Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
2.
Sci Rep ; 9(1): 17620, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31772273

ABSTRACT

Inactivation of the protein complex 'mechanistic target of rapamycin complex 1' (mTORC1) can increase the nuclear content of transcriptional regulators of metabolism and apoptosis. Previous studies established that nuclear import of signal transducer and activator of transcription-1 (STAT1) requires the mTORC1-associated adaptor karyopherin-α1 (KPNA1) when mTORC1 activity is reduced. However, the role of other mTORC1-interacting proteins in the complex, including 'protein kinase C delta' (PKCδ), have not been well characterized. In this study, we demonstrate that PKCδ, a STAT1 kinase, contains a functional 'target of rapamycin signaling' (TOS) motif that directs its interaction with mTORC1. Depletion of KPNA1 by RNAi prevented the nuclear import of PKCδ in cells exposed to the mTORC1 inhibitor rapamycin or amino acid restriction. Mutation of the TOS motif in PKCδ led to its loss of regulation by mTORC1 or karyopherin-α1, resulting in increased constitutive nuclear content. In cells expressing wild-type PKCδ, STAT1 activity and apoptosis were increased by rapamycin or interferon-ß. Those expressing the PKCδ TOS mutant exhibited increased STAT1 activity and apoptosis; further enhancement by rapamycin or interferon-ß, however, was lost. Therefore, the TOS motif in PKCδ is a novel structural mechanism by which mTORC1 prevents PKCδ and STAT1 nuclear import, and apoptosis.


Subject(s)
Cell Nucleus/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Kinase C-delta/metabolism , Active Transport, Cell Nucleus/drug effects , Amino Acid Motifs , Apoptosis/drug effects , Apoptosis/physiology , Cell Line , Humans , Models, Molecular , Mutation, Missense , Point Mutation , Protein Conformation , Protein Interaction Mapping , Protein Kinase C-delta/chemistry , Protein Kinase C-delta/genetics , RNA Interference , RNA, Small Interfering/genetics , Recombinant Proteins/metabolism , Regulatory-Associated Protein of mTOR/metabolism , STAT1 Transcription Factor/biosynthesis , Sequence Alignment , Sirolimus/pharmacology , alpha Karyopherins/antagonists & inhibitors , alpha Karyopherins/metabolism
3.
PLoS One ; 7(5): e33984, 2012.
Article in English | MEDLINE | ID: mdl-22685525

ABSTRACT

The mammalian target of rapamycin (mTOR) modulates immune responses and cellular proliferation. The objective of this study was to assess whether inhibition of mTOR with rapamycin modifies disease severity in two experimental murine models of house dust mite (HDM)-induced asthma. In an induction model, rapamycin was administered to BALB/c mice coincident with nasal HDM challenges for 3 weeks. In a treatment model, nasal HDM challenges were performed for 6 weeks and rapamycin treatment was administered during weeks 4 through 6. In the induction model, rapamycin significantly attenuated airway inflammation, airway hyperreactivity (AHR) and goblet cell hyperplasia. In contrast, treatment of established HDM-induced asthma with rapamycin exacerbated AHR and airway inflammation, whereas goblet cell hyperplasia was not modified. Phosphorylation of the S6 ribosomal protein, which is downstream of mTORC1, was increased after 3 weeks, but not 6 weeks of HDM-challenge. Rapamycin reduced S6 phosphorylation in HDM-challenged mice in both the induction and treatment models. Thus, the paradoxical effects of rapamycin on asthma severity paralleled the activation of mTOR signaling. Lastly, mediastinal lymph node re-stimulation experiments showed that treatment of rapamycin-naive T cells with ex vivo rapamycin decreased antigen-specific Th2 cytokine production, whereas prior exposure to in vivo rapamycin rendered T cells refractory to the suppressive effects of ex vivo rapamycin. We conclude that rapamycin had paradoxical effects on the pathogenesis of experimental HDM-induced asthma. Thus, consistent with the context-dependent effects of rapamycin on inflammation, the timing of mTOR inhibition may be an important determinant of efficacy and toxicity in HDM-induced asthma.


Subject(s)
Asthma/drug therapy , Asthma/etiology , Pyroglyphidae/immunology , Sirolimus/therapeutic use , Animals , Asthma/immunology , Asthma/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Female , Inflammation/drug therapy , Inflammation/etiology , Inflammation/immunology , Inflammation/metabolism , Lung/drug effects , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
4.
J Immunol ; 188(9): 4535-42, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22450807

ABSTRACT

Acute lung injury during bacterial infection is associated with neutrophilic inflammation, epithelial cell apoptosis, and disruption of the alveolar-capillary barrier. TLR4 is required for lung injury in animals exposed to bacterial LPS and initiates proinflammatory responses in part via the transcription factor NF-κB. Ligation of TLR4 also initiates a proapoptotic response by activating IFN-ß and STAT1-dependent genes. We recently demonstrated that mammalian target of rapamycin (mTOR), a key controller of cell growth and survival, can physically interact with STAT1 and suppress the induction of STAT1-dependent apoptosis genes. We therefore hypothesized that the mTOR inhibitor rapamycin would increase LPS-induced apoptosis and lung injury in vivo. Rapamycin increased lung injury and cellular apoptosis in C57BL/6J mice exposed to intratracheal LPS for 24 h. Rapamycin also augmented STAT1 activation, and the induction of STAT1-dependent genes that mediate cellular apoptosis (i.e., Fas, caspase-3). LPS-induced lung injury was attenuated in STAT1 knockout mice. In addition, LPS and IFN-ß-induced apoptosis was absent in cultured cells lacking STAT1, and, unlike in wild-type cells, a permissive effect of rapamycin was not observed. In contrast to its effect on STAT1, rapamycin inhibited NF-κB activation in vivo and reduced selected markers of inflammation (i.e., neutrophils in the bronchoalveolar lavage fluid, TNF-α). Therefore, although it inhibits NF-κB and neutrophilic inflammation, rapamycin augments LPS-induced lung injury and apoptosis in a mechanism that involves STAT1 and the induction of STAT1-dependent apoptosis genes.


Subject(s)
Acute Lung Injury/immunology , Apoptosis/drug effects , Lipopolysaccharides/toxicity , TOR Serine-Threonine Kinases/immunology , Toll-Like Receptor 4/immunology , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Anti-Bacterial Agents/pharmacology , Apoptosis/genetics , Apoptosis/immunology , Bronchoalveolar Lavage , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Interferon-beta/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
5.
J Biol Chem ; 287(18): 14325-35, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22399302

ABSTRACT

Under conditions of reduced mitogen or nutritional substrate levels, the serine/threonine kinase target of rapamycin can augment the nuclear content of distinct transcription factors and promote the induction of stress response genes. In its latent (i.e., unphosphorylated) form, the transcription factor STAT1 regulates a subset of genes involved in immune modulation and apoptosis. Based on previous work indicating a functional relationship between mammalian target of rapamycin (mTOR) and the nuclear content of latent STAT1, we investigated the mechanism by which mTOR controls STAT1 nuclear import. By fluorescence confocal microscopy, inactivation of mTOR with rapamycin promoted the nuclear translocation of unphosphorylated STAT1, but not that of a STAT1 mutant incapable of binding its nuclear import adaptor karyopherin-α1 (KPNA1). By immunoprecipitation, KPNA1 was physically associated with mTOR and STAT1 in a complex that translocated to the nucleus in response to rapamycin. Although mTOR is not a kinase for KPNA1, the mTOR-associated phosphatase protein phosphatase 2A catalytic interacted directly with KPNA1 and regulated nuclear import of the mTOR-KPNA1 complex. KPNA1, or its interaction with STAT1, was required for the nuclear import of latent STAT1, transcriptional induction of the STAT1 gene, and caspase-3 activation under conditions of reduced mTOR activity (i.e. rapamycin, glucose starvation, serum withdrawal). Therefore, at low mitogen or nutrient levels, mTOR and protein phosphatase 2A catalytically control the constitutive nuclear import of latent STAT1 by KPNA1, which are key modulators of STAT1 expression and apoptosis.


Subject(s)
Cell Nucleus/metabolism , TOR Serine-Threonine Kinases/metabolism , alpha Karyopherins/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Apoptosis/physiology , Caspase 3/physiology , Cell Nucleus/genetics , Enzyme Activation/physiology , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Mice , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation , Phosphorylation/physiology , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , TOR Serine-Threonine Kinases/genetics , alpha Karyopherins/genetics
6.
J Biol Chem ; 284(36): 24341-53, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19553685

ABSTRACT

Target of rapamycin (TOR) is a highly conserved serine/threonine kinase that controls cell growth, primarily via regulation of protein synthesis. In Saccharomyces cerevisiae, TOR can also suppress the transcription of stress response genes by a mechanism involving Tap42, a serine/threonine phosphatase subunit, and the transcription factor Msn2. A physical association between mammalian TOR (mTOR) and the transcription factor signal transducer and activator of transcription-1 (STAT1) was recently identified in human cells, suggesting a similar role for mTOR in the transcription of interferon-gamma-stimulated genes. In the current study, we identified a macromolecular protein complex composed of mTOR, STAT1, the Tap42 homologue alpha4, and the protein phosphatase 2A catalytic subunit (PP2Ac). Inactivation of mTOR enhanced its association with STAT1 and increased STAT1 nuclear content in PP2Ac-dependent fashion. Depletion of alpha4, PP2A, or mTOR enhanced the induction of early (i.e. IRF-1) and late (i.e. caspase-1, hiNOS, and Fas) STAT1-dependent genes. The regulation of IRF-1 or caspase-1 by mTOR was independent of other known mTOR effectors p70 S6 kinase and Akt. These results describe a new role for mTOR and alpha4/PP2A in the control of STAT1 nuclear content, and the expression of interferon-gamma-sensitive genes involved in immunity and apoptosis.


Subject(s)
Cell Nucleus/metabolism , Multiprotein Complexes/metabolism , Protein Kinases/metabolism , Protein Phosphatase 2/metabolism , STAT1 Transcription Factor/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/genetics , Apoptosis/immunology , Cell Line , Cell Nucleus/genetics , Cell Nucleus/immunology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Intracellular Signaling Peptides and Proteins , Molecular Chaperones , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Protein Kinases/genetics , Protein Kinases/immunology , Protein Phosphatase 2/genetics , Protein Phosphatase 2/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/immunology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...