Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Nat Cell Biol ; 21(5): 662-663, 2019 05.
Article in English | MEDLINE | ID: mdl-30783264

ABSTRACT

In the version of this Article originally published, the labels for Rictor and mTOR in the whole cell lysate (WCL) blots were swapped in Fig. 3b and the mTOR blot was placed upside down. Unprocessed blots of mTOR were also missing from Supplementary Fig. 9. The corrected Figs are shown below. In addition, control blots for the mTOR antibody (Cell Signalling Technology #2972) were also missing. These are now provided below, as Fig. 9, and show that the lower band is likely non-specific.

3.
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
4.
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
5.
Yale J Biol Med ; 86(3): 287-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24191376
6.
Nat Cell Biol ; 15(11): 1340-50, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24161930

ABSTRACT

The mechanistic target of rapamycin (mTOR) functions as a critical regulator of cellular growth and metabolism by forming multi-component, yet functionally distinct complexes mTORC1 and mTORC2. Although mTORC2 has been implicated in mTORC1 activation, little is known about how mTORC2 is regulated. Here we report that phosphorylation of Sin1 at Thr 86 and Thr 398 suppresses mTORC2 kinase activity by dissociating Sin1 from mTORC2. Importantly, Sin1 phosphorylation, triggered by S6K or Akt, in a cellular context-dependent manner, inhibits not only insulin- or IGF-1-mediated, but also PDGF- or EGF-induced Akt phosphorylation by mTORC2, demonstrating a negative regulation of mTORC2 independent of IRS-1 and Grb10. Finally, a cancer-patient-derived Sin1-R81T mutation impairs Sin1 phosphorylation, leading to hyper-activation of mTORC2 by bypassing this negative regulation. Together, our results reveal a Sin1-phosphorylation-dependent mTORC2 regulation, providing a potential molecular mechanism by which mutations in the mTORC1-S6K-Sin1 signalling axis might cause aberrant hyper-activation of the mTORC2-Akt pathway, which facilitates tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinogenesis , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Humans , Mechanistic Target of Rapamycin Complex 2 , Mutation , Phosphorylation
7.
Int Immunol ; 25(1): 1-10, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23042773

ABSTRACT

Autophagy is an evolutionarily ancient process eukaryotic cells utilize to remove and recycle intracellular material in order to maintain cellular homeostasis. In metazoans, the autophagy machinery not only functions in this capacity but also has evolved to perform a diverse repertoire of intracellular transport and regulatory functions. In response to virus infections, the autophagy machinery degrades viruses, shuttles viral pathogen-associated molecular patterns to endosomes containing Toll-like receptors, facilitates viral-antigen processing for major histocompatibility complex presentation and transports antiviral proteins to viral replication sites. This is accomplished through canonical autophagy or through processes involving distinct subsets of the autophagy-related genes (Atgs). Herein, we discuss how the variable components of the autophagy machinery contribute to antiviral defense and highlight three emerging themes: first, autophagy delivers viral cytosolic components to several distinct endolysosomal compartments; second, Atg proteins act alone, as subgroups or collectively; and third, the specificity of autophagy and the autophagy machinery is achieved by recognition of triggers and selective targeting by adaptors.


Subject(s)
Autophagy/immunology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Virus Diseases/immunology , Viruses/growth & development , Antigen Presentation/genetics , Antigen Presentation/immunology , Autophagy/genetics , Biological Transport , Dendritic Cells/virology , Endosomes/metabolism , Host-Pathogen Interactions , Humans , Immunity, Innate , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Virus Diseases/genetics , Virus Diseases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...