reacted with KSHV positive serum. The immunogenicity of the recombinant ORF73 was further analysed by ELISA and the optimal conditions were determined. The ORF73 ELISA was used to compare the KSHV seroprevalence between Hubei and Xinjiang Han people. The Han people in Xinjiang have significantly higher KSHV seroprevalence than their counterparts in Hubei (6.7% vs 2.9%, P = 0.005).


Subject(s)
Antibodies, Viral/blood , Antigens, Viral , Herpesviridae Infections/diagnosis , Herpesviridae Infections/epidemiology , Herpesvirus 8, Human/isolation & purification , Viral Proteins , Virology/methods , Antigens, Viral/chemistry , Antigens, Viral/genetics , Blotting, Western , China/epidemiology , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay/methods , Escherichia coli/genetics , Gene Expression , Genetic Vectors , Herpesviridae Infections/virology , Herpesvirus 8, Human/immunology , Humans , Molecular Weight , Plasmids , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Seroepidemiologic Studies , Viral Proteins/chemistry , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...