1
|
Bankolé A, Srivastava A, Shihavuddin A, Tighanimine K, Faucourt M, Koka V, Weill S, Nemazanyy I, Nelson AJ, Stokes MP, Delgehyr N, Genovesio A, Meunier A, Fumagalli S, Pende M, Spassky N. mTOR controls ependymal cell differentiation by targeting the alternative cell cycle and centrosomal proteins. EMBO Rep 2025:10.1038/s44319-025-00460-2. [PMID: 40307619 DOI: 10.1038/s44319-025-00460-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Ependymal cells are multiciliated glial cells lining the ventricles of the mammalian brain. Their differentiation from progenitor cells involves cell enlargement and progresses through centriole amplification phases and ciliogenesis. These phases are accompanied by the sharp up-regulation of mTOR Complex 1 activity (mTORC1), a master regulator of macromolecule biosynthesis and cell growth, whose function in ependymal cell differentiation is unknown. We demonstrate that mTORC1 inhibition by rapamycin preserves the progenitor pool by reinforcing quiescence and preventing alternative cell cycle progression for centriole amplification. Overexpressing E2F4 and MCIDAS circumvents mTORC1-regulated processes, enabling centriole amplification despite rapamycin, and enhancing mTORC1 activity through positive feedback. Acute rapamycin treatment in multicentriolar cells during the late phases of differentiation causes centriole regrouping, indicating a direct role of mTORC1 in centriole dynamics. By phosphoproteomic and phosphomutant analysis, we reveal that the mTORC1-mediated phosphorylation of GAS2L1, a centrosomal protein that links actin and microtubule cytoskeletons, participates in centriole disengagement. This multilayered and sequential control of ependymal development by mTORC1, from the progenitor pool to centriolar function, has implications for pathophysiological conditions like aging and hydrocephalus-prone genetic diseases.
Collapse
Affiliation(s)
- Alexia Bankolé
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Ayush Srivastava
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Asm Shihavuddin
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Computational bioimaging and bioinformatics, 75005, Paris, France
- Department of EEE, Presidency University, Dhaka, Bangladesh
| | - Khaled Tighanimine
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Marion Faucourt
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Vonda Koka
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Solene Weill
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Alissa J Nelson
- Cell Signaling Technology INC, 3 Trask Lane, Danvers, MA, 01923, USA
| | - Matthew P Stokes
- Cell Signaling Technology INC, 3 Trask Lane, Danvers, MA, 01923, USA
| | - Nathalie Delgehyr
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Auguste Genovesio
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Computational bioimaging and bioinformatics, 75005, Paris, France
| | - Alice Meunier
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Stefano Fumagalli
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Mario Pende
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France.
| | - Nathalie Spassky
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France.
| |
Collapse
|
2
|
Xiang Y, Liu L, Hou Y, Du S, Xu S, Zhou H, Shao L, Li G, Yu T, Liu Q, Xue M, Yang J, Peng J, Hou M, Shi Y. The mTORC1 pathway participate in hyper-function of B cells in immune thrombocytopenia. Ann Hematol 2023; 102:2317-2327. [PMID: 37421506 DOI: 10.1007/s00277-023-05348-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/29/2023] [Indexed: 07/10/2023]
Abstract
B cell hyper-function plays an important role in the pathogenesis of immune thrombocytopenia (ITP), but the molecular mechanisms underlying such changes remain unclear. We sought to identify regulators of B cell dysfunction in ITP patients through transcriptome sequencing and the use of inhibitors. B cells were isolated from PBMC of 25 ITP patients for B cell function test and transcriptome sequencing. For the potential regulatory factors identified by transcriptome sequencing, the corresponding protein inhibitors were used to explore the regulatory effect of the regulatory factors on B cell dysfunction in vitro. In this study, increased antibody production, enhanced terminal differentiation and highly expressed costimulatory molecules CD80 and CD86 were found in B cells of patients with ITP. In addition, RNA sequencing revealed highly activated mTOR pathway in these pathogenic B cells, indicating that the mTOR pathway may be involved in B cell hyper-function. Furthermore, mTOR inhibitors rapamycin or Torin1 effectively blocked the activation of mTORC1 in B cells, resulting in reduce antibody secretion, impaired differentiation of B cells into plasmablasts and downregulation of costimulatory molecules. Interestingly, as an unspecific inhibitor of mTORC2 besides mTORC1, Torin1 did not show a stronger capacity to modulate B cell function than rapamycin, suggesting that the regulation of B cells by Torin1 may depend on blockade of mTORC1 rather than mTORC2 pathway. These results indicated that the activation of mTORC1 pathway is involved in B cell dysfunction in patients with ITP, and inhibition of mTORC1 pathway might be a potential therapeutic approach for ITP.
Collapse
Affiliation(s)
- Yujiao Xiang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Lu Liu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
- Department of Hematology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Yu Hou
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Jinan, China
- Department of Hematology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Shenghong Du
- Department of Hematology, Taian Central Hospital, Taian, China
| | - Shuqian Xu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Hai Zhou
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Linlin Shao
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Guosheng Li
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Tianshu Yu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Qiang Liu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Meijuan Xue
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Junhui Yang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Jinan, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center in Hematological Diseases, Jinan, China
- Leading Research Group of Scientific Innovation, Department of Science and Technology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Yan Shi
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
3
|
Wang T, Woodman P, Humphrey SJ, Petersen J. Environmental control of Pub1 (NEDD4 family E3 ligase) in Schizosaccharomyces pombe is regulated by TORC2 and Gsk3. Life Sci Alliance 2022; 5:5/5/e202101082. [PMID: 35121625 PMCID: PMC8817228 DOI: 10.26508/lsa.202101082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 11/24/2022] Open
Abstract
The NEDD4 family E3 ligase Pub1 is regulated by the nutrient environment, TORC2, and Gsk3 signalling pathway to control the level of amino acid transporters on the plasma membrane and thus nutrient uptake. Cells respond to changing nutrient environments by adjusting the abundance of surface nutrient transporters and receptors. This can be achieved by modulating ubiquitin-dependent endocytosis, which in part is regulated by the NEDD4 family of E3 ligases. Here we report novel regulation of Pub1, a fission yeast Schizosaccharomyces pombe member of the NEDD4-family of E3 ligases. We show that nitrogen stress inhibits Pub1 function, thereby increasing the abundance of the amino acid transporter Aat1 at the plasma membrane and enhancing sensitivity to the toxic arginine analogue canavanine. We show that TOR complex 2 (TORC2) signalling negatively regulates Pub1, thus TORC2 mutants under nutrient stress have decreased Aat1 at the plasma membrane and are resistant to canavanine. Inhibition of TORC2 signalling increases Pub1 phosphorylation, and this is dependent on Gsk3 activity. Addition of the Tor inhibitor Torin1 increases phosphorylation of Pub1 at serine 199 (S199) by 2.5-fold, and Pub1 protein levels in S199A phospho-ablated mutants are reduced. S199 is conserved in NEDD4 and is located immediately upstream of a WW domain required for protein interaction. Together, we describe how the major TORC2 nutrient-sensing signalling network regulates environmental control of Pub1 to modulate the abundance of nutrient transporters.
Collapse
Affiliation(s)
- Tingting Wang
- Flinders Health and Medical Research Institute, Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, Australia
| | - Philip Woodman
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Camperdown, Australia
| | - Janni Petersen
- Flinders Health and Medical Research Institute, Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, Australia
| |
Collapse
|
4
|
Segreto R, Bazafkan H, Millinger J, Schenk M, Atanasova L, Doppler M, Büschl C, Boeckstaens M, Soto Diaz S, Schreiner U, Sillo F, Balestrini R, Schuhmacher R, Zeilinger S. The TOR kinase pathway is relevant for nitrogen signaling and antagonism of the mycoparasite Trichoderma atroviride. PLoS One 2022; 16:e0262180. [PMID: 34972198 PMCID: PMC8719763 DOI: 10.1371/journal.pone.0262180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/18/2021] [Indexed: 01/14/2023] Open
Abstract
Trichoderma atroviride (Ascomycota, Sordariomycetes) is a well-known mycoparasite applied for protecting plants against fungal pathogens. Its mycoparasitic activity involves processes shared with plant and human pathogenic fungi such as the production of cell wall degrading enzymes and secondary metabolites and is tightly regulated by environmental cues. In eukaryotes, the conserved Target of Rapamycin (TOR) kinase serves as a central regulator of cellular growth in response to nutrient availability. Here we describe how alteration of the activity of TOR1, the single and essential TOR kinase of T. atroviride, by treatment with chemical TOR inhibitors or by genetic manipulation of selected TOR pathway components affected various cellular functions. Loss of TSC1 and TSC2, that are negative regulators of TOR complex 1 (TORC1) in mammalian cells, resulted in altered nitrogen source-dependent growth of T. atroviride, reduced mycoparasitic overgrowth and, in the case of Δtsc1, a diminished production of numerous secondary metabolites. Deletion of the gene encoding the GTPase RHE2, whose mammalian orthologue activates mTORC1, led to rapamycin hypersensitivity and altered secondary metabolism, but had an only minor effect on vegetative growth and mycoparasitic overgrowth. The latter also applied to mutants missing the npr1-1 gene that encodes a fungus-specific kinase known as TOR target in yeast. Genome-wide transcriptome analysis confirmed TOR1 as a regulatory hub that governs T. atroviride metabolism and processes associated to ribosome biogenesis, gene expression and translation. In addition, mycoparasitism-relevant genes encoding terpenoid and polyketide synthases, peptidases, glycoside hydrolases, small secreted cysteine-rich proteins, and G protein coupled receptors emerged as TOR1 targets. Our results provide the first in-depth insights into TOR signaling in a fungal mycoparasite and emphasize its importance in the regulation of processes that critically contribute to the antagonistic activity of T. atroviride.
Collapse
Affiliation(s)
- Rossana Segreto
- Department of Microbiology, University of Innsbruck, Innsbruck, Austria
| | - Hoda Bazafkan
- Department of Microbiology, University of Innsbruck, Innsbruck, Austria
| | - Julia Millinger
- Department of Microbiology, University of Innsbruck, Innsbruck, Austria
| | - Martina Schenk
- Department of Microbiology, University of Innsbruck, Innsbruck, Austria
| | - Lea Atanasova
- Institute of Food Technology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Maria Doppler
- Department of Agrobiotechnology IFA-Tulln, Center for Analytical Chemistry, University of Natural, Resources and Life Sciences, Vienna (BOKU), Tulln, Austria
| | - Christoph Büschl
- Department of Agrobiotechnology IFA-Tulln, Center for Analytical Chemistry, University of Natural, Resources and Life Sciences, Vienna (BOKU), Tulln, Austria
| | - Mélanie Boeckstaens
- Département de Biologie Moléculaire, Laboratory of Biology of Membrane Transport, Université Libre de Bruxelles, Gosselies, Belgium
| | - Silvia Soto Diaz
- Département de Biologie Moléculaire, Laboratory of Biology of Membrane Transport, Université Libre de Bruxelles, Gosselies, Belgium
| | - Ulrike Schreiner
- Department of Microbiology, University of Innsbruck, Innsbruck, Austria
| | | | | | - Rainer Schuhmacher
- Department of Agrobiotechnology IFA-Tulln, Center for Analytical Chemistry, University of Natural, Resources and Life Sciences, Vienna (BOKU), Tulln, Austria
| | - Susanne Zeilinger
- Department of Microbiology, University of Innsbruck, Innsbruck, Austria
- * E-mail:
| |
Collapse
|
5
|
Li L, Zhu T, Song Y, Luo X, Datla R, Ren M. Target of rapamycin controls hyphal growth and pathogenicity through FoTIP4 in Fusarium oxysporum. MOLECULAR PLANT PATHOLOGY 2021; 22:1239-1255. [PMID: 34288333 PMCID: PMC8435236 DOI: 10.1111/mpp.13108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 05/07/2023]
Abstract
Fusarium oxysporum is the causal agent of the devastating Fusarium wilt by invading and colonizing the vascular system in various plants, resulting in substantial economic losses worldwide. Target of rapamycin (TOR) is a central regulator that controls intracellular metabolism, cell growth, and stress responses in eukaryotes, but little is known about TOR signalling in F. oxysporum. In this study, we identified conserved FoTOR signalling pathway components including FoTORC1 and FoTORC2. Pharmacological assays showed that F. oxysporum is hypersensitive to rapamycin in the presence of FoFKBP12 while the deletion mutant strain ΔFofkbp12 is insensitive to rapamycin. Transcriptomic data indicated that FoTOR signalling controls multiple metabolic processes including ribosome biogenesis and cell wall-degrading enzymes (CWDEs). Genetic analysis revealed that FoTOR1 interacting protein 4 (FoTIP4) acts as a new component of FoTOR signalling to regulate hyphal growth and pathogenicity of F. oxysporum. Importantly, transcript levels of genes associated with ribosome biogenesis and CWDEs were dramatically downregulated in the ΔFotip4 mutant strain. Electrophoretic mobility shift assays showed that FoTIP4 can bind to the promoters of ribosome biogenesis- and CWDE-related genes to positively regulate the expression of these genes. These results suggest that FoTOR signalling plays central roles in regulating hyphal growth and pathogenicity of F. oxysporum and provide new insights into FoTOR1 as a target for controlling and preventing Fusarium wilt in plants.
Collapse
Affiliation(s)
- Linxuan Li
- Institute of Urban AgricultureChinese Academy of Agricultural SciencesChengdu National Agricultural Science and Technology CenterChengduChina
| | - Tingting Zhu
- Institute of Urban AgricultureChinese Academy of Agricultural SciencesChengdu National Agricultural Science and Technology CenterChengduChina
| | - Yun Song
- Zhengzhou Research BaseState Key Laboratory of Cotton BiologyZhengzhou UniversityZhengzhouChina
- School of Life SciencesLiaocheng UniversityLiaochengChina
| | - Xiumei Luo
- Institute of Urban AgricultureChinese Academy of Agricultural SciencesChengdu National Agricultural Science and Technology CenterChengduChina
| | - Raju Datla
- Global Institute for Food Security in SaskatoonUniversity of SaskatchewanSaskatoonCanada
| | - Maozhi Ren
- Institute of Urban AgricultureChinese Academy of Agricultural SciencesChengdu National Agricultural Science and Technology CenterChengduChina
- Zhengzhou Research BaseState Key Laboratory of Cotton BiologyZhengzhou UniversityZhengzhouChina
| |
Collapse
|
6
|
Yang G, Francis D, Krycer JR, Larance M, Zhang Z, Novotny CJ, Diaz-Vegas A, Shokat KM, James DE. Dissecting the biology of mTORC1 beyond rapamycin. Sci Signal 2021; 14:eabe0161. [PMID: 34546793 DOI: 10.1126/scisignal.abe0161] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Guang Yang
- University of Sydney, School of life and Environmental Sciences, Charles Perkins Centre, Sydney, New South Wales 2006, Australia
| | - Deanne Francis
- University of Sydney, School of life and Environmental Sciences, Charles Perkins Centre, Sydney, New South Wales 2006, Australia
| | - James R Krycer
- University of Sydney, School of life and Environmental Sciences, Charles Perkins Centre, Sydney, New South Wales 2006, Australia
| | - Mark Larance
- University of Sydney, School of life and Environmental Sciences, Charles Perkins Centre, Sydney, New South Wales 2006, Australia
| | - Ziyang Zhang
- Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, 600 16th Street, San Francisco, CA 94143, USA
| | - Chris J Novotny
- Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, 600 16th Street, San Francisco, CA 94143, USA
| | - Alexis Diaz-Vegas
- University of Sydney, School of life and Environmental Sciences, Charles Perkins Centre, Sydney, New South Wales 2006, Australia
| | - Kevan M Shokat
- Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, 600 16th Street, San Francisco, CA 94143, USA
| | - David E James
- University of Sydney, School of life and Environmental Sciences, Charles Perkins Centre, Sydney, New South Wales 2006, Australia.,University of Sydney, Sydney Medical School, Sydney, New South Wales 2006, Australia
| |
Collapse
|
7
|
Mak T, Jones AW, Nurse P. The TOR-dependent phosphoproteome and regulation of cellular protein synthesis. EMBO J 2021; 40:e107911. [PMID: 34296454 PMCID: PMC8365262 DOI: 10.15252/embj.2021107911] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/28/2021] [Accepted: 06/09/2021] [Indexed: 01/21/2023] Open
Abstract
Cell growth is orchestrated by a number of interlinking cellular processes. Components of the TOR pathway have been proposed as potential regulators of cell growth, but little is known about their immediate effects on protein synthesis in response to TOR-dependent growth inhibition. Here, we present a resource providing an in-depth characterisation of Schizosaccharomyces pombe phosphoproteome in relation to changes observed in global cellular protein synthesis upon TOR inhibition. We find that after TOR inhibition, the rate of protein synthesis is rapidly reduced and that notable phosphorylation changes are observed in proteins involved in a range of cellular processes. We show that this reduction in protein synthesis rates upon TOR inhibition is not dependent on S6K activity, but is partially dependent on the S. pombe homologue of eIF4G, Tif471. Our study demonstrates the impact of TOR-dependent phospho-regulation on the rate of protein synthesis and establishes a foundational resource for further investigation of additional TOR-regulated targets both in fission yeast and other eukaryotes.
Collapse
Affiliation(s)
- Tiffany Mak
- Cell Cycle LaboratoryThe Francis Crick InstituteLondonUK
- Present address:
The Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkKgs. LyngbyDenmark
| | - Andrew W Jones
- Cell Cycle LaboratoryThe Francis Crick InstituteLondonUK
- Protein Analysis and Proteomics PlatformThe Francis Crick InstituteLondonUK
| | - Paul Nurse
- Cell Cycle LaboratoryThe Francis Crick InstituteLondonUK
- Laboratory of Yeast Genetics and Cell BiologyRockefeller UniversityNew YorkNYUSA
| |
Collapse
|
8
|
Halova L, Cobley D, Franz-Wachtel M, Wang T, Morrison KR, Krug K, Nalpas N, Maček B, Hagan IM, Humphrey SJ, Petersen J. A TOR (target of rapamycin) and nutritional phosphoproteome of fission yeast reveals novel targets in networks conserved in humans. Open Biol 2021; 11:200405. [PMID: 33823663 PMCID: PMC8025308 DOI: 10.1098/rsob.200405] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/05/2021] [Indexed: 12/21/2022] Open
Abstract
Fluctuations in TOR, AMPK and MAP-kinase signalling maintain cellular homeostasis and coordinate growth and division with environmental context. We have applied quantitative, SILAC mass spectrometry to map TOR and nutrient-controlled signalling in the fission yeast Schizosaccharomyces pombe. Phosphorylation levels at more than 1000 sites were altered following nitrogen stress or Torin1 inhibition of the TORC1 and TORC2 networks that comprise TOR signalling. One hundred and thirty of these sites were regulated by both perturbations, and the majority of these (119) new targets have not previously been linked to either nutritional or TOR control in either yeasts or humans. Elimination of AMPK inhibition of TORC1, by removal of AMPKα (ssp2::ura4+), identified phosphosites where nitrogen stress-induced changes were independent of TOR control. Using a yeast strain with an ATP analogue-sensitized Cdc2 kinase, we excluded sites that were changed as an indirect consequence of mitotic control modulation by nitrogen stress or TOR signalling. Nutritional control of gene expression was reflected in multiple targets in RNA metabolism, while significant modulation of actin cytoskeletal components points to adaptations in morphogenesis and cell integrity networks. Reduced phosphorylation of the MAPKK Byr1, at a site whose human equivalent controls docking between MEK and ERK, prevented sexual differentiation when resources were sparse but not eliminated.
Collapse
Affiliation(s)
- Lenka Halova
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- Cancer Research UK Manchester Institute, Alderley Park, Macclesfield SK10 4TG, UK
| | - David Cobley
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Mirita Franz-Wachtel
- Proteome Center Tuebingen, University of Tuebingen, Auf der Morgenstelle 15, 72076 Tuebingen, Germany
| | - Tingting Wang
- Flinders Health and Medical Research Institute, Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, South Australia 5042, Australia
| | - Kaitlin R. Morrison
- Flinders Health and Medical Research Institute, Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, South Australia 5042, Australia
| | - Karsten Krug
- Proteome Center Tuebingen, University of Tuebingen, Auf der Morgenstelle 15, 72076 Tuebingen, Germany
| | - Nicolas Nalpas
- Proteome Center Tuebingen, University of Tuebingen, Auf der Morgenstelle 15, 72076 Tuebingen, Germany
| | - Boris Maček
- Proteome Center Tuebingen, University of Tuebingen, Auf der Morgenstelle 15, 72076 Tuebingen, Germany
| | - Iain M. Hagan
- Cancer Research UK Manchester Institute, Alderley Park, Macclesfield SK10 4TG, UK
| | - Sean J. Humphrey
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Camperdown, New South Wales, Australia
| | - Janni Petersen
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- Flinders Health and Medical Research Institute, Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, South Australia 5042, Australia
- Nutrition and Metabolism, South Australia Health and Medical Research Institute, North Terrace, Adelaide, South Australia 5000, Australia
| |
Collapse
|
9
|
Alao JP, Legon L, Rallis C. Crosstalk between the mTOR and DNA Damage Response Pathways in Fission Yeast. Cells 2021; 10:cells10020305. [PMID: 33540829 PMCID: PMC7913062 DOI: 10.3390/cells10020305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
Cells have developed response systems to constantly monitor environmental changes and accordingly adjust growth, differentiation, and cellular stress programs. The evolutionarily conserved, nutrient-responsive, mechanistic target of rapamycin signaling (mTOR) pathway coordinates basic anabolic and catabolic cellular processes such as gene transcription, protein translation, autophagy, and metabolism, and is directly implicated in cellular and organismal aging as well as age-related diseases. mTOR mediates these processes in response to a broad range of inputs such as oxygen, amino acids, hormones, and energy levels, as well as stresses, including DNA damage. Here, we briefly summarize data relating to the interplays of the mTOR pathway with DNA damage response pathways in fission yeast, a favorite model in cell biology, and how these interactions shape cell decisions, growth, and cell-cycle progression. We, especially, comment on the roles of caffeine-mediated DNA-damage override. Understanding the biology of nutrient response, DNA damage and related pharmacological treatments can lead to the design of interventions towards improved cellular and organismal fitness, health, and survival.
Collapse
Affiliation(s)
- John-Patrick Alao
- ZEAB Therapeutic, University of East London, Stratford Campus, Water Lane, Stratford, London E15 4LZ, UK;
| | - Luc Legon
- School of Health, Sport and Bioscience, University of East London, Stratford Campus, Water Lane, Stratford, London E15 4LZ, UK;
| | - Charalampos Rallis
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
- Correspondence:
| |
Collapse
|
10
|
Alao JP, Johansson-Sjölander J, Rallis C, Sunnerhagen P. Caffeine Stabilises Fission Yeast Wee1 in a Rad24-Dependent Manner but Attenuates Its Expression in Response to DNA Damage. Microorganisms 2020; 8:microorganisms8101512. [PMID: 33008060 PMCID: PMC7600152 DOI: 10.3390/microorganisms8101512] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
The widely consumed neuroactive compound caffeine has generated much interest due to its ability to override the DNA damage and replication checkpoints. Previously Rad3 and its homologues was thought to be the target of caffeine’s inhibitory activity. Later findings indicate that the Target of Rapamycin Complex 1 (TORC1) is the preferred target of caffeine. Effective Cdc2 inhibition requires both the activation of the Wee1 kinase and inhibition of the Cdc25 phosphatase. The TORC1, DNA damage, and environmental stress response pathways all converge on Cdc25 and Wee1. We previously demonstrated that caffeine overrides DNA damage checkpoints by modulating Cdc25 stability. The effect of caffeine on cell cycle progression resembles that of TORC1 inhibition. Furthermore, caffeine activates the Sty1 regulated environmental stress response. Caffeine may thus modulate multiple signalling pathways that regulate Cdc25 and Wee1 levels, localisation and activity. Here we show that the activity of caffeine stabilises both Cdc25 and Wee1. The stabilising effect of caffeine and genotoxic agents on Wee1 was dependent on the Rad24 chaperone. Interestingly, caffeine inhibited the accumulation of Wee1 in response to DNA damage. Caffeine may modulate cell cycle progression through increased Cdc25 activity and Wee1 repression following DNA damage via TORC1 inhibition, as TORC1 inhibition increased DNA damage sensitivity.
Collapse
Affiliation(s)
- John P Alao
- School of Health, Sports and Bioscience, University of East London, Stratford Campus, London E15 4LZ, UK
- Department of Chemistry and Molecular Biology, University of Gothenburg, P.O. Box 462, SE-405 30 Gothenburg, Sweden
| | - Johanna Johansson-Sjölander
- Department of Chemistry and Molecular Biology, University of Gothenburg, P.O. Box 462, SE-405 30 Gothenburg, Sweden
| | - Charalampos Rallis
- School of Health, Sports and Bioscience, University of East London, Stratford Campus, London E15 4LZ, UK
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| | - Per Sunnerhagen
- Department of Chemistry and Molecular Biology, University of Gothenburg, P.O. Box 462, SE-405 30 Gothenburg, Sweden
| |
Collapse
|
11
|
Feng YQ, Li BA, Feng F, Chen YS, Ren YX, Zhang H, Cao S. Novel mTOR Inhibitor Enhances the Sensitivity of Hepatocellular Carcinoma Cells to Molecular Targeting Agents. Onco Targets Ther 2020; 13:7165-7176. [PMID: 32801748 PMCID: PMC7394584 DOI: 10.2147/ott.s244474] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 07/02/2020] [Indexed: 12/14/2022] Open
Abstract
Background Although molecular-targeted agents are still the first choice for advanced hepatocellular carcinoma (HCC) treatment, the therapeutic efficacy of these agents is not satisfactory. Recently, the mammalian target of rapamycin (mTOR) is considered to be a promising molecular target that can enhance the sensitivity of HCC cells to antitumor therapy. However, the reported mTOR inhibitors have some shortcomings, and novel mTOR inhibitors need to be developed to enhance the antitumor effect of molecularly targeted agents on advanced HCC. Methods In this study, five small-molecular compounds that could serve as potential mTOR-specific inhibitors were identified by virtual screening. The activity of tert-butyl (4-(9-(2-(1,3-dioxolan-2-yl)ethyl)-6-morpholino-9H-purin-2-yl)phenyl)carbamate (compound 4) was measured by enzyme test and Western blot, and its antitumor effect on HCC was examined in nude mice subcutaneous tumor model. Results The results showed that 4 is the most effective one in inhibiting the activation of mTOR kinase (mTOR IC50 = 17.52±3.67 nmol/L) among the five lead compounds. Further research in this study indicated that treatment with 4 enhanced the sensitivity of HCC cells to the molecular-targeted agents, such as sorafenib, regorafenib, lenvatinib, anlotinib, and apatinib. In addition, this research indicated that mTOR was correlated with the poor prognosis in patients with advanced HCC who received sorafenib. Conclusion Our study identified a new type of small-molecular inhibitors of mTOR and confirmed their ability to enhance the antitumor effect of molecular-targeted agents on advanced HCC.
Collapse
Affiliation(s)
- Ying-Qi Feng
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Bo-An Li
- Center for Clinical Laboratory, The Fifth Medical Center, General Hospital of Chinese PLA, Beijing 100039, People's Republic of China
| | - Fan Feng
- Center for Clinical Laboratory, The Fifth Medical Center, General Hospital of Chinese PLA, Beijing 100039, People's Republic of China
| | - Yong-Shou Chen
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Yi-Xin Ren
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Heng Zhang
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Shuang Cao
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| |
Collapse
|
12
|
Alao JP, Sunnerhagen P. Caffeine as a tool for investigating the integration of Cdc25 phosphorylation, activity and ubiquitin-dependent degradation in Schizosaccharomyces pombe. Cell Div 2020; 15:10. [PMID: 32612670 PMCID: PMC7322915 DOI: 10.1186/s13008-020-00066-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/08/2020] [Indexed: 12/27/2022] Open
Abstract
The evolutionarily conserved Cdc25 phosphatase is an essential protein that removes inhibitory phosphorylation moieties on the mitotic regulator Cdc2. Together with the Wee1 kinase, a negative regulator of Cdc2 activity, Cdc25 is thus a central regulator of cell cycle progression in Schizosaccharomyces pombe. The expression and activity of Cdc25 is dependent on the activity of the Target of Rapamycin Complex 1 (TORC1). TORC1 inhibition leads to the activation of Cdc25 and repression of Wee1, leading to advanced entry into mitosis. Withdrawal of nitrogen leads to rapid Cdc25 degradation via the ubiquitin- dependent degradation pathway by the Pub1 E3- ligase. Caffeine is believed to mediate the override of DNA damage checkpoint signalling, by inhibiting the activity of the ataxia telangiectasia mutated (ATM)/Rad3 homologues. This model remains controversial, as TORC1 appears to be the preferred target of caffeine in vivo. Recent studies suggest that caffeine induces DNA damage checkpoint override by inducing the nuclear accumulation of Cdc25 in S. pombe. Caffeine may thus modulate Cdc25 activity and stability via inhibition of TORC1. A clearer understanding of the mechanisms by which caffeine stabilises Cdc25, may provide novel insights into how TORC1 and DNA damage signalling is integrated.
Collapse
Affiliation(s)
- John P Alao
- School of Health, Sports and Bioscience, University of East London, Stratford Campus, London, E15 4LZ UK.,Department of Chemistry and Molecular Biology, University of Gothenburg, Box 462, Gothenburg, SE- 405 30 Sweden
| | - Per Sunnerhagen
- Department of Chemistry and Molecular Biology, University of Gothenburg, Box 462, Gothenburg, SE- 405 30 Sweden
| |
Collapse
|
13
|
Feng YQ, Gu SX, Chen YS, Gao XD, Ren YX, Chen JC, Lu YY, Zhang H, Cao S. Virtual Screening and Optimization of Novel mTOR Inhibitors for Radiosensitization of Hepatocellular Carcinoma. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1779-1798. [PMID: 32440103 PMCID: PMC7220363 DOI: 10.2147/dddt.s249156] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/09/2020] [Indexed: 12/12/2022]
Abstract
Background Radiotherapy has an ameliorative effect on a wide variety of tumors, but hepatocellular carcinoma (HCC) is insensitive to this treatment. Overactivated mammalian target of rapamycin (mTOR) plays an important part in the resistance of HCC to radiotherapy; thus, mTOR inhibitors have potential as novel radiosensitizers to enhance the efficacy of radiotherapy for HCC. Methods A lead compound was found based on pharmacophore modeling and molecular docking, and optimized according to the differences between the ATP-binding pockets of mTOR and PI3K. The radiosensitizing effect of the optimized compound (2a) was confirmed by colony formation assays and DNA double-strand break assays in vitro. The discovery and preclinical characteristics of this compound are described. Results The key amino acid residues in mTOR were identified, and a precise virtual screening model was constructed. Compound 2a, with a 4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidine scaffold, exhibited promising potency against mTOR (mTOR IC50=7.1 nmol/L (nM)) with 126-fold selectivity over PI3Kα. Moreover, 2a significantly enhanced the sensitivity of HCC to radiotherapy in vitro in a dose-dependent manner. Conclusion A new class of selective mTOR inhibitors was developed and their radiosensitization effects were confirmed. This study also provides a basis for developing mTOR-specific inhibitors for use as radiosensitizers for HCC radiotherapy.
Collapse
Affiliation(s)
- Ying-Qi Feng
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Shuang-Xi Gu
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Yong-Shou Chen
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Xu-Dong Gao
- Comprehensive Liver Cancer Department, The Fifth Medical Center, Chinese PLA General Hospital, Beijing 100039, People's Republic of China
| | - Yi-Xin Ren
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Jian-Chao Chen
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Ministry of Education, Shenyang 110016, People's Republic of China
| | - Yin-Ying Lu
- Comprehensive Liver Cancer Department, The Fifth Medical Center, Chinese PLA General Hospital, Beijing 100039, People's Republic of China
| | - Heng Zhang
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Shuang Cao
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China.,National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| |
Collapse
|
14
|
Rodríguez-López M, Gonzalez S, Hillson O, Tunnacliffe E, Codlin S, Tallada VA, Bähler J, Rallis C. The GATA Transcription Factor Gaf1 Represses tRNAs, Inhibits Growth, and Extends Chronological Lifespan Downstream of Fission Yeast TORC1. Cell Rep 2020; 30:3240-3249.e4. [PMID: 32160533 PMCID: PMC7068653 DOI: 10.1016/j.celrep.2020.02.058] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 12/17/2019] [Accepted: 02/13/2020] [Indexed: 12/13/2022] Open
Abstract
Target of Rapamycin Complex 1 (TORC1) signaling promotes growth and aging. Inhibition of TORC1 leads to reduced protein translation, which promotes longevity. TORC1-dependent post-transcriptional regulation of protein translation has been well studied, while analogous transcriptional regulation is less understood. Here we screen fission yeast mutants for resistance to Torin1, which inhibits TORC1 and cell growth. Cells lacking the GATA factor Gaf1 (gaf1Δ) grow normally even in high doses of Torin1. The gaf1Δ mutation shortens the chronological lifespan of non-dividing cells and diminishes Torin1-mediated longevity. Expression profiling and genome-wide binding experiments show that upon TORC1 inhibition, Gaf1 directly upregulates genes for small-molecule metabolic pathways and indirectly represses genes for protein translation. Surprisingly, Gaf1 binds to and downregulates the tRNA genes, so it also functions as a transcription factor for RNA polymerase III. Thus, Gaf1 controls the transcription of both protein-coding and tRNA genes to inhibit translation and growth downstream of TORC1.
Collapse
Affiliation(s)
- María Rodríguez-López
- Institute of Healthy Ageing and Department of Genetics, Evolution & Environment, University College London, London WC1E 6BT, UK
| | - Suam Gonzalez
- School of Health, Sport and Bioscience, University of East London, Stratford Campus, London E14 4LZ, UK
| | - Olivia Hillson
- School of Health, Sport and Bioscience, University of East London, Stratford Campus, London E14 4LZ, UK
| | - Edward Tunnacliffe
- Institute of Healthy Ageing and Department of Genetics, Evolution & Environment, University College London, London WC1E 6BT, UK
| | - Sandra Codlin
- Institute of Healthy Ageing and Department of Genetics, Evolution & Environment, University College London, London WC1E 6BT, UK
| | - Victor A Tallada
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide/CSIC, 41013 Sevilla, Spain
| | - Jürg Bähler
- Institute of Healthy Ageing and Department of Genetics, Evolution & Environment, University College London, London WC1E 6BT, UK.
| | - Charalampos Rallis
- Institute of Healthy Ageing and Department of Genetics, Evolution & Environment, University College London, London WC1E 6BT, UK; School of Health, Sport and Bioscience, University of East London, Stratford Campus, London E14 4LZ, UK; School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK.
| |
Collapse
|
15
|
Zhang Z, Hou H, Yu S, Zhou C, Zhang X, Li N, Zhang S, Song K, Lu Y, Liu D, Lu H, Xu H. Inflammation-induced mammalian target of rapamycin signaling is essential for retina regeneration. Glia 2019; 68:111-127. [PMID: 31444939 DOI: 10.1002/glia.23707] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 12/18/2022]
Abstract
Upon retina injury, Müller glia in the zebrafish retina respond by generating multipotent progenitors to repair the retina. However, the complete mechanisms underlying retina regeneration remain elusive. Here we report inflammation-induced mammalian target of rapamycin (mTOR) signaling in the Müller glia is essential for retina regeneration in adult zebrafish. We show after a stab injury, mTOR is rapidly activated in Müller glia and later Müller glia-derived progenitor cells (MGPCs). Importantly, mTOR is required for Müller glia dedifferentiation, as well as the proliferation of Müller glia and MGPCs. Interestingly, transient mTOR inhibition by rapamycin only reversibly suppresses MGPC proliferation, while its longer suppression by knocking down Raptor significantly inhibits the regeneration of retinal neurons. We further show mTOR promotes retina regeneration by regulating the mRNA expression of key reprogramming factors ascl1a and lin-28a, cell cycle-related genes and critical cytokines. Surprisingly, we identify microglia/macrophage-mediated inflammation as an important upstream regulator of mTOR in the Müller glia and it promotes retina regeneration through mTOR. Our study not only demonstrates the important functions of mTOR but also reveals an interesting link between inflammation and the mTOR signaling during retina regeneration.
Collapse
Affiliation(s)
- Zhiqiang Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu Province, China
| | - Haitao Hou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu Province, China
| | - Shuguang Yu
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Cuiping Zhou
- Department of Ophthalmology, Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xiaoli Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu Province, China
| | - Na Li
- Department of Ophthalmology, Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Shuqiang Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu Province, China
| | - Kaida Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu Province, China
| | - Ying Lu
- Department of Ophthalmology, Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Dong Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu Province, China
| | - Hong Lu
- Department of Ophthalmology, Eye Institute, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Hui Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu Province, China
| |
Collapse
|
16
|
Oya E, Durand-Dubief M, Cohen A, Maksimov V, Schurra C, Nakayama JI, Weisman R, Arcangioli B, Ekwall K. Leo1 is essential for the dynamic regulation of heterochromatin and gene expression during cellular quiescence. Epigenetics Chromatin 2019; 12:45. [PMID: 31315658 PMCID: PMC6636030 DOI: 10.1186/s13072-019-0292-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 07/10/2019] [Indexed: 11/16/2022] Open
Abstract
Background Cellular quiescence is a reversible differentiation state during which cells modify their gene expression program to inhibit metabolic functions and adapt to a new cellular environment. The epigenetic changes accompanying these alterations are not well understood. We used fission yeast cells as a model to study the regulation of quiescence. When these cells are starved for nitrogen, the cell cycle is arrested in G1, and the cells enter quiescence (G0). A gene regulatory program is initiated, including downregulation of thousands of genes—for example, those related to cell proliferation—and upregulation of specific genes—for example, autophagy genes—needed to adapt to the physiological challenge. These changes in gene expression are accompanied by a marked alteration of nuclear organization and chromatin structure. Results Here, we investigated the role of Leo1, a subunit of the conserved RNA polymerase-associated factor 1 (Paf1) complex, in the quiescence process using fission yeast as the model organism. Heterochromatic regions became very dynamic in fission yeast in G0 during nitrogen starvation. The reduction of heterochromatin in early G0 was correlated with reduced target of rapamycin complex 2 (TORC2) signaling. We demonstrated that cells lacking Leo1 show reduced survival in G0. In these cells, heterochromatic regions, including subtelomeres, were stabilized, and the expression of many genes, including membrane transport genes, was abrogated. TOR inhibition mimics the effect of nitrogen starvation, leading to the expression of subtelomeric genes, and this effect was suppressed by genetic deletion of leo1. Conclusions We identified a protein, Leo1, necessary for survival during quiescence. Leo1 is part of a conserved protein complex, Paf1C, linked to RNA polymerase II. We showed that Leo1, acting downstream of TOR, is crucial for the dynamic reorganization of chromosomes and the regulation of gene expression during cellular quiescence. Genes encoding membrane transporters are not expressed in quiescent leo1 mutant cells, and cells die after 2 weeks of nitrogen starvation. Taken together, our results suggest that Leo1 is essential for the dynamic regulation of heterochromatin and gene expression during cellular quiescence. Electronic supplementary material The online version of this article (10.1186/s13072-019-0292-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eriko Oya
- Department of Biosciences and Nutrition, Karolinska Institutet, NEO Building, 141 83, Huddinge, Sweden
| | - Mickaël Durand-Dubief
- Department of Biosciences and Nutrition, Karolinska Institutet, NEO Building, 141 83, Huddinge, Sweden
| | - Adiel Cohen
- Department of Natural and Life Sciences, The Open University of Israel, Ra'anana, Israel
| | - Vladimir Maksimov
- Department of Biosciences and Nutrition, Karolinska Institutet, NEO Building, 141 83, Huddinge, Sweden
| | - Catherine Schurra
- Unite Dynamique du Génome, Département Génomes et Génétique, Pasteur Institute, Paris, France
| | - Jun-Ichi Nakayama
- Division of Chromatin Regulation, National Institute for Basic Biology, Okazaki, Japan
| | - Ronit Weisman
- Department of Natural and Life Sciences, The Open University of Israel, Ra'anana, Israel
| | - Benoit Arcangioli
- Unite Dynamique du Génome, Département Génomes et Génétique, Pasteur Institute, Paris, France
| | - Karl Ekwall
- Department of Biosciences and Nutrition, Karolinska Institutet, NEO Building, 141 83, Huddinge, Sweden.
| |
Collapse
|
17
|
Son O, Kim S, Kim D, Hur YS, Kim J, Cheon CI. Involvement of TOR signaling motif in the regulation of plant autophagy. Biochem Biophys Res Commun 2019; 501:643-647. [PMID: 29738770 DOI: 10.1016/j.bbrc.2018.05.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/04/2018] [Indexed: 01/22/2023]
Abstract
In our previous studies, we have demonstrated that a stretch of amino-acid sequences identified from Arabidopsis ribosomal S6 kinase 1 (AtS6K1) provided a plant version of the TOS (TOR-signaling) motif, mediating the interaction with the Raptor protein in the TOR (Target of Rapamycin) kinase complex. Here we report the presence of same element in Arabidopsis Autophagy related-13 (AtATG13) protein, which is a key component of the plant autophagy response. Its composition is nearly identical to that found in the AtS6K1 in the five-amino-acid core sequence, and the presence of this five-amino-acid sequence was found to be essential for its interaction with the Raptor protein. A mutant AtATG13 protein lacking this five-amino-acid element conferred an elevated autophagy response and could not effectively phosphorylated by TOR kinase activity, demonstrating its role in mediating the TOR signaling to the components that carry it as a possible TOS motif. A ligand-binding simulation model using the MM-PBSA method indicates that both of the five-amino-acid sequence elements of AtS6K1 and AtATG13 have strong probability of making stable interface with the Raptor binding pocket, corroborating our proposition for this element as the plant TOS motif.
Collapse
Affiliation(s)
- Ora Son
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, South Korea
| | - Sunghan Kim
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, South Korea
| | - Dooil Kim
- Department of Biological Science, Chungbuk National University, Cheongju, 28644, South Korea
| | - Yoon-Sun Hur
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, South Korea
| | - Jiyoung Kim
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, South Korea
| | - Choong-Ill Cheon
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, South Korea.
| |
Collapse
|
18
|
Ahmad Z, Magyar Z, Bögre L, Papdi C. Cell cycle control by the target of rapamycin signalling pathway in plants. JOURNAL OF EXPERIMENTAL BOTANY 2019; 70:2275-2284. [PMID: 30918972 DOI: 10.1093/jxb/erz140] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/15/2019] [Indexed: 06/09/2023]
Abstract
Cells need to ensure a sufficient nutrient and energy supply before committing to proliferate. In response to positive mitogenic signals, such as light, sugar availability, and hormones, the target of rapamycin (TOR) signalling pathway promotes cell growth that connects to the entry and passage through the cell division cycle via multiple signalling mechanisms. Here, we summarize current understanding of cell cycle regulation by the RBR-E2F regulatory hub and the DREAM-like complexes, and highlight possible functional relationships between these regulators and TOR signalling. A genetic screen recently uncovered a downstream signalling component to TOR that regulates cell proliferation, YAK1, a member of the dual specificity tyrosine phosphorylation-regulated kinase (DYRK) family. YAK1 activates the plant-specific SIAMESE-RELATED (SMR) cyclin-dependent kinase inhibitors and therefore could be important to regulate both the CDKA-RBR-E2F pathway to control the G1/S transition and the mitotic CDKB1;1 to control the G2/M transition. TOR, as a master regulator of both protein synthesis-driven cell growth and cell proliferation is also central for cell size homeostasis. We conclude the review by briefly highlighting the potential applications of combining TOR and cell cycle knowledge in the context of ensuring future food security.
Collapse
Affiliation(s)
- Zaki Ahmad
- School of Biological Sciences, Bourne Laboratory. Royal Holloway, University of London, Egham, Surrey, UK
| | - Zoltán Magyar
- Institute of Plant Biology, Biological Research Centre, Hungarian Academy of Sciences Szeged, Hungary
| | - László Bögre
- School of Biological Sciences, Bourne Laboratory. Royal Holloway, University of London, Egham, Surrey, UK
| | - Csaba Papdi
- School of Biological Sciences, Bourne Laboratory. Royal Holloway, University of London, Egham, Surrey, UK
| |
Collapse
|
19
|
Lie S, Banks P, Lawless C, Lydall D, Petersen J. The contribution of non-essential Schizosaccharomyces pombe genes to fitness in response to altered nutrient supply and target of rapamycin activity. Open Biol 2019; 8:rsob.180015. [PMID: 29720420 PMCID: PMC5990653 DOI: 10.1098/rsob.180015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/06/2018] [Indexed: 12/12/2022] Open
Abstract
Nutrient fluctuations in the cellular environment promote changes in cell metabolism and growth to adapt cell proliferation accordingly. The target of rapamycin (TOR) signalling network plays a key role in the coordination of growth and cell proliferation with the nutrient environment and, importantly, nutrient limitation reduces TOR complex 1 (TORC1) signalling. We have performed global quantitative fitness profiling of the collection of Schizosaccharomyces pombe strains from which non-essential genes have been deleted. We identified genes that regulate fitness when cells are grown in a nutrient-rich environment compared with minimal environments, with varying nitrogen sources including ammonium, glutamate and proline. In addition, we have performed the first global screen for genes that regulate fitness when both TORC1 and TORC2 signalling is reduced by Torin1. Analysis of genes whose deletions altered fitness when nutrients were limited, or when TOR signalling was compromised, identified a large number of genes that regulate transmembrane transport, transcription and chromatin organization/regulation and vesicle-mediated transport. The ability to tolerate reduced TOR signalling placed demands upon a large number of biological processes including autophagy, mRNA metabolic processing and nucleocytoplasmic transport. Importantly, novel biological processes and all processes known to be regulated by TOR were identified in our screens. In addition, deletion of 62 genes conserved in humans gave rise to strong sensitivity or resistance to Torin1, and 29 of these 62 genes have novel links to TOR signalling. The identification of chromatin and transcriptional regulation, nutritional uptake and transport pathways in this powerful genetic model now paves the way for a molecular understanding of how cells adapt to the chronic and acute fluctuations in nutrient supply that all eukaryotes experience at some stage, and which is a key feature of cancer cells within solid tumours.
Collapse
Affiliation(s)
- Shervi Lie
- Flinders Centre for Innovation in Cancer, College of Medicine & Public Health, Flinders University, Bedford Park, Adelaide, South Australia 5042, Australia
| | - Peter Banks
- High Throughput Screening Facility, Newcastle Biomedicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Conor Lawless
- Institute for Cell & Molecular Biosciences, Newcastle University Medical School, Newcastle upon Tyne NE2 4HH, UK
| | - David Lydall
- Institute for Cell & Molecular Biosciences, Newcastle University Medical School, Newcastle upon Tyne NE2 4HH, UK
| | - Janni Petersen
- Flinders Centre for Innovation in Cancer, College of Medicine & Public Health, Flinders University, Bedford Park, Adelaide, South Australia 5042, Australia .,South Australia Health and Medical Research Institute, North Terrace, PO Box 11060, Adelaide, South Australia 5000, Australia
| |
Collapse
|
20
|
Kumar P, Awasthi A, Nain V, Issac B, Puria R. Novel insights into TOR signalling in Saccharomyces cerevisiae through Torin2. Gene 2018; 669:15-27. [DOI: 10.1016/j.gene.2018.05.081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/06/2018] [Accepted: 05/21/2018] [Indexed: 12/18/2022]
|
21
|
Guo JC, Li J, Zhou L, Yang JY, Zhang ZG, Liang ZY, Zhou WX, You L, Zhang TP, Zhao YP. CXCL12-CXCR7 axis contributes to the invasive phenotype of pancreatic cancer. Oncotarget 2018; 7:62006-62018. [PMID: 27542220 PMCID: PMC5308707 DOI: 10.18632/oncotarget.11330] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/27/2016] [Indexed: 12/26/2022] Open
Abstract
Chemokine (C-X-C motif) receptor 7 (CXCR7) and its ligand, chemokine (C-X-C motif) ligand 12 (CXCL12), were established to be involved in biological behaviors and associated with prognosis in many cancers. However, effects, underlying mechanisms of CXCL12-CXCR7 axis in invasive phenotype of pancreatic cancer (PC) and its clinicopathologic significances have not been comprehensively explored. In the present study, it was first found by tissue microarray-based immunohistochemistry that CXCL12 and CXCR7 staining scores were significantly associated with vessel invasion and overall survival in two independent cohorts of PC. Besides, co-expression of these proteins was an independent prognosticator in multivariate analysis in both cohorts. Then, migration and invasion, but not proliferation, were decreased in CXCR7-stably silenced PC cells, whereas opposite changes were observed in CXCR7-stably overexpressed cells, accompanied by alterations of mTOR and Rho/ROCK pathways. CXCL12 stimulated migration, invasion, CXCR7 expression and phosphorylation of key mTOR proteins. AMD3100 did not influence effects of CXCL12. Two mTOR inhibitors, rapamycin and Torin1, reversed enhanced invasive phenotypes and mTOR phosphorylation in CXCR7-overexpressed cells. Moreover, CXCR7 directly interacts with mTOR. Finally, liver metastasis, but not growth, was affected by CXCR7 status in orthotopically-implanted PC models in nude mice. Collectively, CXCL12-CXCR7 axis accelerates migration and invasion of PC cells through mTOR and Rho/ROCK pathways, and predicts poor prognosis of PC.
Collapse
Affiliation(s)
- Jun-Chao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Jian Li
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China.,Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Jian-Yu Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhi-Yong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Wei-Xun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Tai-Ping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| | - Yu-Pei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
22
|
Navarro FJ, Chakravarty P, Nurse P. Phosphorylation of the RNA-binding protein Zfs1 modulates sexual differentiation in fission yeast. J Cell Sci 2017; 130:4144-4154. [PMID: 29084823 PMCID: PMC5769579 DOI: 10.1242/jcs.208066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/24/2017] [Indexed: 12/21/2022] Open
Abstract
Sexual differentiation in the fission yeast Schizosaccharomyces pombe promotes cell cycle arrest and extensive changes in gene expression, resulting in cell-to-cell fusion, the exchange of hereditary material and specialized cell division. These events are detrimental to the cell if they are triggered in inappropriate conditions, and therefore the decision to differentiate must be precisely controlled. Here, we investigated the role of the RNA-binding protein Zfs1 in this process by identifying its targets and characterizing novel post-translational regulatory mechanisms. We found that Zfs1 negatively regulates the G1 cyclin Puc1, and deregulated Puc1 levels inhibit differentiation in the zfs1Δ mutant. We also found that Zfs1 undergoes phosphorylation, which is stimulated upon nitrogen depletion or inhibition of the TOR pathway. Phosphorylation of Zfs1 modulates accumulation of Puc1 and plays an important role in the response of the cell to sexual differentiation signals. We propose that Zfs1 functions as an integrator of nutrient information to modulate sexual differentiation, contributing to the establishment of the differentiation-activating threshold.
Collapse
Affiliation(s)
| | - Probir Chakravarty
- Bioinformatics, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Paul Nurse
- Cell Cycle Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| |
Collapse
|
23
|
Laboucarié T, Detilleux D, Rodriguez-Mias RA, Faux C, Romeo Y, Franz-Wachtel M, Krug K, Maček B, Villén J, Petersen J, Helmlinger D. TORC1 and TORC2 converge to regulate the SAGA co-activator in response to nutrient availability. EMBO Rep 2017; 18:2197-2218. [PMID: 29079657 DOI: 10.15252/embr.201744942] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 08/31/2017] [Accepted: 09/07/2017] [Indexed: 12/19/2022] Open
Abstract
Gene expression regulation is essential for cells to adapt to changes in their environment. Co-activator complexes have well-established roles in transcriptional regulation, but less is known about how they sense and respond to signaling cues. We have previously shown that, in fission yeast, one such co-activator, the SAGA complex, controls gene expression and the switch from proliferation to differentiation in response to nutrient availability. Here, using a combination of genetic, biochemical, and proteomic approaches, we show that SAGA responds to nutrients through the differential phosphorylation of its Taf12 component, downstream of both the TORC1 and TORC2 pathways. Taf12 phosphorylation increases early upon starvation and is controlled by the opposing activities of the PP2A phosphatase, which is activated by TORC1, and the TORC2-activated Gad8AKT kinase. Mutational analyses suggest that Taf12 phosphorylation prevents cells from committing to differentiation until starvation reaches a critical level. Overall, our work reveals that SAGA is a direct target of nutrient-sensing pathways and has uncovered a mechanism by which TORC1 and TORC2 converge to control gene expression and cell fate decisions.
Collapse
Affiliation(s)
| | | | | | - Céline Faux
- CRBM, CNRS, University of Montpellier, Montpellier, France
| | - Yves Romeo
- CRBM, CNRS, University of Montpellier, Montpellier, France
| | | | | | - Boris Maček
- Proteome Center Tübingen, Tuebingen, Germany
| | - Judit Villén
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Janni Petersen
- Flinders Centre for Innovation in Cancer, School of Medicine, Faculty of Health Science, Flinders University, Adelaide, SA, Australia
| | | |
Collapse
|
24
|
Host Serine/Threonine Kinases mTOR and Protein Kinase C-α Promote InlB-Mediated Entry of Listeria monocytogenes. Infect Immun 2017; 85:IAI.00087-17. [PMID: 28461391 DOI: 10.1128/iai.00087-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/25/2017] [Indexed: 01/27/2023] Open
Abstract
The bacterial pathogen Listeria monocytogenes causes foodborne illnesses resulting in gastroenteritis, meningitis, or abortion. Listeria induces its internalization into some human cells through interaction of the bacterial surface protein InlB with the host receptor tyrosine kinase Met. InlB-dependent entry requires localized polymerization of the host actin cytoskeleton. The signal transduction pathways that act downstream of Met to regulate actin filament assembly or other processes during Listeria uptake remain incompletely characterized. Here, we demonstrate important roles for the human serine/threonine kinases mTOR and protein kinase C-α (PKC-α) in InlB-dependent entry. Experiments involving RNA interference (RNAi) indicated that two multiprotein complexes containing mTOR, mTORC1 and mTORC2, are each needed for efficient internalization of Listeria into cells of the human cell line HeLa. InlB stimulated Met-dependent phosphorylation of mTORC1 or mTORC2 substrates, demonstrating activation of both mTOR-containing complexes. RNAi studies indicated that the mTORC1 effectors 4E-BP1 and hypoxia-inducible factor 1α (HIF-1α) and the mTORC2 substrate PKC-α each control Listeria uptake. Genetic or pharmacological inhibition of PKC-α reduced the internalization of Listeria and the accumulation of actin filaments that normally accompanies InlB-mediated entry. Collectively, our results identify mTOR and PKC-α to be host factors exploited by Listeria to promote infection. PKC-α controls Listeria entry, at least in part, by regulating the actin cytoskeleton downstream of the Met receptor.
Collapse
|
25
|
Cobley D, Hálová L, Schauries M, Kaczmarek A, Franz-Wachtel M, Du W, Krug K, Maček B, Petersen J. Ste12/Fab1 phosphatidylinositol-3-phosphate 5-kinase is required for nitrogen-regulated mitotic commitment and cell size control. PLoS One 2017; 12:e0172740. [PMID: 28273166 PMCID: PMC5342193 DOI: 10.1371/journal.pone.0172740] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/08/2017] [Indexed: 11/18/2022] Open
Abstract
Tight coupling of cell growth and cell cycle progression enable cells to adjust their rate of division, and therefore size, to the demands of proliferation in varying nutritional environments. Nutrient stress promotes inhibition of Target Of Rapamycin Complex 1 (TORC1) activity. In fission yeast, reduced TORC1 activity advances mitotic onset and switches growth to a sustained proliferation at reduced cell size. A screen for mutants, that failed to advance mitosis upon nitrogen stress, identified a mutant in the PIKFYVE 1-phosphatidylinositol-3-phosphate 5-kinase fission yeast homolog Ste12. Ste12PIKFYVE deficient mutants were unable to advance the cell cycle to reduce cell size after a nitrogen downshift to poor nitrogen (proline) growth conditions. While it is well established that PI(3,5)P2 signalling is required for autophagy and that Ste12PIKFYVE mutants have enlarged vacuoles (yeast lysosomes), neither a block to autophagy or mutants that independently have enlarged vacuoles had any impact upon nitrogen control of mitotic commitment. The addition of rapamycin to Ste12PIKFYVE deficient mutants reduced cell size at division to suggest that Ste12PIKFYVE possibly functions upstream of TORC1. ste12 mutants display increased Torin1 (TOR inhibitor) sensitivity. However, no major impact on TORC1 or TORC2 activity was observed in the ste12 deficient mutants. In summary, Ste12PIKFYVE is required for nitrogen-stress mediated advancement of mitosis to reduce cell size at division.
Collapse
Affiliation(s)
- David Cobley
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Lenka Hálová
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Marie Schauries
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Adrian Kaczmarek
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, SA, Australia
| | | | - Wei Du
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Karsten Krug
- Proteome Center Tübingen, Auf der Morgenstelle, Tuebingen, Germany
| | - Boris Maček
- Proteome Center Tübingen, Auf der Morgenstelle, Tuebingen, Germany
| | - Janni Petersen
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, SA, Australia
- South Australia Health and Medical Research Institute, North Terrace, Adelaide SA Australia
- * E-mail:
| |
Collapse
|
26
|
Multiple crosstalk between TOR and the cell integrity MAPK signaling pathway in fission yeast. Sci Rep 2016; 6:37515. [PMID: 27876895 PMCID: PMC5120329 DOI: 10.1038/srep37515] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 10/28/2016] [Indexed: 02/07/2023] Open
Abstract
In eukaryotic cells, the highly conserved Target of Rapamycin (TOR) and the Mitogen Activated Protein Kinase (MAPK) signaling pathways elicit adaptive responses to extra- and intracellular conditions by regulating essential cellular functions. However, the nature of the functional relationships between both pathways is not fully understood. In the fission yeast Schizosaccharomyces pombe the cell integrity MAPK pathway (CIP) regulates morphogenesis, cell wall structure and ionic homeostasis. We show that the Rab GTPase Ryh1, a TORC2 complex activator, cross-activates the CIP and its core member, the MAPK Pmk1, by two distinct mechanisms. The first one involves TORC2 and its downstream effector, Akt ortholog Gad8, which together with TORC1 target Psk1 increase protein levels of the PKC ortholog Pck2 during cell wall stress or glucose starvation. Also, Ryh1 activates Pmk1 in a TORC2-independent fashion by prompting plasma membrane trafficking and stabilization of upstream activators of the MAPK cascade, including PDK ortholog Ksg1 or Rho1 GEF Rgf1. Besides, stress-activated Pmk1 cross-inhibits Ryh1 signaling by decreasing the GTPase activation cycle, and this ensures cell growth during alterations in phosphoinositide metabolism. Our results reveal a highly intricate cross-regulatory relationship between both pathways that warrants adequate cell adaptation and survival in response to environmental changes.
Collapse
|
27
|
Ke B, Tian M, Li J, Liu B, He G. Targeting Programmed Cell Death Using Small-Molecule Compounds to Improve Potential Cancer Therapy. Med Res Rev 2016; 36:983-1035. [PMID: 27357603 DOI: 10.1002/med.21398] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 05/04/2016] [Accepted: 05/28/2016] [Indexed: 02/05/2023]
Affiliation(s)
- Bowen Ke
- Department of Anesthesiology, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Mao Tian
- Department of Anesthesiology, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Jingjing Li
- Department of Anesthesiology, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Bo Liu
- Department of Anesthesiology, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital, Sichuan University; Chengdu 610041 China
| | - Gu He
- Department of Anesthesiology, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy; West China Hospital, Sichuan University; Chengdu 610041 China
| |
Collapse
|
28
|
Ma N, Ma Y, Nakashima A, Kikkawa U, Furuyashiki T. The Loss of Lam2 and Npr2-Npr3 Diminishes the Vacuolar Localization of Gtr1-Gtr2 and Disinhibits TORC1 Activity in Fission Yeast. PLoS One 2016; 11:e0156239. [PMID: 27227887 PMCID: PMC4881991 DOI: 10.1371/journal.pone.0156239] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 05/11/2016] [Indexed: 12/13/2022] Open
Abstract
In mammalian cells, mTORC1 activity is regulated by Rag GTPases. It is thought that the Ragulator complex and the GATOR (GAP activity towards Rags) complex regulate RagA/B as its GDP/GTP exchange factor (GEF) and GTPase-activating protein (GAP), respectively. However, the functions of components in these complexes remain elusive. Using fission yeast as a model organism, here we found that the loss of Lam2 (SPBC1778.05c), a homolog of a Ragulator component LAMTOR2, as well as the loss of Gtr1 or Gtr2 phenocopies the loss of Npr2 or Npr3, homologs of GATOR components Nprl2 or Nprl3, respectively. These phenotypes were rescued by TORC1 inhibition using pharmacological or genetic means, and the loss of Lam2, Gtr1, Gtr2, Npr2 or Npr3 disinhibited TORC1 activity under nitrogen depletion, as measured by Rps6 phosphorylation. Consistently, overexpression of GDP-locked Gtr1S20L or GTP-locked Gtr2Q60L, which suppress TORC1 activity in budding yeast, rescued the growth defect of Δgtr1 cells or Δgtr2 cells, respectively, and the loss of Lam2, Npr2 or Npr3 similarly diminished the vacuolar localization and the protein levels of Gtr1 and Gtr2. Furthermore, Lam2 physically interacted with Npr2 and Gtr1. These findings suggest that Lam2 and Npr2-Npr3 function together as a tether for GDP-bound Gtr1 to the vacuolar membrane, thereby suppressing TORC1 activity for multiple cellular functions.
Collapse
Affiliation(s)
- Ning Ma
- Division of Pharmacology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yan Ma
- Division of Pharmacology, Graduate School of Medicine, Kobe University, Kobe, Japan
- * E-mail:
| | | | - Ushio Kikkawa
- Biosignal Research Center, Kobe University, Kobe, Japan
| | - Tomoyuki Furuyashiki
- Division of Pharmacology, Graduate School of Medicine, Kobe University, Kobe, Japan
| |
Collapse
|
29
|
Baker K, Kirkham S, Halova L, Atkin J, Franz-Wachtel M, Cobley D, Krug K, Maček B, Mulvihill DP, Petersen J. TOR complex 2 localises to the cytokinetic actomyosin ring and controls the fidelity of cytokinesis. J Cell Sci 2016; 129:2613-24. [PMID: 27206859 PMCID: PMC4958305 DOI: 10.1242/jcs.190124] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/06/2016] [Indexed: 01/30/2023] Open
Abstract
The timing of cell division is controlled by the coupled regulation of growth and division. The target of rapamycin (TOR) signalling network synchronises these processes with the environmental setting. Here, we describe a novel interaction of the fission yeast TOR complex 2 (TORC2) with the cytokinetic actomyosin ring (CAR), and a novel role for TORC2 in regulating the timing and fidelity of cytokinesis. Disruption of TORC2 or its localisation results in defects in CAR morphology and constriction. We provide evidence that the myosin II protein Myp2 and the myosin V protein Myo51 play roles in recruiting TORC2 to the CAR. We show that Myp2 and TORC2 are co-dependent upon each other for their normal localisation to the cytokinetic machinery. We go on to show that TORC2-dependent phosphorylation of actin-capping protein 1 (Acp1, a known regulator of cytokinesis) controls CAR stability, modulates Acp1-Acp2 (the equivalent of the mammalian CAPZA-CAPZB) heterodimer formation and is essential for survival upon stress. Thus, TORC2 localisation to the CAR, and TORC2-dependent Acp1 phosphorylation contributes to timely control and the fidelity of cytokinesis and cell division.
Collapse
Affiliation(s)
- Karen Baker
- School of Biosciences, University of Kent, Giles Lane, Canterbury, Kent CT2 7NJ, UK
| | - Sara Kirkham
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Lenka Halova
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Jane Atkin
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | | | - David Cobley
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Karsten Krug
- Proteome Center Tübingen, Auf der Morgenstelle 15, Tübingen 72076, Germany
| | - Boris Maček
- Proteome Center Tübingen, Auf der Morgenstelle 15, Tübingen 72076, Germany
| | - Daniel P Mulvihill
- School of Biosciences, University of Kent, Giles Lane, Canterbury, Kent CT2 7NJ, UK
| | - Janni Petersen
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, SA 5001, Australia South Australia Health and Medical Research Institute, North Terrace, PO Box 11060, Adelaide, SA 5000, Australia
| |
Collapse
|
30
|
Xia Y, Chen J, Gong C, Chen H, Sun J. α-Mangostin, a Natural Agent, Enhances the Response of NRAS Mutant Melanoma to Retinoic Acid. Med Sci Monit 2016; 22:1360-7. [PMID: 27104669 PMCID: PMC4844330 DOI: 10.12659/msm.898204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The identification and use of novel compounds alone or in combination hold promise for the fight against NRAS mutant melanoma. MATERIAL AND METHODS We screened a kinase-specific inhibitor library through combining it with α-Mangostin in NRAS mutant melanoma cell line, and verified the enhancing effect of α-Mangostin through inhibition of the tumorigenesis pathway. RESULTS Within the kinase inhibitors, retinoic acid showed a significant synergistic effect with α-Mangostin. α-Mangostin also can reverse the drug resistance of retinoic acid in RARa siRNA-transduced sk-mel-2 cells. Colony assay, TUNEL staining, and the expressions of several apoptosis-related genes revealed that a-Mangostin enhanced the effect of retinoic acid-induced apoptosis. The combination treatment resulted in marked induction of ROS generation and inhibition of the AKT/S6 pathway. CONCLUSIONS These results indicate that the combination of these novel natural agents with retinoid acid may be clinically effective in NRAS mutant melanoma.
Collapse
Affiliation(s)
- Yun Xia
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Jing Chen
- Department of Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Chongwen Gong
- Department of Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Hongxiang Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| |
Collapse
|
31
|
Ma Y, Ma N, Liu Q, Qi Y, Manabe RI, Furuyashiki T. Tor Signaling Regulates Transcription of Amino Acid Permeases through a GATA Transcription Factor Gaf1 in Fission Yeast. PLoS One 2015; 10:e0144677. [PMID: 26689777 PMCID: PMC4686964 DOI: 10.1371/journal.pone.0144677] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/20/2015] [Indexed: 01/17/2023] Open
Abstract
In the fission yeast, two Tor isoforms, Tor1 and Tor2, oppositely regulate gene expression of amino acid permeases. To elucidate the transcriptional machinery for these regulations, here we have employed the cap analysis of gene expression (CAGE), a method of analyzing expression profiles and identifying transcriptional start sites (TSSs). The loss of Tor1 decreased, and Tor2 inhibition by its temperature sensitive mutation increased, mRNA expression of isp5+, per1+, put4+ and SPBPB2B2.01. In contrast, the loss of Tor1 increased, and Tor2 inhibition decreased, the expression of cat1+. These changes were confirmed by semi-quantitative RT-PCR. These opposite effects by the loss of Tor1 and Tor2 inhibition appeared to occur evenly across multiple TSSs for the respective genes. The motif discovery analysis based on the CAGE results identified the GATA motifs as a potential cis-regulatory element for Tor-mediated regulation. In the luciferase reporter assay, the loss of Tor1 reduced, and Tor2 inhibition and nitrogen depletion increased, the activity of isp5+ promoter as well as that of a GATAAG reporter. One of the GATAAG motifs in isp5+ promoter was critical for its transcriptional activity, and a GATA transcription factor Gaf1 was critical for the activities of isp5+ promoter and the GATAAG reporter. Furthermore, Tor2 inhibition and nitrogen depletion induced nuclear localization of Gaf1 from the cytosol and its dephosphorylation. These results suggest that Tor2 inhibition, which is known to be induced by nitrogen depletion, promotes nuclear localization of Gaf1, thereby inducing isp5+ transcription through Gaf1 binding to the GATAAG motif in its promoter. Since Gaf1 was also critical for transcription of per1+ and put4+, Tor-Gaf1 signaling may coordinate transcription of multiple amino acid permeases according to nutrient availability.
Collapse
Affiliation(s)
- Yan Ma
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
- * E-mail:
| | - Ning Ma
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Qingbin Liu
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yao Qi
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ri-ichiroh Manabe
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Japan
| | - Tomoyuki Furuyashiki
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
32
|
Liu Q, Ma Y, Zhou X, Furuyashiki T. Constitutive Tor2 Activity Promotes Retention of the Amino Acid Transporter Agp3 at Trans-Golgi/Endosomes in Fission Yeast. PLoS One 2015; 10:e0139045. [PMID: 26447710 PMCID: PMC4598100 DOI: 10.1371/journal.pone.0139045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/07/2015] [Indexed: 12/13/2022] Open
Abstract
Amino acid transporters are located at specific subcellular compartments, and their localizations are regulated by the extracellular availability of amino acids. In yeast, target of rapamycin (TOR) activation induces the internalization of amino acid transporters located at the plasma membrane. However, whether and how TOR signaling regulates other amino acid transporters located at intracellular compartments remains unknown. Here, we demonstrate that in the fission yeast, the TOR inhibitor Torin-1 induces the transfer of several yellow fluorescent protein (YFP)-fused intracellular amino acid transporters, including Agp3, Isp5, Aat1, and Put4, from trans-Golgi/endosomes into the vacuoles. By contrast, the localizations of YFP-fused Can1, Fnx1, and Fnx2 transporter proteins were unaffected upon Torin-1 treatment. There are two TOR isoforms in fission yeast, Tor1 and Tor2. Whereas tor1 deletion did not affect the Torin-1-induced transfer of Agp3-YFP, Tor2 inhibition using a temperature-sensitive mutant induced the transfer of Agp3-YFP to the vacuolar lumen, similar to the effects of Torin-1 treatment. Tor2 inhibition also induced the transfer of the YFP-fused Isp5, Aat1, and Put4 transporter proteins to the vacuoles, although only partial transfer of the latter two transporters was observed. Under nitrogen depletion accompanied by reduced Tor2 activity, Agp3-YFP was transferred from the trans-Golgi/endosomes to the plasma membrane and then to the vacuoles, where it was degraded by the vacuolar proteases Isp6 and Psp3. Mutants with constitutively active Tor2 showed delayed transfer of Agp3-YFP to the plasma membrane upon nitrogen depletion. Cells lacking Tsc2, a negative regulator of Tor2, also showed a delay in this process in a Tor2-dependent manner. Taken together, these findings suggest that constitutive Tor2 activity is critical for the retention of amino acid transporters at trans-Golgi/endosomes. Moreover, nitrogen depletion suppresses Tor2 activity through Tsc2, thereby promoting the surface expression of these transporters.
Collapse
Affiliation(s)
- Qingbin Liu
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yan Ma
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
- * E-mail:
| | - Xin Zhou
- Department of Oncology, the First Affiliated Hospital of Liaoning Medical University, Jinzhou, China
| | - Tomoyuki Furuyashiki
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
33
|
Abstract
In this article, we will discuss the biochemistry of mitosis in eukaryotic cells. We will focus on conserved principles that, importantly, are adapted to the biology of the organism. It is vital to bear in mind that the structural requirements for division in a rapidly dividing syncytial Drosophila embryo, for example, are markedly different from those in a unicellular yeast cell. Nevertheless, division in both systems is driven by conserved modules of antagonistic protein kinases and phosphatases, underpinned by ubiquitin-mediated proteolysis, which create molecular switches to drive each stage of division forward. These conserved control modules combine with the self-organizing properties of the subcellular architecture to meet the specific needs of the cell. Our discussion will draw on discoveries in several model systems that have been important in the long history of research on mitosis, and we will try to point out those principles that appear to apply to all cells, compared with those in which the biochemistry has been specifically adapted in a particular organism.
Collapse
Affiliation(s)
- Samuel Wieser
- The Gurdon Institute, Cambridge CB2 1QN, United Kingdom
| | | |
Collapse
|
34
|
Davie E, Forte GMA, Petersen J. Nitrogen regulates AMPK to control TORC1 signaling. Curr Biol 2015; 25:445-54. [PMID: 25639242 PMCID: PMC4331286 DOI: 10.1016/j.cub.2014.12.034] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 11/06/2014] [Accepted: 12/10/2014] [Indexed: 10/29/2022]
Abstract
BACKGROUND Cell growth and cell-cycle progression are tightly coordinated to enable cells to adjust their size (timing of division) to the demands of proliferation in varying nutritional environments. In fission yeast, nitrogen stress results in sustained proliferation at a reduced size. RESULTS Here, we show that cells can sense nitrogen stress to reduce target of rapamycin complex-1 (TORC1) activity. Nitrogen-stress-induced TORC1 inhibition differs from amino-acid-dependent control of TORC1 and requires the Ssp2 (AMPKα) kinase, the Tsc1/2 complex, and Rhb1 GTPase. Importantly, the β and γ regulatory subunits of AMPK are not required to control cell division in response to nitrogen stress, providing evidence for a nitrogen-sensing mechanism that is independent of changes in intracellular ATP/AMP levels. The CaMKK homolog Ssp1 is constitutively required for phosphorylation of the AMPKα(Ssp2) T loop. However, we find that a second homolog CaMKK(Ppk34) is specifically required to stimulate AMPKα(Ssp2) activation in response to nitrogen stress. Finally, ammonia also controls mTORC1 activity in human cells; mTORC1 is activated upon the addition of ammonium to glutamine-starved Hep3B cancer cells. CONCLUSIONS The alternative nitrogen source ammonia can simulate TORC1 activity to support growth and division under challenging nutrient settings, a situation often seen in cancer.
Collapse
Affiliation(s)
- Elizabeth Davie
- Faculty of Life Sciences, University of Manchester, C.4255 Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Gabriella M A Forte
- Faculty of Life Sciences, University of Manchester, C.4255 Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Janni Petersen
- Faculty of Life Sciences, University of Manchester, C.4255 Michael Smith Building, Oxford Road, Manchester M13 9PT, UK; Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, SA 5001, Australia.
| |
Collapse
|