Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
2.
Mol Ther Nucleic Acids ; 2: e141, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24399205

ABSTRACT

Selective gene silencing by RNA interference (RNAi) involves double-stranded small interfering RNA (ds siRNA) composed of single-stranded (ss) guide and passenger RNAs. siRNA is recognized and processed by Ago2 and C3PO, endonucleases of the RNA-induced silencing complex (RISC). RISC cleaves passenger RNA, exposing the guide RNA for base-pairing with its homologous mRNA target. Remarkably, the 3' end of passenger RNA can accommodate a DNA extension of 19-nucleotides without loss of RNAi function. This construct is termed passenger-3'-DNA/ds siRNA and includes a 3'-nuclease-resistant mini-hairpin structure. To test this novel modification further, we have now compared the following constructs: (I) guide-3'-DNA/ds siRNA, (II) passenger-3'-DNA/ds siRNA, (III) guide-3'-DNA/ss siRNA, and (IV) passenger-3'-DNA/ss siRNA. The RNAi target was SIRT1, a cancer-specific survival factor. Constructs I-III each induced selective knock-down of SIRT1 mRNA and protein in both noncancer and cancer cells, accompanied by apoptotic cell death in the cancer cells. Construct IV, which lacks the SIRT1 guide strand, had no effect. Importantly, the 3'-DNA mini-hairpin conferred nuclease resistance to constructs I and II. Resistance required the double-stranded RNA structure since single-stranded guide-3'-DNA/ss siRNA (construct III) was susceptible to serum nucleases with associated loss of RNAi activity. The potential applications of 3'-DNA/siRNA constructs are discussed.Molecular Therapy-Nucleic Acids (2014) 2, e141; doi:10.1038/mtna.2013.68; published online 7 January 2014.

3.
Open Biol ; 3(11): 130130, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24258275

ABSTRACT

The NAD(+)-dependent deacetylase SIRT1 is involved in diverse cellular processes, and has also been linked with multiple disease states. Among these, SIRT1 expression negatively correlates with cancer survival in both laboratory and clinical studies. Active regulator of SIRT1 (AROS) was the first reported post-transcriptional regulator of SIRT1 activity, enhancing SIRT1-mediated deacetylation and downregulation of the SIRT1 target p53. However, little is known regarding the role of AROS in regulation of SIRT1 during disease. Here, we report the cellular and molecular effects of RNAi-mediated AROS suppression, comparing this with the role of SIRT1 in a panel of human cell lines of both cancerous and non-cancerous origins. Unexpectedly, AROS is found to vary in its modulation of p53 acetylation according to cell context. AROS suppresses p53 acetylation only following the application of cell damaging stress, whereas SIRT1 suppresses p53 under all conditions analysed. This supplements the original characterization of AROS but indicates that SIRT1 activity can persist following suppression of AROS. We also demonstrate that knockdown of AROS induces apoptosis in three cancer cell lines, independent of p53 activation. Importantly, AROS is not required for the viability of three non-cancer cell lines indicating a putative role for AROS in specifically promoting cancer cell survival.


Subject(s)
Cell Survival , Neoplasms/drug therapy , Nuclear Proteins/physiology , RNA Interference , Sirtuin 1/physiology , Transcription Factors/physiology , Acetylation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HCT116 Cells , Humans , MCF-7 Cells , Neoplasms/genetics , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
4.
Nucleic Acids Res ; 41(7): 4185-97, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23462953

ABSTRACT

Active regulator of SIRT1 (AROS) binds and upregulates SIRT1, an NAD(+)-dependent deacetylase. In addition, AROS binds RPS19, a structural ribosomal protein, which also functions in ribosome biogenesis and is implicated in multiple disease states. The significance of AROS in relation to ribosome biogenesis and function is unknown. Using human cells, we now show that AROS localizes to (i) the nucleolus and (ii) cytoplasmic ribosomes. Co-localization with nucleolar proteins was verified by confocal immunofluorescence of endogenous protein and confirmed by AROS depletion using RNAi. AROS association with cytoplasmic ribosomes was analysed by sucrose density fractionation and immunoprecipitation, revealing that AROS selectively associates with 40S ribosomal subunits and also with polysomes. RNAi-mediated depletion of AROS leads to deficient ribosome biogenesis with aberrant precursor ribosomal RNA processing, reduced 40S subunit ribosomal RNA and 40S ribosomal proteins (including RPS19). Together, this results in a reduction in 40S subunits and translating polysomes, correlating with reduced overall cellular protein synthesis. Interestingly, knockdown of AROS also results in a functionally significant increase in eIF2α phosphorylation. Overall, our results identify AROS as a factor with a role in both ribosome biogenesis and ribosomal function.


Subject(s)
Nuclear Proteins/physiology , Protein Biosynthesis , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Transcription Factors/physiology , Amino Acid Sequence , Cell Line, Tumor , Cell Nucleolus/chemistry , Cytoplasm/chemistry , Eukaryotic Initiation Factor-2/metabolism , Humans , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA, Ribosomal/metabolism , Ribosome Subunits, Small, Eukaryotic/metabolism , Ribosomes/chemistry , Transcription Factors/chemistry , Transcription Factors/metabolism
5.
Curr Opin Oncol ; 24(1): 68-75, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22080944

ABSTRACT

PURPOSE OF REVIEW: SIRT1 impacts upon diverse cellular processes via its roles in the determination of chromatin structure, chromatin remodelling and gene expression. This review covers the recent discoveries linking SIRT1 with the regulation of mammalian metabolism and considers ways in which abnormal metabolism in disease may, in turn, impact upon SIRT1 because of SIRT1's functional dependency upon NAD. RECENT FINDINGS: Diverse signalling pathways are integrated to regulate energy metabolism and homeostasis. Such pathways involve intracellular networks and mitochondria, and also intercellular signalling within and between tissues to co-ordinate adaptive metabolic responses within the organism as a whole. Here, we outline the recent studies exploring the regulatory links between SIRT1 and mitochondrial biogenesis, cellular redox and associated metabolic pathways, and angiogenesis/Notch signalling. These links are effected by the SIRT1-mediated deacetylation of transcriptional regulators and enzymes with key roles in metabolism. SUMMARY: SIRT1 activity is directly coupled with homeostasis and metabolism. SIRT1 is also a metabolic sensor. It follows that disease-related metabolic abnormalities are likely to impinge upon SIRT1 functioning. Disease-related functions of SIRT1, in their turn, offer potential targets for the development of novel SIRT1-based therapies. In cancer, for example, the survival function of SIRT1 may reflect abnormal cancer metabolism and identifies SIRT1 as a target for anticancer therapy.


Subject(s)
Energy Metabolism/physiology , Neoplasms/metabolism , Sirtuin 1/metabolism , Cell Hypoxia/physiology , Glycolysis/physiology , Humans , Isoenzymes/metabolism , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5 , Mitochondria/metabolism , NAD/metabolism , Poly(ADP-ribose) Polymerases/metabolism
6.
Mol Cell Biol ; 32(3): 704-16, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22124156

ABSTRACT

SIRT1 is an NAD-dependent deacetylase and epigenetic regulator essential for normal mammalian development and homeostasis. Here we describe a human SIRT1 splice variant, designated SIRT1-Δ2/9, in which the deacetylase coding sequence is lost due to splicing between exons 2 and 9. This work aimed to determine if SIRT1-Δ2/9 is a novel functional product of the SIRT1 gene. Endogenous SIRT1-Δ2/9 protein was identified in human cell lysate by immunoblotting and splice variant-specific RNA interference (RNAi). SIRT1-Δ2/9 mRNA is bound by CUGBP2, which downregulates its translation. Using pulldown assays, we demonstrate that SIRT1-Δ2/9 binds p53 protein. SIRT1-Δ2/9 maintains basal p53 protein levels and supports p53 function in response to DNA damage, as evidenced by RNAi-mediated depletion of SIRT1-Δ2/9 prior to damage. In turn, basal p53 downregulates SIRT1-Δ2/9 RNA levels, while stress-activated p53 eliminates SIRT1-Δ2/9. Loss of wild-type (wt) p53 has been correlated with overexpression of SIRT1-Δ2/9 in a range of human cancers. Exogenous SIRT1-Δ2/9 protein associates with specific promoters in chromatin and can regulate cancer-related gene expression, as evidenced by chromatin immunoprecipitation analysis and RNAi/genomic array data. SIRT1 is of major therapeutic importance, and potential therapeutic drugs are screened against SIRT1 deacetylase activity. Our discovery of SIRT1-Δ2/9 identifies a new, deacetylase-independent therapeutic target for SIRT1-related diseases, including cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Sirtuin 1/genetics , Tumor Suppressor Protein p53/metabolism , Alternative Splicing , Cell Line, Tumor , DNA Damage , Gene Deletion , Humans , Neoplasms/metabolism , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/metabolism , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/genetics
7.
Cancer Biol Ther ; 12(12): 1059-68, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22157150

ABSTRACT

The serine/threonine protein kinase Aurora A is known to interact with and phosphorylate tumor suppressor p53 at Serine 215 (S215), inhibiting the transcriptional activity of p53. We show that Aurora A positively regulates human p53 protein levels and, using isogenic p53 wild-type and p53-null colorectal carcinoma cells, further show that p53 regulates human Aurora A protein expression. S215 is located in the DNA-binding core of p53 and at the center of the cryptic epitope for PAb240 antibody, which is used to detect mutant and denatured p53. Following denaturing SDS PAGE, the PAb240 epitope was detectable by immunoblotting in only two out of eight cell lines. The efficacy of novel p53-targeted anticancer therapies may be influenced by the conformational state of p53, therefore, the initial determination of p53 status may be relevant. We found no correlation between phosphorylation of p53 at S215 and PAb240 antibody recognition. However, phosphorylation at S37 was positively associated with PAb240 reactivity. More importantly, we provide the first evidence of Aurora A-mediated cross-talk between N- and C-terminal p53 post-translational modifications. As p53 and Aurora A are targets for anticancer therapy the impact of their reciprocal relationship and Aurora A-induced post-translational modification of p53 should be considered.


Subject(s)
Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Antibodies, Monoclonal/immunology , Antibody Specificity , Aurora Kinases , Cell Line, Tumor , Epitopes/immunology , Epitopes/metabolism , HCT116 Cells , Humans , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transcriptional Activation , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
8.
9.
PLoS One ; 5(10): e13502, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20975832

ABSTRACT

BACKGROUND: The NAD-dependent deacetylase SIRT1 is a nutrient-sensitive coordinator of stress-tolerance, multiple homeostatic processes and healthspan, while p53 is a stress-responsive transcription factor and our paramount tumour suppressor. Thus, SIRT1-mediated inhibition of p53 has been identified as a key node in the common biology of cancer, metabolism, development and ageing. However, precisely how SIRT1 integrates such diverse processes remains to be elucidated. METHODOLOGY/PRINCIPAL FINDINGS: Here we report that SIRT1 is alternatively spliced in mammals, generating a novel SIRT1 isoform: SIRT1-ΔExon8. We show that SIRT1-ΔExon8 is expressed widely throughout normal human and mouse tissues, suggesting evolutionary conservation and critical function. Further studies demonstrate that the SIRT1-ΔExon8 isoform retains minimal deacetylase activity and exhibits distinct stress sensitivity, RNA/protein stability, and protein-protein interactions compared to classical SIRT1-Full-Length (SIRT1-FL). We also identify an auto-regulatory loop whereby SIRT1-ΔExon8 can regulate p53, while in reciprocal p53 can influence SIRT1 splice variation. CONCLUSIONS/SIGNIFICANCE: We characterize the first alternative isoform of SIRT1 and demonstrate its evolutionary conservation in mammalian tissues. The results also reveal a new level of inter-dependency between p53 and SIRT1, two master regulators of multiple phenomena. Thus, previously-attributed SIRT1 functions may in fact be distributed between SIRT1 isoforms, with important implications for SIRT1 functional studies and the current search for SIRT1-activating therapeutics to combat age-related decline.


Subject(s)
Alternative Splicing , Sirtuin 1/physiology , Tumor Suppressor Protein p53/physiology , Acetylation , Animals , Exons , Humans , Mice , Reverse Transcriptase Polymerase Chain Reaction , Sirtuin 1/genetics
10.
J Neurooncol ; 97(3): 373-82, 2010 May.
Article in English | MEDLINE | ID: mdl-19907922

ABSTRACT

Medulloblastomas are highly malignant, poorly differentiated childhood tumours arising in the cerebellum. These tumors rarely lose TP53, which is the most commonly mutated gene in cancer. Recent work has shown that the basal level of p53 plays an important role in maternal reproduction by maintaining the expression of LIF in the uterus. Since LIF can maintain the undifferentiated state of stem cells we set out to ask if p53 regulates LIF in the human medulloblastoma cell lines DAOY and D283MED. We also used p53-/- and p53+/+ isogenic HCT116 colorectal carcinoma cell lines, already reported to exhibit p53-dependent expression of the LIF D transcript, to establish the extent of p53-dependency for LIF M and T alternative transcripts. Whilst all three known, full-length alternative transcripts are more abundant in p53+/+ cells, the alternative LIF M and T transcripts appear particularly sensitive to p53. In the p53 wild-type medulloblastoma cell line D283MED chromatin immunoprecipitation experiments showed p53 binding to the LIF gene. The mutant p53 expressed in line DAOY did not bind to this region or to the p21(WAF1) p53 binding site. RNA interference against either WIP1 or SIRT1 stabilized p53 and enhanced the transcription of LIF in D283MED cells. Interestingly, siRNA against WIP1 or SIRT1 also induced increased apoptosis in the medulloblastoma line D283MED and, over a longer time period, in DAOY cells. We speculate that suppression of p53 function by combined WIP1-mediated dephosphorylation and SIRT1 deacetylation enables medulloblastoma cell survival but p53-dependent and independent apoptotic pathways remain intact. Thus small molecule inhibitors of SIRT1 may be useful in treatment of medulloblastoma.


Subject(s)
Apoptosis/physiology , Gene Expression Regulation, Neoplastic/physiology , Leukemia Inhibitory Factor/metabolism , Medulloblastoma/metabolism , Tumor Suppressor Protein p53/physiology , Apoptosis/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation/methods , Flow Cytometry/methods , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , Phosphoprotein Phosphatases/metabolism , Protein Binding/genetics , Protein Phosphatase 2C , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Transfection/methods , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
11.
PLoS One ; 4(10): e7305, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19806201

ABSTRACT

BACKGROUND: The regulation of apoptosis under basal (non-stress) conditions is crucial for normal mammalian development and also for normal cellular turnover in different tissues throughout life. Deficient regulation of basal apoptosis, or its perturbation, can result in impaired development and/or disease states including cancer. In contrast to stress-induced apoptosis the regulation of apoptosis under basal conditions is poorly understood. To address this issue we have compared basal- and stress-induced apoptosis in human epithelial cells of normal and cancerous origins. For this purpose we focussed our study on the opposing pro-apoptotic JNK/anti-apoptotic NFkappaB pathways. METHODOLOGY/PRINCIPAL FINDINGS: Combinatorial RNAi plus gene knockout were employed to access and map basal regulatory pathways of apoptosis. Follow-on, in-depth analyses included exogenous expression of phosphorylation mutants and chromatin immunoprecipitation. We demonstrate that basal apoptosis is constitutively suppressed by JNK2 in a range of human cancer cell lines. This effect was not observed in non-cancer cells. Silencing JNK2 by RNAi resulted in JNK1-dependent apoptosis of cancer cells via up-regulation of the AP-1 factor c-Jun. Unexpectedly we discovered that JNK1 and c-Jun promote basal apoptosis in the absence of "activating phosphorylations" typically induced by stress. Hypo-phosphorylated c-Jun accumulated to high levels following JNK2 silencing, auto-regulated its own expression and suppressed expression of Bcl-3, an unusual IkappaB protein and regulator of NFkappaB. Basal apoptosis was mediated by components of the TNFalpha response pathway but was mechanistically distinct from TNFalpha-induced apoptosis. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that mechanistically distinct pathways operate to regulate apoptosis in mammalian cells under basal (physiological) versus stress-induced conditions. We also describe a novel apoptotic network which governs the basal survival of cancer cells. Such information is crucial for understanding normal cellular turnover during mammalian development and subsequently throughout life. This information also opens new avenues for therapeutic intervention in human proliferative disease states including cancer.


Subject(s)
Apoptosis , Gene Expression Regulation, Neoplastic , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , B-Cell Lymphoma 3 Protein , Cell Line, Tumor , Cell Survival , Gene Silencing , Humans , NF-kappa B/metabolism , Neoplasms/metabolism , Phosphorylation , RNA Interference , Tumor Necrosis Factor-alpha/metabolism
12.
Curr Pharm Des ; 15(1): 39-44, 2009.
Article in English | MEDLINE | ID: mdl-19149601

ABSTRACT

The intersection between regulatory pathways responsive to metabolic fluctuation on one hand, and to cellular stress on the other, is a fascinating area within which NAD/NADH responsive proteins play a major role [1, 2]. A key player amongst these is SIRT1, a member of the mammalian sirtuin family (SIRT1-7). SIRT1 is an NAD-dependent deacetylase with critical functions in the maintenance of homeostasis and cell survival. In this review I shall focus upon (i) the cellular regulation of SIRT1 expression and (ii) the cellular regulation of SIRT1 activity. In addition the distinction between basal and stress-induced functions will be addressed: do they simply reflect a sliding scale of response, or are they mechanistically distinct? Elevated levels of SIRT1 are evident in cancer and SIRT1 can function as a cancer-specific survival factor in human cell lines. However, in a mouse model SIRT1 is reported to function as a tumour suppressor. Possible explanations for this apparent discrepancy will be considered. Given the high profile of SIRT1 as a potential therapeutic target it is clearly important to clarify its basal functioning in relation to differentiation, cell type, intercellular communication, and to age-related disease states including neurodegeneration and cancer.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Sirtuins/metabolism , Gene Expression Regulation, Neoplastic/physiology , Humans , Neoplasms/metabolism , Sirtuin 1
13.
Aging (Albany NY) ; 1(3): 316-27, 2009 Mar 02.
Article in English | MEDLINE | ID: mdl-20157519

ABSTRACT

Senescence is blocked in human cervical keratinocytes infected with high risk human papillomavirus (e.g. HPV type16). Viral oncoproteins HPV E6 and HPV E7 access the cell cycle via cellular p53 and retinoblastoma proteins respectively. Previously we have shown that HPV E7, not HPV E6, is also responsible for cervical cancer cell survival (SiHa cells; HPV type16). We now present evidence that SIRT1, an aging-related NAD-dependent deacetylase, mediates HPV E7 survival function in SiHa cervical cancer cells. Moreover, HPV E7 up-regulates SIRT1 protein when expressed in primary human keratinocytes. Conversely, SIRT1 levels decrease following RNAi-mediated silencing of HPV E7 in SiHa cells. Silencing HPV E6 has no effect on SIRT1 but, as expected, causes marked accumulation of p53 protein accompanied by p53-mediated up-regulation of p21. However, p53 acetylation (K382Ac) was barely detectable. Since p53 is a known SIRT1 substrate we propose that elevated SIRT1 levels (induced by HPV E7) attenuate p53 pro-apoptotic capacity via its de-acetylation. Our discovery that HPV E7 up-regulates SIRT1 links a clinically important oncogenic virus with the multi-functional SIRT1 protein. This link may open the way for a more in-depth understanding of the process of HPV-induced malignant transformation and also of the inter-relationships between aging and cancer.


Subject(s)
Papillomavirus E7 Proteins/physiology , Sirtuin 1/biosynthesis , Up-Regulation , Uterine Cervical Neoplasms/metabolism , Acetylation , Aging/metabolism , Aurora Kinases , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Gene Expression Regulation , Host-Pathogen Interactions , Human papillomavirus 16/physiology , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/metabolism , Oncogene Proteins, Viral/physiology , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/physiology , Survivin , Tumor Suppressor Protein p53/metabolism , Uterine Cervical Neoplasms/virology
14.
Cell Cycle ; 7(19): 3091-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18838864

ABSTRACT

Mammalian SIRT1 is an NAD-dependent deacetylase with critical roles in the maintenance of homeostasis and cell survival. Elevated levels of SIRT1 protein are evident in cancer in which SIRT1 can function as a cancer-specific survival factor. Here we demonstrate that elevated SIRT1 protein in human cells is not attributable to increased SIRT1 mRNA levels but, instead, reflects SIRT1 protein stability. RNAi-mediated depletion of JNK2 reduced the half-life of SIRT1 protein from >9 h to <2 h and this correlated with lack of SIRT1 protein phosphorylation at serine 27. In contrast, depletion of JNK1 had no effect upon SIRT1 protein stability and SIRT1 phosphorylation at serine 47 showed no correlation with SIRT1 protein stability. Thus we show that JNK2 is linked, directly or indirectly, with SIRT1 protein stability and that this function is coupled with SIRT1 phosphorylation at serine 27. Our observations identify a route for therapeutic modulation of SIRT1 protein levels in SIRT1-linked diseases including cancer, neurodegeneration and diabetes.


Subject(s)
Mitogen-Activated Protein Kinase 9/metabolism , Sirtuins/genetics , Cell Line, Tumor , Gene Expression Regulation , HCT116 Cells , Humans , Phosphorylation , RNA, Messenger/metabolism , Serine/genetics , Serine/metabolism , Sirtuin 1 , Sirtuins/metabolism
15.
Cancer Biol Ther ; 7(9): 1481-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18769132

ABSTRACT

The tumor suppressor protein p53 is considered the "Guardian of the Genome", crucial for cell cycle control and mutated in over 50% of human cancers. Following cellular stress, post-translational modifications such as phosphorylation and acetylation stabilise and activate p53 for cell cycle arrest, DNA repair, apoptosis or senescence. p53 protein functions as a tetramer and we have shown that loss of tetramerisation and changes at the N-terminus influence the recovery of wild type p53 'status'. To investigate the relationship between tetramerisation and post-translational modifications we examined a range of site-specific modifications in wild type and dimeric mutant (M340Q/L344R) murine p53 expressed in MEFs p53(-/-) and in wild type, monomeric (L344P) and dimeric (M340Q/L344R) human p53 expressed in HCT116 p53(-/-) cells. Using site-specific antibodies we demonstrate that in murine p53, S15 is phosphorylated in a tetramerisation-dependent manner. In contrast, human p53 S15 phosphorylation is not tetramerisation-dependent. Inability to form tetramers in human p53 proteins reduced site-specific N-terminal phosphorylation at S6, S9 and S46 and reduced C-terminal phosphorylation and acetylation at S315 and K382 respectively. In addition, p53 tetramerisation is required for efficient p21 and hdm2 transcription and protein expression and recruitment of p53 to specific promoter regions of p21 and hdm2.


Subject(s)
Fibroblasts/metabolism , Protein Processing, Post-Translational , Tumor Suppressor Protein p53/metabolism , Acetylation , Animals , Cells, Cultured , Dimerization , Embryo, Mammalian/cytology , HCT116 Cells , Humans , Mice , Models, Molecular , Phosphorylation , Protein Structure, Quaternary , Protein Structure, Tertiary , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/ultrastructure
16.
Cell Cycle ; 6(21): 2669-77, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17957139

ABSTRACT

SIRT3, one of seven mammalian sirtuins, is a NAD-dependent deacetylase. SIRT3 localizes to mitochondria where it deacetylates and thus activates acetyl-CoA synthetase 2 (AceCS2), indicating a role for SIRT3 in metabolism. Here we provide evidence that SIRT3 also impacts upon apoptosis and cell growth control. Using RNAi under basal (non-stress) conditions we show that SIRT3 is required for apoptosis induced by selective silencing of Bcl-2 in HCT116 human epithelial cancer cells. Identical treatment of ARPE19 epithelial non-cancer cells induces G(1) growth arrest which also proved to be SIRT3-dependent. Previously we have identified SIRT1 and JNK2 as constitutive suppressors of apoptosis in HCT116 cells. We now demonstrate that SIRT3 functions in JNK2-regulated apoptosis but is dispensable for SIRT1-regulated apoptosis. SIRT3 is also dispensable for stress-induced apoptosis. Thus the pro-apoptotic functioning of SIRT3 is selectively coupled with defined pathways regulating cell survival under basal conditions.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/physiology , Mitochondrial Proteins/physiology , Signal Transduction/physiology , Sirtuins/physiology , Cell Line , Cell Survival/genetics , Cell Survival/physiology , Gene Silencing/physiology , Growth Inhibitors/physiology , HCT116 Cells , Humans , RNA, Small Interfering/physiology , Signal Transduction/genetics , Sirtuin 3
17.
EMBO J ; 25(21): 5191-200, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17053786

ABSTRACT

p53 major tumour suppressor protein has presented a challenge for structural biology for two decades. The intact and complete p53 molecule has eluded previous attempts to obtain its structure, largely due to the intrinsic flexibility of the protein. Using ATP-stabilised p53, we have employed cryoelectron microscopy and single particle analysis to solve the first three-dimensional structure of the full-length p53 tetramer (resolution 13.7 A). The p53 molecule is a D2 tetramer, resembling a hollow skewed cube with node-like vertices of two sizes. Four larger nodes accommodate central core domains, as was demonstrated by fitting of its X-ray structure. The p53 monomers are connected via their juxtaposed N- and C-termini within smaller N/C nodes to form dimers. The dimers form tetramers through the contacts between core nodes and N/C nodes. This structure revolutionises existing concepts of p53's molecular organisation and resolves conflicting data relating to its biochemical properties. This architecture of p53 in toto suggests novel mechanisms for structural plasticity, which enables the protein to bind variably spaced DNA target sequences, essential for p53 transactivation and tumour suppressor functions.


Subject(s)
Models, Molecular , Tumor Suppressor Protein p53/chemistry , Cryoelectron Microscopy/methods , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , Humans , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Tumor Suppressor Protein p53/metabolism
18.
Cancer Res ; 65(22): 10457-63, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16288037

ABSTRACT

SIRT1 is a conserved NAD-dependent deacetylase that regulates life span in accord with nutritional provision. In mammalian cells, SIRT1 also down-regulates stress-induced p53 and FoxO pathways for apoptosis, thus favoring survival under stress. The functioning of SIRT1 under normal, nonstressed conditions of cell growth is unknown. Here we have asked if SIRT1 has the capacity to influence cell viability in the absence of applied stress. For this purpose we used synthetic small interfering RNA to silence SIRT1 gene expression by RNA interference (RNAi). We show that the process of RNAi, by itself, does not affect cell growth and is not sufficient to activate a cellular stress response (indicated by lack of activation of endogenous p53). We also show that, in the absence of applied stress, SIRT1 silencing induces growth arrest and/or apoptosis in human epithelial cancer cells. In contrast, normal human epithelial cells and normal human diploid fibroblasts seem to be refractory to SIRT1 silencing. Combined gene knockout with RNAi cosilencing experiments indicate that SIRT1 and Bcl-2 may suppress separable apoptotic pathways in the same cell lineage and that the SIRT1-regulated pathway is independent of p53, Bax, and caspase-2. Alternatively, SIRT1 may suppress apoptosis downstream from these apoptotic factors. In either case, we show that FoxO4 (but not FoxO3) is required as proapoptotic mediator. We further identify caspase-3 and caspase-7 as downstream executioners of SIRT1/FoxO4-regulated apoptosis. Our work identifies SIRT1 as a novel target for selective killing of cancer versus noncancer epithelial cells.


Subject(s)
Histone Deacetylases/physiology , Sirtuins/physiology , Apoptosis/physiology , Caspase 3 , Caspase 7 , Caspases/metabolism , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Survival/physiology , Gene Silencing , HCT116 Cells , Histone Deacetylase Inhibitors , Histone Deacetylases/genetics , Humans , Proto-Oncogene Proteins c-bcl-2/physiology , RNA Interference , RNA, Small Interfering/genetics , Sirtuin 1 , Sirtuins/antagonists & inhibitors , Sirtuins/genetics , Tumor Suppressor Protein p53/physiology
19.
Nucleic Acids Res ; 33(18): e151, 2005 Oct 07.
Article in English | MEDLINE | ID: mdl-16214804

ABSTRACT

RNA interference (RNAi) is a process of post-transcriptional gene silencing initiated by double-stranded RNAs, including short interfering RNA (siRNA). Silencing is sequence-specific and RNAi has rapidly become central to the study of gene function. RNAi also carries promise for selective silencing of viral and endogenous genes causal for disease. To detect the very low levels of siRNA effective for RNAi we modified the 3' end of the sense strand of siRNA with a nuclease-resistant DNA hairpin. We show that the modified siRNA-DNA construct (termed 'crook' siRNA) functions as a primer for the PCR and describe a novel, yet simple PCR protocol for its quantification (amolar levels/cell). When transfected into mammalian cells, crook siRNA induces selective mRNA knock-down equivalent to its unmodified siRNA counterpart. This new bifunctional siRNA construct will enable future in vivo studies on the uptake, distribution and pharmacokinetics of siRNA, and is particularly important for the development of siRNA-based therapeutics. More generally, PCR-based detection of siRNA carries wide-ranging applications for RNAi reverse genetics.


Subject(s)
Polymerase Chain Reaction/methods , RNA Interference , RNA, Small Interfering/analysis , RNA, Small Interfering/chemistry , Cell Line , DNA Primers , Humans , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Phenotype , RNA, Messenger/metabolism , Templates, Genetic , Transfection
20.
Proc Natl Acad Sci U S A ; 102(43): 15412-7, 2005 Oct 25.
Article in English | MEDLINE | ID: mdl-16234232

ABSTRACT

One of many protein-protein interactions modulated upon DNA damage is that of the single-stranded DNA-binding protein, replication protein A (RPA), with the p53 tumor suppressor. Here we report the crystal structure of RPA residues 1-120 (RPA70N) bound to the N-terminal transactivation domain of p53 (residues 37-57; p53N) and, by using NMR spectroscopy, characterize two mechanisms by which the RPA/p53 interaction can be modulated. RPA70N forms an oligonucleotide/oligosaccharide-binding fold, similar to that previously observed for the ssDNA-binding domains of RPA. In contrast, the N-terminal p53 transactivation domain is largely disordered in solution, but residues 37-57 fold into two amphipathic helices, H1 and H2, upon binding with RPA70N. The H2 helix of p53 structurally mimics the binding of ssDNA to the oligonucleotide/oligosaccharide-binding fold. NMR experiments confirmed that both ssDNA and an acidic peptide mimicking a phosphorylated form of RPA32N can independently compete the acidic p53N out of the binding site. Taken together, our data suggest a mechanism for DNA damage signaling that can explain a threshold response to DNA damage.


Subject(s)
DNA, Single-Stranded/chemistry , Replication Protein A/chemistry , Transcriptional Activation , Tumor Suppressor Protein p53/chemistry , Amino Acid Sequence , Binding Sites , Binding, Competitive , DNA Damage , Humans , Molecular Sequence Data , Phosphorylation , Protein Folding , Protein Structure, Tertiary , Replication Protein A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...