Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Leukoc Biol ; 116(4): 779-792, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38652699

ABSTRACT

The mammalian target of rapamycin (mTOR) pathway plays a key role in determining immune cells function through modulation of their metabolic status. By specific deletion of Rictor in CD11c+ myeloid cells (referred to here as CD11cRicΔ/Δ), we investigated the role of mTOR complex 2 (mTORC2) signaling in dendritic cells (DCs) function in mice. We showed that upon dextran sulfate sodium-induced colitis, the lack of mTORC2 signaling CD11c+ cells diminishes the colitis score and abrogates DC migration to the mesenteric lymph nodes, thereby diminishing the infiltration of T helper 17 cells in the lamina propria and subsequent inflammation. These findings corroborate with the abrogation of cytoskeleton organization and the decreased activation of Rac1 and Cdc42 GTPases observed in CD11c+-mTORC2-deficient cells. Meta-analysis on colonic samples from ulcerative colitis patients revealed increased gene expression of proinflammatory cytokines, which coincided with augmented expression of the mTOR pathway, a positive correlation between the DC marker ITGAX and interleukin-6, the expression of RICTOR, and CDC42. Together, this work proposes that targeting mTORC2 on DCs offers a key to hamper inflammatory responses, and this way, ameliorates the progression and severity of intestinal inflammatory diseases.


Subject(s)
Cell Movement , Colitis , Dendritic Cells , Dextran Sulfate , Mechanistic Target of Rapamycin Complex 2 , Myeloid Cells , Signal Transduction , Animals , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Dendritic Cells/immunology , Dendritic Cells/metabolism , Colitis/pathology , Colitis/chemically induced , Colitis/immunology , Myeloid Cells/metabolism , Myeloid Cells/immunology , Dextran Sulfate/toxicity , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/genetics , CD11c Antigen/metabolism , cdc42 GTP-Binding Protein/metabolism , Humans , rac1 GTP-Binding Protein/metabolism , Mice, Inbred C57BL , Disease Models, Animal , Mice, Knockout , Neuropeptides , CD11 Antigens
2.
Int J Mol Sci ; 24(15)2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37569368

ABSTRACT

TOR proteins, also known as targets of rapamycin, are serine/threonine kinases involved in various signaling pathways that regulate cell growth. The protozoan parasite Giardia lamblia is the causative agent of giardiasis, a neglected infectious disease in humans. In this study, we used a bioinformatics approach to examine the structural features of GTOR, a G. lamblia TOR-like protein, and predict functional associations. Our findings confirmed that it shares significant similarities with functional TOR kinases, including a binding domain for the FKBP-rapamycin complex and a kinase domain resembling that of phosphatidylinositol 3-kinase-related kinases. In addition, it can form multiprotein complexes such as TORC1 and TORC2. These results provide valuable insights into the structure-function relationship of GTOR, highlighting its potential as a molecular target for controlling G. lamblia cell proliferation. Furthermore, our study represents a step toward rational drug design for specific anti-giardiasis therapeutic agents.


Subject(s)
Giardia lamblia , Giardiasis , Humans , Sirolimus/pharmacology , Giardia lamblia/metabolism , TOR Serine-Threonine Kinases/metabolism , Signal Transduction , Mechanistic Target of Rapamycin Complex 2/metabolism
3.
Front Immunol ; 13: 945980, 2022.
Article in English | MEDLINE | ID: mdl-35983057

ABSTRACT

CD8+ T lymphocytes are one of the main effector cells of the immune system, they protect the organism against intracellular threats such as viruses and bacteria, as well as neoplasms. It is currently well established that CD8+ T cells have distinct immune responses, given by their phenotypes Tc1, Tc2, Tc17, and TcReg. The cellular plasticity of such phenotypes depends on the presence of different combinations of cytokines in the extracellular medium. It is known that metabolic imbalances play an important role in immune response, but the precise role of metabolic disturbances on the differentiation and function of CD8+ T cells, however, has not been explored. In this work, we used a computational model to explore the potential effect of metabolic alterations such as hyperglycemia, high alcohol consumption, dyslipidemia, and diabetes on CD8+ T cell differentiation. Our model predicts that metabolic alterations preclude the effector function of all CD8+ T cell phenotypes except for TcReg cells. It also suggests that such inhibition originates from the increase of reactive oxygen species in response to metabolic stressors. Finally, we simulated the outcome of treating metabolic-inhibited CD8+ T cells with drugs targeting key molecules such as mTORC1, mTORC2, Akt, and others. We found that overstimulation of mTORC2 may restore cell differentiation and functions of all effector phenotypes, even in diabetic patients. These findings highlight the importance of our predictive model to find potential targets to strengthen immunosuppressed patients in chronic diseases, like diabetes.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocyte Activation , Cell Differentiation , Cytokines/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism
4.
Am J Physiol Endocrinol Metab ; 321(5): E592-E605, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34541875

ABSTRACT

Deletion of mechanistic target of rapamycin complex 2 (mTORC2) essential component rapamycin insensitive companion of mTOR (Rictor) by a Cre recombinase under control of the broad, nonadipocyte-specific aP2/FABP4 promoter impairs thermoregulation and brown adipose tissue (BAT) glucose uptake on acute cold exposure. We investigated herein whether adipocyte-specific mTORC2 deficiency affects BAT and inguinal white adipose tissue (iWAT) signaling, metabolism, and thermogenesis in cold-acclimated mice. For this, 8-wk-old male mice bearing Rictor deletion and therefore mTORC2 deficiency in adipocytes (adiponectin-Cre) and littermates controls were either kept at thermoneutrality (30 ± 1°C) or cold-acclimated (10 ± 1°C) for 14 days and evaluated for BAT and iWAT signaling, metabolism, and thermogenesis. Cold acclimation inhibited mTORC2 in BAT and iWAT, but its residual activity is still required for the cold-induced increases in BAT adipocyte number, total UCP-1 content and mRNA levels of proliferation markers Ki67 and cyclin 1 D, and de novo lipogenesis enzymes ATP-citrate lyase and acetyl-CoA carboxylase. In iWAT, mTORC2 residual activity is partially required for the cold-induced increases in multilocular adipocytes, mitochondrial mass, and uncoupling protein 1 (UCP-1) content. Conversely, BAT mTORC1 activity and BAT and iWAT glucose uptake were upregulated by cold independently of mTORC2. Noteworthy, the impairment in BAT and iWAT total UCP-1 content and thermogenic capacity induced by adipocyte mTORC2 deficiency had no major impact on whole body energy expenditure in cold-acclimated mice due to a compensatory activation of muscle shivering. In conclusion, adipocyte mTORC2 deficiency impairs, through different mechanisms, BAT and iWAT total UCP-1 content and thermogenic capacity in cold-acclimated mice, without affecting glucose uptake and whole body energy expenditure.NEW & NOTEWORTHY BAT and iWAT mTORC2 is inhibited by cold acclimation, but its residual activity is required for cold-induced increases in total UCP-1 content and thermogenic capacity, but not glucose uptake and mTORC1 activity. The impaired BAT and iWAT total UCP-1 content and thermogenic capacity induced by adipocyte mTORC2 deficiency are compensated by activation of muscle shivering in cold-acclimated mice.


Subject(s)
Acclimatization/physiology , Adipocytes/metabolism , Adipose Tissue, Brown/physiology , Adipose Tissue, White/physiology , Energy Metabolism/physiology , Glucose/metabolism , Mechanistic Target of Rapamycin Complex 2/deficiency , Thermogenesis/genetics , Animals , Cold Temperature , Gene Deletion , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Male , Mechanistic Target of Rapamycin Complex 2/genetics , Mice , Mice, Inbred C57BL , Uncoupling Protein 1
5.
Neoplasia ; 23(9): 951-965, 2021 09.
Article in English | MEDLINE | ID: mdl-34343821

ABSTRACT

The Hippo and mTOR signaling cascades are major regulators of cell growth and division. Aberrant regulation of these pathways has been demonstrated to contribute to gliomagenesis and result in enhanced glioblastoma proliferation and invasive characteristics. Several crosstalk mechanisms have been described between these two pathways, although a complete picture of these signaling interactions is lacking and is required for effective therapeutic targeting. Here we report the ability of mTORC2 to directly phosphorylate YAP at serine 436 (Ser436) positively regulating YAP activity. We show that mTORC2 activity enhances YAP transcriptional activity and the induction of YAP-dependent target gene expression while its ablation via genetic or pharmacological means has the opposite affects on YAP function. mTORC2 interacts with YAP via Sin1 and mutational analysis of serine 436 demonstrates that this phosphorylation event affects several properties of YAP leading to enhanced transactivation potential. Moreover, YAP serine 436 mutants display altered glioblastoma growth, migratory capacity and invasiveness both in vitro and in xenograft experiments. We further demonstrate that mTORC2 is able to regulate a Hippo pathway resistant allele of YAP suggesting that mTORC2 can regulate YAP independent of Hippo signaling. Correlative associations between the expression of these components in GBM patient samples also supported the presence of this signaling relationship. These results advance a direct mTORC2/YAP signaling axis driving GBM growth, motility and invasiveness.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Tumor Burden/physiology , YAP-Signaling Proteins/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Female , Glioblastoma/genetics , Glioblastoma/pathology , Hippo Signaling Pathway/physiology , Humans , Mechanistic Target of Rapamycin Complex 2/genetics , Mice , Mice, SCID , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphorylation/physiology , Xenograft Model Antitumor Assays/methods , YAP-Signaling Proteins/genetics
6.
Mol Nutr Food Res ; 65(1): e1900768, 2021 01.
Article in English | MEDLINE | ID: mdl-32103588

ABSTRACT

SCOPE: Evidence gathered in the last decades suggests that lipotoxicity and inflammation are the main factors connecting adipose tissue dysfunction to the development of metabolic diseases such as insulin resistance, nonalcoholic fatty liver disease (NAFLD), cardiovascular disease, and certain types of cancer, among others. The mechanistic target of rapamycin (mTOR) is a serine threonine kinase that functions as the catalytic entity of two multiprotein complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). These complexes are important components of signaling pathways activated by nutrients, growth factors, and inflammatory mediators and are therefore directly involved in the regulation of adipocyte and macrophage metabolism and function. METHODS AND RESULTS: In this article, studies that evaluate the involvement of mTORC1 and 2 in the regulation of macrophage and adipocyte function and their implication in the development of metabolic-disease-associated adipose tissue dysfunction are reviewed. CONCLUSION: In adipocytes, optimal levels of mTORC1 activity are required for its pro-lipogenic actions, while in macrophages, mTORC1 regulates features of both M1 and M2 polarization. mTORC2, on the other hand, promotes glucose uptake and de novo lipogenesis in adipocytes and counteracts macrophage inflammatory response.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, White/pathology , Macrophages/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Adipose Tissue, White/metabolism , Animals , Humans , Inflammation Mediators/metabolism , Metabolic Diseases/metabolism , Obesity/metabolism , Obesity/pathology
7.
PLoS Genet ; 16(5): e1008255, 2020 05.
Article in English | MEDLINE | ID: mdl-32392211

ABSTRACT

mTOR, a serine/threonine protein kinase that is involved in a series of critical cellular processes, can be found in two functionally distinct complexes, mTORC1 and mTORC2. In contrast to mTORC1, little is known about the mechanisms that regulate mTORC2. Here we show that mTORC2 activity is reduced in mice with a hypomorphic mutation of the Ric-8B gene. Ric-8B is a highly conserved protein that acts as a non-canonical guanine nucleotide exchange factor (GEF) for heterotrimeric Gαs/olf type subunits. We found that Ric-8B hypomorph embryos are smaller than their wild type littermates, fail to close the neural tube in the cephalic region and die during mid-embryogenesis. Comparative transcriptome analysis revealed that signaling pathways involving GPCRs and G proteins are dysregulated in the Ric-8B mutant embryos. Interestingly, this analysis also revealed an unexpected impairment of the mTOR signaling pathway. Phosphorylation of Akt at Ser473 is downregulated in the Ric-8B mutant embryos, indicating a decreased activity of mTORC2. Knockdown of the endogenous Ric-8B gene in cultured cell lines leads to reduced phosphorylation levels of Akt (Ser473), further supporting the involvement of Ric-8B in mTORC2 activity. Our results reveal a crucial role for Ric-8B in development and provide novel insights into the signals that regulate mTORC2.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Animals , Cells, Cultured , Down-Regulation/genetics , Embryo, Mammalian , Embryonic Development/genetics , Female , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Developmental , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics
8.
PLoS One ; 14(4): e0215871, 2019.
Article in English | MEDLINE | ID: mdl-31002704

ABSTRACT

Tubule-interstitial injury (TII) is a critical step in the progression of renal disease. It has been proposed that changes in proximal tubule (PT) albumin endocytosis plays an important role in the development of TII. Some reports have shown protective effects of lithium on kidney injury animal models that was correlated to proteinuria. We tested the hypothesis that lithium treatment ameliorates the development of TII due to changes in albumin endocytosis. Two experimental models were used: (1) TII induced by albumin overload in an animal model; (2) LLC-PK1 cells, a PT cell line. Lithium treatment ameliorates TII induced by albumin overload measured by (1) proteinuria; (2) collagen deposition; (3) area of tubule-interstitial space, and (4) macrophage infiltration. Lithium treatment increased mTORC2 activity leading to the phosphorylation of protein kinase B (PKB) at Ser473 and its activation. This mechanism enhanced albumin endocytosis in PT cells, which decreased the proteinuria observed in TII induced by albumin overload. This effect did not involve changes in the expression of megalin, a PT albumin receptor. In addition, activation of this pathway decreased apoptosis in LLC-PK1 cells, a PT cell line, induced by higher albumin concentration, similar to that found in pathophysiologic conditions. Our results indicate that the protective role of lithium treatment on TII induced by albumin overload involves an increase in PT albumin endocytosis due to activation of the mTORC2/PKB pathway. These results open new possibilities in understanding the effects of lithium on the progression of renal disease.


Subject(s)
Kidney Tubules, Proximal/drug effects , Lithium Carbonate/pharmacology , Mechanistic Target of Rapamycin Complex 2/genetics , Protective Agents/pharmacology , Proteinuria/drug therapy , Proto-Oncogene Proteins c-akt/genetics , Albumins/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cell Movement/drug effects , Endocytosis/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Humans , Kidney Tubules, Proximal/injuries , Kidney Tubules, Proximal/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mechanistic Target of Rapamycin Complex 2/agonists , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred BALB C , Proteinuria/metabolism , Proteinuria/physiopathology , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
9.
Mol Cell Endocrinol ; 479: 78-86, 2019 01 05.
Article in English | MEDLINE | ID: mdl-30217602

ABSTRACT

Maternal diabetes programs cardiovascular alterations in the adult offspring but the mechanisms involved remain unclarified. Here, we addresed whether maternal diabetes programs cardiac alterations related to extracellular matrix remodeling in the adult offspring, as well as the role of forkhead box transcription factor 1 (FOXO1) in the induction of these alterations. The heart from adult offspring from control and streptozotocin-induced diabetic rats was evaluated. Increased glycemia, triglyceridemia and insulinemia and markers of cardiomyopathy were found in the offspring from diabetic rats. In the heart, an increase in active FOXO1 and mRNA levels of its target genes, Mmp-2 and Ctgf, genes related to an altered extracellular matrix remodeling, together with an increase in collagen deposition and a decrease in the connexin43 levels, were found in the offspring from diabetic rats. Altogether, these results suggest an important role of FOXO1 activation in the cardiac alterations induced by intrauterine programming in maternal diabetes.


Subject(s)
Cardiovascular Diseases/metabolism , Diabetes, Gestational/metabolism , Nerve Tissue Proteins/metabolism , Animals , Biomarkers/metabolism , Body Weight , Collagen/metabolism , Connexin 43/metabolism , Extracellular Matrix/metabolism , Female , Matrix Metalloproteinase 2/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Myocardium/metabolism , Myocardium/pathology , Organ Size , Phosphorylation , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , TOR Serine-Threonine Kinases/metabolism
10.
Nutrition ; 54: 94-99, 2018 10.
Article in English | MEDLINE | ID: mdl-29778908

ABSTRACT

OBJECTIVES: After bariatric surgery, modifications to signaling pathway networks including those of the metabolic regulator called mammalian or mechanistic target of rapamycin (mTOR) may lead to molecular alterations related to energy source availability, systemic nutrients, and catabolic and anabolic cellular processes. This study aimed to identify gene expression changes with regard to the mTOR complex 2 subunit signaling pathway in obese patients before and after bariatric surgery. METHODS: The experimental group included 13 obese women who were examined before (preoperative) and 6 mo after (postoperative) Roux-en-Y gastric bypass (RYGB) surgery. The control group included nine apparently eutrophic women matched by age and without any other metabolic diseases (i.e., no diabetes and no liver or kidney diseases). Peripheral blood mononuclear cell samples were collected for RNA extraction and subsequent microarray analysis. RESULTS: After this methodological procedure, we identified 47 000 differentially expressed genes. A subsequent bioinformatic analysis showed that three diferentially expressed genes (rapamycin-insensitive companion of mTOR [RICTOR], phosphoinositide-3-kinase regulatory subunit 1 [PIK3 R1], and hypoxia inducible factor 1 alpha subunit 1A [HIF1 A]) participated in the mTOR signaling pathway. Real-time quantitative polymerase chain reaction revealed that RICTOR, PIK3 R1, and HIF1 A were upregulated 6 mo after RYGB surgery (P <0.05). In addition, patients in the experimental group lost weight significantly and presented significant improvement in biochemical/metabolic variables. CONCLUSIONS: The weight loss that was induced by RYGB surgery alters the mTOR signaling pathway and specifically the mTOR complex 2 subunit. The increased expression of genes that act in this pathway such as RICTOR, PIK3 R1, and HIF1 A reflects the induced weight loss and improved metabolic indicators (e.g., insulin resistance and lipolysis) that are evidenced in this study.


Subject(s)
Gastric Bypass , Mechanistic Target of Rapamycin Complex 2/metabolism , Obesity/genetics , Signal Transduction/genetics , Weight Loss/genetics , Adult , Female , Humans , Leukocytes, Mononuclear/metabolism , Longitudinal Studies , Middle Aged , Obesity/surgery , Postoperative Period , Preoperative Period , Treatment Outcome
11.
Ann Neurol ; 83(2): 311-327, 2018 02.
Article in English | MEDLINE | ID: mdl-29331082

ABSTRACT

OBJECTIVE: Temporal lobe epilepsy (TLE) is a chronic epilepsy syndrome defined by seizures and progressive neurological disabilities, including cognitive impairments, anxiety, and depression. Here, human TLE specimens were investigated focusing on the mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) and complex 2 (mTORC2) activities in the brain, given that both pathways may represent unique targets for treatment. METHODS: Surgically resected hippocampal and temporal lobe samples from therapy-resistant TLE patients were analyzed by western blotting to quantify the expression of established mTORC1 and mTORC2 activity markers and upstream or downstream signaling pathways involving the two complexes. Histological and immunohistochemical techniques were used to assess hippocampal and neocortical structural abnormalities and cell-specific expression of individual biomarkers. Samples from patients with focal cortical dysplasia (FCD) type II served as positive controls. RESULTS: We found significantly increased expression of phospho-mTOR (Ser2448), phospho-S6 (Ser235/236), phospho-S6 (Ser240/244), and phospho-Akt (Ser473) in TLE samples compared to controls, consistent with activation of both mTORC1 and mTORC2. Our work identified the phosphoinositide 3-kinase and Ras/extracellular signal-regulated kinase signaling pathways as potential mTORC1 and mTORC2 upstream activators. In addition, we found that overactive mTORC2 signaling was accompanied by induction of two protein kinase B-dependent prosurvival pathways, as evidenced by increased inhibitory phosphorylation of forkhead box class O3a (Ser253) and glycogen synthase kinase 3 beta (Ser9). INTERPRETATION: Our data demonstrate that mTOR signaling is significantly dysregulated in human TLE, offering new targets for pharmacological interventions. Specifically, clinically available drugs that suppress mTORC1 without compromising mTOR2 signaling, such as rapamycin and its analogs, may represent a new group of antiepileptogenic agents in TLE patients. Ann Neurol 2018;83:311-327.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Signal Transduction/physiology , Adult , Brain/metabolism , Epilepsy, Temporal Lobe/physiopathology , Female , Humans , Male , Middle Aged , TOR Serine-Threonine Kinases/metabolism , Young Adult
12.
Transplantation ; 101(12): 2830-2840, 2017 12.
Article in English | MEDLINE | ID: mdl-28885497

ABSTRACT

BACKGROUND: Little is known about how new-generation adenosine triphosphate-competitive mechanistic target of rapamycin (mTOR) kinase inhibitors affect immunity and allograft rejection. METHODS: mTOR complex (C) 1 and 2 signaling in dendritic cells and T cells was analyzed by Western blotting, whereas immune cell populations in normal and heart allograft recipient mice were analyzed by flow cytometry. Alloreactive T cell proliferation was quantified in mixed leukocyte reaction; intracellular cytokine production and serum antidonor IgG levels were determined by flow analysis and immunofluorescence staining used to detect IgG in allografts. RESULTS: The novel target of rapamycin kinase inhibitor AZD2014 impaired dendritic cell differentiation and T cell proliferation in vitro and depressed immune cells and allospecific T cell responses in vivo. A 9-day course of AZD2014 (10 mg/kg, intraperitoneally, twice daily) or rapamycin (RAPA) (1 mg/kg, intraperitoneally, daily) prolonged median heart allograft survival time significantly (25 days for AZD2014, 100 days for RAPA, 9.5 days for control). Like RAPA, AZD2014 suppressed graft mononuclear cell infiltration, increased regulatory T cell to effector memory T cell ratios and reduced T follicular helper and B cells 7 days posttransplant. By 21 days (10 days after drug withdrawal), however, T follicular helper and B cells and donor-specific IgG1 and IgG2c antibody titers were significantly lower in RAPA-treated compared with AZD2014-treated mice. Elevated regulatory T cell to effector memory T cell ratios were maintained after RAPA, but not AZD2014 withdrawal. CONCLUSIONS: Immunomodulatory effects of AZD2014, unlike those of RAPA, were not sustained after drug withdrawal, possibly reflecting distinct pharmacokinetics or/and inhibitory effects of AZD2014 on mTORC2.


Subject(s)
Adenosine Triphosphate/chemistry , Graft Rejection , Heart Transplantation , Immune System/drug effects , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Morpholines/pharmacology , Animals , Benzamides , Cell Proliferation , Dendritic Cells/cytology , Graft Survival/drug effects , Immunoglobulin G/chemistry , Immunosuppressive Agents/pharmacology , Male , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Protein Kinase Inhibitors/pharmacology , Pyrimidines , Sirolimus/pharmacology , T-Lymphocytes/cytology , Transplantation, Homologous
13.
Exp Cell Res ; 339(1): 67-80, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26453937

ABSTRACT

The lack of a successful treatment for triple-negative breast cancer demands the study of the heterogeneity of cells that constitute these tumors. With this aim, two clones from triple negative breast MDA-MB-231 cancer cells were isolated: One with fibroblast-like appearance (F) and another with semi-epithelial (SE) morphology. Cells of the F clone have a higher migration and tumorigenesis capacity than SE cells, suggesting that these cells are in a more advanced stage of epithelial to mesenchymal transformation. In agreement, F cells have a diminished expression of the tight junction proteins claudins 1 and 4, and an increased content of ß-catenin. The latter is due to an augmented activity of the canonical Wnt route and of the EGFR/PI3K/mTORC2/AKT pathway favoring the cytoplasmic accumulation of ß-catenin and its transcriptional activity. In addition, F cells display increased phosphorylation of ß-catenin at Tyr654 by Src. These changes favor in F cells, the over-expression of Snail that promotes EMT. Finally, we observe that both F and SE cells display markers of cancer stem cells, which are more abundant in the F clone.


Subject(s)
ErbB Receptors/metabolism , Multiprotein Complexes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Triple Negative Breast Neoplasms/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Chemotaxis , Epithelial-Mesenchymal Transition , Female , Fluorescent Antibody Technique , Humans , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Nude , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Phosphorylation , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Am J Transplant ; 15(4): 891-902, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25737114

ABSTRACT

Since the discovery of Rapamycin (RAPA) and its immunosuppressive properties, enormous progress has been made in characterizing the mechanistic target of rapamycin (mTOR). Use of RAPA and its analogues (rapalogs) as anti-rejection agents has been accompanied by extensive investigation of how targeting of mTOR complex 1 (mTORC1), the principal target of RAPA, and more recently mTORC2, affects the function of immune cells, as well as vascular endothelial cells, that play crucial roles in regulation of allograft rejection. While considerable knowledge has accumulated on the function of mTORC1 and 2 in T cells, understanding of the differential roles of these complexes in antigen-presenting cells, NK cells and B cells/plasma cells is only beginning to emerge. Immune cell-specific targeting of mTORC1 or mTORC2, together with use of novel, second generation, dual mTORC kinase inhibitors (TORKinibs) have started to play an important role in elucidating the roles of these complexes and their potential for targeting in transplantation. Much remains unknown about the role of mTOR complexes and the consequences of mTOR targeting on immune reactivity in clinical transplantation. Here we address recent advances in understanding and evolving perspectives of the role of mTOR complexes and mTOR targeting in immunity, with extrapolation to transplantation.


Subject(s)
Immune System/physiology , Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , Transplantation , Animals , Humans , Immunosuppressive Agents/administration & dosage , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2
16.
PLoS One ; 9(4): e95432, 2014.
Article in English | MEDLINE | ID: mdl-24740015

ABSTRACT

The phosphoinositide-3-kinase (PI3K)/protein kinase B (Akt) axis plays a central role in attenuating inflammation upon macrophage stimulation with toll-like receptor (TLR) ligands. The mechanistic target of rapamycin complex 2 (mTORC2) relays signal from PI3K to Akt but its role in modulating inflammation in vivo has never been investigated. To evaluate the role of mTORC2 in the regulation of inflammation in vivo, we have generated a mouse model lacking Rictor, an essential mTORC2 component, in myeloid cells. Primary macrophages isolated from myeloid-specific Rictor null mice exhibited an exaggerated response to TLRs ligands, and expressed high levels of M1 genes and lower levels of M2 markers. To determine whether the loss of Rictor similarly affected inflammation in vivo, mice were either fed a high fat diet, a situation promoting chronic but low-grade inflammation, or were injected with lipopolysaccharide (LPS), which mimics an acute, severe septic inflammatory condition. Although high fat feeding contributed to promote obesity, inflammation, macrophage infiltration in adipose tissue and systemic insulin resistance, we did not observe a significant impact of Rictor loss on these parameters. However, mice lacking Rictor exhibited a higher sensitivity to septic shock when injected with LPS. Altogether, these results indicate that mTORC2 is a key negative regulator of macrophages TLR signalling and that its role in modulating inflammation is particularly important in the context of severe inflammatory challenges. These observations suggest that approaches aimed at modulating mTORC2 activity may represent a possible therapeutic approach for diseases linked to excessive inflammation.


Subject(s)
Carrier Proteins/genetics , Gene Deletion , Macrophages, Peritoneal/pathology , Obesity/pathology , Animals , Carrier Proteins/immunology , Diet, High-Fat , Fibroblasts/immunology , Fibroblasts/pathology , Gene Expression Regulation , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Insulin Resistance , Lipopolysaccharides , Macrophages, Peritoneal/immunology , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Obesity/chemically induced , Obesity/genetics , Obesity/immunology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Rapamycin-Insensitive Companion of mTOR Protein , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
17.
Biochem Biophys Res Commun ; 444(2): 218-23, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24462769

ABSTRACT

Diverse G protein-coupled receptors depend on Gßγ heterodimers to promote cell polarization and survival via direct activation of PI3Kγ and potentially other effectors. These events involve full activation of AKT via its phosphorylation at Ser473, suggesting that mTORC2, the kinase that phosphorylates AKT at Ser473, is activated downstream of Gßγ. Thus, we tested the hypothesis that Gßγ directly contributes to mTOR signaling. Here, we demonstrate that endogenous mTOR interacts with Gßγ. Cell stimulation with serum modulates Gßγ interaction with mTOR. The carboxyl terminal region of mTOR, expressed as a GST-fusion protein, including the serine/threonine kinase domain, binds Gßγ heterodimers containing different Gß subunits, except Gß4. Both, mTORC1 and mTORC2 complexes interact with Gß1γ2 which promotes phosphorylation of their respective substrates, p70S6K and AKT. In addition, chronic treatment with rapamycin, a condition known to interfere with assembly of mTORC2, reduces the interaction between Gßγ and mTOR and the phosphorylation of AKT; whereas overexpression of Gαi interfered with the effect of Gßγ as promoter of p70S6K and AKT phosphorylation. Altogether, our results suggest that Gßγ positively regulates mTOR signaling via direct interactions and provide further support to emerging strategies based on the therapeutical potential of inhibiting different Gßγ signaling interfaces.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , TOR Serine-Threonine Kinases/metabolism , Blotting, Western , Enzyme Activation/drug effects , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein gamma Subunits/genetics , HEK293 Cells , Humans , Immunoprecipitation , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , Two-Hybrid System Techniques
18.
Cell Signal ; 25(4): 829-38, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23291002

ABSTRACT

Epithelial-mesenchymal transition (EMT), proliferation and migration of RPE cells characterize the development of proliferative vitreoretinopathy (PVR) and other fibro-proliferative eye diseases leading to blindness. A common event in these pathologies is the alteration of the BRB which allows the interaction of RPE cells with thrombin, a pro-inflammatory protease contained in serum. Thrombin promotion of cytoskeletal reorganization, proliferation, and migration has been reported in different cell types, although the molecular mechanisms involved in these processes remain poorly understood. Our previous work demonstrated that thrombin promotes RPE cell proliferation, cytoskeletal remodeling and migration, hallmark processes in the development of PVR. Thrombin induction of RPE cell proliferation requires PI3K, PDK1, and Akt/PKB (Akt) signaling leading to cyclin D1 gene expression. Since Akt functions as an upstream activator of mechanistic target of rapamycin complex 1 (mTORC1) and is also a downstream target for mTORC2, the aim of this work was to determine whether mTOR is involved in thrombin-induced RPE cell proliferation by regulating cyclin D1 expression in immortalized rat RPE-J cell line. Results demonstrate that thrombin-induced cyclin D1 expression and cell proliferation require Akt-independent phosphorylation/activation of mTOR at Ser 2448 mediated by PI3K/PKC-ζ/ERK1/2 signaling, concomitant to Akt-dependent activation of p70S6K carried by mTORC1.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Multiprotein Complexes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Thrombin/pharmacology , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cell Proliferation/drug effects , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Cyclin D1/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA Interference , RNA, Small Interfering , Rapamycin-Insensitive Companion of mTOR Protein , Rats , Regulatory-Associated Protein of mTOR , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
19.
J Hypertens ; 31(2): 303-11, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23303355

ABSTRACT

OBJECTIVES: Insulin is recognized to increase renal salt reabsorption in the distal nephron and hyperinsulinemic states have been shown to be associated with increased expression of the renal NaCl cotransporter (NCC). However, the effect of insulin on NCC functional activity has not been reported. METHODS: Using a heterologous expression system of Xenopus laevis oocytes, a mouse distal convoluted cell line, mDCT15 cells, endogenously expressing NCC, and an ex-vivo kidney perfusion technique, we assessed the effect of insulin on the activity and phosphorylation of NCC. The signaling pathway involved was analyzed. RESULTS: In Xenopus oocytes insulin increases the activity of NCC together with its phosphorylation at threonine residue 58. Activation of NCC by insulin was also observed in mDCT15 cells. Additionally, insulin increased the NCC phosphorylation in kidney under the ex-vivo perfusion technique. In oocytes and mDCT15 cells, insulin effect on NCC was prevented with inhibitors of phosphatidylinositol 3-kinase (PI3K), mTORC2, and AKT1 kinases, but not by inhibitors of MAP or mTORC1 kinases, suggesting that PI3K-mTORC2-AKT1 is the intracellular pathway required. Additionally, activation of NCC by insulin was not affected by wild-type or mutant versions of with no lysine kinase 1, with no lysine kinase 4, or serum glucocorticoid kinase 1, but it was no longer observed in the presence of wild-type or the dominant negative, catalytically inactive with no lysine kinase 3, implicating this kinase in the process. CONCLUSION: Insulin induces activation and phosphorylation of NCC. This effect could play an important role in arterial hypertension associated with hyperinsulinemic states, such as obesity, metabolic syndrome, or type 2 diabetes mellitus.


Subject(s)
Insulin/pharmacology , Kidney/drug effects , Sodium Chloride Symporters/metabolism , Animals , Blotting, Western , Cells, Cultured , Kidney/metabolism , Mechanistic Target of Rapamycin Complex 2 , Mice , Multiprotein Complexes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Xenopus laevis
20.
Oxid Med Cell Longev ; 2013: 963217, 2013.
Article in English | MEDLINE | ID: mdl-24489988

ABSTRACT

Resveratrol, a phenolic compound found in various plants, including grapes, berries, and peanuts, shows promise for the treatment of cancer, aging, type 2 diabetes, and cardiovascular diseases. Resveratrol can promote transcription factor nuclear factor-erythroid 2-related factor 2 (Nrf2) activation, increase the expression level of SIRT-1, which is a sirtuin family protein, and reduce mTOR pathway signaling. This compound has anti-inflammatory properties in that it inhibits or antagonizes the nuclear factor-κB (NF-κB) activity, which is a redox-sensitive transcription factor that coordinates the inflammatory response. Inflammation and oxidative stress, which are common features in patients with chronic kidney disease (CKD), are interrelated and associated with cardiovascular disease and the progression of CKD itself. Because of the modulation of the mechanisms involved in the inflammatory-oxidative stress cycle, resveratrol could play an important role in controlling CKD-related metabolic derangements. Although resveratrol supplementation in theory is a promising therapy in this patient group, there are no studies evaluating its effects. Thus, the present review aims to describe the role of resveratrol in inflammation and oxidative stress modulation and its possible benefits to patients with CKD.


Subject(s)
Renal Insufficiency, Chronic/drug therapy , Stilbenes/therapeutic use , Humans , Inflammation/pathology , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/metabolism , Oxidative Stress/drug effects , Resveratrol , Stilbenes/chemistry , Stilbenes/pharmacology , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL