Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
J Allergy Clin Immunol ; 145(1): 283-300.e8, 2020 01.
Article in English | MEDLINE | ID: mdl-31401286

ABSTRACT

BACKGROUND: Perturbation of epidermal barrier formation will profoundly compromise overall skin function, leading to a dry and scaly, ichthyosis-like skin phenotype that is the hallmark of a broad range of skin diseases, including ichthyosis, atopic dermatitis, and a multitude of clinical eczema variants. An overarching molecular mechanism that orchestrates the multitude of factors controlling epidermal barrier formation and homeostasis remains to be elucidated. OBJECTIVE: Here we highlight a specific role of mammalian target of rapamycin complex 2 (mTORC2) signaling in epidermal barrier formation. METHODS: Epidermal mTORC2 signaling was specifically disrupted by deleting rapamycin-insensitive companion of target of rapamycin (Rictor), encoding an essential subunit of mTORC2 in mouse epidermis (epidermis-specific homozygous Rictor deletion [RicEKO] mice). Epidermal structure and barrier function were investigated through a combination of gene expression, biochemical, morphological and functional analysis in RicEKO and control mice. RESULTS: RicEKO newborns displayed an ichthyosis-like phenotype characterized by dysregulated epidermal de novo lipid synthesis, altered lipid lamellae structure, and aberrant filaggrin (FLG) processing. Despite a compensatory transcriptional epidermal repair response, the protective epidermal function was impaired in RicEKO mice, as revealed by increased transepidermal water loss, enhanced corneocyte fragility, decreased dendritic epidermal T cells, and an exaggerated percutaneous immune response. Restoration of Akt-Ser473 phosphorylation in mTORC2-deficient keratinocytes through expression of constitutive Akt rescued FLG processing. CONCLUSION: Our findings reveal a critical metabolic signaling relay of barrier formation in which epidermal mTORC2 activity controls FLG processing and de novo epidermal lipid synthesis during cornification. Our findings provide novel mechanistic insights into epidermal barrier formation and could open up new therapeutic opportunities to restore defective epidermal barrier conditions.


Subject(s)
Epidermis , Intermediate Filament Proteins , Lipids , Protein Processing, Post-Translational/immunology , Rapamycin-Insensitive Companion of mTOR Protein , Signal Transduction/immunology , Animals , Epidermis/immunology , Epidermis/metabolism , Filaggrin Proteins , Ichthyosis/genetics , Ichthyosis/immunology , Ichthyosis/metabolism , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/immunology , Intermediate Filament Proteins/metabolism , Lipids/biosynthesis , Lipids/genetics , Lipids/immunology , Mice , Mice, Knockout , Protein Processing, Post-Translational/genetics , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Rapamycin-Insensitive Companion of mTOR Protein/immunology , Signal Transduction/genetics
2.
Nat Commun ; 9(1): 4874, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451838

ABSTRACT

The metabolic checkpoint kinase mechanistic/mammalian target of rapamycin (mTOR) regulates natural killer (NK) cell development and function, but the exact underlying mechanisms remain unclear. Here, we show, via conditional deletion of Raptor (mTORC1) or Rictor (mTORC2), that mTORC1 and mTORC2 promote NK cell maturation in a cooperative and non-redundant manner, mainly by controlling the expression of Tbx21 and Eomes. Intriguingly, mTORC1 and mTORC2 regulate cytolytic function in an opposing way, exhibiting promoting and inhibitory effects on the anti-tumor ability and metabolism, respectively. mTORC1 sustains mTORC2 activity by maintaining CD122-mediated IL-15 signaling, whereas mTORC2 represses mTORC1-modulated NK cell effector functions by restraining STAT5-mediated SLC7A5 expression. These positive and negative crosstalks between mTORC1 and mTORC2 signaling thus variegate the magnitudes and kinetics of NK cell activation, and help define a paradigm for the modulation of NK maturation and effector functions.


Subject(s)
Killer Cells, Natural/immunology , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Regulatory-Associated Protein of mTOR/genetics , T-Box Domain Proteins/genetics , Animals , Cell Differentiation , Gene Expression Regulation , Humans , Interleukin-15/genetics , Interleukin-15/immunology , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/immunology , Killer Cells, Natural/cytology , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rapamycin-Insensitive Companion of mTOR Protein/deficiency , Rapamycin-Insensitive Companion of mTOR Protein/immunology , Regulatory-Associated Protein of mTOR/deficiency , Regulatory-Associated Protein of mTOR/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction , T-Box Domain Proteins/immunology
3.
Front Immunol ; 9: 215, 2018.
Article in English | MEDLINE | ID: mdl-29487597

ABSTRACT

Background: Rictor is an essential component of mammalian target of rapamycin (mTOR) complex 2 (mTORC2), a conserved serine/threonine kinase that may play a role in cell proliferation, survival and innate or adaptive immune responses. Genetic loss of Rictor inactivates mTORC2, which directly activates Akt S473 phosphorylation and promotes pro-survival cell signaling and proliferation. Methods and results: To study the role of mTORC2 signaling in monocytes and macrophages, we generated mice with myeloid lineage-specific Rictor deletion (MRictor-/-). These MRictor-/- mice exhibited dramatic reductions of white blood cells, B-cells, T-cells, and monocytes but had similar levels of neutrophils compared to control Rictor flox-flox (Rictorfl/fl) mice. MRictor-/- bone marrow monocytes and peritoneal macrophages expressed reduced levels of mTORC2 signaling and decreased Akt S473 phosphorylation, and they displayed significantly less proliferation than control Rictorfl/fl cells. In addition, blood monocytes and peritoneal macrophages isolated from MRictor-/- mice were significantly more sensitive to pro-apoptotic stimuli. In response to LPS, MRictor-/- macrophages exhibited the M1 phenotype with higher levels of pro-inflammatory gene expression and lower levels of Il10 gene expression than control Rictorfl/fl cells. Further suppression of LPS-stimulated Akt signaling with a low dose of an Akt inhibitor, increased inflammatory gene expression in macrophages, but genetic inactivation of Raptor reversed this rise, indicating that mTORC1 mediates this increase of inflammatory gene expression. Next, to elucidate whether mTORC2 has an impact on atherosclerosis in vivo, female and male Ldlr null mice were reconstituted with bone marrow from MRictor-/- or Rictorfl/fl mice. After 10 weeks of the Western diet, there were no differences between the recipients of the same gender in body weight, blood glucose or plasma lipid levels. However, both female and male MRictor-/- → Ldlr-/- mice developed smaller atherosclerotic lesions in the distal and proximal aorta. These lesions contained less macrophage area and more apoptosis than lesions of control Rictorfl/fl → Ldlr-/- mice. Thus, loss of Rictor and, consequently, mTORC2 significantly compromised monocyte/macrophage survival, and this markedly diminished early atherosclerosis in Ldlr-/- mice. Conclusion: Our results demonstrate that mTORC2 is a key signaling regulator of macrophage survival and its depletion suppresses early atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Macrophages/immunology , Monocytes/immunology , Rapamycin-Insensitive Companion of mTOR Protein/immunology , Receptors, LDL/genetics , Animals , Aorta/immunology , Aorta/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Proliferation , Cell Survival/immunology , Diet, Western/adverse effects , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rapamycin-Insensitive Companion of mTOR Protein/genetics
4.
Nat Commun ; 8: 14208, 2017 01 27.
Article in English | MEDLINE | ID: mdl-28128208

ABSTRACT

Alternatively activated macrophages (M2) have an important function in innate immune responses to parasitic helminths, and emerging evidence also indicates these cells are regulators of systemic metabolism. Here we show a critical role for mTORC2 signalling in the generation of M2 macrophages. Abrogation of mTORC2 signalling in macrophages by selective conditional deletion of the adaptor molecule Rictor inhibits the generation of M2 macrophages while leaving the generation of classically activated macrophages (M1) intact. Selective deletion of Rictor in macrophages prevents M2 differentiation and clearance of a parasitic helminth infection in mice, and also abrogates the ability of mice to regulate brown fat and maintain core body temperature. Our findings define a role for mTORC2 in macrophages in integrating signals from the immune microenvironment to promote innate type 2 immunity, and also to integrate systemic metabolic and thermogenic responses.


Subject(s)
Macrophages/physiology , Mechanistic Target of Rapamycin Complex 2/immunology , Strongylida Infections/immunology , Thermogenesis/physiology , Animals , Cell Differentiation/immunology , Cells, Cultured , Disease Models, Animal , Female , Gene Knockout Techniques , Helminthiasis, Animal/immunology , Humans , Immunity, Innate/physiology , Male , Mechanistic Target of Rapamycin Complex 2/genetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred C57BL , Nippostrongylus/immunology , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Rapamycin-Insensitive Companion of mTOR Protein/immunology , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Signal Transduction/physiology , Strongylida Infections/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL