1
|
Abstract
Rapamycin inhibits cell proliferation, yet preserves (re)-proliferative potential (RPP). RPP is a potential of quiescent cells that is lost in senescent cells. mTOR drives conversion from quiescence to senescence (geroconversion). By suppressing geroconversion, rapamycin preserves RPP. Geroconversion is characterized by proliferation-like levels of phospho-S6K/S6/4E-BP1 in nonproliferating cells arrested by p16 and/or p21. mTOR-driven geroconversion is associated with cellular hyperfunction, which in turn leads to organismal aging manifested by age-related diseases.
Collapse
|
2
|
Kisko M, Bouain N, Safi A, Medici A, Akkers RC, Secco D, Fouret G, Krouk G, Aarts MGM, Busch W, Rouached H. LPCAT1 controls phosphate homeostasis in a zinc-dependent manner. eLife 2018; 7:e32077. [PMID: 29453864 PMCID: PMC5826268 DOI: 10.7554/elife.32077] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 02/15/2018] [Indexed: 12/25/2022] Open
Abstract
All living organisms require a variety of essential elements for their basic biological functions. While the homeostasis of nutrients is highly intertwined, the molecular and genetic mechanisms of these dependencies remain poorly understood. Here, we report a discovery of a molecular pathway that controls phosphate (Pi) accumulation in plants under Zn deficiency. Using genome-wide association studies, we first identified allelic variation of the Lyso-PhosphatidylCholine (PC) AcylTransferase 1 (LPCAT1) gene as the key determinant of shoot Pi accumulation under Zn deficiency. We then show that regulatory variation at the LPCAT1 locus contributes significantly to this natural variation and we further demonstrate that the regulation of LPCAT1 expression involves bZIP23 TF, for which we identified a new binding site sequence. Finally, we show that in Zn deficient conditions loss of function of LPCAT1 increases the phospholipid Lyso-PhosphatidylCholine/PhosphatidylCholine ratio, the expression of the Pi transporter PHT1;1, and that this leads to shoot Pi accumulation.
Collapse
Affiliation(s)
- Mushtak Kisko
- BPMP, Univ Montpellier, CNRS, INRA, SupAgroMontpellierFrance
| | - Nadia Bouain
- BPMP, Univ Montpellier, CNRS, INRA, SupAgroMontpellierFrance
| | - Alaeddine Safi
- BPMP, Univ Montpellier, CNRS, INRA, SupAgroMontpellierFrance
| | - Anna Medici
- BPMP, Univ Montpellier, CNRS, INRA, SupAgroMontpellierFrance
| | - Robert C Akkers
- Laboratory of GeneticsWageningen UniversityWageningenNetherlands
| | - David Secco
- BPMP, Univ Montpellier, CNRS, INRA, SupAgroMontpellierFrance
| | | | - Gabriel Krouk
- BPMP, Univ Montpellier, CNRS, INRA, SupAgroMontpellierFrance
| | - Mark GM Aarts
- Laboratory of GeneticsWageningen UniversityWageningenNetherlands
| | - Wolfgang Busch
- Gregor Mendel InstituteAustrian Academy of Sciences, Vienna BiocenterViennaAustria
- Plant Molecular and Cellular Biology LaboratorySalk Institute for Biological StudiesLa JollaUnited States
| | - Hatem Rouached
- BPMP, Univ Montpellier, CNRS, INRA, SupAgroMontpellierFrance
| |
Collapse
|
3
|
Kisko M, Bouain N, Safi A, Medici A, Akkers RC, Secco D, Fouret G, Krouk G, Aarts MG, Busch W, Rouached H. LPCAT1 controls phosphate homeostasis in a zinc-dependent manner. eLife 2018; 7:32077. [PMID: 29453864 DOI: 10.7554/elife.32077.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 02/15/2018] [Indexed: 05/22/2023] Open
Abstract
All living organisms require a variety of essential elements for their basic biological functions. While the homeostasis of nutrients is highly intertwined, the molecular and genetic mechanisms of these dependencies remain poorly understood. Here, we report a discovery of a molecular pathway that controls phosphate (Pi) accumulation in plants under Zn deficiency. Using genome-wide association studies, we first identified allelic variation of the Lyso-PhosphatidylCholine (PC) AcylTransferase 1 (LPCAT1) gene as the key determinant of shoot Pi accumulation under Zn deficiency. We then show that regulatory variation at the LPCAT1 locus contributes significantly to this natural variation and we further demonstrate that the regulation of LPCAT1 expression involves bZIP23 TF, for which we identified a new binding site sequence. Finally, we show that in Zn deficient conditions loss of function of LPCAT1 increases the phospholipid Lyso-PhosphatidylCholine/PhosphatidylCholine ratio, the expression of the Pi transporter PHT1;1, and that this leads to shoot Pi accumulation.
Collapse
Affiliation(s)
- Mushtak Kisko
- BPMP, Univ Montpellier, CNRS, INRA, SupAgro, Montpellier, France
| | - Nadia Bouain
- BPMP, Univ Montpellier, CNRS, INRA, SupAgro, Montpellier, France
| | - Alaeddine Safi
- BPMP, Univ Montpellier, CNRS, INRA, SupAgro, Montpellier, France
| | - Anna Medici
- BPMP, Univ Montpellier, CNRS, INRA, SupAgro, Montpellier, France
| | - Robert C Akkers
- Laboratory of Genetics, Wageningen University, Wageningen, Netherlands
| | - David Secco
- BPMP, Univ Montpellier, CNRS, INRA, SupAgro, Montpellier, France
| | | | - Gabriel Krouk
- BPMP, Univ Montpellier, CNRS, INRA, SupAgro, Montpellier, France
| | - Mark Gm Aarts
- Laboratory of Genetics, Wageningen University, Wageningen, Netherlands
| | - Wolfgang Busch
- Gregor Mendel Institute, Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
- Plant Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, United States
| | - Hatem Rouached
- BPMP, Univ Montpellier, CNRS, INRA, SupAgro, Montpellier, France
| |
Collapse
|
4
|
Soliman MA, Berardi P, Pastyryeva S, Bonnefin P, Feng X, Colina A, Young D, Riabowol K. ING1a expression increases during replicative senescence and induces a senescent phenotype. Aging Cell 2008; 7:783-94. [PMID: 18691180 DOI: 10.1111/j.1474-9726.2008.00427.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The ING family of tumor suppressor proteins affects cell growth, apoptosis and response to DNA damage by modulating chromatin structure through association with different HAT and HDAC complexes. The major splicing isoforms of the ING1 locus are ING1a and INGlb. While INGlb plays a role in inducing apoptosis, the function of ING1a is currently unknown. Here we show that alternative splicing of the ING1 message alters the INGla:INGlb ratio by approximately 30-fold in senescent compared to low passage primary fibroblasts. INGla antagonizes INGlb function in apoptosis, induces the formation of structures resembling senescence-associated heterochromatic foci containing heterochromatin protein 1 gamma, the accumulation of senescence-associated beta-galactosidase activity and promotes senescent cell morphology and cell cycle arrest. Phenotypic effects may result from differential effects on gene expression since ING1a increases levels of both retinoblastoma and the p16 cyclin-dependent kinase inhibitor and ING1a and ING1b have opposite effects on the expression of proliferating nuclear cell antigen (PCNA), which is required for cell growth. Gene expression appears to be altered by targeting of HDAC complexes to gene promoters since INGla associates with several-fold higher levels of HDAC1 in senescent, compared to replication-competent cells and ING1 is found on the PCNA promoter by chromatin immunoprecipitation analysis. These data demonstrate a novel role for the ING1 proteins in differentially regulating senescence-associated chromatin remodeling vs. apoptosis and support the idea that altered ratios of the ING1 splicing isoforms may contribute to establishing the senescent phenotype through HDAC and HAT complex-mediated effects on chromatin structure.
Collapse
Affiliation(s)
- Mohamed A Soliman
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Chrétien A, Piront N, Delaive E, Demazy C, Ninane N, Toussaint O. Increased abundance of cytoplasmic and nuclear caveolin 1 in human diploid fibroblasts in H2O2-induced premature senescence and interplay with p38αMAPK. FEBS Lett 2008; 582:1685-92. [DOI: 10.1016/j.febslet.2008.04.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Accepted: 04/15/2008] [Indexed: 11/24/2022]
|
6
|
Chen GL, Vallender EJ, Miller GM. Functional characterization of the human TPH2 5' regulatory region: untranslated region and polymorphisms modulate gene expression in vitro. Hum Genet 2007; 122:645-57. [PMID: 17972101 DOI: 10.1007/s00439-007-0443-y] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Accepted: 10/23/2007] [Indexed: 12/19/2022]
Abstract
Tryptophan hydroxylase-2 (TPH2) is a recently identified TPH isoform responsible for neuronal serotonin (5-HT) synthesis, and TPH2 polymorphisms are associated with a range of behavioral traits and psychiatric disorders. This study characterized cis-acting elements and three common polymorphisms (-703G/T, -473T/A, and 90A/G) in the 5' regulatory region of human TPH2 by using luciferase reporter assay, quantitative real-time PCR, and electrophoretic mobility shift assay (EMSA). The core promoter of human TPH2 was localized to the region between -107 and +7, and the segment of +8 to +53 within the 5'-UTR was found to exert a potent inhibitory effect on gene expression at both transcriptional and post-transcriptional levels. In both RN46A and HEK-293 cell lines, the TTA (-703T/-473T/90A) haplotype of the three polymorphisms showed the lowest gene expression compared with other haplotypes, and the -703G/T and -473T/A polymorphisms tended to exert a synergic effect on gene expression dependent upon the sequence of the 5'-UTR. In RN46A, the 90A/G polymorphism significantly increased luciferase activity and mRNA level irrespective of the other two polymorphisms, while in HEK-293 cells the effect of 90A/G was dependent on the alleles at loci -703 and -473. EMSA showed that all the three polymorphisms potentially alter DNA-protein interactions, while the 90A/G polymorphism predictably alters the 5'-UTR secondary structure of mRNA and influences RNA-protein interactions. In conclusion, our present study demonstrates that both the 5'-UTR and common polymorphisms (especially the 90A/G) in the 5' regulatory region of human TPH2 have a significant impact on gene expression.
Collapse
Affiliation(s)
- Guo-Lin Chen
- New England Primate Research Center, Division of Neurochemistry, Harvard Medical School, One Pine Hill Drive, Southborough, MA, 01772-9102, USA.
| | | | | |
Collapse
|
7
|
Lu XF, Jiang XG, Lu YB, Bai JH, Mao ZB. Characterization of a novel positive transcription regulatory element that differentially regulates the insulin-like growth factor binding protein-3 (IGFBP-3) gene in senescent cells. J Biol Chem 2005; 280:22606-15. [PMID: 15817480 DOI: 10.1074/jbc.m412073200] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Insulin-like growth factor binding protein-3 (IGFBP-3) is a well documented growth inhibitor and pro-apoptotic factor. IGFBP-3 mRNA and its protein are overexpressed by senescent human diploid fibroblasts. However, the mechanism responsible for the up-regulation of its expression is still unclear. This report describes a novel transcriptional regulatory element, IGFBP-3 enhancer element (IEE), identified in the 5' untranslated region of the IGFBP-3 gene. This element differentially activates IGFBP-3 expression in senescent versus young fibroblasts. Electrophoretic mobility shift assays revealed abundant complexes in senescent cell nuclear extracts compared with young cell nuclear extracts. Similar to young proliferative cells, young quiescent cells showed reduced binding activity; enhancement of this activity was specific to senescent cells and not an effect of cell cycle arrest. The DNase I footprint revealed the protein-binding core sequence within the IEE through which the protein binds the IEE. Site-directed mutagenesis within IEE abolished binding activity and selectively decreased IGFBP-3 promoter activity in senescent (but not young) cells. Furthermore, introduction of an IEE decoy suppressed the endogenous IGFBP-3 gene expression specifically in senescent cells. These results point to the IEE as being a positive transcription regulatory element that contributes to the up-regulation of IGFBP-3 during cellular senescence.
Collapse
MESH Headings
- 5' Untranslated Regions
- Base Sequence
- Blotting, Northern
- Cell Nucleus/metabolism
- Cell Proliferation
- Cells, Cultured
- Cellular Senescence
- Cloning, Molecular
- Cross-Linking Reagents/pharmacology
- Deoxyribonuclease I/metabolism
- Down-Regulation
- Enhancer Elements, Genetic
- Fibroblasts/metabolism
- Gene Deletion
- Gene Expression Regulation
- Genes, Regulator
- Genes, Reporter
- Humans
- Insulin-Like Growth Factor Binding Protein 3/biosynthesis
- Insulin-Like Growth Factor Binding Protein 3/genetics
- Luciferases/metabolism
- Models, Genetic
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Mutation
- Oligonucleotides/chemistry
- Promoter Regions, Genetic
- Protein Binding
- RNA, Messenger/metabolism
- Transfection
- Ultraviolet Rays
- Up-Regulation
Collapse
Affiliation(s)
- Xiao Feng Lu
- Department of Biochemistry and Molecular Biology, Health Science Center, Peking University, Beijing, China
| | | | | | | | | |
Collapse
|
8
|
Scott IS, Heath TM, Morris LS, Rushbrook SM, Bird K, Vowler SL, Arends MJ, Coleman N. A novel immunohistochemical method for estimating cell cycle phase distribution in ovarian serous neoplasms: implications for the histopathological assessment of paraffin-embedded specimens. Br J Cancer 2004; 90:1583-90. [PMID: 15083189 PMCID: PMC2409706 DOI: 10.1038/sj.bjc.6601660] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We have investigated whether immunohistochemical markers can identify differences in cell cycle phase distribution in ovarian serous neoplasms, including borderline tumours of different grades. Sections of normal ovary (n=18), serous cystadenoma (n=21), borderline serous tumours (n=21) and serous cystadenocarcinoma (n=15) were analysed by immunohistochemistry using markers of cell cycle entry (Mcm-2) and cell cycle phase, including cyclin D1 (mid-to-late G1), cyclin A (S phase), cyclin B1 (G2 phase) and phosphohistone H3 (mitosis). Double-labelling confocal microscopy confirmed marker phase specificity and phase estimations were corroborated by flow cytometry. On progression from normal ovary through serous cystadenoma and borderline tumours to cystadenocarcinomas, expression of Mcm-2 (P<0.0001), cyclin D1 (P=0.002), cyclin A (P<0.0001), cyclin B1 (P<0.0001) and phosphohistone H3 (P<0.0001) increased, paralleled by an increase in the S-phase fraction (cyclin A : Mcm-2 ratio; P=0.002). Borderline tumours of increasing grade also showed increased Mcm-2 and cyclin A expression, together with an increase in the S-phase fraction. Immunohistochemistry can be used to estimate cell cycle phase distribution in ovarian serous neoplasms, giving results similar to flow cytometric analysis and enabling direct assessment of tumour heterogeneity. Immunohistochemical estimates of the S-phase fraction may identify serous borderline tumours likely to exhibit malignant progression and/or select serous cystadenocarcinomas likely to respond to adjuvant therapy.
Collapse
Affiliation(s)
- I S Scott
- MRC Cancer Cell Unit, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 2XZ, UK.
| | | | | | | | | | | | | | | |
Collapse
|