97 results on '"MAMMALIAN TARGET"'
Search Results
2. Combinations of Cytotoxic Drugs, Ionizing Radiation, and Mammalian Target of Rapamycin (mTOR) Inhibitors
- Author
-
Sarkaria, Jann N., Brady, L. W., editor, Heilmann, H. -P., editor, Molls, M., editor, Brown, J. Martin, editor, Mehta, Minesh P., editor, and Nieder, Carsten, editor
- Published
- 2006
- Full Text
- View/download PDF
3. Multiple myeloma metabolism – a treasure trove of therapeutic targets?
- Author
-
Roman-Trufero, M, Auner, HW, Edwards, CM, and Cancer Research UK
- Subjects
KINASE GCN2 ,plasma cell ,Immunology ,Bone Marrow ,1108 Medical Microbiology ,Tumor Microenvironment ,Humans ,Immunology and Allergy ,BONE-MARROW MICROENVIRONMENT ,cancer biology ,Science & Technology ,proteostasis ,bone marrow (BM) adipocytes ,UNFOLDED PROTEIN RESPONSE ,SECRETORY APPARATUS ,ADIPOCYTES ,multiple myeloma ,proteasome ,MAMMALIAN TARGET ,PROTEASOME INHIBITION ,PLASMA-CELLS ,1107 Immunology ,B-CELLS ,AUTOPHAGY ,Proteasome Inhibitors ,Life Sciences & Biomedicine ,metabolism - Abstract
Multiple myeloma is an incurable cancer of plasma cells that is predominantly located in the bone marrow. Multiple myeloma cells are characterized by distinctive biological features that are intricately linked to their core function, the assembly and secretion of large amounts of antibodies, and their diverse interactions with the bone marrow microenvironment. Here, we provide a concise and introductory discussion of major metabolic hallmarks of plasma cells and myeloma cells, their roles in myeloma development and progression, and how they could be exploited for therapeutic purposes. We review the role of glucose consumption and catabolism, assess the dependency on glutamine to support key metabolic processes, and consider metabolic adaptations in drug-resistant myeloma cells. Finally, we examine the complex metabolic effects of proteasome inhibitors on myeloma cells and the extracellular matrix, and we explore the complex relationship between myeloma cells and bone marrow adipocytes.
- Published
- 2022
- Full Text
- View/download PDF
4. Mammalian target of rapamycin: Immunosuppressive drugs offer new insights into cell growth regulation
- Author
-
Abraham, Robert T., Parnham, Michael J., editor, Letts, L. Gordon, editor, and Morgan, Douglas W., editor
- Published
- 2000
- Full Text
- View/download PDF
5. The mTOR pathway is necessary for survival of mice with short telomeres
- Author
-
Maria A. Blasco, Carmen Blanco-Aparicio, Kurt Whittemore, Paula Martinez, Sarita Saraswati, Lydia Thelma Poluha, Iole Ferrara-Romeo, Osvaldo Graña-Castro, Juana M. Flores, Elena Hernández-Encinas, Rosa Serrano, Ministerio de Economía y Competitividad (España), Comunidad de Madrid (España), World Cancer Research Fund International, and Botín Foundation
- Subjects
0301 basic medicine ,Male ,Telomerase ,Aging ,General Physics and Astronomy ,PROTEIN ,Diseases ,mTORC1 ,0302 clinical medicine ,Neoplasms ,LENGTH ,TUBEROUS SCLEROSIS ,Phosphorylation ,lcsh:Science ,Mice, Knockout ,Multidisciplinary ,Kinase ,TOR Serine-Threonine Kinases ,Telomere ,CANCER ,Survival Rate ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,S6 KINASE ,Female ,EXPRESSION ,Cell biology ,Science ,EXTENDS LIFE-SPAN ,Longevity ,Biology ,Ribosomal Protein S6 Kinases, 90-kDa ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,medicine ,Animals ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Sirolimus ,Skeletal muscle ,General Chemistry ,GENE ,Mice, Inbred C57BL ,030104 developmental biology ,MAMMALIAN TARGET ,biology.protein ,Cancer research ,RNA ,lcsh:Q ,RAPAMYCIN ,DNA Damage - Abstract
Telomerase deficiency leads to age-related diseases and shorter lifespans. Inhibition of the mechanistic target of rapamycin (mTOR) delays aging and age-related pathologies. Here, we show that telomerase deficient mice with short telomeres (G2-Terc−/−) have an hyper-activated mTOR pathway with increased levels of phosphorylated ribosomal S6 protein in liver, skeletal muscle and heart, a target of mTORC1. Transcriptional profiling confirms mTOR activation in G2-Terc−/− livers. Treatment of G2-Terc−/− mice with rapamycin, an inhibitor of mTORC1, decreases survival, in contrast to lifespan extension in wild-type controls. Deletion of mTORC1 downstream S6 kinase 1 in G3-Terc−/− mice also decreases longevity, in contrast to lifespan extension in single S6K1−/− female mice. These findings demonstrate that mTOR is important for survival in the context of short telomeres, and that its inhibition is deleterious in this setting. These results are of clinical interest in the case of human syndromes characterized by critically short telomeres., Telomerase deficiency leads to age-related diseases and shortened lifespan, while inhibition of the mTOR pathway delays aging. Here, the authors show that inhibition of mTORC1 signaling shortens the lifespan of telomerase deficient mice.
- Published
- 2020
6. mTORC1 Inhibition Protects Human Regulatory T Cells From Granzyme-B-Induced Apoptosis
- Author
-
Siawosh K. Eskandari, Hazim Allos, Basmah S. Al Dulaijan, Gandolina Melhem, Ina Sulkaj, Juliano B. Alhaddad, Anis J. Saad, Christa Deban, Philip Chu, John Y. Choi, Branislav Kollar, Bohdan Pomahac, Leonardo V. Riella, Stefan P. Berger, Jan S. F. Sanders, Judy Lieberman, Li Li, Jamil R. Azzi, Groningen Kidney Center (GKC), and Groningen Institute for Organ Transplantation (GIOT)
- Subjects
EXPRESSION ,FOXP3 ,Immunology ,Receptors, Antigen, T-Cell ,RAPAMYCIN INHIBITION ,Apoptosis ,Mechanistic Target of Rapamycin Complex 1 ,T-Lymphocytes, Regulatory ,Granzymes ,CALCINEURIN ,Mice ,granzyme B ,grapoptosis ,KINASE ,Immunology and Allergy ,Animals ,Humans ,treg homeostasis ,MODULATION ,MEDIATED SUPPRESSION ,mTORC1 ,rapamycin ,INDUCTION ,PERFORIN ,human tregs ,MAMMALIAN TARGET - Abstract
Regulatory T cells (Tregs) have shown great promise as a means of cellular therapy in a multitude of allo- and auto-immune diseases—due in part to their immunosuppressive potency. Nevertheless, the clinical efficacy of human Tregsin patients has been limited by their poorin vivohomeostasis. To avert apoptosis, Tregsrequire stable antigenic (CD3ζ/T-cell-receptor-mediated), co-stimulatory (CD28-driven), and cytokine (IL-2-dependent) signaling. Notably, this sequence of signals supports an activated Tregphenotype that includes a high expression of granzymes, particularly granzyme B (GrB). Previously, we have shown that aside from the functional effects of GrB in lysing target cells to modulate allo-immunity, GrB can leak out of the intracellular lysosomal granules of host Tregs, initiating pro-apoptotic pathways. Here, we assessed the role of inhibiting mechanistic target of rapamycin complex 1 (mTORC1), a recently favored drug target in the transplant field, in regulating human TregapoptosisviaGrB. Usingex vivomodels of human Tregculture and a humanized mouse model of human skin allotransplantation, we found that by inhibiting mTORC1 using rapamycin, intracytoplasmic expression and functionality of GrB diminished in host Tregs; lowering human Tregapoptosis by in part decreasing the phosphorylation of S6K and c-Jun. These findings support the already clinically validated effects of mTORC1 inhibition in patients, most notably their stabilization of Tregbioactivity andin vivohomeostasis.
- Published
- 2022
7. Lysosomes at the Crossroads of Cell Metabolism, Cell Cycle, and Stemness
- Author
-
Arnaud Besson and Ada Nowosad
- Subjects
nutrient sensing ,Biochemistry & Molecular Biology ,Chemistry, Multidisciplinary ,CDK ,P70 S6 KINASE ,TRANSFER-RNA SYNTHETASE ,RAPAMYCIN COMPLEX 1 ,self-renewal ,Catalysis ,Inorganic Chemistry ,stemness ,cell metabolism ,cell signaling ,Animals ,Humans ,quiescence ,TUMOR-SUPPRESSOR ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy ,Science & Technology ,Organic Chemistry ,Cell Cycle ,RAG GTPASES ,Cell Differentiation ,General Medicine ,MTORC1 ACTIVATION ,Computer Science Applications ,Chemistry ,Adult Stem Cells ,HEMATOPOIETIC STEM ,MAMMALIAN TARGET ,VACUOLAR H+-ATPASE ,Physical Sciences ,lysosome ,mTOR ,AMINO-ACID TRANSPORTERS ,cell cycle ,Lysosomes ,Life Sciences & Biomedicine ,Metabolic Networks and Pathways ,Signal Transduction - Abstract
Initially described as lytic bodies due to their degradative and recycling functions, lysosomes play a critical role in metabolic adaptation to nutrient availability. More recently, the contribution of lysosomal proteins to cell signaling has been established, and lysosomes have emerged as signaling hubs that regulate diverse cellular processes, including cell proliferation and cell fate. Deciphering these signaling pathways has revealed an extensive crosstalk between the lysosomal and cell cycle machineries that is only beginning to be understood. Recent studies also indicate that a number of lysosomal proteins are involved in the regulation of embryonic and adult stem cell fate and identity. In this review, we will focus on the role of the lysosome as a signaling platform with an emphasis on its function in integrating nutrient sensing with proliferation and cell cycle progression, as well as in stemness-related features, such as self-renewal and quiescence. ispartof: INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES vol:23 issue:4 ispartof: location:Switzerland status: published
- Published
- 2022
8. A low aromatic amino-acid diet improves renal function and prevent kidney fibrosis in mice with chronic kidney disease
- Author
-
Barba, Christophe, Benoit, Bérengère, Bres, Emilie, Chanon, Stéphanie, Vieille-Marchiset, Aurélie, Pinteur, Claudie, Pesenti, Sandra, Glorieux, Griet, Picard, Cécile, Fouque, Denis, Soulage, Christophe O., Koppe, Laetitia, Cardiovasculaire, métabolisme, diabétologie et nutrition (CarMeN), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Hospices Civils de Lyon (HCL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre Hospitalier Lyon Sud [CHU - HCL] (CHLS), Hospices Civils de Lyon (HCL), Ghent University Hospital, and ROSSI, Sabine
- Subjects
Male ,030309 nutrition & dietetics ,P-CRESYL SULFATE ,Science ,030209 endocrinology & metabolism ,Kidney ,GLOMERULAR-FILTRATION-RATE ,Article ,MECHANISMS ,Amino Acids, Aromatic ,Mice ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Diet, Protein-Restricted ,Medicine and Health Sciences ,Animals ,Humans ,Uremic Toxins ,Renal Insufficiency, Chronic ,2. Zero hunger ,DAMAGE ,0303 health sciences ,Kidney diseases ,Malnutrition ,GUT MICROBIOTA ,LOW-PROTEIN DIET ,Fibrosis ,3. Good health ,Disease Models, Animal ,[SDV.AEN] Life Sciences [q-bio]/Food and Nutrition ,MAMMALIAN TARGET ,Nephrology ,Medicine ,[SDV.AEN]Life Sciences [q-bio]/Food and Nutrition - Abstract
International audience; Abstract Despite decades of use of low protein diets (LPD) in the management of chronic kidney disease (CKD), their mechanisms of action are unclear. A reduced production of uremic toxins could contribute to the benefits of LPDs. Aromatic amino-acids (AA) are precursors of major uremic toxins such as p-cresyl sulfate (PCS) and indoxyl sulfate (IS). We hypothesize that a low aromatic amino acid diet (LA-AAD, namely a low intake of tyrosine, tryptophan and phenylalanine) while being normoproteic, could be as effective as a LPD, through the decreased production of uremic toxins. Kidney failure was chemically induced in mice with a diet containing 0.25% (w/w) of adenine. Mice received three different diets for six weeks: normoproteic diet (NPD: 14.7% proteins, aromatic AAs 0.019%), LPD (5% proteins, aromatic AAs 0.007%) and LA-AAD (14% proteins, aromatic AAs 0.007%). Both LPD and LA-AAD significantly reduced proteinuria, kidney fibrosis and inflammation. While LPD only slightly decreased plasma free PCS and free IS compared to NPD; free fractions of both compounds were significantly decreased by LA-AAD. These results suggest that a LA-AAD confers similar benefits of a LPD in delaying the progression of CKD through a reduction in some key uremic toxins production (such as PCS and IS), with a lower risk of malnutrition.
- Published
- 2021
- Full Text
- View/download PDF
9. Rigosertib elicits potent anti-tumor responses in colorectal cancer by inhibiting Ras signaling pathway
- Author
-
Majid Ghayour Mobarhan, Farnaz Barneh, Elisa Giovannetti, Majid Khazaei, Milad Hashemzehi, Fereshteh Asgharzadeh, Gordon A. Ferns, Mohieddin Jafari, Reyhaneh Moradi Marjaneh, Seyed Mahdi Hassanian, Amir Avan, Mohammad Reza Ahmadian, Mohammad-Reza Parizadeh, Farzad Rahmani, Mikhail Ryzhikov, Amir R Afshari, Atena Soleimani, Medical oncology laboratory, CCA - Cancer biology and immunology, Amsterdam Gastroenterology Endocrinology Metabolism, Research Program in Systems Oncology, and Institute for Molecular Medicine Finland
- Subjects
0301 basic medicine ,Cell type ,Beta-catenin ,Angiogenesis ,Cell ,LEUKEMIA CELLS ,Glycine ,Apoptosis ,BETA-CATENIN ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,OXALIPLATIN ,Sulfones ,Ras signaling ,CYCLIN D1 ,Cell Proliferation ,biology ,Cell growth ,fungi ,Rigosertib ,Cell Biology ,IN-VITRO ,Xenograft Model Antitumor Assays ,3. Good health ,Colon cancer ,030104 developmental biology ,medicine.anatomical_structure ,PHASE-I ,Ras Signaling Pathway ,MAMMALIAN TARGET ,DNA-DAMAGE ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Mdm2 ,1182 Biochemistry, cell and molecular biology ,sense organs ,Rigoserib ,MESSENGER-RNA ,Colorectal Neoplasms ,Signal Transduction - Abstract
Background The therapeutic potency of Rigosertib (RGS) in the treatment of the myelodysplastic syndrome has been investigated previously, but little is known about its mechanisms of action. Methods The present study integrates systems and molecular biology approaches to investigate the mechanisms of the anti-tumor effects of RGS, either alone or in combination with 5-FU in cellular and animal models of colorectal cancer (CRC). Results The effects of RGS were more pronounced in dedifferentiated CRC cell types, compared to cell types that were epithelial-like. RGS inhibited cell proliferation and cell cycle progression in a cell-type specific manner, and that was dependent on the presence of mutations in KRAS, or its down-stream effectors. RGS increased both early and late apoptosis, by regulating the expression of p53, BAX and MDM2 in tumor model. We also found that RGS induced cell senescence in tumor tissues by increasing ROS generation, and impairing oxidant/anti-oxidant balance. RGS also inhibited angiogenesis and metastatic behavior of CRC cells, by regulating the expression of CD31, E-cadherin, and matrix metalloproteinases-2 and 9. Conclusion Our findings support the therapeutic potential of this potent RAS signaling inhibitor either alone or in combination with standard regimens for the management of patients with CRC.
- Published
- 2021
- Full Text
- View/download PDF
10. Podocytes maintain high basal levels of autophagy independent of mtor signaling
- Author
-
Bjoern Hartleben, Wei Liang, Shuya Liu, Kosuke Yamahara, Ketan Patel, Kathrin Thedieck, Tillmann Bork, Olivia Lenoir, Philipp Lee, Christoph Schell, Tobias B. Huber, Zhejia Tian, Pierre-Louise Tharaux, Department of Medicine IV [Freiburg, Germany] (Faculty of Medicine), University of Freiburg [Freiburg], Division of Nephrology [Wuhan, China], Wuhan University [China]-Renmin Hospital [Wuhan, China], Department of Medicine [Otsu, Japan], Shiga University of Medical Science, Department of Medicine [Hamburg, Germany], Universitaetsklinikum Hamburg-Eppendorf = University Medical Center Hamburg-Eppendorf [Hamburg] (UKE), Center for Molecular Biosciences Innsbruck [Innsbruck, Austria] (Institute of Biochemistry), University of Innsbruck, University Medical Center Groningen [Groningen] (UMCG), University of Oldenburg, Hannover Medical School [Hannover] (MHH), Freiburg Institute for Advanced Studies [Freiburg, Germany], Albert-Ludwigs-Universität Freiburg, School of Biological Sciences [Reading], University of Reading (UOR), Paris-Centre de Recherche Cardiovasculaire (PARCC - UMR-S U970), Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Service de néphrologie [Hôpital Européen Georges Pompidou - APHP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), This work was supported by the German Research Foundation [CRC1140, CRC1192], European Foundation for the Study of Diabetes (EFSD), German Research Foundation Heisenberg Program [HU1016/ 5-1], German Research Foundation Heisenberg Program [HU1016/8-2], EC | European Research Council (ERC), H2020-IMI2 BEAt-DKD [115974], BMBF-STOP-FSGS [01GM1518C], Excellence Initiative of the German Federal and State Governments BIOSS, FRIAS Freiburg Institute of Advanced Studies, Alexander von Humboldt Foundation, German Research Foundation [CRC992], National Natural Science Foundation of China (NSFC) [81470912], Berta Ottenstein Program, Else Kroener Fresenius Foundation NAKSYS, BMBF GlioPATH [01ZX1402B], BMBF MAPTor-NET [031A426B], German Research Foundation [TH 1358/3-1], MESI-STRAT [754688], Uehara Memorial Foundation, German TS Foundation, Stichting TSC Fonds (calls 2015 and 2017), PoLiMeR Innovative Training Network (Marie SklodowskaCurie grant agreement No. 812616, Rosalind-Franklin Fellowship of the Universitiy of Groningen., Tharaux, Pierre-Louis, Leopold Franzens Universität Innsbruck - University of Innsbruck, and Center for Liver, Digestive and Metabolic Diseases (CLDM)
- Subjects
AMPK ,0301 basic medicine ,podocyte ,Research Paper - Basic Science ,PROTEIN ,glomerulus ,Podocyte ,ACTIVATION ,LC3 ,PHOSPHORYLATION ,ULK1 ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,CELL-LINE ,biology ,Podocytes ,TOR Serine-Threonine Kinases ,MTOR ,Raptor ,Cell biology ,medicine.anatomical_structure ,signaling ,Signal Transduction ,Research Article ,autophagy ,kidney ,Mice, Transgenic ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Mechanistic Target of Rapamycin Complex 1 ,DIABETIC-NEPHROPATHY ,Nephrin ,03 medical and health sciences ,medicine ,Animals ,Humans ,Protein kinase A ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,Molecular Biology ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Sirolimus ,030102 biochemistry & molecular biology ,rapamycin ,RPTOR ,Autophagy ,KIDNEY-DISEASE ,Cell Biology ,Tsc1 ,030104 developmental biology ,MAMMALIAN TARGET ,biology.protein ,Podocin ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
International audience; While constant basal levels of macroautophagy/autophagy are a prerequisite to preserve long-lived podocytes at the filtration barrier, MTOR regulates at the same time podocyte size and compensatory hypertrophy. Since MTOR is known to generally suppress autophagy, the apparently independent regulation of these two key pathways of glomerular maintenance remained puzzling. We now report that long-term genetic manipulation of MTOR activity does in fact not influence high basal levels of autophagy in podocytes either in vitro or in vivo. Instead we present data showing that autophagy in podocytes is mainly controlled by AMP-activated protein kinase (AMPK) and ULK1 (unc-51 like kinase 1). Pharmacological inhibition of MTOR further shows that the uncoupling of MTOR activity and autophagy is time dependent. Together, our data reveal a novel and unexpected cell-specific mechanism, which permits concurrent MTOR activity as well as high basal autophagy rates in podocytes. Thus, these data indicate manipulation of the AMPK-ULK1 axis rather than inhibition of MTOR as a promising therapeutic intervention to enhance autophagy and preserve podocyte homeostasis in glomerular diseases.Abbreviations: AICAR: 5-aminoimidazole-4-carboxamide ribonucleotide; AMPK: AMP-activated protein kinase; ATG: autophagy related; BW: body weight; Cq: chloroquine; ER: endoplasmic reticulum; ESRD: end stage renal disease; FACS: fluorescence activated cell sorting; GFP: green fluorescent protein; i.p.: intra peritoneal; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; NPHS1: nephrosis 1, nephrin; NPHS2: nephrosis 2, podocin; PLA: proximity-ligation assay; PRKAA: 5'-AMP-activated protein kinase catalytic subunit alpha; RPTOR/RAPTOR: regulatory associated protein of MTOR, complex 1; RFP: red fluorescent protein; TSC1: tuberous sclerosis 1; ULK1: unc-51 like kinase 1.
- Published
- 2019
- Full Text
- View/download PDF
11. Delta(9)-Tetrahydrocannabinol Promotes Oligodendrocyte Development and CNS Myelination in Vivo
- Author
-
Neurociencias, Neurozientziak, Huerga Gómez, Alba, Aguado, Tania, Sánchez de la Torre, Anibal, Bernal Chico, Ana, Matute Almau, Carlos José, Mato Santos, Susana, Guzmán, Manuel, Galve Roperh, Ismael, Palazuelos, Javier, Neurociencias, Neurozientziak, Huerga Gómez, Alba, Aguado, Tania, Sánchez de la Torre, Anibal, Bernal Chico, Ana, Matute Almau, Carlos José, Mato Santos, Susana, Guzmán, Manuel, Galve Roperh, Ismael, and Palazuelos, Javier
- Abstract
Delta(9)-Tetrahydrocannabinol (THC), the main bioactive compound found in the plantCannabis sativa, exerts its effects by activating cannabinoid receptors present in many neural cells. Cannabinoid receptors are also physiologically engaged by endogenous cannabinoid compounds, the so-called endocannabinoids. Specifically, the endocannabinoid 2-arachidonoylglycerol has been highlighted as an important modulator of oligodendrocyte (OL) development at embryonic stages and in animal models of demyelination. However, the potential impact of THC exposure on OL lineage progression during the critical periods of postnatal myelination has never been explored. Here, we show that acute THC administration at early postnatal ages in mice enhanced OL development and CNS myelination in the subcortical white matter by promoting oligodendrocyte precursor cell cycle exit and differentiation. Mechanistically, THC-induced-myelination was mediated by CB(1)and CB(2)cannabinoid receptors, as demonstrated by the blockade of THC actions by selective receptor antagonists. Moreover, the THC-mediated modulation of oligodendroglial differentiation relied on the activation of the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway, as mTORC1 pharmacological inhibition prevented the THC effects. Our study identifies THC as an effective pharmacological strategy to enhance oligodendrogenesis and CNS myelination in vivo
- Published
- 2021
12. Deletion of mTOR in liver epithelial cells enhances hepatic metastasis of colon cancer
- Author
-
Roman Eickhoff, Long Jiao, Antje Egners, Lara R. Heij, Ulf P. Neumann, Hans Duimel, Johanna Roth, Sandra Jumpertz, Carmen López-Iglesias, Maximilian Schmeding, David Meierhofer, Andreas Kroh, Pilar Caro, Thorsten Cramer, Merve Erdem, RS: M4I - Maastricht MultiModal Molecular Imaging Institute, M4I, Microscopy CORE Lab, Surgery, and RS: NUTRIM - R2 - Liver and digestive health
- Subjects
HOMEOSTASIS ,Colorectal cancer ,NF-KAPPA-B ,necroptosis ,Inflammation ,METABOLISM ,Pathology and Forensic Medicine ,Metastasis ,ACTIVATION ,PATHWAY ,Mice ,REGENERATION ,medicine ,Animals ,Neoplasm Metastasis ,liver regeneration ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Mice, Knockout ,INSULIN-RESISTANCE ,biology ,business.industry ,TOR Serine-Threonine Kinases ,Liver Neoplasms ,NF-kappa B ,DEATH ,Cancer ,medicine.disease ,Liver regeneration ,liver metastasis ,colon cancer ,MAMMALIAN TARGET ,inflammation ,Cancer cell ,Colonic Neoplasms ,Cancer research ,biology.protein ,mTOR ,Hepatocytes ,medicine.symptom ,business ,RAPAMYCIN - Abstract
Activation of the mechanistic target of rapamycin (mTOR) pathway is frequently found in cancer, but mTOR inhibitors have thus far failed to demonstrate significant antiproliferative efficacy in the majority of cancer types. Besides cancer cell-intrinsic resistance mechanisms, it is conceivable that mTOR inhibitors impact on non-malignant host cells in a manner that ultimately supports resistance of cancer cells. Against this background, we sought to analyze the functional consequences of mTOR inhibition in hepatocytes for the growth of metastatic colon cancer. To this end, we established liver epithelial cell (LEC)-specific knockout (KO) of mTOR (mTOR(LEC)) mice. We used these mice to characterize the growth of colorectal liver metastases with or without partial hepatectomy to model different clinical settings. Although the LEC-specific loss of mTOR remained without effect on metastasis growth in intact liver, partial liver resection resulted in the formation of larger metastases in mTOR(LEC) mice compared with wildtype controls. This was accompanied by significantly enhanced inflammatory activity in LEC-specific mTOR KO livers after partial liver resection. Analysis of NF-kappa B target gene expression and immunohistochemistry of p65 displayed a significant activation of NF-kappa B in mTOR(LEC) mice, suggesting a functional importance of this pathway for the observed inflammatory phenotype. Taken together, we show an unexpected acceleration of liver metastases upon deletion of mTOR in LECs. Our results support the notion that non-malignant host cells can contribute to resistance against mTOR inhibitors and encourage testing whether anti-inflammatory drugs are able to improve the efficacy of mTOR inhibitors for cancer therapy. (c) 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
- Published
- 2021
13. Evaluating Targeted Therapies in Ovarian Cancer Metabolism: Novel Role for PCSK9 and Second Generation mTOR Inhibitors
- Author
-
Sanz, Dafne Jacome, Raivola, Juuli, Karvonen, Hanna, Arjama, Mariliina, Barker, Harlan, Murumägi, Astrid, Ungureanu, Daniela, Tampere University, BioMediTech, Clinical Medicine, Genome-Scale Biology (GSB) Research Program, ATG - Applied Tumor Genomics, Faculty of Medicine, Institute for Molecular Medicine Finland, and Precision Systems Medicine
- Subjects
STRUCTURAL BASIS ,ovarian cancers ,CELL-SURVIVAL ,rapalogs ,3122 Cancers ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,ENDOMETRIAL ,DEGRADATION ,NVP-BEZ235 ,Article ,PCSK9 ,MAMMALIAN TARGET ,DISCOVERY ,omentum ,METASTASIS ,mTOR ,GROWTH ,3111 Biomedicine ,OPTIMIZATION ,drug testing ,metabolism ,RC254-282 - Abstract
Simple Summary Ovarian cancer (OC) is known for its poor prognosis, due to the absence of reliable biomarkers and its late diagnosis, since the early-stage disease is almost asymptomatic. Lipid metabolism plays an important role in OC progression due to the development of omental metastasis in the abdominal cavity. The aim of our study was to assess the therapeutic role of various enzymes involved in lipid metabolism regulation or synthesis, in different subtypes of OC represented by cell lines as well as patient-derived cancer cell cultures (PDCs). We show that proprotein convertase subtilisin/kexin type 9 (PCSK9), a cholesterol-regulating enzyme, plays a pro-survival role in OC and targeting its expression impairs cancer cell growth. We also tested a small library of metabolic and mTOR-targeting drugs to identify drug vulnerabilities specific to various subtypes of OC. Our results show that in OC cell lines and PDCs the second generation of mTOR inhibitors such as AZD8055, vistusertib, dactolisib and sapanisertib, have higher cytotoxic activity compared to the first generation mTOR inhibitors such as rapalogs. These results suggest that, in the era of precision medicine, it is possible to target the metabolic pathway in OC and identify subtype-specific drug vulnerabilities that could be advanced to the clinic. Background: Dysregulated lipid metabolism is emerging as a hallmark in several malignancies, including ovarian cancer (OC). Specifically, metastatic OC is highly dependent on lipid-rich omentum. We aimed to investigate the therapeutic value of targeting lipid metabolism in OC. For this purpose, we studied the role of PCSK9, a cholesterol-regulating enzyme, in OC cell survival and its downstream signaling. We also investigated the cytotoxic efficacy of a small library of metabolic (n = 11) and mTOR (n = 10) inhibitors using OC cell lines (n = 8) and ex vivo patient-derived cell cultures (PDCs, n = 5) to identify clinically suitable drug vulnerabilities. Targeting PCSK9 expression with siRNA or PCSK9 specific inhibitor (PF-06446846) impaired OC cell survival. In addition, overexpression of PCSK9 induced robust AKT phosphorylation along with increased expression of ERK1/2 and MEK1/2, suggesting a pro-survival role of PCSK9 in OC cells. Moreover, our drug testing revealed marked differences in cytotoxic responses to drugs targeting metabolic pathways of high-grade serous ovarian cancer (HGSOC) and low-grade serous ovarian cancer (LGSOC) PDCs. Our results show that targeting PCSK9 expression could impair OC cell survival, which warrants further investigation to address the dependency of this cancer on lipogenesis and omental metastasis. Moreover, the differences in metabolic gene expression and drug responses of OC PDCs indicate the existence of a metabolic heterogeneity within OC subtypes, which should be further explored for therapeutic improvements.
- Published
- 2021
14. Assessment of PI3K/mTOR/AKT Pathway Elements to Serve as Biomarkers and Therapeutic Targets in Penile Cancer
- Author
-
Jindrich Cinatl, Stephan Macher-Goeppinger, Jaroslav Cinatl, Maarten Albersen, Olesya Vakhrusheva, Katrin E. Tagscherer, Mario Schindeldecker, Florian Rothweiler, Andreas Neisius, Wilfried Roth, Philipp Stenzel, Igor Tsaur, Eva Juengel, Robert Dotzauer, Anita Thomas, Axel Haferkamp, Sascha Reetz, and Martin Michaelis
- Subjects
0301 basic medicine ,Cancer Research ,RM ,medicine.medical_treatment ,P70-S6 Kinase 1 ,cell lines ,BREAST ,Article ,Targeted therapy ,RC0254 ,03 medical and health sciences ,0302 clinical medicine ,PROGNOSTIC-FACTORS ,PIK3CA GENE ,Medicine ,Penile cancer ,PROSTATE ,ddc:610 ,Viability assay ,AKT EXPRESSION ,TISSUE MICROARRAY ,Protein kinase B ,PI3K/AKT/mTOR pathway ,RC254-282 ,HIGH-FREQUENCY ,Science & Technology ,Akt/PKB signaling pathway ,business.industry ,MUTATIONS ,AKT ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,penile cancer ,targeted therapy ,030104 developmental biology ,Oncology ,MAMMALIAN TARGET ,030220 oncology & carcinogenesis ,Cancer research ,mTOR ,Immunohistochemistry ,biomarker ,SQUAMOUS-CELL CARCINOMA ,business ,Life Sciences & Biomedicine - Abstract
Simple Summary: Penile cancer is a rare but aggressive malignancy characterized by rapid tumor growth as well as prompt metastasis in groin lymphatics. While localized diseases can be successfully cured by surgery in most cases, no truly effective treatment options have been established for metastatic diseases as of yet. In the current investigation, we assessed the value of selected members of the PI3K/mTOR/AKT pathway to serve as tumor markers or therapeutic targets for this disease. Higher expression of AKT was significantly more prevalent in high-grade tumors and independently predictive of the worse survival parameters, while increased expression of pmTOR was associated with an inferior prognosis as well. Treatment with the pan-AKT inhibitor capivasertib in PeCa cell lines induced significant reduction of cell viability and movement capacity. These findings might aid in the understanding of the molecular tumor background as well as development of novel treatment options for advanced penile cancer. Abstract: The PI3K/mTOR/AKT pathway might represent an intriguing option for treatment of penile cancer (PeCa). We aimed to assess whether members of this pathway might serve as biomarkers and targets for systemic therapy. Tissue of primary cancer from treatment-naïve PeCa patients was used for tissue microarray analysis. Immunohistochemical staining was performed with antibodies against AKT, pAKT, mTOR, pmTOR, pS6, pPRAS, p4EBP1, S6K1 and pp70S6K. Protein expression was correlated with clinicopathological characteristics as well as overall survival (OS), disease-specific survival (DSS), recurrence-free survival (RFS) and metastasis-free survival (MFS). AKT inhibition was tested in two primarily established, treatment-naïve PeCa cell lines by treatment with capivasertib and analysis of cell viability and chemotaxis. A total of 76 patients surgically treated for invasive PeCa were included. Higher expression of AKT was significantly more prevalent in high-grade tumors and predictive of DSS and OS in the Kaplan–Meier analysis, and an independent predictor of worse OS and DSS in the multivariate regression analysis. Treatment with pan-AKT inhibitor capivasertib in PeCa cell lines induced a significant downregulation of both total AKT and pAKT as well as decreased cell viability and chemotaxis. Selected protein candidates of the mTOR/AKT signaling pathway demonstrate association with histological and survival parameters of PeCa patients, whereas AKT appears to be the most promising one.
- Published
- 2021
15. Metformin inhibits polyphosphate-induced hyper-permeability and inflammation
- Author
-
Saeedeh Mehraban, Milad Hashemzehi, Reyhaneh Behnam-Rassouli, Mohammad-Hassan Arjmand, Farzad Rahmani, Maryam Fakhraie, Seyed Mahdi Hassanian, Mikhail Ryzhikov, Sayyed-Hadi Sayyed-Hosseinian, Farnaz Barneh, Mohieddin Jafari, Najmeh Jaberi, Mohammad Shabani, Seyede leili Adel barkhordar, Majid Khazaei, Gordon A. Ferns, Fatemeh Ariakia, Fereshteh Asgharzadeh, Amir Avan, Atena Soleimani, Research Program in Systems Oncology, and Research Programs Unit
- Subjects
Male ,Anti-Inflammatory Agents ,Vascular permeability ,030204 cardiovascular system & hematology ,VASCULAR-PERMEABILITY ,COMPLEMENT ,ACTIVATION ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,AMP-Activated Protein Kinase Kinases ,Cell Movement ,Polyphosphates ,Immunology and Allergy ,0303 health sciences ,Factor XII ,Mice, Inbred BALB C ,MOLECULAR-MECHANISMS ,Chemistry ,TOR Serine-Threonine Kinases ,PLATELETS ,Metformin ,3. Good health ,FACTOR-KAPPA-B ,317 Pharmacy ,medicine.symptom ,Oxidation-Reduction ,medicine.drug ,Signal Transduction ,Immunology ,COAGULATION ,Bradykinin ,Inflammation ,Capillary Permeability ,03 medical and health sciences ,Sepsis ,medicine ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Platelet activation ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Pharmacology ,AMPK ,digestive system diseases ,INORGANIC POLYPHOSPHATE ,Disease Models, Animal ,Oxidative Stress ,MAMMALIAN TARGET ,Cancer research ,AMPK signaling ,RESPONSES - Abstract
Circulating inflammatory factor inorganic polyphosphate (polyP) released from activated platelets could enhance factor XII and bradykinin resulted in increased capillary leakage and vascular permeability. PolyP induce inflammatory responses through mTOR pathway in endothelial cells, which is being reported in several diseases including atherosclerosis, thrombosis, sepsis, and cancer. Systems and molecular biology approaches were used to explore the regulatory role of the AMPK activator, metformin, on polyP-induced hyper-permeability in different organs in three different models of polyP-induced hyper-permeability including local, systemic shortand systemic long-term approaches in murine models. Our results showed that polyP disrupts endothelial barrier integrity in skin, liver, kidney, brain, heart, and lung in all three study models and metformin abrogates the disruptive effect of polyP. We also showed that activation of AMPK signaling pathway, regulation of oxidant/ anti-oxidant balance, as well as decrease in inflammatory cell infiltration constitute a set of molecular mechanisms through which metformin elicits it's protective responses against polyP-induced hyper-permeability. These results support the clinical values of AMPK activators including the FDA-approved metformin in attenuating vascular damage in polyP-associated inflammatory diseases.
- Published
- 2021
16. Finding new edges: systems approaches to MTOR signaling
- Author
-
Alexander Martin Heberle, Maria Rodríguez Peiris, Kathrin Thedieck, and Ulrike Rehbein
- Subjects
Computer science ,Bioinformatics ,Systems biology ,Cellular homeostasis ,Network topology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,BINDING PARTNER ,Animals ,Homeostasis ,Humans ,TUMOR-SUPPRESSOR ,Gene Regulatory Networks ,Protein Interaction Maps ,Systems Biology & Networks ,Mechanistic target of rapamycin ,Protein kinase B ,Review Articles ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,INSULIN-RESISTANCE ,0303 health sciences ,amino acids ,biology ,TOR Serine-Threonine Kinases ,RAG GTPASES ,Feed forward ,PHOSPHOPROTEOME REVEALS ,Computational Biology ,systems biology ,Systems approaches ,TSC1-TSC2 COMPLEX ,Signaling ,Systems Integration ,RICH AKT SUBSTRATE ,Metabolism ,MAMMALIAN TARGET ,030220 oncology & carcinogenesis ,computational models ,biology.protein ,KINASE-ACTIVITY ,protein kinase B ,GENE-PRODUCTS ,mechanistic target of rapamycin ,Neuroscience ,Signal Transduction - Abstract
Cells have evolved highly intertwined kinase networks to finely tune cellular homeostasis to the environment. The network converging on the mechanistic target of rapamycin (MTOR) kinase constitutes a central hub that integrates metabolic signals and adapts cellular metabolism and functions to nutritional changes and stress. Feedforward and feedback loops, crosstalks and a plethora of modulators finely balance MTOR-driven anabolic and catabolic processes. This complexity renders it difficult — if not impossible — to intuitively decipher signaling dynamics and network topology. Over the last two decades, systems approaches have emerged as powerful tools to simulate signaling network dynamics and responses. In this review, we discuss the contribution of systems studies to the discovery of novel edges and modulators in the MTOR network in healthy cells and in disease.
- Published
- 2021
- Full Text
- View/download PDF
17. ORP2, a cholesterol transporter, regulates angiogenic signaling in endothelial cells
- Author
-
Guoping Pan, Otto K. Kari, Riikka Kosonen, Arthur Ralko, Annika Koponen, Vesa M. Olkkonen, Annukka M. Kivelä, Daoguang Yan, Amita Arora, Elina Ikonen, Wonhwa Cho, Juuso H. Taskinen, Joseph Ndika, Medicum, Faculty of Pharmacy, Research Services, Biopharmaceutics Group, Division of Pharmaceutical Biosciences, Drug Delivery Unit, Drug Research Program, HUMI - Human Microbiome Research, Faculty of Medicine, Lipid Trafficking Lab, Department of Anatomy, Doctoral Programme in Biomedicine, Doctoral Programme in Integrative Life Science, Doctoral Programme in Drug Research, STEMM - Stem Cells and Metabolism Research Program, and Biosciences
- Subjects
Phosphatidylinositol 4,5-Diphosphate ,0301 basic medicine ,Receptors, Steroid ,Angiogenesis ,Osbpl2 ,UP-REGULATION ,Biochemistry ,angiogenesis ,0302 clinical medicine ,Cell Movement ,Lipid raft ,Receptors, Notch ,Chemistry ,TOR Serine-Threonine Kinases ,VEGF receptor ,Sterol homeostasis ,Cadherins ,Cell biology ,cholesterol trafficking ,Signal transduction ,Oxysterol-binding protein ,Signal Transduction ,Biotechnology ,GROWTH-FACTOR ,SUPPRESSES ANGIOGENESIS ,Nitric Oxide Synthase Type III ,Notch signaling pathway ,Neovascularization, Physiologic ,VE-CADHERIN ,Endosomes ,Caveolins ,03 medical and health sciences ,Antigens, CD ,Human Umbilical Vein Endothelial Cells ,Genetics ,Humans ,TRAFFICKING ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,RECEPTOR ,Cell Membrane ,Actins ,Matrix Metalloproteinases ,LIPID RAFTS ,Receptors, Vascular Endothelial Growth Factor ,030104 developmental biology ,MAMMALIAN TARGET ,1182 Biochemistry, cell and molecular biology ,OXYSTEROL-BINDING-PROTEIN ,3111 Biomedicine ,MEMBRANE ,Proto-Oncogene Proteins c-akt ,030217 neurology & neurosurgery - Abstract
https://doi.org/10.1096/fj.202000202R Oxysterol-binding protein-related protein 2 (ORP2), a cholesterol-PI(4,5)P(2)countercurrent transporter, was recently identified as a novel regulator of plasma membrane (PM) cholesterol and PI(4,5)P(2)content in HeLa cells. Here, we investigate the role of ORP2 in endothelial cell (EC) cholesterol and PI(4,5)P(2)distribution, angiogenic signaling, and angiogenesis. We show that ORP2 knock-down modifies the distribution of cholesterol accessible to a D4H probe, between late endosomes and the PM. Depletion of ORP2 from ECs inhibits their angiogenic tube formation capacity, alters the gene expression of angiogenic signaling pathways such as VEGFR2, Akt, mTOR, eNOS, and Notch, and reduces EC migration, proliferation, and cell viability. We show that ORP2 regulates the integrity of VEGFR2 at the PM in a cholesterol-dependent manner, the depletion of ORP2 resulting in proteolytic cleavage by matrix metalloproteinases, and reduced activity of VEGFR2 and its downstream signaling. We demonstrate that ORP2 depletion increases the PM PI(4,5)P(2)coincident with altered F-actin morphology, and reduces both VEGFR2 and cholesterol in buoyant raft membranes. Moreover, ORP2 knock-down suppresses the expression of the lipid raft-associated proteins VE-cadherin and caveolin-1. Analysis of the retinal microvasculature in ORP2 knock-out mice generated during this study demonstrates the subtle alterations of morphology characterized by reduced vessel length and increased density of tip cells and perpendicular sprouts. Gene expression changes in the retina suggest disturbance of sterol homeostasis, downregulation of VE-cadherin, and a putative disturbance of Notch signaling. Our data identifies ORP2 as a novel regulator of EC cholesterol and PI(4,5)P(2)homeostasis and cholesterol-dependent angiogenic signaling.
- Published
- 2020
18. E-selectin targeted immunoliposomes for rapamycin delivery to activated endothelial cells
- Author
-
Gholizadeh Soltani, Shima, Visweswaran, Ganesh Ram R, Storm, G, Hennink, Wim E., Kamps, Jan A A M, Kok, Robbert J., Afd Pharmaceutics, Pharmaceutics, Afd Pharmaceutics, Pharmaceutics, and Nanotechnology and Biophysics in Medicine (NANOBIOMED)
- Subjects
0301 basic medicine ,CANCER-THERAPY ,EXPERIMENTAL ARTHRITIS ,Cell Survival ,media_common.quotation_subject ,Endothelial cells ,Pharmaceutical Science ,02 engineering and technology ,Mice ,03 medical and health sciences ,Drug Delivery Systems ,E-selectin ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Humans ,Rapamycin ,Internalization ,LIPID-BILAYERS ,Protein kinase B ,PI3K/AKT/mTOR pathway ,media_common ,Sirolimus ,Dose-Response Relationship, Drug ,biology ,Immunoliposomes ,MTOR ,technology, industry, and agriculture ,ACTIN CYTOSKELETON ,IN-VITRO ,021001 nanoscience & nanotechnology ,Actin cytoskeleton ,Molecular biology ,TNF-ALPHA ,In vitro ,Anti-Bacterial Agents ,PROTEIN-KINASE B ,RHEUMATOID-ARTHRITIS ,030104 developmental biology ,MAMMALIAN TARGET ,Liposomes ,biology.protein ,Cancer research ,Tumor necrosis factor alpha ,Targeted delivery ,E-Selectin ,0210 nano-technology ,medicine.drug - Abstract
Activated endothelial cells play a pivotal role in the pathology of inflammatory disorders and thus present a target for therapeutic intervention by drugs that intervene in inflammatory signaling cascades, such as rapamycin (mammalian target of rapamycin (mTOR) inhibitor). In this study we developed anti-E-selectin immunoliposomes for targeted delivery to E-selectin over-expressing tumor necrosis factor-alpha (TNF-alpha) activated endothelial cells. Liposomes composed of 1,2-dipalmitoyl-sn-glycero-3.; hosphocholine (DPPC), Cholesterol, and 1,2-Distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethyleneglycol)-2000]-maleimide (DSPE-PEG- Mal) were loaded with rapamycin via lipid film hydration, after which they were further functionalized by coupling N-succinimidyl-S-acetylthioacetate (SATA)-modified mouse anti human E-selectin antibodies to the distal ends of the maleimidyl (Mal)-PEG groups. In cell binding assays, these immunoliposomes bound specifically to TNF-alpha activated endothelial cells. Upon internalization, rapamycin loaded immunoliposomes inhibited proliferation and migration of endothelial cells, as well as expression of inflammatory mediators. Our findings demonstrate that rapamycin-loaded immunoliposomes can specifically inhibit inflammatory responses in inflamed endothelial cells.
- Published
- 2018
19. Environmental arginine controls multinuclear giant cell metabolism and formation
- Author
-
Universitat Rovira i Virgili, Brunner, Julia S.; Vulliard, Loan; Hofmann, Melanie; Kieler, Markus; Lercher, Alexander; Vogel, Andrea; Russier, Marion; Brueggenthies, Johanna B.; Kerndl, Martina; Saferding, Victoria; Niederreiter, Birgit; Junza, Alexandra; Frauenstein, Annika; Scholtysek, Carina; Mikami, Yohei; Klavins, Kristaps; Kroenke, Gerhard; Bergthaler, Andreas; O'Shea, John J.; Weichhart, Thomas; Meissner, Felix; Smolen, Josef S.; Cheng, Paul; Yanes, Oscar; Menche, Joerg; Murray, Peter J.; Sharif, Omar; Blueml, Stephan; Schabbauer, Gernot, Universitat Rovira i Virgili, and Brunner, Julia S.; Vulliard, Loan; Hofmann, Melanie; Kieler, Markus; Lercher, Alexander; Vogel, Andrea; Russier, Marion; Brueggenthies, Johanna B.; Kerndl, Martina; Saferding, Victoria; Niederreiter, Birgit; Junza, Alexandra; Frauenstein, Annika; Scholtysek, Carina; Mikami, Yohei; Klavins, Kristaps; Kroenke, Gerhard; Bergthaler, Andreas; O'Shea, John J.; Weichhart, Thomas; Meissner, Felix; Smolen, Josef S.; Cheng, Paul; Yanes, Oscar; Menche, Joerg; Murray, Peter J.; Sharif, Omar; Blueml, Stephan; Schabbauer, Gernot
- Abstract
Multinucleated giant cells (MGCs) are implicated in many diseases including schistosomiasis, sarcoidosis and arthritis. MGC generation is energy intensive to enforce membrane fusion and cytoplasmic expansion. Using receptor activator of nuclear factor kappa-Beta ligand (RANKL) induced osteoclastogenesis to model MGC formation, here we report RANKL cellular programming requires extracellular arginine. Systemic arginine restriction improves outcome in multiple murine arthritis models and its removal induces preosteoclast metabolic quiescence, associated with impaired tricarboxylic acid (TCA) cycle function and metabolite induction. Effects of arginine deprivation on osteoclastogenesis are independent of mTORC1 activity or global transcriptional and translational inhibition. Arginine scarcity also dampens generation of IL-4 induced MGCs. Strikingly, in extracellular arginine absence, both cell types display flexibility as their formation can be restored with select arginine precursors. These data establish how environmental amino acids control the metabolic fate of polykaryons and suggest metabolic ways to manipulate MGC-associated pathologies and bone remodelling. Multinucleated giant cells (MGCs) are important in the pathogenesis of various diseases. Here, the authors demonstrate that extracellular presence of the amino acid arginine is required for MGC formation and metabolism, suggesting a translational impact for strategies utilizing systemic arginine depletion in MGC-mediated diseases.
- Published
- 2020
20. MTOR
- Author
-
Preedy, Victor R., editor and Watson, Ronald R., editor
- Published
- 2010
- Full Text
- View/download PDF
21. GSK3β Controls mTOR and Prosurvival Signaling in Neurons
- Author
-
Iwona A. Cymerman, Fred Van Leuven, Malgorzata Urbanska, Agata Gozdz, Matylda Macias, Herman Devijver, Ilona Kondratiuk, Ewa Liszewska, Jacek Jaworski, and Benoit Lechat
- Subjects
0301 basic medicine ,Regulator ,Apoptosis ,mTORC1 ,mTORC2 ,Neuronal survival ,Glycogen synthase kinases-3 ,TEMPORAL-LOBE EPILEPSY ,Phosphorylation ,IN-VIVO ,Cells, Cultured ,Neurons ,Ribosomal Protein S6 ,Mammalian target of rapamycin ,Kainic Acid ,TOR Serine-Threonine Kinases ,CONDITIONAL TRANSGENIC MICE ,CORTICAL-NEURONS ,INDUCED SEIZURES ,Brain ,Cell Differentiation ,PROTEIN TRANSLATION ,Cell biology ,Isoenzymes ,Crosstalk (biology) ,Neurology ,Signal transduction ,Life Sciences & Biomedicine ,Signal Transduction ,Cell Survival ,LYSOSOMAL ACIDIFICATION ,Neuroscience (miscellaneous) ,Mice, Transgenic ,Biology ,Neuroprotection ,CELL-PROLIFERATION ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Animals ,Excitotoxicity ,PI3K/AKT/mTOR pathway ,Science & Technology ,Glycogen Synthase Kinase 3 beta ,HIPPOCAMPAL-NEURONS ,RPTOR ,Neurosciences ,030104 developmental biology ,MAMMALIAN TARGET ,nervous system ,Neurosciences & Neurology ,Proto-Oncogene Proteins c-akt - Abstract
Glycogen synthase kinases-3β (GSK3β) is a key regulator of cell homeostasis. In neurons, GSK3β contributes to control of neuronal transmission and plasticity. Despite extensive studies in non-neuronal cells, crosstalk between GSK3β and other signaling pathways remains not well defined in neurons. In the present study, we report that GSK3β positively affected the activity of effectors of mammalian target of rapamycin complex 1 (mTORC1) and complex 2 (mTORC2), in mature neurons in vitro and in vivo. GSK3β also promoted prosurvival signaling and attenuated kainic acid-induced apoptosis. Our study identified GSK3β as a positive regulator of prosurvival signaling, including the mTOR pathway, and indicates the possible neuroprotective role of GSK3β in models of pharmacologically induced excitotoxicity. ispartof: MOLECULAR NEUROBIOLOGY vol:55 issue:7 pages:6050-6062 ispartof: location:United States status: published
- Published
- 2017
- Full Text
- View/download PDF
22. Advances in epilepsy gene discovery and implications for epilepsy diagnosis and treatment
- Author
-
Joseph D. Symonds, Sameer M. Zuberi, Marvin Johnson, and Imperial College Healthcare NHS Trust- BRC Funding
- Subjects
0301 basic medicine ,Potassium Channels ,PHENOTYPIC SPECTRUM ,ILAE COMMISSION ,Bioinformatics ,Epilepsy ,0302 clinical medicine ,OF-FUNCTION MUTATIONS ,Medicine ,genetics ,ENCEPHALOPATHY ,Phenotype ,Neurology ,SEVERE DEVELOPMENTAL DELAY ,Life Sciences & Biomedicine ,systems genetics ,precision medicine ,Clinical Neurology ,MEDLINE ,drug discovery ,03 medical and health sciences ,GRIN2A MUTATIONS ,Stratified medicine ,FOCAL CORTICAL DYSPLASIA ,Animals ,Humans ,Epilepsy therapy ,Genetic Association Studies ,De novo mutations ,Science & Technology ,Neurology & Neurosurgery ,business.industry ,Neurosciences ,1103 Clinical Sciences ,SOMATIC MUTATIONS ,1702 Cognitive Science ,medicine.disease ,Precision medicine ,Clinical neurology ,030104 developmental biology ,MAMMALIAN TARGET ,DE-NOVO MUTATIONS ,networks ,Mutation ,next-generation sequencing ,Neurosciences & Neurology ,Neurology (clinical) ,1109 Neurosciences ,business ,030217 neurology & neurosurgery ,Gene Discovery - Abstract
Purpose of review: Epilepsy genetics is shifting from the academic pursuit of gene discovery to a clinical discipline based on molecular diagnosis and stratified medicine. We consider the latest developments in epilepsy genetics and review how gene discovery in epilepsy is influencing the clinical classification of epilepsy and informing new therapeutic approaches and drug discovery. Recent findings: Recent studies highlighting the importance of mutation in GABA receptors, NMDA receptors, potassium channels, G-protein coupled receptors, mammalian target of rapamycin pathway and chromatin remodeling are discussed. Examples of precision medicine in epilepsy targeting gain-of-function mutations in KCNT1, GRIN2A, GRIN2D and SCN8A are presented. Potential reasons for the paucity of examples of precision medicine for loss-of-function mutations or in non-ion channel epilepsy genes are explored. We highlight how systems genetics and gene network analyses have suggested that pathways disrupted in epilepsy overlap with those of other neurodevelopmental traits including human cognition. We review how network-based computational approaches are now being applied to epilepsy drug discovery. Summary: We are living in an unparalleled era of epilepsy gene discovery. Advances in clinical care from this progress are already materializing through improved clinical diagnosis and stratified medicine. The application of targeted drug repurposing based on single gene defects has shown promise for epilepsy arising from gain-of-function mutations in ion-channel subunit genes, but important barriers remain to translating these approaches to non-ion channel epilepsy genes and loss-of-function mutations. Gene network analysis offers opportunities to discover new pathways for epilepsy, to decipher epilepsy's relationship to other neurodevelopmental traits and to frame a new approach to epilepsy drug discovery.
- Published
- 2017
- Full Text
- View/download PDF
23. The Impact of Hypoxia on Neutrophil Degranulation and Consequences for the Host
- Author
-
Alison M. Condliffe, Wei Li, Katharine M Lodge, Andrew S. Cowburn, Li, Wei [0000-0002-1924-3120], and Apollo - University of Cambridge Repository
- Subjects
BACTERICIDAL CAPACITY ,Chemistry, Multidisciplinary ,INDUCIBLE FACTOR-1-ALPHA ,STREPTOCOCCUS-PNEUMONIAE ,Review ,PHOSPHOINOSITIDE 3-KINASE ,Extracellular Traps ,Cell Degranulation ,Neutrophil Activation ,PULMONARY-HYPERTENSION ,lcsh:Chemistry ,0302 clinical medicine ,neutrophils ,WEIBEL-PALADE BODIES ,lcsh:QH301-705.5 ,Spectroscopy ,0303 health sciences ,INFLAMMATORY CELLS ,Degranulation ,General Medicine ,Cell Hypoxia ,Computer Science Applications ,Chemistry ,030220 oncology & carcinogenesis ,Physical Sciences ,medicine.symptom ,Life Sciences & Biomedicine ,Proteases ,Biochemistry & Molecular Biology ,Phagocytosis ,0699 Other Biological Sciences ,Inflammation ,Infections ,Catalysis ,ENDOTHELIAL-GROWTH-FACTOR ,Inorganic Chemistry ,03 medical and health sciences ,0399 Other Chemical Sciences ,medicine ,Animals ,Humans ,Secretion ,Physical and Theoretical Chemistry ,Molecular Biology ,030304 developmental biology ,degranulation ,0604 Genetics ,Innate immune system ,Science & Technology ,Chemical Physics ,hypoxia ,Secretory Vesicles ,Organic Chemistry ,Neutrophil extracellular traps ,Immunity, Innate ,MAMMALIAN TARGET ,lcsh:Biology (General) ,lcsh:QD1-999 ,Immunology ,Neutrophil degranulation - Abstract
Neutrophils are key effector cells of innate immunity, rapidly recruited to defend the host against invading pathogens. Neutrophils may kill pathogens intracellularly, following phagocytosis, or extracellularly, by degranulation and the release of neutrophil extracellular traps; all of these microbicidal strategies require the deployment of cytotoxic proteins and proteases, packaged during neutrophil development within cytoplasmic granules. Neutrophils operate in infected and inflamed tissues, which can be profoundly hypoxic. Neutrophilic infiltration of hypoxic tissues characterises a myriad of acute and chronic infectious and inflammatory diseases, and as well as potentially protecting the host from pathogens, neutrophil granule products have been implicated in causing collateral tissue damage in these scenarios. This review discusses the evidence for the enhanced secretion of destructive neutrophil granule contents observed in hypoxic environments and the potential mechanisms for this heightened granule exocytosis, highlighting implications for the host. Understanding the dichotomy of the beneficial and detrimental consequences of neutrophil degranulation in hypoxic environments is crucial to inform potential neutrophil-directed therapeutics in order to limit persistent, excessive, or inappropriate inflammation.
- Published
- 2020
24. Δ9-Tetrahydrocannabinol promotes oligodendrocyte development and CNS myelination in vivo
- Author
-
Alba Huerga-Gómez, Susana Mato, Tania Aguado, Ana Bernal-Chico, Manuel Guzmán, Aníbal Sánchez-de la Torre, Javier Palazuelos, Ismael Galve-Roperh, and Carlos Matute
- Subjects
0301 basic medicine ,Bioquímica ,Cannabinoid receptor ,cell-proliferation ,Neurociencias ,oligodendrocyte precursor cells ,mTORC1 ,Biology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,cannabinoids ,0302 clinical medicine ,cannabinoid receptors ,In vivo ,mental disorders ,medicine ,Rapamycin mTOR ,marijuana use ,Receptor ,Tetrahydrocannabinol ,white-matter ,organic chemicals ,myelination ,differentiation ,Endocannabinoid system ,CB1 ,Oligodendrocyte ,CB2 ,mammalian target ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,remyelination ,Neurology ,exposure ,lipids (amino acids, peptides, and proteins) ,Signal transduction ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Delta(9)-Tetrahydrocannabinol (THC), the main bioactive compound found in the plantCannabis sativa, exerts its effects by activating cannabinoid receptors present in many neural cells. Cannabinoid receptors are also physiologically engaged by endogenous cannabinoid compounds, the so-called endocannabinoids. Specifically, the endocannabinoid 2-arachidonoylglycerol has been highlighted as an important modulator of oligodendrocyte (OL) development at embryonic stages and in animal models of demyelination. However, the potential impact of THC exposure on OL lineage progression during the critical periods of postnatal myelination has never been explored. Here, we show that acute THC administration at early postnatal ages in mice enhanced OL development and CNS myelination in the subcortical white matter by promoting oligodendrocyte precursor cell cycle exit and differentiation. Mechanistically, THC-induced-myelination was mediated by CB(1)and CB(2)cannabinoid receptors, as demonstrated by the blockade of THC actions by selective receptor antagonists. Moreover, the THC-mediated modulation of oligodendroglial differentiation relied on the activation of the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway, as mTORC1 pharmacological inhibition prevented the THC effects. Our study identifies THC as an effective pharmacological strategy to enhance oligodendrogenesis and CNS myelination in vivo Centro de Investigacion Biomedica en Red Enfermedades Neurodegenerativas, Grant/Award Numbers: CB06/0005/0076, CB06/05/0005; Comunidad de Madrid, Grant/Award Numbers: 2016-T1/BMD-1060, PEJD2017-PRE/BMD-3703; Eusko Jaurlaritza, Grant/Award Numbers: IT1203-19, PIBA19-0059; Fundacion Tatiana Perez de Guzman el Bueno; FEDER and Instituto de Salud Carlos III, Grant/Award Numbers: PI18-00941, PI18/00513; Ministerio de Economia y Competitividad, Grant/Award Numbers: RTI2018-095311-B-I00, SAF2016-75292-R, SAF2017-83516-R
- Published
- 2020
25. A Newly Established Murine Cell Line as a Model for Hepatocellular Cancer in Non-Alcoholic Steatohepatitis
- Author
-
Jochen Nolting, Ulf P. Neumann, Tom Florian Ulmer, Athanassios Fragoulis, Roman Eickhoff, Herdit M. Schüler, Daniel Heise, Jeanette Walter, Thorsten Cramer, Andreas Kroh, Surgery, and RS: NUTRIM - R2 - Liver and digestive health
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,SCORING SYSTEM ,Cell ,HEPATITIS-C ,Mice ,0302 clinical medicine ,Liver Neoplasms, Experimental ,Non-alcoholic Fatty Liver Disease ,Spectroscopy ,biology ,General Medicine ,hepatocellular carcinoma ,cell line ,Cell cycle ,Computer Science Applications ,GLUTAMINE ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,ddc:540 ,LIVER-TRANSPLANT RECIPIENTS ,Neoplastic Stem Cells ,mTOR ,non-alcoholic steatohepatitis ,RENAL-FUNCTION ,Carcinoma, Hepatocellular ,CARCINOMA ,Morpholines ,Catalysis ,Article ,Inorganic Chemistry ,03 medical and health sciences ,Cancer stem cell ,Cell Line, Tumor ,medicine ,Biomarkers, Tumor ,Animals ,Everolimus ,Physical and Theoretical Chemistry ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Cell growth ,Organic Chemistry ,CD44 ,KU-0063794 ,Antigens, Differentiation ,digestive system diseases ,030104 developmental biology ,Pyrimidines ,MAMMALIAN TARGET ,Cancer cell ,biology.protein ,Cancer research ,RISK-FACTORS - Abstract
International journal of molecular sciences 20(22), 5658 (2019). doi:10.3390/ijms20225658, Published by Molecular Diversity Preservation International, Basel
- Published
- 2019
- Full Text
- View/download PDF
26. The TBC/RabGAP Armus Coordinates Rac1 and Rab7 Functions during Autophagy
- Author
-
Reiko Daigaku, Mattéa J. Finelli, Noor Faizah Mohd-Naim, Richard E. Francis, Clare E. Futter, Louis Perdios, Ivan Dikic, Vania M.M. Braga, Marieke A. M. Frasa, Sigrid B. Thoresen, Bernadette Carroll, Filipe C. Maximiano, Jessica McCormack, Medical Research Council (MRC), and Cancer Research UK
- Subjects
Keratinocytes ,rac1 GTP-Binding Protein ,GTPase-activating protein ,Endocytic cycle ,Fluorescent Antibody Technique ,GTPase ,Guanosine triphosphate ,CELL BIOLOGY ,CELL-MIGRATION ,ACTIVATION ,chemistry.chemical_compound ,0302 clinical medicine ,Phagosomes ,Protein Interaction Mapping ,0303 health sciences ,GTPase-Activating Proteins ,11 Medical And Health Sciences ,CANCER ,Cell biology ,Vesicular transport protein ,Protein Transport ,DEVELOPMENTAL BIOLOGY ,030220 oncology & carcinogenesis ,Guanosine Triphosphate ,Life Sciences & Biomedicine ,ATG8 ,Microtubule-Associated Proteins ,Protein Binding ,Signal Transduction ,PROTEINS ,Molecular Sequence Data ,ENDOPLASMIC-RETICULUM ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,MATURATION ,03 medical and health sciences ,Autophagy ,Humans ,Amino Acid Sequence ,Molecular Biology ,030304 developmental biology ,Science & Technology ,COMPLEX ,Infant, Newborn ,rab7 GTP-Binding Proteins ,06 Biological Sciences ,Culture Media ,Enzyme Activation ,chemistry ,MAMMALIAN TARGET ,Guanosine diphosphate ,rab GTP-Binding Proteins ,Lysosomes ,GTPASE - Abstract
Summary Autophagy is an evolutionarily conserved process that enables catabolic and degradative pathways. These pathways commonly depend on vesicular transport controlled by Rabs, small GTPases inactivated by TBC/RabGAPs. The Rac1 effector TBC/RabGAP Armus (TBC1D2A) is known to inhibit Rab7, a key regulator of lysosomal function. However, the precise coordination of signaling and intracellular trafficking that regulates autophagy is poorly understood. We find that overexpression of Armus induces the accumulation of enlarged autophagosomes, while Armus depletion significantly delays autophagic flux. Upon starvation-induced autophagy, Rab7 is transiently activated. This spatiotemporal regulation of Rab7 guanosine triphosphate/guanosine diphosphate cycling occurs by Armus recruitment to autophagosomes via interaction with LC3, a core autophagy regulator. Interestingly, autophagy potently inactivates Rac1. Active Rac1 competes with LC3 for interaction with Armus and thus prevents its appropriate recruitment to autophagosomes. The precise coordination between Rac1 and Rab7 activities during starvation suggests that Armus integrates autophagy with signaling and endocytic trafficking., Graphical Abstract Highlights ► Autophagy strongly inactivates Rac1 GTPase and recruits the TBC/RabGAP Armus ► Appropriate Armus localization at autophagosomes requires binding to LC3 ► Armus inhibition delays autophagic flux and increases levels of Rab7·GTP ► Rac1, Armus, and Rab7 coordinate efficient lysosome fusion with autophagosomes, The Rac1 effector Armus is a GTPase-activating protein that regulates Rab7 function and thus endosomal trafficking to lysosomes. Carroll et al. find that Armus binds directly to the core autophagy factor LC3, facilitates fusion of autophagosomes with lysosomes, and integrates local Rac1 activity into the control of autophagy.
- Published
- 2019
- Full Text
- View/download PDF
27. The mTOR and PP2A Pathways Regulate PHD2 Phosphorylation to Fine-Tune HIF1α Levels and Colorectal Cancer Cell Survival under Hypoxia
- Author
-
Kris Gevaert, Mario Di Matteo, René P. Zahedi, Manuel Ehling, Thomas Kietzmann, Stefan Loroch, Giusy Di Conza, Massimiliano Mazzone, Hans Prenen, Sarah Trusso Cafarello, Fabiola Moretti, Albert Sickmann, Daniela Mennerich, Sofie Deschoemaeker, and Cell Biology and Histology
- Subjects
0301 basic medicine ,PROTEIN ,Medicine and Health Sciences ,Protein Phosphatase 2 ,Phosphorylation ,lcsh:QH301-705.5 ,Tumor ,biology ,Kinase ,TOR Serine-Threonine Kinases ,REDD1 ,Ribosomal Protein S6 Kinases, 70-kDa ,Cell Hypoxia ,3. Good health ,Cell biology ,PP2A ,INDUCIBLE FACTOR ,colon cancer ,AUTOPHAGY ,Hypoxia-Inducible Factor 1 ,Colorectal Neoplasms ,Life Sciences & Biomedicine ,HT29 Cells ,Signal Transduction ,EXPRESSION ,Cell Survival ,HIF1 ,PROLYL HYDROXYLASES ,INHIBITION ,alpha Subunit ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,Hypoxia-Inducible Factor-Proline Dioxygenases ,03 medical and health sciences ,Cell Line, Tumor ,HIF ,Humans ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Science & Technology ,hypoxia ,HEK293 Cells ,Hypoxia-Inducible Factor 1, alpha Subunit ,Ribosomal Protein S6 Kinases ,HEK 293 cells ,Autophagy ,RPTOR ,Biology and Life Sciences ,Protein phosphatase 2 ,Cell Biology ,DEGRADATION ,70-kDa ,030104 developmental biology ,MAMMALIAN TARGET ,lcsh:Biology (General) ,biology.protein ,Human medicine ,metabolism - Abstract
Summary Oxygen-dependent HIF1α hydroxylation and degradation are strictly controlled by PHD2. In hypoxia, HIF1α partly escapes degradation because of low oxygen availability. Here, we show that PHD2 is phosphorylated on serine 125 (S125) by the mechanistic target of rapamycin (mTOR) downstream kinase P70S6K and that this phosphorylation increases its ability to degrade HIF1α. mTOR blockade in hypoxia by REDD1 restrains P70S6K and unleashes PP2A phosphatase activity. Through its regulatory subunit B55α, PP2A directly dephosphorylates PHD2 on S125, resulting in a further reduction of PHD2 activity that ultimately boosts HIF1α accumulation. These events promote autophagy-mediated cell survival in colorectal cancer (CRC) cells. B55α knockdown blocks neoplastic growth of CRC cells in vitro and in vivo in a PHD2-dependent manner. In patients, CRC tissue expresses higher levels of REDD1, B55α, and HIF1α but has lower phospho-S125 PHD2 compared with a healthy colon. Our data disclose a mechanism of PHD2 regulation that involves the mTOR and PP2A pathways and controls tumor growth., Graphical Abstract, Highlights • PHD2 is phosphorylated at Ser125 by P70S6K and dephosphorylated by PP2A/B55α • PHD2 dephosphorylation impairs its function, resulting in increased HIF1α accumulation • HIF1α promotes CRC survival in hypoxia via autophagy in a PHD2/B55α-dependent fashion • B55α silencing blocks CRC tumor growth in vitro and in vivo; this is PHD2 dependent, Di Conza et al. find that PP2A/B55α dephosphorylates and partly inactivates PHD2, leading to augmented HIF1α and CRC cell survival in hypoxia through autophagy. Dephosphorylated PHD2 and B55α accumulate in CRC human specimens versus normal colon, and B55α targeting impairs CRC neoplastic growth in vitro and in mice.
- Published
- 2017
- Full Text
- View/download PDF
28. Systematic drug sensitivity testing reveals synergistic growth inhibition by dasatinib or mTOR inhibitors with paclitaxel in ovarian granulosa cell tumor cells
- Author
-
Bhagwan Yadav, Arto Leminen, Matti Kankainen, Anniina Färkkilä, Noora Andersson, Annika Riska, Jing Tang, Evgeny Kulesskiy, Leila Unkila-Kallio, Ulla-Maija Haltia, Olli Kallioniemi, Ralf Bützow, Mikko Anttonen, Krister Wennerberg, Markku Heikinheimo, Tero Aittokallio, Department of Obstetrics and Gynecology, University of Helsinki, Clinicum, Children's Hospital, Precision Cancer Pathology, Institute for Molecular Medicine Finland, HUSLAB, Department of Pathology, Medicum, Developmental and tumor biology research group, Olli-Pekka Kallioniemi / Principal Investigator, Krister Wennerberg / Principal Investigator, Tero Aittokallio / Principal Investigator, Bioinformatics, Department of Clinical Chemistry and Hematology, HUS Gynecology and Obstetrics, HUS Children and Adolescents, and Precision Systems Medicine
- Subjects
0301 basic medicine ,Dasatinib ,Pharmacology ,chemistry.chemical_compound ,0302 clinical medicine ,3123 Gynaecology and paediatrics ,Antineoplastic Combined Chemotherapy Protocols ,Granulosa Cell Tumor ,Aged, 80 and over ,Ovarian Neoplasms ,TOR Serine-Threonine Kinases ,Obstetrics and Gynecology ,BREAST-CANCER CELLS ,Drug Synergism ,CHEMOTHERAPY ,BLEOMYCIN ,Middle Aged ,SOLID TUMORS ,3. Good health ,Oncology ,Paclitaxel ,030220 oncology & carcinogenesis ,Female ,medicine.drug ,EXPRESSION ,Granulosa cell ,3122 Cancers ,Bleomycin ,03 medical and health sciences ,Ovarian cancer ,Cell Line, Tumor ,medicine ,Humans ,MYELOID-LEUKEMIA ,COMBINATION ,PI3K/AKT/mTOR pathway ,TERM-FOLLOW-UP ,Aged ,Everolimus ,business.industry ,ta3121 ,medicine.disease ,ta3122 ,ta3123 ,030104 developmental biology ,MRNA Sequencing ,chemistry ,MAMMALIAN TARGET ,Kinase inhibitors ,Screening strategies ,Cancer research ,ESTABLISHMENT ,Drug Screening Assays, Antitumor ,business - Abstract
Objective. Resistance to standard chemotherapy poses a major clinical problem in the treatment of ovarian cancer patients. Adult-type granulosa cell tumor (AGCT) is a unique ovarian cancer subtype for which efficient treatment options are lacking in advanced disease. To this end, systematic drug response and transcriptomics profiling were performed to uncover new therapy options for AGCTs. Methods. The responses of three primary and four recurrent AGCTs to 230 anticancer compounds were screened in vitro using a systematic drug sensitivity and resistance testing (DSRT) platform, coupled with mRNA sequencing. The responses of the AGCTs were compared with those of human granulosa luteal cells and bone marrow mononuclear cells. Results. Patient-derived AGCT cells showed selective sensitivity to the Src family tyrosine kinase inhibitor dasatinib. A combination of either dasatinib or an mTOR-inhibitor everolimus with paclitaxel resulted in synergistic inhibition of AGCT cell viability. The key kinase targets of dasatinib and members of the mTOR pathway were constantly expressed at mRNA and protein levels, indicating multikinase signal addictions in the AGCT cells. Transcriptomic characterization of the tumors revealed no known oncogenic mutations, suggesting that the drug sensitivity of AGCTs was rather conveyed by selective target expression. Conclusions. We used a systematic functional approach to reveal novel treatment options for a unique gynecological cancer. The selective synergy found between taxanes and dasatinib or mTOR inhibitors warrants further clinical investigations of these combinations in relapsed or aggressive AGCTs and demonstrate that high throughput drug screening and molecular profiling can provide an effective approach to uncover new therapy options. (C) 2016 Elsevier Inc. All rights reserved.
- Published
- 2017
- Full Text
- View/download PDF
29. CD98hc (SLC3A2) sustains amino acid and nucleotide availability for cell cycle progression
- Author
-
Universitat Rovira i Virgili, Cano-Crespo S, Chillarón J, Junza A, Fernández-Miranda G, García J, Polte C, R de la Ballina L, Ignatova Z, Yanes Ó, Zorzano A, Stephan-Otto Attolini C, Palacín M, Universitat Rovira i Virgili, and Cano-Crespo S, Chillarón J, Junza A, Fernández-Miranda G, García J, Polte C, R de la Ballina L, Ignatova Z, Yanes Ó, Zorzano A, Stephan-Otto Attolini C, Palacín M
- Abstract
CD98 heavy chain (CD98hc) forms heteromeric amino acid (AA) transporters by interacting with different light chains. Cancer cells overexpress CD98hc-transporters in order to meet their increased nutritional and antioxidant demands, since they provide branched-chain AA (BCAA) and aromatic AA (AAA) availability while protecting cells from oxidative stress. Here we show that BCAA and AAA shortage phenocopies the inhibition of mTORC1 signalling, protein synthesis and cell proliferation caused by CD98hc ablation. Furthermore, our data indicate that CD98hc sustains glucose uptake and glycolysis, and, as a consequence, the pentose phosphate pathway (PPP). Thus, loss of CD98hc triggers a dramatic reduction in the nucleotide pool, which leads to replicative stress in these cells, as evidenced by the enhanced DNA Damage Response (DDR), S-phase delay and diminished rate of mitosis, all recovered by nucleoside supplementation. In addition, proper BCAA and AAA availability sustains the expression of the enzyme ribonucleotide reductase. In this regard, BCAA and AAA shortage results in decreased content of deoxynucleotides that triggers replicative stress, also recovered by nucleoside supplementation. On the basis of our findings, we conclude that CD98hc plays a central role in AA and glucose cellular nutrition, redox homeostasis and nucleotide availability, all key for cell proliferation.
- Published
- 2019
30. A systems study reveals concurrent activation of AMPK and mTOR by amino acids
- Subjects
CA2+-SENSING RECEPTOR ,BETA-CELLS ,MAMMALIAN TARGET ,INSULIN-RECEPTOR SUBSTRATE-1 ,IN-VITRO ,PROTEIN-KINASE ,PHOSPHOINOSITIDE 3-KINASE ,PHOSPHORYLATION ,RAPAMYCIN ,LKB1-INDEPENDENT MANNER - Abstract
Amino acids (aa) are not only building blocks for proteins, but also signalling molecules, with the mammalian target of rapamycin complex 1 (mTORC1) acting as a key mediator. However, little is known about whether aa, independently of mTORC1, activate other kinases of the mTOR signalling network. To delineate aa-stimulated mTOR network dynamics, we here combine a computational-experimental approach with text mining-enhanced quantitative proteomics. We report that AMP-activated protein kinase (AMPK), phosphatidylinositide 3-kinase (PI3K) and mTOR complex 2 (mTORC2) are acutely activated by aa-readdition in an mTORC1-independent manner. AMPK activation by aa is mediated by Ca2+/calmodulin-dependent protein kinase kinase beta (CaMKK beta). In response, AMPK impinges on the autophagy regulators Unc-51-like kinase-1 (ULK1) and c-Jun. AMPK is widely recognized as an mTORC1 antagonist that is activated by starvation. We find that aa acutely activate AMPK concurrently with mTOR. We show that AMPK under aa sufficiency acts to sustain autophagy. This may be required to maintain protein homoeostasis and deliver metabolite intermediates for biosynthetic processes.
- Published
- 2016
- Full Text
- View/download PDF
31. A systems study reveals concurrent activation of AMPK and mTOR by amino acids
- Author
-
Pezze, Piero Dalle, Ruf, Stefanie, Sonntag, Annika G, Langelaar-Makkinje, Miriam, Hall, Philip, Heberle, Alexander M, Navas, Patricia Razquin, van Eunen, Karen, Tölle, Regine C, Schwarz, Jennifer J, Wiese, Heike, Warscheid, Bettina, Deitersen, Jana, Stork, Björn, Fäßler, Erik, Schäuble, Sascha, Hahn, Udo, Horvatovich, Peter, Shanley, Daryl P, Thedieck, Kathrin, Analytical Biochemistry, and Medicinal Chemistry and Bioanalysis (MCB)
- Subjects
INSULIN-RECEPTOR SUBSTRATE-1 ,Science ,Calcium-Calmodulin-Dependent Protein Kinase Kinase ,Mechanistic Target of Rapamycin Complex 2 ,PHOSPHOINOSITIDE 3-KINASE ,AMP-Activated Protein Kinases ,Mechanistic Target of Rapamycin Complex 1 ,Models, Biological ,LKB1-INDEPENDENT MANNER ,Article ,Cell Line ,Phosphatidylinositol 3-Kinases ,BETA-CELLS ,Autophagy-Related Protein-1 Homolog ,Humans ,Amino Acids ,PHOSPHORYLATION ,CA2+-SENSING RECEPTOR ,TOR Serine-Threonine Kinases ,Intracellular Signaling Peptides and Proteins ,IN-VITRO ,PROTEIN-KINASE ,MAMMALIAN TARGET ,Gene Expression Regulation ,biological phenomena, cell phenomena, and immunity ,RAPAMYCIN ,Proto-Oncogene Proteins c-akt - Abstract
Amino acids (aa) are not only building blocks for proteins, but also signalling molecules, with the mammalian target of rapamycin complex 1 (mTORC1) acting as a key mediator. However, little is known about whether aa, independently of mTORC1, activate other kinases of the mTOR signalling network. To delineate aa-stimulated mTOR network dynamics, we here combine a computational–experimental approach with text mining-enhanced quantitative proteomics. We report that AMP-activated protein kinase (AMPK), phosphatidylinositide 3-kinase (PI3K) and mTOR complex 2 (mTORC2) are acutely activated by aa-readdition in an mTORC1-independent manner. AMPK activation by aa is mediated by Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ). In response, AMPK impinges on the autophagy regulators Unc-51-like kinase-1 (ULK1) and c-Jun. AMPK is widely recognized as an mTORC1 antagonist that is activated by starvation. We find that aa acutely activate AMPK concurrently with mTOR. We show that AMPK under aa sufficiency acts to sustain autophagy. This may be required to maintain protein homoeostasis and deliver metabolite intermediates for biosynthetic processes., mTORC1 is known to mediate the signalling activity of amino acids. Here, the authors combine modelling with experiments and find that amino acids acutely stimulate mTORC2, IRS/PI3K and AMPK, independently of mTORC1. AMPK activation through CaMKKβ sustains autophagy under non-starvation conditions.
- Published
- 2016
32. mTOR is a fine tuning molecule in CDK inhibitors-induced distinct cell death mechanisms via PI3K/AKT/mTOR signaling axis in prostate cancer cells
- Author
-
Pınar Obakan-Yerlikaya, Özge Berrak, Narcin Palavan-Unsal, Elif Damla Arisan, and Ajda Coker-Gurkan
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Programmed cell death ,Mediated Apoptosis ,Clinical Biochemistry ,Cyclin-Dependent Kinase ,Pharmaceutical Science ,Apoptosis ,urologic and male genital diseases ,03 medical and health sciences ,DU145 ,Cyclin-dependent kinase ,Mammalian Target ,Cell Line, Tumor ,LNCaP ,Autophagy ,Roscovitine ,Humans ,Rapamycin ,Phosphorylation ,Protein Kinase Inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Pharmacology ,biology ,Cell growth ,Chemistry ,TOR Serine-Threonine Kinases ,Cell Cycle ,Biochemistry (medical) ,RPTOR ,Purvalanol ,Induced Apoptosis ,Prostatic Neoplasms ,Cell Biology ,3. Good health ,Cell biology ,030104 developmental biology ,Purines ,Receptors, Androgen ,Androgen Receptor ,mTOR ,biology.protein ,Proto-Oncogene Proteins c-akt ,Transcription Factors ,Pathway ,Signal Transduction - Abstract
Purvalanol and roscovitine are cyclin dependent kinase (CDK) inhibitors that induce cell cycle arrest and apoptosis in various cancer cells. We further hypothesized that co-treatment of CDK inhibitors with rapamycin, an mTOR inhibitor, would be an effective combinatory strategy for the inhibition of prostate cancer regard to androgen receptor (AR) status due to inhibition of proliferative pathway, PI3K/AKT/mTOR, and induction of cell death mechanisms. Androgen responsive (AR+), PTEN-/- LNCaP and androgen independent (AR-), PTEN+/- DU145 prostate cancer cells were exposed to purvalanol (20 A mu M) and roscovitine (30 A mu M) with or without rapamycin for 24 h. Cell viability assay, immunoblotting, flow cytometry and fluorescence microscopy was used to define the effect of CDK inhibitors with or without rapamycin on proliferative pathway and cell death mechanisms in LNCaP and DU145 prostate cancer cells. Co-treatment of rapamycin modulated CDK inhibitors-induced cytotoxicity and apoptosis that CDK inhibitors were more potent to induce cell death in AR (+) LNCaP cells than AR (-) DU145 cells. CDK inhibitors in the presence or absence of rapamycin induced cell death via modulating upstream PI3K/AKT/mTOR signaling pathway in LNCaP cells, exclusively only treatment of purvalanol have strong potential to inhibit both upstream and downstream targets of mTOR in LNCaP and DU145 cells. However, co-treatment of rapamycin with CDK inhibitors protects DU145 cells from apoptosis via induction of autophagy mechanism. We confirmed that purvalanol and roscovitine were strong apoptotic and autophagy inducers that based on regulation of PI3K/AKT/mTOR signaling pathway. Co-treatment of rapamycin with purvalanol and roscovitine exerted different effects on cell survival and death mechanisms in LNCaP and DU145 cell due to their AR receptor status. Our studies show that co-treatment of rapamycin with CDK inhibitors inhibit prostate cancer cell viability more effectively than either agent alone, in part, by targeting the mTOR signaling cascade in AR (+) LNCaP cells. In this point, mTOR is a fine-tuning player in purvalanol and roscovitine-induced apoptosis and autophagy via regulation of PI3K/AKT and the downstream targets, which related with cell proliferation.
- Published
- 2016
- Full Text
- View/download PDF
33. Hypoxia-inducible factor 1 alpha is required for the tumourigenic and aggressive phenotype associated with Rab25 expression in ovarian cancer
- Subjects
CARCINOMA ,INCREASES ,HIF-1 alpha ,TUMOR-SUPPRESSOR PROTEIN ,FACTOR 1-ALPHA ,tumourigenic ,intraperitoneal ,UP-REGULATION ,HYPOXIA-INDUCIBLE-FACTOR-1-ALPHA ,Rab25 ,ovarian cancer ,HUMAN-BREAST-CANCER ,MAMMALIAN TARGET ,ENDOTHELIAL GROWTH-FACTOR ,STEM-CELLS - Abstract
The small GTPase Rab25 has been functionally linked to tumour progression and aggressiveness in ovarian cancer and promotes invasion in three-dimensional environments. This type of migration has been shown to require the expression of the hypoxia-inducible factor 1 alpha (HIF-1a). In this report we demonstrate that Rab25 regulates HIF-1a protein expression in an oxygen independent manner in a panel of cancer cell lines. Regulation of HIF-1a protein expression by Rab25 did not require transcriptional upregulation, but was dependent on de novo protein synthesis through the Erbb2/ERK1/2 and p70S6K/mTOR pathways. Rab25 expression induced HIF-1 transcriptional activity, increased cisplatin resistance, and conferred intraperitoneal growth to the A2780 cell line in immunocompromised mice. Targeting HIF1 activity by silencing HIF-1 beta re-sensitised cells to cisplatin in vitro and reduced tumour formation of A2780-Rab25 expressing cells in vivo in a mouse ovarian peritoneal carcinomatosis model. Similar effects on cisplatin resistance in vitro and intraperitoneal tumourigenesis in vivo were obtained after HIF1b knockdown in the ovarian cancer cell line SKOV3, which expresses endogenous Rab25 and HIF-1a at atmospheric oxygen concentrations. Our results suggest that Rab25 tumourigenic potential and chemoresistance relies on HIF1 activity in aggressive and metastatic ovarian cancer. Targeting HIF-1 activity may potentially be effective either alone or in combination with standard chemotherapy for aggressive metastatic ovarian cancer.
- Published
- 2016
34. Everolimus Exposure and Early Metabolic Response as Predictors of Treatment Outcomes in Breast Cancer Patients Treated with Everolimus and Exemestane
- Author
-
Marianne A. Jonker, Maaike de Boer, Nielka P. van Erp, Winald R. Gerritsen, Erik H.J.G. Aarntzen, Willem Grootjans, Yvonne Kamm, Sasja F. Mulder, Paul C. de Jong, Annelieke E.C.A.B. Willemsen, Johannes W. B. de Groot, Lioe-Fee de Geus-Oei, Jolien Tol, A. Vos, Carla M.L. van Herpen, Biomedical Photonic Imaging, Interne Geneeskunde, MUMC+: MA Medische Oncologie (9), and RS: GROW - R3 - Innovative Cancer Diagnostics & Therapy
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Breast Neoplasms ,ENDOCRINE MONOTHERAPY ,PLUS EXEMESTANE ,Standardized uptake value ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,All institutes and research themes of the Radboud University Medical Center ,Pharmacokinetics ,Exemestane ,Internal medicine ,medicine ,Humans ,Pharmacology (medical) ,Everolimus ,Aged ,Chemotherapy ,business.industry ,Middle Aged ,CHEMOTHERAPY ,EFFICACY ,medicine.disease ,SOLID TUMORS ,MTOR INHIBITOR EVEROLIMUS ,Women's cancers Radboud Institute for Health Sciences [Radboudumc 17] ,Androstadienes ,PHASE-I ,PET ,030104 developmental biology ,MAMMALIAN TARGET ,chemistry ,SAFETY ,030220 oncology & carcinogenesis ,Urological cancers Radboud Institute for Health Sciences [Radboudumc 15] ,Toxicity ,Female ,business ,Progressive disease ,medicine.drug ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] - Abstract
Background: Treating breast cancer patients with everolimus and exemestane can be challenging due to toxicity and suboptimal treatment responses. Objective: We investigated whether everolimus exposure and early metabolic response are predictors for toxicity and effectiveness in these patients. Patients and Methods: We performed pharmacokinetic assessments 14 and 35 days after starting treatment. [18F]fluorodeoxyglucose-positron emission tomography (18F-FDG-PET) was performed at baseline, and 14 and 35 days after the start of the therapy. We recorded toxicity, defined as dose interventions within 3 months, and progression-free survival (PFS). Results: Among 44 evaluable patients, the geometric mean (GM) Ctrough was higher in patients with toxicity compared to patients without (17.4 versus 12.3 μg/L (p = 0.02)). The optimal cut-off value to predict toxicity was Ctrough > 19.2 μg/L. GM Ctrough of patients with and without progressive disease (PD) within 3 months was not significantly different (12.0 versus 15.2 μg/L (p = 0.118)). In 28 evaluable patients, PD within 3 months could best be predicted using the percentage decrease in peak standardized uptake value normalized by lean body mass of the lesion with highest FDG uptake (SULpeak high) at day 14. Patients with 11% decrease in SULpeak high at day 14 had a median PFS of 90 days versus 411 days, respectively (p = 0.0013) and more frequently had PD within 3 months: 70 vs 11%, respectively. Conclusions: Our results show that everolimus toxicity is related to everolimus Ctrough. No relation was observed between everolimus exposure and treatment effectiveness. An early FDG-PET can identify patients at high risk of nonresponse. These results warrant further validation. Clinicaltrials.gov identifier: NCT01948960.
- Published
- 2018
35. Weighing In on mTOR Complex 2 Signaling: The Expanding Role in Cell Metabolism
- Author
-
Guannan Li, Yongting Luo, Wenyi Xu, and Wei Cui
- Subjects
0301 basic medicine ,Aging ,HYDROPHOBIC MOTIF PHOSPHORYLATION ,mTORC1 ,GLUTAMINE-METABOLISM ,Mechanistic Target of Rapamycin Complex 2 ,Review Article ,Biochemistry ,mTORC2 ,03 medical and health sciences ,Metabolic Diseases ,CRYO-EM STRUCTURE ,Animals ,Homeostasis ,Humans ,PROTEIN-KINASE-C ,lcsh:QH573-671 ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,DE-NOVO LIPOGENESIS ,POSITIVE FEEDBACK LOOP ,LIFE-SPAN ,INSULIN-RESISTANCE ,Science & Technology ,biology ,Cell growth ,lcsh:Cytology ,General Medicine ,Cell Biology ,Cell biology ,030104 developmental biology ,Cell metabolism ,ADIPOSE-TISSUE ,Structural biology ,MAMMALIAN TARGET ,biology.protein ,Signal transduction ,Life Sciences & Biomedicine ,Signal Transduction - Abstract
In all eukaryotes, the mechanistic target of rapamycin (mTOR) signaling emerges as a master regulator of homeostasis, which integrates environmental inputs, including nutrients, energy, and growth factors, to regulate many fundamental cellular processes such as cell growth and metabolism. mTOR signaling functions through two structurally and functionally distinct complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), which correspond to two major branches of signal output. While mTORC1 is well characterized for its structure, regulation, and function in the last decade, information of mTORC2 signaling is only rapidly expanding in recent years, from structural biology, signaling network, to functional impact. Here we review the recent advances in many aspects of the mTORC2 signaling, with particular focus on its involvement in the control of cell metabolism and its physiological implications in metabolic diseases and aging.
- Published
- 2018
36. GCN2 contributes to mTORC1 inhibition by leucine deprivation through an ATF4 independent mechanism
- Author
-
Florent Mesclon, Christopher G. Proud, Céline Jousse, Laurent Parry, Anne Catherine Maurin, Valérie Carraro, Julien Averous, Alain Bruhat, Pierre Fafournoux, Sarah Lambert-Langlais, Philippe Pierre, Unité de Nutrition Humaine - Clermont Auvergne (UNH), Institut National de la Recherche Agronomique (INRA)-Université Clermont Auvergne (UCA), Centre d'Immunologie de Marseille - Luminy (CIML), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU), Aix Marseille Université (AMU), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Institute for Research in Biomedicine, University of Aveiro, South Australian Health and Medical Research Institute [ Adelaide] (SAHMRI), University of Adelaide, Unité de Nutrition Humaine (UNH), Université d'Auvergne - Clermont-Ferrand I (UdA)-Clermont Université-Institut National de la Recherche Agronomique (INRA), Institute for Research in Biomedicine ( iBiMED), Universidade de Aveiro, School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia, Institut National de la Recherche Agronomique (INRA) Fondation ARC pour la Recherche sur le CancerIlidio Pinho foundation, Institut National de la Recherche Agronomique (INRA)-Université d'Auvergne - Clermont-Ferrand I (UdA)-Clermont Université, Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), University of South Australia [Adelaide], Institut National de la Recherche Agronomique (INRA), Fondation ARC pour la Recherche sur le Cancer, Ilidio Pinho foundation, Institut National de la Recherche Agronomique (INRA)-Université Clermont Auvergne [2017-2020] (UCA [2017-2020]), ProdInra, Archive Ouverte, Unité de Nutrition Humaine - Clermont Auvergne ( UNH ), Université Clermont Auvergne ( UCA ) -Institut national de la recherche agronomique [Auvergne/Rhône-Alpes] ( INRA Auvergne/Rhône-Alpes ), U1104, UMR 7280, Centre d'Immunologie de Marseille-Luminy ( CIML ), Aix Marseille Université ( AMU ), Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique ( CNRS ), South Australian Health and Medical Research Institute ( SAHMRI ), and VAFFIDES, Chantal
- Subjects
0301 basic medicine ,Arginine ,[ SDV.AEN ] Life Sciences [q-bio]/Food and Nutrition ,Eukaryotic Initiation Factor-2 ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Mechanistic Target of Rapamycin Complex 1 ,Protein Serine-Threonine Kinases ,Biology ,Activating Transcription Factor 4 ,Article ,Mice ,03 medical and health sciences ,Leucine ,preinitiation complex ,translational control ,pathway ,mammalian target ,[SDV.BBM] Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Animals ,Food and Nutrition ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Phosphorylation ,chemistry.chemical_classification ,ATF3 ,Multidisciplinary ,synthèse de protéine ,ATF4 ,Translation (biology) ,muscle squelettique ,Fibroblasts ,Embryo, Mammalian ,Amino acid ,[SDV.AEN] Life Sciences [q-bio]/Food and Nutrition ,acide aminé ,030104 developmental biology ,voluntary muscle ,Biochemistry ,chemistry ,Alimentation et Nutrition ,biological phenomena, cell phenomena, and immunity ,[SDV.AEN]Life Sciences [q-bio]/Food and Nutrition ,amino acid ,Signal Transduction ,expression des gènes - Abstract
It is well known that the GCN2 and mTORC1 signaling pathways are regulated by amino acids and share common functions, in particular the control of translation. The regulation of GCN2 activity by amino acid availability relies on the capacity of GCN2 to sense the increased levels of uncharged tRNAs upon amino acid scarcity. In contrast, despite recent progress in the understanding of the regulation of mTORC1 by amino acids, key aspects of this process remain unsolved. In particular, while leucine is well known to be a potent regulator of mTORC1, the mechanisms by which this amino acid is sensed and control mTORC1 activity are not well defined. Our data establish that GCN2 is involved in the inhibition of mTORC1 upon leucine or arginine deprivation. However, the activation of GCN2 alone is not sufficient to inhibit mTORC1 activity, indicating that leucine and arginine exert regulation via additional mechanisms. While the mechanism by which GCN2 contributes to the initial step of mTORC1 inhibition involves the phosphorylation of eIF2α, we show that it is independent of the downstream transcription factor ATF4. These data point to a novel role for GCN2 and phosphorylation of eIF2α in the control of mTORC1 by certain amino acids.
- Published
- 2018
- Full Text
- View/download PDF
37. 90th Anniversary Commentary: The mTORC1 Complex-A Central Player in the Control and Regulation of Amino Acid Sufficiency
- Author
-
Daniel Tomé, Physiologie de la Nutrition et du Comportement Alimentaire (PNCA), and Institut National de la Recherche Agronomique (INRA)-AgroParisTech
- Subjects
0301 basic medicine ,postabsorptive rats ,Literature, Modern ,[SDV]Life Sciences [q-bio] ,Medicine (miscellaneous) ,Nutritional Status ,Computational biology ,mTORC1 ,transporters ,Mechanistic Target of Rapamycin Complex 1 ,translation initiation ,History, 21st Century ,03 medical and health sciences ,inhibits translation ,Animals ,Humans ,skeletal-muscle ,Amino Acids ,Control (linguistics) ,chemistry.chemical_classification ,Nutrition and Dietetics ,pathway ,TOR Serine-Threonine Kinases ,Nutritional Requirements ,Nutritional status ,protein-synthesis ,mammalian target ,Amino acid ,030104 developmental biology ,chemistry ,Multiprotein Complexes ,leucine ,metabolism ,Signal Transduction - Abstract
International audience; The ability of cells to respond to nutrient availability and to changes in environmental conditions by appropriate adaptation of anabolic and catabolic processes, cell growth, and proliferation is of fundamental importance for survival. Numerous studies have been conducted during the last few decades to identify signals and cellular mechanisms involved in these means of regulation. The pioneering paper by Anthony et al. (1) published in The Journal of Nutrition in 2000 showed that the BCAA leucine can stimulate protein synthesis in skeletal muscle in vivo in food-deprived rats and that the leucine-dependent signaling pathways involved in this stimulation include the protein kinase mechanistic target of rapamycin (mTOR) pathway.
- Published
- 2018
- Full Text
- View/download PDF
38. Unmasking the impact of Rictor in cancer: novel insights of mTORC2 complex
- Author
-
Giampaolo Tortora, Michele Milella, A. Gkountakos, Caterina Vicentini, Michele Simbolo, Vincenzo Corbo, Emilio Bria, Sara Pilotto, Aldo Scarpa, and Andrea Mafficini
- Subjects
0301 basic medicine ,Cancer Research ,Carcinogenesis ,AKT PHOSPHORYLATION ,mTORC1 ,medicine.disease_cause ,mTORC2 ,COLORECTAL-CANCER ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,HEPATOCELLULAR-CARCINOMA ,Neoplasms ,Phosphorylation ,CELL LUNG CANCERS ,Regulation of gene expression ,Clinical Trials as Topic ,biology ,TOR Serine-Threonine Kinases ,digestive, oral, and skin physiology ,General Medicine ,PROSTATE-CANCER ,Gene Expression Regulation, Neoplastic ,Treatment Outcome ,030220 oncology & carcinogenesis ,PIVOTAL ROLE ,COMPONENT RICTOR ,MAMMALIAN TARGET ,3T3-L1 ADIPOCYTES ,PTEN LOSS ,Signal Transduction ,DNA Copy Number Variations ,Antineoplastic Agents ,Mechanistic Target of Rapamycin Complex 2 ,Mechanistic Target of Rapamycin Complex 1 ,03 medical and health sciences ,microRNA ,medicine ,Animals ,Humans ,Epigenetics ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Gene Amplification ,030104 developmental biology ,Rapamycin-Insensitive Companion of mTOR Protein ,Cancer research ,biology.protein ,Proto-Oncogene Proteins c-akt - Abstract
Genomic alterations affecting components of the mechanistic target of rapamycin (mTOR) pathway are found rather frequently in cancers, suggesting that aberrant pathway activity is implicated in oncogenesis of different tumor types. mTOR functions as the core catalytic kinase of two distinct complexes, mTOR complex 1 (mTORC1) and 2 (mTORC2), which control numerous vital cellular processes. There is growing evidence indicating that Rictor, an essential subunit of the mTORC2 complex, is inappropriately overexpressed across numerous cancer types and this is associated with poor survival. To date, the candidate mechanisms responsible for aberrant Rictor expression described in cancer are two: (i) gene amplification and (ii) epigenetic regulation, mainly by microRNAs. Moreover, different mTOR-independent Rictor-containing complexes with oncogenic role have been documented, revealing alternative routes of Rictor-driven tumorigenesis, but simultaneously, paving the way for identifying novel biomarkers and therapeutic targets. Here, we review the main preclinical and clinical data regarding the role of Rictor in carcinogenesis and metastatic behavior as well as the potentiality of its alteration as a target.
- Published
- 2018
39. TSC1 Activates TGF-β-Smad2/3 Signaling in Growth Arrest and Epithelial-to-Mesenchymal Transition
- Author
-
Sushma Nagaraja Grellscheid, Ralf Baumeister, Michael Reth, Stefanie Ruf, Ineke Kuper, Mirja Tamara Prentzell, Gerd Walz, Kathrin Thedieck, Christopher Boehlke, Birgit Holzwarth, Roland Nitschke, Kathrin Kläsener, Elke Neumann-Haefelin, Lars Maerz, Antje Thien, Annika G Sonntag, and Center for Liver, Digestive and Metabolic Diseases (CLDM)
- Subjects
Receptor complex ,Fluorescent Antibody Technique ,Apoptosis ,Smad2 Protein ,SMAD ,mTORC1 ,Tuberous Sclerosis Complex 1 Protein ,Immunoenzyme Techniques ,TGF-BETA RECEPTOR ,CELL-CYCLE ARREST ,0302 clinical medicine ,Transforming Growth Factor beta ,TUBEROUS SCLEROSIS COMPLEX ,Phosphorylation ,MEDIATED PHOSPHORYLATION ,Cells, Cultured ,0303 health sciences ,TOR Serine-Threonine Kinases ,Flow Cytometry ,3. Good health ,Cell biology ,GENE-PRODUCTS ,Signal transduction ,Signal Transduction ,congenital, hereditary, and neonatal diseases and abnormalities ,Epithelial-Mesenchymal Transition ,Blotting, Western ,DISTINCT ROLES ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mothers against decapentaplegic homolog 3 ,Tuberous Sclerosis Complex 2 Protein ,Humans ,Immunoprecipitation ,Smad3 Protein ,FYVE DOMAIN PROTEIN ,CANCER-CELLS ,Molecular Biology ,Protein kinase B ,030304 developmental biology ,Cell Proliferation ,SMAD PROTEINS ,Tumor Suppressor Proteins ,Cell Biology ,MAMMALIAN TARGET ,Multiprotein Complexes ,Proto-Oncogene Proteins c-akt ,Receptors, Transforming Growth Factor beta ,030217 neurology & neurosurgery ,Transforming growth factor ,Developmental Biology - Abstract
The tuberous sclerosis proteins TSC1 and TSC2 are key integrators of growth factor signaling. They suppress cell growth and proliferation by acting in a heteromeric complex to inhibit the mammalian target of rapamycin complex 1 (mTORC1). In this study, we identify TSC1 as a component of the transforming growth factor beta (TGF-beta)-Smad2/3 pathway. Here, TSC1 functions independently of TSC2. TSC1 interacts with the TGF-beta receptor complex and Smad2/3 and is required for their association with one another. TSC1 regulates TGF-beta-induced Smad2/3 phosphorylation and target gene expression and controls TGF-beta- induced growth arrest and epithelial-to-mesenchymal transition (EMT). Hyperactive Akt specifically activates TSC1-dependent cytostatic Smad signaling to induce growth arrest. Thus, TSC1 couples Akt activity to TGF-beta-Smad2/3 signaling. This has implications for cancer treatments targeting phosphoinositide 3-kinases and Akt because they may impair tumor-suppressive cytostatic TGF-beta signaling by inhibiting Akt-and TSC1-dependent Smad activation.
- Published
- 2015
- Full Text
- View/download PDF
40. Glucocorticoid Receptor Signaling Impairs Protein Turnover Regulation in Hypoxia-Induced Muscle Atrophy in Male Mice
- Author
-
Theije, Chiel C., Theije, Chiel C., Schols, Annemie M. W. J., Lamers, Wouter H., Ceelen, Judith J. M., van Gorp, Rick H., Hermans, J. J. Rob, Kohler, S. Elonore, Langen, Ramon C. J., Theije, Chiel C., Theije, Chiel C., Schols, Annemie M. W. J., Lamers, Wouter H., Ceelen, Judith J. M., van Gorp, Rick H., Hermans, J. J. Rob, Kohler, S. Elonore, and Langen, Ramon C. J.
- Abstract
Hypoxemia may contribute to muscle wasting in conditions such as chronic obstructive pulmonary disease. Muscle wasting develops when muscle proteolysis exceeds protein synthesis. Hypoxia induces skeletal muscle atrophy in mice, which can in part be attributed to reduced food intake. We hypothesized that hypoxia elevates circulating corticosterone concentrations by reduced food intake and enhances glucocorticoid receptor (GR) signaling in muscle, which causes elevated protein degradation signaling and dysregulates protein synthesis signaling during hypoxia-induced muscle atrophy. Muscle-specific GR knockout and control mice were subjected to normoxia, normobaric hypoxia (8% oxygen), or pair-feeding to the hypoxia group for 4 days. Plasma corticosterone and muscle GR signaling increased after hypoxia and pair-feeding. GR deficiency prevented muscle atrophy by pair-feeding but not by hypoxia. GR deficiency differentially affected activation of ubiquitin 26S-proteasome and autophagy proteolytic systems by pair-feeding and hypoxia. Reduced food intake suppressed mammalian target of rapamycin complex 1 (mTORC1) activity under normoxic but not hypoxic conditions, and this retained mTORC1 activity was mediated by GR. We conclude that GR signaling is required for muscle atrophy and increased expression of proteolysis-associated genes induced by decreased food intake under normoxic conditions. Under hypoxic conditions, muscle atrophy and elevated gene expression of the ubiquitin proteasomal system-associated E3 ligases Murf1 and Atrogin-1 are mostly independent of GR signaling. Furthermore, impaired inhibition of mTORC1 activity is GR-dependent in hypoxia-induced muscle atrophy.
- Published
- 2018
41. Glucocorticoid Receptor Signaling Impairs Protein Turnover Regulation in Hypoxia-Induced Muscle Atrophy in Male Mice
- Author
-
Wouter H. Lamers, Rick H. van Gorp, Chiel C. de Theije, J. J. Rob Hermans, Judith J.M. Ceelen, Ramon C. J. Langen, Annemie M. W. J. Schols, S Elonore Köhler, Biobank, RS: NUTRIM - R3 - Respiratory & Age-related Health, Ondersteunend personeel NTM, Pulmonologie, Anatomie & Embryologie, Promovendi NTM, Biochemie, Promovendi CD, RS: CARIM - R1.02 - Vascular aspects thrombosis and haemostasis, Pharmacology and Personalised Medicine, RS: SHE - R1 - Research (OvO), and RS: NUTRIM - R2 - Liver and digestive health
- Subjects
Male ,0301 basic medicine ,mTORC1 ,UP-REGULATION ,PATHWAY ,Random Allocation ,Endocrinology ,Glucocorticoid receptor ,Hypoxia ,GENE-EXPRESSION ,Mice, Knockout ,Chemistry ,MTOR ,EPITHELIAL-CELLS ,Muscle atrophy ,Muscular Atrophy ,medicine.anatomical_structure ,Muscle Fibers, Fast-Twitch ,SKELETAL-MUSCLE ,medicine.symptom ,Signal Transduction ,Proteasome Endopeptidase Complex ,medicine.medical_specialty ,WEIGHT-LOSS ,Mice, Transgenic ,Protein degradation ,OBSTRUCTIVE PULMONARY-DISEASE ,Gene Expression Regulation, Enzymologic ,03 medical and health sciences ,Receptors, Glucocorticoid ,Atrophy ,GLUTAMINE-SYNTHETASE ,Internal medicine ,Autophagy ,medicine ,Animals ,Muscle, Skeletal ,Glucocorticoids ,Crosses, Genetic ,Cell Size ,Protein turnover ,Skeletal muscle ,Hypoxia (medical) ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,MAMMALIAN TARGET ,Proteolysis ,Corticosterone - Abstract
Hypoxemia may contribute to muscle wasting in conditions such as chronic obstructive pulmonary disease. Muscle wasting develops when muscle proteolysis exceeds protein synthesis. Hypoxia induces skeletal muscle atrophy in mice, which can in part be attributed to reduced food intake. We hypothesized that hypoxia elevates circulating corticosterone concentrations by reduced food intake and enhances glucocorticoid receptor (GR) signaling in muscle, which causes elevated protein degradation signaling and dysregulates protein synthesis signaling during hypoxia-induced muscle atrophy. Muscle-specific GR knockout and control mice were subjected to normoxia, normobaric hypoxia (8% oxygen), or pair-feeding to the hypoxia group for 4 days. Plasma corticosterone and muscle GR signaling increased after hypoxia and pair-feeding. GR deficiency prevented muscle atrophy by pair-feeding but not by hypoxia. GR deficiency differentially affected activation of ubiquitin 26S-proteasome and autophagy proteolytic systems by pair-feeding and hypoxia. Reduced food intake suppressed mammalian target of rapamycin complex 1 (mTORC1) activity under normoxic but not hypoxic conditions, and this retained mTORC1 activity was mediated by GR. We conclude that GR signaling is required for muscle atrophy and increased expression of proteolysis-associated genes induced by decreased food intake under normoxic conditions. Under hypoxic conditions, muscle atrophy and elevated gene expression of the ubiquitin proteasomal system–associated E3 ligases Murf1 and Atrogin-1 are mostly independent of GR signaling. Furthermore, impaired inhibition of mTORC1 activity is GR-dependent in hypoxia-induced muscle atrophy.
- Published
- 2018
42. Guideline-based and bioinformatic reassessment of lesion-associated gene and variant pathogenicity in focal human epilepsies
- Author
-
Dennis Lal, Michael Nothnagel, Peter Nürnberg, Mark J. Daly, Patrick May, Ingmar Blümcke, Aarno Palotie, Lisa-Marie Niestroj, Juanjiangmeng Du, Centre of Excellence in Complex Disease Genetics, Aarno Palotie / Principal Investigator, Institute for Molecular Medicine Finland, University of Helsinki, Genomics of Neurological and Neuropsychiatric Disorders, and Luxembourg Centre for Systems Biomedicine (LCSB): Bioinformatics Core (R. Schneider Group) [research center]
- Subjects
0301 basic medicine ,Male ,Neurology [D14] [Human health sciences] ,Disease ,3124 Neurology and psychiatry ,Germline ,0302 clinical medicine ,DIAGNOSTIC METHODS ,Databases, Genetic ,Missense mutation ,Genetics ,gene pathogenicity ,low-grade epilepsy associated tumors ,3. Good health ,CORTICAL DYSPLASIA ,Neurology ,Medical genetics ,Female ,Genetics & genetic processes [F10] [Life sciences] ,Génétique & processus génétiques [F10] [Sciences du vivant] ,focal cortical dysplasia ,PATHWAY MUTATIONS CAUSE ,STANDARDS ,medicine.medical_specialty ,In silico ,Genomics ,focal epilepsies ,Biology ,low-grade epilepsy-associated tumors ,03 medical and health sciences ,variant pathogenicity ,medicine ,Humans ,MALFORMATIONS ,Genetic Predisposition to Disease ,Genetic Testing ,Gene ,SPECTRUM ,Neurologie [D14] [Sciences de la santé humaine] ,Epilepsy ,3112 Neurosciences ,Computational Biology ,Genetic Variation ,SOMATIC MUTATIONS ,Cortical dysplasia ,INTERNATIONAL CONSENSUS CLASSIFICATION ,medicine.disease ,030104 developmental biology ,MAMMALIAN TARGET ,Neurology (clinical) ,030217 neurology & neurosurgery - Abstract
Objective: Increasing availability of surgically resected brain tissue from patients with focal epilepsy and Focal Cortical Dysplasia (FCD) or low-grade glio-neuronal tumors has fostered large-scale genetic examination. However, assessment of pathogenicity of germline and somatic variants remains difficult. Here, we present a state of the art evaluation of reported genes and variants associated with epileptic brain lesions. Methods: We critically re-evaluated the pathogenicity for all neuropathology-associated variants reported to date in PubMed and ClinVar databases including 101 neuropathology-associated missense variants encompassing 11 disease-related genes. We assessed gene variant tolerance and classified all identified missense variants according to guidelines from the American College of Medical Genetics and Genomics (ACMG). We further extended the bioinformatic variant prediction by introducing a novel gene-specific deleteriousness ranking for prediction scores. Results: Application of ACMG guidelines and in silico gene variant tolerance analysis classified only seven out of 11 genes to be likely disease-associated according to the reported a disease mechanism, while 61 (60.4%) of 101 variants of those genes were classified as of uncertain significance (VUS), 37 (36.6%) as being likely pathogenic (LP) and 3 (3%) as being pathogenic (P). Significance: We concluded that the majority of neuropathology-associated variants reported to date do not have enough evidence to be classified as pathogenic. Interpretation of lesion-associated variants remains challenging and application of current ACMG guidelines is recommended for interpretation and prediction.
- Published
- 2017
43. A Drosophila genetic screen for suppressors of S6kinase-dependent growth identifies the F-box subunit Archipelago/FBXW7
- Author
-
Muhammad-Kashif Zahoor, Mickael Poidevin, Jacques Montagne, Damien Garrido, Caroline Lecerf, Institut de Biologie Intégrative de la Cellule (I2BC), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), Département Biologie Cellulaire (BioCell), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), Croissance et métabolisme chez la Drosophile (METABO), and Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie Intégrative de la Cellule (I2BC)
- Subjects
0106 biological sciences ,0301 basic medicine ,F-Box-WD Repeat-Containing Protein 7 ,[SDV]Life Sciences [q-bio] ,mTORC1 ,fbxw7 ,Genetic screen ,01 natural sciences ,Animals, Genetically Modified ,ubiquitin ligases ,RNA interference ,mtor ,Drosophila Proteins ,Phosphorylation ,S2 cells ,Ago/FBXW7 ,Gene knockdown ,Schneider 2 cells ,Effector ,binding pocket ,Gene Expression Regulation, Developmental ,General Medicine ,cellular growth ,tumor-suppressor ,mammalian target ,Cell biology ,Drosophila melanogaster ,Larva ,RNA Interference ,Signal Transduction ,P70-S6 Kinase 1 ,Biology ,Protein degradation ,Mechanistic Target of Rapamycin Complex 1 ,Cell Line ,03 medical and health sciences ,Genetics ,Animals ,Molecular Biology ,p70 s6 kinase ,Ago ,F-Box Proteins ,Ribosomal Protein S6 Kinases ,Signaling ,030104 developmental biology ,activation ,protein ,010606 plant biology & botany - Abstract
International audience; This study was designed to identify novel negative regulators of the Drosophila S6kinase (dS6K). S6K is a downstream effector of the growth-regulatory complex mTORC1 (mechanistic-Target-of-Rapamycin complex 1). Nutrients activate mTORC1, which in turn induces the phosphorylation of S6K to promote cell growth, whereas fasting represses mTORC1 activity. Here, we screened 11,000 RNA-interfering (RNAi) lines and retained those that enhanced a dS6K-dependent growth phenotype. Since RNAi induces gene knockdown, enhanced tissue growth supports the idea that the targeted gene acts as a growth suppressor. To validate the resulting candidate genes, we monitored dS6K phosphorylation and protein levels in double-stranded RNAi-treated S2 cells. We identified novel dS6K negative regulators, including gene products implicated in basal cellular functions, suggesting that feedback inputs modulate mTORC1/dS6K signaling. We also identified Archipelago (Ago), the Drosophila homologue of FBXW7, which is an E3-ubiquitin-ligase subunit that loads ubiquitin units onto target substrates for proteasome-mediated degradation. Despite a previous report showing an interaction between Ago/FBXW7 and dS6K in a yeast two-hybrid assay and the presence of an Ago/FBXW7-consensus motif in the dS6K polypeptide, we could not see a direct interaction in immunoprecipitation assay. Nevertheless, we observed that loss-of-ago/fbxw7 in larvae resulted in an increase in dS6K protein levels, but no change in the levels of phosphorylated dS6K or dS6K transcripts, suggesting that Ago/FBXW7 indirectly controls dS6K translation or stability. Through the identification of novel negative regulators of the downstream target, dS6K, our study may help deciphering the underlying mechanisms driving deregulations of mTORC1, which underlies several human diseases.
- Published
- 2017
- Full Text
- View/download PDF
44. Variation in a range of mTOR-related genes associates with intracranial volume and intellectual disability
- Author
-
M. van Gastel, Tjitske Kleefstra, Eric Smeets, Alexander P.A. Stegmann, R. Pfundt, Nicholas Katsanis, Christian Gilissen, Ype Elgersma, Servi J. C. Stevens, M. Proietti-Onori, G. M. S. Mancini, Barbara Franke, G. M. van Woerden, Janita Bralten, Margot R.F. Reijnders, Stefan H. Lelieveld, Maria Kousi, T. van Essen, Perciliz L. Tan, Han G. Brunner, Marieke Klein, MUMC+: DA KG Lab Centraal Lab (9), Klinische Genetica, RS: GROW - R4 - Reproductive and Perinatal Medicine, MUMC+: DA Klinische Genetica (5), Neurosciences, and Clinical Genetics
- Subjects
0301 basic medicine ,General Physics and Astronomy ,SET ANALYSIS ,0302 clinical medicine ,MIGRATION DEFICITS ,Cell Movement ,Megalencephaly ,TUBEROUS SCLEROSIS COMPLEX ,INTRACTABLE EPILEPSY ,lcsh:Science ,Cells, Cultured ,Zebrafish ,IN-VIVO ,Genetics ,Neurons ,education.field_of_study ,Multidisciplinary ,biology ,TOR Serine-Threonine Kinases ,Brain ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Human brain ,Organ Size ,medicine.anatomical_structure ,MESSENGER-RNA TRANSLATION ,Brain size ,RHEB ,Signal Transduction ,Science ,Population ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Seizures ,Intellectual Disability ,CEREBRAL-CORTEX ,medicine ,Animals ,Humans ,Allele ,education ,Gene ,PI3K/AKT/mTOR pathway ,Cell Size ,Sirolimus ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,CORTICAL DEVELOPMENT ,General Chemistry ,SOMATIC MUTATIONS ,medicine.disease ,030104 developmental biology ,MAMMALIAN TARGET ,Mutation ,biology.protein ,lcsh:Q ,Ras Homolog Enriched in Brain Protein ,030217 neurology & neurosurgery - Abstract
De novo mutations in specific mTOR pathway genes cause brain overgrowth in the context of intellectual disability (ID). By analyzing 101 mMTOR-related genes in a large ID patient cohort and two independent population cohorts, we show that these genes modulate brain growth in health and disease. We report the mTOR activator gene RHEB as an ID gene that is associated with megalencephaly when mutated. Functional testing of mutant RHEB in vertebrate animal models indicates pathway hyperactivation with a concomitant increase in cell and head size, aberrant neuronal migration, and induction of seizures, concordant with the human phenotype. This study reveals that tight control of brain volume is exerted through a large community of mTOR-related genes. Human brain volume can be altered, by either rare disruptive events causing hyperactivation of the pathway, or through the collective effects of common alleles., The mTOR pathway is a key regulator of normal brain development. Here, the authors identify de novo mutations in RHEB, an mTOR activator protein, in patients with intellectual disability associated with megalencephaly and find a role for RHEB in regulating neuronal soma size and migration in vitro and in vivo.
- Published
- 2017
- Full Text
- View/download PDF
45. Interactions of (2S,6S;2R,6R)-Hydroxynorketamine, a Secondary Metabolite of (R,S)-Ketamine, with Morphine
- Author
-
Tuomas Lilius, Eija Kalso, Mikko Niemi, Viljami Jokinen, Pekka Rauhala, Hanna Viisanen, Medicum, Department of Pharmacology, Department of Clinical Pharmacology, Clinicum, Mikko Olavi Niemi / Principal Investigator, Eija Kalso / Principal Investigator, Anestesiologian yksikkö, Department of Diagnostics and Therapeutics, Pekka Rauhala / Principal Investigator, and HUS Perioperative, Intensive Care and Pain Medicine
- Subjects
0301 basic medicine ,Male ,Nociception ,LIVER MICROSOMAL PREPARATIONS ,Hydroxynorketamine ,Pharmacology ,Toxicology ,Nociceptive Pain ,Rats, Sprague-Dawley ,0302 clinical medicine ,Drug tolerance ,Opioid receptor ,Drug Interactions ,Receptor ,Behavior, Animal ,Morphine ,Brain ,General Medicine ,Drug Tolerance ,3. Good health ,Analgesics, Opioid ,317 Pharmacy ,NMDA receptor ,Ketamine ,SPINAL-CORD ,medicine.drug ,Pain Threshold ,medicine.drug_class ,Motor Activity ,NMDA RECEPTOR ANTAGONIST ,OPIOID RECEPTOR ,03 medical and health sciences ,ARRIVE GUIDELINES ,medicine ,Animals ,UDP-GLUCURONOSYLTRANSFERASES ,SPRAGUE-DAWLEY RATS ,Anesthetics, Dissociative ,business.industry ,IN-VITRO ,CANCER PAIN ,Disease Models, Animal ,030104 developmental biology ,MAMMALIAN TARGET ,3111 Biomedicine ,business ,030217 neurology & neurosurgery - Abstract
Ketamine and its primary metabolite norketamine attenuate morphine tolerance by antagonising N-methyl-d-aspartate (NMDA) receptors. Ketamine is extensively metabolized to several other metabolites. The major secondary metabolite (2S,6S;2R,6R)-hydroxynorketamine (6-hydroxynorketamine) is not an NMDA antagonist. However, it may modulate nociception through negative allosteric modulation of 7 nicotinic acetylcholine receptors. We studied whether 6-hydroxynorketamine could affect nociception or the effects of morphine in acute or chronic administration settings. Male Sprague Dawley rats received subcutaneous 6-hydroxynorketamine or ketamine alone or in combination with morphine, as a cotreatment during induction of morphine tolerance, and after the development of tolerance induced by subcutaneous minipumps administering 9.6 mg morphine daily. Tail flick, hot plate, paw pressure and rotarod tests were used. Brain and serum drug concentrations were quantified with high-performance liquid chromatography-tandem mass spectrometry. Ketamine (10 mg/kg), but not 6-hydroxynorketamine (10 and 30 mg/kg), enhanced antinociception and decreased rotarod performance following acute administration either alone or combined with morphine. Ketamine efficiently attenuated morphine tolerance. Acutely administered 6-hydroxynorketamine increased the brain concentration of morphine (by 60%), and brain and serum concentrations of 6-hydroxynorketamine were doubled by morphine pre-treatment. This pharmacokinetic interaction did not, however, lead to altered morphine tolerance. Co-administration of 6-hydroxynorketamine 20 mg/kg twice daily did not influence development of morphine tolerance. Even though morphine and 6-hydroxynorketamine brain concentrations were increased after co-administration, the pharmacokinetic interaction had no effect on acute morphine nociception or tolerance. These results indicate that 6-hydroxynorketamine does not have antinociceptive properties or attenuate opioid tolerance in a similar way as ketamine.
- Published
- 2017
46. Potential Targets' Analysis Reveals Dual PI3K/mTOR Pathway Inhibition as a Promising Therapeutic Strategy for Uterine Leiomyosarcomas-an ENITEC Group Initiative
- Author
-
Mariël Brinkhuis, Miriam Mints, Tjalling Bosse, Koen Van de Vijver, Debby Thomas, R.P.F.M. Kruitwagen, Ronald P. Zweemer, Helga B. Salvesen, Michal Zikan, Tine Cuppens, Carole Mestdagh, Jone Trovik, Ellen Gomme, Pavel Dundr, Philippe Moerman, Godelieve Verbist, Jutta Huvila, Michael R. Mallmann, Maciej Stukan, Natalja T. ter Haar, Farid Moinfar, Els Hermans, Daniela Annibali, Johannes Haybaeck, Angel Garcia-Jimenez, Frédéric Amant, Olli Carpén, An Coosemans, Eva Wardelmann, Eva Colas, Leonardus F. Massuger, Amsterdam Reproduction & Development (AR&D), Other departments, Medicum, Department of Pathology, Precision Cancer Pathology, MUMC+: MA Arts Assistenten Obstetrie Gynaecologie (9), MUMC+: MA Medische Staf Obstetrie Gynaecologie (9), RS: GROW - R2 - Basic and Translational Cancer Biology, and Obstetrie & Gynaecologie
- Subjects
0301 basic medicine ,Leiomyosarcoma ,Cancer Research ,Pathology ,AKT-MTOR PATHWAY ,ENDOMETRIAL STROMAL SARCOMA ,Uterine sarcoma ,Mice ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Molecular Targeted Therapy ,Phosphorylation ,CYCLIN D1 ,Phosphoinositide-3 Kinase Inhibitors ,Ribosomal Protein S6 ,Women's cancers Radboud Institute for Molecular Life Sciences [Radboudumc 17] ,biology ,Soft tissue sarcoma ,TOR Serine-Threonine Kinases ,Prognosis ,Immunohistochemistry ,TUMORS ,Gene Expression Regulation, Neoplastic ,PI3K/mTOR pathway ,SOFT-TISSUE SARCOMA ,Oncology ,030220 oncology & carcinogenesis ,Uterine Neoplasms ,TRIAL ,Female ,Signal Transduction ,EXPRESSION ,medicine.medical_specialty ,Stromal cell ,3122 Cancers ,Disease-Free Survival ,03 medical and health sciences ,medicine ,Biomarkers, Tumor ,PTEN ,BREAST-CANCER ,Animals ,Humans ,PI3K/AKT/mTOR pathway ,Endometrial stromal sarcoma ,business.industry ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,MAMMALIAN TARGET ,Patient-derived xenograft models ,Cancer research ,biology.protein ,business ,GROWTH-FACTOR RECEPTOR - Abstract
Purpose: Uterine sarcomas are rare and heterogeneous tumors characterized by an aggressive clinical behavior. Their high rates of recurrence and mortality point to the urgent need for novel targeted therapies and alternative treatment strategies. However, no molecular prognostic or predictive biomarkers are available so far to guide choice and modality of treatment. Experimental Design: We investigated the expression of several druggable targets (phospho-S6S240 ribosomal protein, PTEN, PDGFR-α, ERBB2, and EGFR) in a large cohort of human uterine sarcoma samples (288), including leiomyosarcomas, low-grade and high-grade endometrial stromal sarcomas, undifferentiated uterine sarcomas, and adenosarcomas, together with 15 smooth muscle tumors of uncertain malignant potential (STUMP), 52 benign uterine stromal tumors, and 41 normal uterine tissues. The potential therapeutic value of the most promising target, p-S6S240, was tested in patient-derived xenograft (PDX) leiomyosarcoma models. Results: In uterine sarcomas and STUMPs, S6S240 phosphorylation (reflecting mTOR pathway activation) was associated with higher grade (P = 0.001) and recurrence (P = 0.019), as shown by logistic regression. In addition, p-S6S240 correlated with shorter progression-free survival (P = 0.034). Treatment with a dual PI3K/mTOR inhibitor significantly reduced tumor growth in 4 of 5 leiomyosarcoma PDX models (with tumor shrinkage in 2 models). Remarkably, the 4 responding models showed basal p-S6S240 expression, whereas the nonresponding model was scored as negative, suggesting a role for p-S6S240 in response prediction to PI3K/mTOR inhibition. Conclusions: Dual PI3K/mTOR inhibition represents an effective therapeutic strategy in uterine leiomyosarcoma, and p-S6S240 expression is a potential predictive biomarker for response to treatment. Clin Cancer Res; 23(5); 1274–85. ©2017 AACR.
- Published
- 2017
47. High dietary protein intake, reducing or eliciting insulin resistance?
- Author
-
Marco Mensink, Jessica Schwarz, Daniel Tomé, Annemarie Rietman, Frans J. Kok, Division of Human Nutrition, Wageningen University and Research Centre [Wageningen] (WUR), Physiologie de la Nutrition et du Comportement Alimentaire (PNCA), AgroParisTech-Institut National de la Recherche Agronomique (INRA), Wageningen University and Research [Wageningen] (WUR), and Institut National de la Recherche Agronomique (INRA)-AgroParisTech
- Subjects
Blood Glucose ,030309 nutrition & dietetics ,[SDV]Life Sciences [q-bio] ,medicine.medical_treatment ,Medicine (miscellaneous) ,metabolic risk-factors ,Type 2 diabetes ,Voeding, Metabolisme en Genomica ,body-composition ,0302 clinical medicine ,Insulin ,2. Zero hunger ,chemistry.chemical_classification ,0303 health sciences ,Nutrition and Dietetics ,TOR Serine-Threonine Kinases ,low-fat diets ,Metabolism and Genomics ,mammalian target ,Amino acid ,weight-loss diet ,Metabolisme en Genomica ,Nutrition, Metabolism and Genomics ,Dietary Proteins ,medicine.medical_specialty ,Diabetes risk ,low-carbohydrate-diet ,030209 endocrinology & metabolism ,Biology ,03 medical and health sciences ,Insulin resistance ,Voeding ,Internal medicine ,Diabetes mellitus ,medicine ,Animals ,Humans ,skeletal-muscle ,chain amino-acids ,Nutrition ,VLAG ,Global Nutrition ,Wereldvoeding ,blood-glucose control ,Insulin sensitivity ,medicine.disease ,Diet ,Endocrinology ,Diabetes Mellitus, Type 2 ,chemistry ,glycemic control ,Insulin Resistance ,Amino Acids, Branched-Chain ,Dietary protein intake - Abstract
Dietary proteins have an insulinotropic effect and thus promote insulin secretion, which indeed leads to enhanced glucose clearance from the blood. In the long term, however, a high dietary protein intake is associated with an increased risk of type 2 diabetes. Moreover, branched-chain amino acids (BCAA), a prominent group of amino acids, were recently identified to be associated with diabetes. Observational data and intervention studies do not point in the same direction regarding the effect of protein intake on insulin sensitivity and diabetes risk. Therefore, the first aim of this review will be to discuss human studies addressing high dietary protein intake and insulin action, with special attention for BCAA. In the second part, we will highlight the (patho) physiological consequences of high-protein diets regarding insulin action, in particular the role of the mechanistic target of the rapamycin pathway.
- Published
- 2014
- Full Text
- View/download PDF
48. PI3K-p110-alpha-subtype signalling mediates survival, proliferation and neurogenesis of cortical progenitor cells via activation of mTORC2
- Author
-
Clemens Kreutz, Stefan Christopher Weise, Tanja Vogel, Shalaka Wahane, Jens Timmer, Nicole Hellbach, Kathrin Thedieck, Riccardo Vezzali, Kerstin Krieglstein, Mirja Tamara Prentzell, and Center for Liver, Digestive and Metabolic Diseases (CLDM)
- Subjects
medicine.medical_treatment ,mTORC1 ,AKT PHOSPHORYLATION ,Biochemistry ,PI3K-subunit ,Receptor, IGF Type 1 ,Mice ,Phosphatidylinositol 3-Kinases ,Neural Stem Cells ,Pregnancy ,Transforming Growth Factor beta ,Neurotrophic factors ,PI3K P110-ALPHA ,Insulin-Like Growth Factor I ,INDUCED APOPTOSIS ,biology ,TOR Serine-Threonine Kinases ,Neurogenesis ,Immunohistochemistry ,Cell biology ,embryonic structures ,cerebral cortex ,PHOSPHATIDYLINOSITOL 3-KINASE ,Female ,Signal Transduction ,insulin ,Cell Survival ,Class I Phosphatidylinositol 3-Kinases ,brain ,Blotting, Western ,Primary Cell Culture ,CROSS-TALK ,Mechanistic Target of Rapamycin Complex 2 ,Cellular and Molecular Neuroscience ,GROWTH-FACTOR-I ,medicine ,Animals ,Progenitor cell ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,MESENCHYMAL TRANSITION ,HIPPOCAMPAL-NEURONS ,Growth factor ,Transforming growth factor beta ,Microarray Analysis ,neuron ,PI3K-isoform ,MAMMALIAN TARGET ,Multiprotein Complexes ,biology.protein ,Proto-Oncogene Proteins c-akt ,NEUROTROPHIC FACTOR - Abstract
Development of the cerebral cortex is controlled by growth factors among which transforming growth factor beta (TGFβ) and insulin-like growth factor 1 (IGF1) have a central role. The TGFβ- and IGF1-pathways cross-talk and share signalling molecules, but in the central nervous system putative points of intersection remain unknown. We studied the biological effects and down-stream molecules of TGFβ and IGF1 in cells derived from the mouse cerebral cortex at two developmental time points, E13.5 and E16.5. IGF1 induces PI3K, AKT and the mammalian target of rapamycin complexes (mTORC1/mTORC2) primarily in E13.5-derived cells, resulting in proliferation, survival and neuronal differentiation, but has small impact on E16.5-derived cells. TGFβ has little effect at E13.5. It does not activate the PI3K- and mTOR-signalling network directly, but requires its activity to mediate neuronal differentiation specifically at E16.5. Our data indicate a central role of mTORC2 in survival, proliferation as well as neuronal differentiation of E16.5-derived cortical cells. mTORC2 promotes these cellular processes and is under control of PI3K-p110-alpha signalling. PI3K-p110-beta signalling activates mTORC2 in E16.5-derived cells but it does not influence cell survival, proliferation and differentiation. This finding indicates that different mTORC2 subtypes may be implicated in cortical development and that these subtypes are under control of different PI3K isoforms. Within developing cortical cells TGFβ- and IGF-signalling activities are timely separated. TGFβ dominates in E16.5-derived cells and drives neuronal differentiation. IGF influences survival, proliferation and neuronal differentiation in E13.5-derived cells. mTORC2-signalling in E16.5-derived cells influences survival, proliferation and differentiation, activated through PI3K-p110-alpha. PI3K-p110-beta-signalling activates a different mTORC2. Both PI3K/mTORC2-signalling pathways are required but not directly activated in TGFβ-mediated neuronal differentiation.
- Published
- 2014
- Full Text
- View/download PDF
49. T cell receptor-mediated activation is a potent inducer of macroautophagy in human CD8+CD28+ T cells but not in CD8+CD28− T cells
- Author
-
Stefan Brunner, Theresa Pritz, Beatrix Grubeck-Loebenstein, Carina Knabb, Willi Salvenmoser, Kathrin Thedieck, Birgit Holzwarth, Christoph R. Arnold, and Center for Liver, Digestive and Metabolic Diseases (CLDM)
- Subjects
Male ,Aging ,ENERGY-METABOLISM ,mTORC1 ,CD8-Positive T-Lymphocytes ,Biochemistry ,GRAVES-DISEASE ,Interleukin 21 ,0302 clinical medicine ,Endocrinology ,Cytotoxic T cell ,IL-2 receptor ,Cells, Cultured ,0303 health sciences ,TOR Serine-Threonine Kinases ,ZAP70 ,CD28 ,hemic and immune systems ,Flow Cytometry ,Natural killer T cell ,Cell biology ,mTOR ,Female ,Immunosuppressive Agents ,Human ,Receptors, Antigen, T-Cell ,T cells ,chemical and pharmacologic phenomena ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,SIGNALING PATHWAYS ,03 medical and health sciences ,CD28 Antigens ,Autophagy ,Genetics ,Humans ,Molecular Biology ,Aged ,030304 developmental biology ,Sirolimus ,JNK1-MEDIATED PHOSPHORYLATION ,IMMUNE RISK PHENOTYPE ,PROTEIN-KINASE COMPLEX ,Cell Biology ,EARLY ATHEROSCLEROTIC DAMAGE ,RHEUMATOID-ARTHRITIS ,MAMMALIAN TARGET ,Multiprotein Complexes ,Leukocytes, Mononuclear ,Cancer research ,HUMAN BONE-MARROW ,CD8 ,030215 immunology - Abstract
A key feature of the aged human immune system is the accumulation of highly differentiated CD8(+)CD28(-) T cells, a phenomenon that negatively influences immune function in the elderly. However, the mechanisms that regulate survival or death of CD8(+)CD28(-) T cells remain incompletely understood. Macroautophagy has been shown to protect cells from unfavorable environmental conditions and extend lifespan of various cells and organisms. In this study, we investigated autophagy in CD8(+)CD28(+) and CD8(+)CD28(-) T cells following T cell receptor (TCR) engagement. We demonstrate that TCR-mediated activation led to a potent induction of autophagy in CD8(+)CD28(+) T cells which was accompanied by an increased activity of the mammalian target of rapamycin complex 1 (mTORC1). This was surprising, as mTORC1 is generally perceived as an inhibitor of autophagy. Inhibition of mTORC1 by rapamycin could still enhance activation-induced autophagy. In contrast, CD8(+)CD28(-) T cells induced autophagy to a significantly lower extent in response to TCR engagement compared to CD8(+)CD28(+) T cells and failed to increase autophagy upon mTORC1 inhibition. In conclusion, we describe for the first time the induction of autophagy in human CD8(+) T cells following TCR engagement and the decreased ability of CD8(+)CD28(-) T cells to induce autophagy, suggesting that they cannot meet the metabolic needs of antigen receptor-mediated activation and are therefore unlikely to survive when confronted by their specific antigens.
- Published
- 2014
- Full Text
- View/download PDF
50. Increased protein aggregation in Zucker Diabetic Fatty rat brain
- Author
-
Robert H. Henning, Veroniek M. van Praag, Sjoerd W. Landheer, Fatemeh Talaei, Mahdi Hamidi Shishavan, Hendrik Buikema, Groningen Kidney Center (GKC), Vascular Ageing Programme (VAP), and Groningen Institute for Organ Transplantation (GIOT)
- Subjects
Male ,Protein aggregates ,COGNITIVE DYSFUNCTION ,Glycosylation ,HIGH GLUCOSE ,endocrine system diseases ,Protein aggregation ,medicine.disease_cause ,NaHS ,ZDF rats ,Gene expression ,Hydrogen Sulfide ,Phosphorylation ,OXIDATIVE STRESS ,Cystathionine beta synthase ,biology ,TOR Serine-Threonine Kinases ,Neurodegeneration ,Brain ,MOUSE MODEL ,AMYLOID-BETA ,Cell biology ,ALZHEIMERS-DISEASE ,mTOR ,Microtubule-Associated Proteins ,Research Article ,Carboxymethyllysine ,medicine.medical_specialty ,Amyloid beta ,tau Proteins ,In Vitro Techniques ,Diabetes Mellitus, Experimental ,Internal medicine ,medicine ,Autophagy ,Animals ,Fibronectin ,Ubiquitination ,nutritional and metabolic diseases ,Cell Biology ,medicine.disease ,Fibronectins ,Rats ,Rats, Zucker ,Disease Models, Animal ,Endocrinology ,Proteostasis ,MAMMALIAN TARGET ,CELL-DEATH ,biology.protein ,TAU ,Reactive oxygen species ,Oxidative stress - Abstract
Background: Diabetes and particularly high blood glucose levels are implicated in neurodegeneration. One of the hallmarks of neurodegeneration is protein aggregation. We investigated the presence of protein aggregation in the frontal brain of Zucker diabetic fatty (ZDF) rats, an animal model for diabetes. Further, the effect of NaHS in suppressing protein aggregation in cultured brain slices from ZDF was assessed.Results: The levels of protein synthesis, protein/gene expression, autophagy and anti-oxidant defense were evaluated in ZDF and control (Lean) brains.Compared to Lean, ZDF brains displayed a significant increase in protein aggregates, p-tau, fibronectin expression and protein glycosylation. Increased phosphorylation of mTOR and S6 ribosomal protein in ZDF indicated higher protein synthesis, while the increase in ubiquitinated proteins and LC3-I in ZDF brains accompanied by lower LC3-II expression and LC3-II/LC3-I levels indicated the blockage of proteolytic pathways. CBS (cystathionine beta synthase) protein and mRNA expression and thiol group levels in ZDF brains were lower compared to Lean. ZDF brains show a higher level of reactive oxygen species. In vitro NaHS treatment normalized proteostasis while counteracting oxidative stress.Conclusion: Our data demonstrate increased protein synthesis and aggregation in the diabetic ZDF rat brain, which was reversible by NaHS treatment. This is the first report on the potential use of NaHS as a novel strategy against protein aggregation in diabetic brain.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.