Skip to main content

An RNA interference screen for identifying downstream effectors of the p53 and pRB tumour suppressor pathways involved in senescence

Abstract

Background

Cellular senescence is an irreversible cell cycle arrest that normal cells undergo in response to progressive shortening of telomeres, changes in telomeric structure, oncogene activation or oxidative stress and acts as an important tumour suppressor mechanism.

Results

To identify the downstream effectors of the p53-p21 and p16-pRB tumour suppressor pathways crucial for mediating entry into senescence, we have carried out a loss-of-function RNA interference screen in conditionally immortalised human fibroblasts that can be induced to rapidly undergo senescence, whereas in primary cultures senescence is stochastic and occurs asynchronously. These cells are immortal but undergo a rapid irreversible arrest upon activation of the p53-p21 and p16-pRB pathways that can be readily bypassed upon their inactivation. The primary screen identified 112 known genes including p53 and another 29 shRNAmirs targetting as yet unidentified loci. Comparison of these known targets with genes known to be up-regulated upon senescence in these cells, by micro-array expression profiling, identified 4 common genes TMEM9B, ATXN10, LAYN and LTBP2/3. Direct silencing of these common genes, using lentiviral shRNAmirs, bypassed senescence in the conditionally immortalised cells.

Conclusion

The senescence bypass screen identified TMEM9B, ATXN10, LAYN and LTBP2/3 as novel downstream effectors of the p53-p21 and p16-pRB tumour suppressor pathways. Although none of them has previously been linked to cellular senescence, TMEM9B has been suggested to be an upstream activator of NF-κB signalling which has been found to have a causal role in promoting senescence. Future studies will focus on determining on how many of the other primary hits also have a casual role in senescence and what is the mechanism of action.

Background

Normal somatic cells undergo a finite number of divisions before entering a state of irreversible growth arrest termed cellular senescence [1]. This is triggered in response to a variety of intrinsic and extrinsic stimuli including progressive telomere shortening or changes in telomeric structure at the ends of chromosomes or other forms of genotoxic stress such as oncogene activation, or DNA damage or oxidative stress, resulting in a DNA damage response and growth arrest via activation of the p53 tumour suppressor pathway [2, 3]. Non-genotoxic stress induces senescence by a telomere independent mechanism involving activation of the p16-pRB pathway by up-regulation of p16INK4a[3, 4].

Cellular senescence acts as an important tumour suppressor mechanism. Overcoming senescence and acquiring a limitless replicative potential has been proposed to be one of the key events required for malignant transformation [5]. Senescence is thought to have evolved as an example of antagonistic pleiotrophy, whereby its beneficial traits in a reproductively active individual have deleterious effects later in life [6, 7]. The underlying mechanisms that control cellular senescence, the signal transduction pathways involved and how the diverse signals that result in senescence are all integrated, remain poorly defined. Moreover the downstream effectors of the p53-p21 and p16-pRB pathways that result in the irreversible growth arrest and entry into senescence are unknown.

The discovery of RNA interference as a mechanism to silence gene expression has revolutionized studies on mammalian gene expression and has permitted loss-of-function genome-wide genetic screens, to identify genes involved in a variety of cellular processes, to be performed [812]. A number of shRNA libraries have been developed for carrying out such genome-wide screens, one of which is the pSM2 Retroviral shRNAmir library [13] (Thermo Scientific Open Biosystems). This library has several unique features that make it very versatile and efficient for large-scale screens particularly the human microRNA-30 (miR30) adapted design which increases knockdown specificity and efficiency [14].

Here we present a RNA interference screen using the human pSM2 retroviral shRNAmir library, carried out in the conditionally immortal HMF3A human fibroblasts, to identify genes whose silencing bypasses senescence arrest induced by activation of the p53-p21 and p16-pRB pathways. The primary screen identified 112 known genes and another 29 shRNAmirs targetting as yet unidentified loci. Comparison of the known targets with genes known to be up-regulated upon senescence by micro-array expression profiling, identified 4 common genes whose expression was reversed when senescence was bypassed upon inactivation of the p53-p21 and p16-pRB pathways.

Results

To directly identify the downstream effectors of the p53-p21 and p16-pRB pathways, we have utilized the conditionally immortal HMF3A human fibroblasts that were derived by immortalising adult human mammary fibroblasts with the catalytic subunit of human telomerase and a thermolabile U19tsA58 mutant of SV40 Large T antigen [15]. These cells are immortal if grown at 34°C but undergo a senescence arrest upon inactivation of the thermolabile U19tsA58 T antigen resulting in the activation of the p53-p21 and p16-pRB pathways [15]. They are stringently temperature sensitive but senescence can be readily bypassed by inactivation of the p53-p21 or the p16-pRB pathway [16]. To facilitate efficient transduction of these cells by retroviral infection, they were transduced with the full length murine ecotropic retroviral receptor and CL3EcoR cells derived, that most closely mirror the parental cells [16]. The temperature dependent senescent arrest of CL3EcoR cells and its bypass upon inactivation of p21CIP1 by silencing or sequestration of the RB family of proteins by HPV16 E7 are shown in Figure 1.

Figure 1
figure 1

Characteristics of CL3EcoR cells. a: CL3EcoR cells are immortal at 34°C but undergo a senescence arrest upon shift up 38°C. b: Senescence is bypassed upon silencing of p21CIP1 using pRSp21F or sequestration of RB family of proteins by HPV16 E7.

The pSM2 library version 1.3 comprising 15,148 constructs targetting 9,392 human cancer associated genes was amplified and each 96 well plate used to prepare a pool of plasmid DNA; each of the 100 pools contained between 150 to 200 different shRNAmir constructs with each gene being represented by 1 to 3 shRNAmirs. To ensure that CL3EcoR cells were sufficiently sensitive to identify a single shRNAmir construct in a pool of 200 shRNAmir constructs, pRSp21F (a p21CIP1 shRNA construct) [17] was mixed in a ratio of 1:200 with pRSLaminA/C and used to assay bypass of senescence in CL3EcoR cells. The pRSp21F construct was used because the pSM2 library version 1.3 did not contain any silencing constructs for p21CIP1. Silencing of LaminA/C did not bypass senescence, very few growing colonies were obtained (Figure 2a) whereas silencing of p21CIP1 was very efficient and produced essentially a confluent monolayer of growing cells (Figure 2b). The 1:200 p21CIP1/LaminA/C mix produced numerous distinct densely growing colonies (Figure 2c) indicating that CL3EcoR cells and the procedure were sufficiently sensitive to generate colonies in which senescence had been overcome.

Figure 2
figure 2

Sensitivity of the screen. CL3EcoR cells were infected with retroviruses prepared from pRSLamin A/C (a), pRSp21F (b), or a 1/200 mix of pRSp21F/pRSLamin A/C (c). After puromycin selection, cells were reseeded at 8.5 × 104 per 15 cm plate or 0.5 × 104 per well in 6-well plates and shifted to 38°C for 3 weeks.

ShRNA interference screen

The formula: ln [1-0.95]/ln [1-1/(Library Size)] recommended for genetic screens by the Nolan lab (http://www.stanford.edu/group/nolan/screens/screens.html), suggested that approximately 1000 independent infectious events would be sufficient for a 99% confidence that all shRNAs within a pool had been sampled. To ensure that the screen would be saturating, virus sufficient to yield 10,000 infectious events was utilised for each pool (shown in Additional File 1). The screen was performed in successive waves of 10 pools. To minimise variation and background reversion, CL3EcoR cells were used at the same passage for every pool. Virus prepared from pRSp21F and pRSLamin A/C was used as positive and negative controls respectively to evaluate the level of background and ensure that the complementation assay worked for each round of the screen. Stably transduced cells were trypsinised and reseeded. Three weeks after reseeding, flasks were examined to identify growing colonies; a representative colony is shown in Figure 3. Each colony was examined microscopically to ensure it comprised growing cells and the number of colonies obtained for each pool determined. The number of stably transduced cells, the number of flasks reseeded and the number of colonies obtained for each flask at 38°C are shown in Additional File 2. The flasks which contained more densely growing/bigger colonies (indicated in red in Additional File 2) were trypsinised, replated and used for extracting genomic DNA when confluent.

Figure 3
figure 3

ShRNA screen. CL3EcoR cells were transduced with ecotropic retroviruses prepared from each of the 100 pools of shRNAmirs, selected with puromycin, reseeded and shifted up to 38°C for 3 weeks. Densely growing colonies were considered to have bypassed senescence and potentially contain candidate shRNAmirs.

34 out of 100 pools yielded healthy growing colonies; pools 13, 78 and 82 particularly gave a higher number of colonies which were also larger. Pools 16, 18, 19, 21 and 80 also yielded colonies but they were smaller. For each pool that contained growing colonies, 1 to 4 flasks containing the highest number of growing colonies, were reseeded for extracting genomic DNA and resulted in a total of 81 sub-pools.

Identification of shRNAmirs

The shRNAmir proviral inserts were by amplified by two rounds of nested PCR using pSM2 specific primers, the amplified products TOPO cloned and plasmid DNA extracted from at least six colonies sequenced to identify all shRNAs present within each pool; for some pools, DNA from many more colonies was sequenced. The hair pin sequence was determined by searching for the miR-30 context and the miR-30 loop that are common to all inserts and frame the hair pin. The sequence of the hair pin was used to identify the target gene by searching the pSM2 data base or by BLASTN analysis of the NCBI human genome data base (http://blast.ncbi.nlm.nih.gov/Blast.cgi). Sequences that could not be linked back to the list of hair pin sequences within the Open Biosystems collection or were not 100% homologous to a gene within Genbank were not pursued and are presented in Additional File 3.

The rescued shRNAmir hair pins identified 112 known genes and another 29 shRNAmirs targetting as yet unidentified loci. For each pool, the number of times that sequence was obtained, the corresponding insert reference, gene name and the number of shRNAmir constructs for that gene within the SM2 library are shown in Table 1. The last column of the table indicates if the recovered insert was a match to a hair pin present in that particular pool ("match") or if it was present within another pool ("listed in pool X"). 14 hair pins were isolated from incorrect pools. Some inserts were detected several times in multiple pools. For example, the hair pin v2HS_119967 (corresponding to LOC155004) listed within pool 52 was also identified from pool 3, 4, 5, 9, 12, 30, 59, 60, 64, 71, 72, 74, 77, and 98 suggesting that there may have been some cross-contamination of the library perhaps during replica plating. However, this was not a problem since the aim of our screen was to identify shRNAmirs that are able to bypass senescence and does not depend upon the identity of the pool. Since this insert was isolated from so many pools, it could be that it is a strong positive or that it was highly represented within the library and likely to be a false positive.

Table 1 Primary screen

Pool 13, 78 and 82 that produced more colonies and colonies that were larger and healthier than others, identified the following genes: Pool 13: DYNLRB1, FARSLB, PPARGC1A, TAOK1, CEND1, ABL1, LOC342404, TMEM168, SIRPG, IYD and SMCR7; Pool 78: PRRX1, SLC25A21, ARMCX2, LOC284804, LOC349839, MRP63, TMEM63B, ZBTB1, KIA0286 and EIF4A1; and Pool 82: EIF4A1, CCDC70, PRICKLE2, PLP1, SAMD4A, RASA4, TP53, ARNTL, BCL2L12, ADNP2, DSG4, LOC345672, LOC349975, LOC349811 and ARHGAP20. Moreover, pool 82, one of the pools that gave the best senescence bypass, contained the only shRNAmir targetting p53, v2HS_93615 identified in the screen, thereby internally validating it.

Overlap of the primary candidates of the shRNA screen with microarray data for genes up-regulated upon senescence in CL3EcoR cells

To prioritise the candidates identified from the primary screen for functional validation, they were compared to genes up-regulated upon senescence and whose expression was reversed when senescence was abrogated upon inactivation of the p53 and pRB pathways [[16]; microarray expression profiling data is available from Gene Expression Omnibus database accession number GSE24810]. This identified 4 common genes, ATXN10, LAYN, LTBP2/3 and TMEM9B. The microarray expression profiling data presented in Table 2 showed that they were all up-regulated upon senescence growth arrest: ATXN10 by 1.3 fold (p-value 1 × 10-3), LAYN by 2 fold (p-value 2 × 10-4), LTBP2/3 by 2.5 (p-value 9 × 10-8), 1.7 (p-value 5 × 10-9), 1.5 (p-value 8 × 10-4) and 1.4 fold (p-value 5 × 10-5) and TMEM9B by 1.4 (p-value 1 × 10-7) and 1.3 fold (p-value 2 × 10-4) respectively. The data in Table 2 further show that up-regulation of these candidates was reversed when senescence was bypassed upon inactivation of the p53-p21 and p16-pRB pathways by silencing p53 (pRS_p53) or p21CIP1 (pRS_p21) or by sequestration of the RB family of proteins by HPV16 E7 or by expression of the dominant negative E2F-DB protein.

Table 2 Microarray expression profiling data for common genes

The identification of TMEM9B was particularly remarkable because the microarray analysis has suggested that senescence growth arrest in CL3EcoR cells is associated with activation of the NF-κB signalling pathway and TMEM9B has previously been shown to be able to activate NF-κB dependent reporter constructs [18, 19]. To determine if silencing of TMEM9B would bypass senescence, 4 GIPZ lentiviral silencing constructs (v2LHS_247318, 58957, 58958 and 58959; Thermo Scientific Open Biosystems) targetting TMEM9B were obtained, pooled and introduced into CL3EcoR cells after packaging as lentiviruses. The GIPZ lentiviral library contains the same hair pins as the retroviral library but is more stable and the constructs are packaged as lentiviruses rather than retroviruses. Lentiviral human GIPZ Lamin A/C shRNAmir (v2LHS_62719) was used as a negative control. Silencing of TMEM9B was clearly able to bypass senescence (Figure 4a). Moreover each of the constructs was individually able to overcome senescence arrest with v2LHS_58957 being the most efficient [16].

Figure 4
figure 4

Silencing targets identified from the primary screen. CL3EcoR cells were infected in triplicate with lentiviruses prepared from the indicated GIPZ shRNAmir constructs targetting TMEM9B (a), LTBP2/3 (b), ATXN10 (c) and LAYN (d). Cells resistant to 6 μg/ml puromycin were isolated, reseeded and assayed for bypass of senescence by culturing at 38°C for 3 weeks. Lentiviruses prepared from the GIPZ shRNAmir Lamin A/C (v2LHS_62719) construct were used as the negative control. For TMEM9B, the mix of constructs comprised v2LHS_247318, 58957, 58958 and 58959.

To determine if ATXN10, LAYN and LTBP2/3 were also able to directly bypass senescence, lentivirus constructs from the GIPZ lentiviral shRNAmir library were used. The complementation assay for LTBP2/3, in Figure 4b, showed that silencing LTBP2/3 with the one available silencing construct (v2LHS_34089) clearly yielded healthy growing colonies. Silencing of ATXN10 was tested using 4 different silencing constructs v2LHS_71735, 71736, 71737 and 71740. All four constructs were able to overcome senescence and yielded more growing colonies than the Lamin A/C negative control (Figure 4c). Silencing of LAYN was tested using 2 different silencing constructs v2LHS_265009 and 118722; both constructs were able to bypass growth arrest and produced growing colonies (Figure 4d).

Taken together our results showed that silencing of TMEM9B, ATXN10, LAYN and LTBP2/3 was able to bypass senescence in the conditionally immortal human fibroblasts.

Discussion

To directly identify the downstream effectors of the p53-p21 and p16-pRB pathways crucial for mediating entry into senescence, we carried out a loss-of-function RNA interference screen in the conditionally immortal HMF3A human fibroblasts. This identified 112 known genes including p53 and another 29 shRNAmirs targetting unidentified loci. Comparison of these known targets with genes up-regulated upon senescence in these cells identified 4 common genes TMEM9B, ATXN10, LAYN and LTBP2/3. Direct silencing of these common genes using lentiviral shRNAmirs bypasses senescence in the HMF3A cells. Although none of these genes has previously been linked to cellular senescence, TMEM9B has been suggested to be an upstream activator of NF-κB signalling which we have found to have a causal role in promoting senescence.

The 112 known targets identified by the shRNA screen comprise a wide variety of genes but most importantly one of them was the only p53 shRNAmir (v2HS_93615 from pool 82) present within this library, thereby internally validating the screen. Moreover all of the primary targets were identified from single shRNAmirs even though we have subsequently shown that other shRNAmirs corresponding to these targets present within the library are able to bypass senescence. It is not clear why other shRNAmirs were not isolated in the screen; however this is exactly what has been observed previously by others such as Westbrook and colleagues [20]. Nevertheless it remains to be demonstrated which of the targets identified by the primary screen are able to bypass senescence when assayed individually.

TMEM9B was one of the 4 genes in common between the shRNA screen and genes known to be up-regulated upon senescence in HMF3A cells [16]. Moreover expression of TMEM9B was down-regulated when senescence was bypassed upon abrogation of the p53-p21 or p16-pRB pathways. TMEM9B is a glycosylated protein that localises to lysosome membranes and partially to early endosomes. It has been shown to be a component of TNF signalling and a module shared between the interleukin-1 and Toll-like receptor pathways. It is also essential for TNF activation of both NF-κB and MAPK pathways by acting downstream of RIP1 and upstream of the MAPK and IκB kinases at the level of the TAK1 complex [19]. TMEM9B was also identified in a large scale study to identify genes activating NF-κB and MAPK signalling pathways [18]. These results are all consistent with our finding that in the conditionally immortal HMF3A cells, senescence growth arrest is associated with an activation of NF-κB signalling and suppression of this pathway bypasses senescence [16].

The latent TGFβ-binding protein 2/3 (LTBP2/3) hair pin sequence was identified from pool 66. Up-regulation of LTBP2/3 expression upon growth arrest was reversed when senescence was overcome. LTBPs are secreted proteins initially identified through their binding to TGFβ and may be involved in their assembly, secretion and targetting [21]. LTBP2/3 in particular has been found to play an essential role in the secretion and targetting of TGFβ1 [22]. Since silencing of LTBP2/3 can bypass senescence in HMF3A cells, it suggests that LTBP2/3 may be linked with the control of cell growth and be playing a role in suppressing tumour progression perhaps through regulation of TGFβ. This is in accordance with the identification of TGFβ as a senescence-inducing factor in the human lung A549 adenocarcinoma cells [23]. It is also in accordance with several other reports suggesting that TGFβ1 is capable of inducing cellular senescence. For instance, stimulation of human diploid fibroblasts with TGFβ1 triggers the appearance of senescence associated-β-galactosidase activity and increases steady state mRNA levels of senescence associated genes including APO J, fibronectin, and M22 [2426]; both APO J (clusterin) and fibronectin are up-regulated in CL3 cells upon senescence arrest and this is reversed when senescence is bypassed [16].

Ataxin (ATXN) 10 was slightly up-regulated upon senescence arrest which was reversed upon silencing of p53 and p21CIP1 or ectopic expression of the dominant negative E2F-DB protein. Spinocerebellar ataxia type 10 (SCA10) is a dominantly inherited disorder characterized by ataxia, seizures and anticipation caused by an intronic ATTCT pentanucleotide repeat expansion. The product of SCA10 encodes the novel protein, ATXN10, previously known as E46L, which is widely expressed in the brain and belongs to the family of armadillo repeat proteins. Although clinical features of the disease are well characterized, very little is known about ATXN10. ATXN10 knock down by RNAi has recently been shown to cause increased apoptosis in primary cerebellar cultures, implicated in SCA10 pathogenesis [27, 28]. This is in contrast to our finding that silencing of ATXN10 in HMF3A cells by four different shRNAmirs did not cause apoptosis but promoted growth and permitted a bypass of senescence.

Layilin (LAYN) identified from pool 58 was 2 fold up-regulated upon senescence arrest, which was reversed upon abrogation of the growth arrest by inactivation of either the p53-p21 or the p16-pRB pathways. Moreover two different LAYN shRNAmirs were found to directly bypass senescence in HMF3A cells. Layilin is a widely expressed integral membrane hyaluronan receptor, originally identified as a binding partner of talin located in membrane ruffles. Talin is responsible, along with its adaptor proteins, for maintaining the cytoskeleton-membrane linkage by binding to integral membrane proteins and to the cytoskeleton. Recently layilin has been suggested to play a crucial role in lymphatic metastasis of lung carcinoma A549 cells [29].

In addition to the genes described above, a number of other interesting genes particularly TAOK1, RAS4A and ARMCX2 were identified. TAOK1 is a micro-tubule affinity-regulating kinase required for both chromosome congression and checkpoint-induced anaphase delay [30]. It is known to activate the p38MAPK pathway through the specific phosphorylation of MKK3. This is a complex pathway responsive to stress stimuli and involved in cell differentiation and apoptosis and has been shown to have an important causative role in senescence [31]. RAS4A encodes a member of the GAP1 family of GTPase-activating proteins that have been identified to suppress the Ras/mitogen-activated protein kinase pathway in response to an elevation of Ca2+ ions. Stimuli that increase intracellular Ca2+ levels result in the translocation of this protein to the plasma membrane, where it activates Ras GTPase activity resulting in Ras being converted from the active GTP-bound state to the inactive GDP-bound state and suppression of downstream signalling [32]. ARMCX2 encodes a member of the ALEX family of proteins and may play a role in tumour suppression. This protein contains a potential N-terminal transmembrane domain and a single Armadillo repeat; armadillo repeat containing proteins are involved in development, maintenance of tissue integrity and suppressing carcinomas [33].

Conclusions

The RNA interference screen has identified 112 known candidate proteins including p53 and another 29 shRNAmirs targetting as yet unidentified loci. Although none of them except p53 had previously been linked to senescence or known to be downstream effectors of the p53-p21 and p16-pRB tumour suppressor pathways, directly silencing four of these candidates, TMEM9B, ATXN10, LAYN and LTBP2/3 bypassed senescence in CL3EcoR cells. It remains to be determined whether direct silencing of any of the other primary candidates will also bypass senescence. Any genes that can bypass senescence upon silencing are novel starting points for identifying the signalling networks that act downstream of p53 and pRB to induce cellular senescence. The genes/proteins identified in the screen are also potential tumour suppressors, and a mechanistic dissection of their mode of action and role in cancer will undoubtedly provide new avenues for further research.

Methods

Cell Culture

CL3EcoR cells were maintained at 34°C ± 0.5°C [15]. Temperature shift experiments were performed at 38°C ± 0.5°C. Phoenix ecotropic and HEK293T cells were obtained from the ATCC and maintained at 37°C. Cells were grown in Dulbecco's modified Eagles medium (DMEM) supplemented with 10% (v/v) heat inactivated foetal bovine serum, 2 mM glutamine, 100 units/ml penicillin and 100 μg/ml streptomycin. All media and components were obtained from Invitrogen.

Viral packaging and infection

Lentiviruses were prepared according to Besnier et al.[34]. Ecotropic viruses were prepared by transfecting 10 μg of retroviral plasmid DNA into phoenix ecotropic cells by FuGENE 6 Transfection reagent (Roche), according to the manufacturer's instructions. 24 hrs post-transfection, media was changed and fresh medium added. 48 hrs post-transfection, retroviral supernatant was harvested, filtered through a 0.45 μm filter and either used immediately or frozen at -80°C. A second harvest was prepared by adding fresh media to the plates and harvesting the virus supernatant the next day.

Cells were infected with virus supernatants for 24 hrs at 34°C. Four days post-infection, antibiotic selection was added (2 μg/ml puromycin for pRS and pSM2 retroviruses or 6 μg/ml puromycin for pGIPZ shRNAmir lentiviruses; Invitrogen). Selection of cells infected with human GIPZ lentiviral shRNAmir constructs in puromycin at 6 μg/ml, enriches for cells with higher levels of shRNAmir expression. For the senescence bypass assay, the stably transduced cells were plated at 5 × 104 cells in T-75 flasks or at 1.6 × 104 cells in T-25 flasks and incubated overnight at 34°C. Next day the medium was changed and the cells shifted to 38°C for 3 weeks. Flasks which contained more densely growing or bigger colonies were trypsinised, replated and used for extracting genomic DNA when confluent.

References

  1. Hayflick L, Moorhead PS: The serial cultivation of human diploid cell strains. Exp Cell Res. 1961, 25: 585-621. 10.1016/0014-4827(61)90192-6.

    Article  CAS  PubMed  Google Scholar 

  2. Ben-Porath I, Weinberg RA: When cells get stressed: an integrative view of cellular senescence. J Clin Invest. 2004, 113: 8-13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Campisi J, d'Adda di Fagagna F: Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol. 2007, 8: 729-40.

    Article  CAS  PubMed  Google Scholar 

  4. Ben-Porath I, Weinberg RA: The signals and pathways activating cellular senescence. Int J Biochem Cell Biol. 2005, 37: 961-76. 10.1016/j.biocel.2004.10.013.

    Article  CAS  PubMed  Google Scholar 

  5. Hanahan D, Weinberg RA: The hallmarks of cancer. Cell. 2000, 100: 57-70. 10.1016/S0092-8674(00)81683-9.

    Article  CAS  PubMed  Google Scholar 

  6. Kirkwood TB, Austad SN: Why do we age?. Nature. 2000, 408: 233-8. 10.1038/35041682.

    Article  CAS  PubMed  Google Scholar 

  7. Campisi J: Senescent cells, tumor suppression, and organismal aging: good citizens, bad neighbors. Cell. 2005, 120: 513-22. 10.1016/j.cell.2005.02.003.

    Article  CAS  PubMed  Google Scholar 

  8. Berns K, Hijmans EM, Mullenders J, Brummelkamp TR, Velds A, Heimerikx M, Kerkhoven RM, Madiredjo M, Nijkamp W, Weigelt B, Agami R, Ge W, Cavet G, Linsley PS, Beijersbergen RL, Bernards R: A large-scale RNAi screen in human cells identifies new components of the p53 pathway. Nature. 2004, 428: 431-7. 10.1038/nature02371.

    Article  CAS  PubMed  Google Scholar 

  9. Iorns E, Lord CJ, Turner N, Ashworth A: Utilizing RNA interference to enhance cancer drug discovery. Nat Rev Drug Discov. 2007, 6: 556-68. 10.1038/nrd2355.

    Article  CAS  PubMed  Google Scholar 

  10. Zender L, Xue W, Zuber J, Semighini CP, Krasnitz A, Ma B, Zender P, Kubicka S, Luk JM, Schirmacher P, McCombie WR, Wigler M, Hicks J, Hannon GJ, Powers S, Lowe SW: An oncogenomics-based in vivo RNAi screen identifies tumor suppressors in liver cancer. Cell. 2008, 135: 852-64. 10.1016/j.cell.2008.09.061.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hu G, Kim J, Xu Q, Leng Y, Orkin SH, Elledge SJ: A genome-wide RNAi screen identifies a new transcriptional module required for self-renewal. Genes Dev. 2009, 23: 837-48. 10.1101/gad.1769609.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhang S, Binari R, Zhou R, Perrimon N: A genome wide RNA interference screen for modifiers of aggregates formation by mutant Huntingtin in Drosophila. Genetics. 2010, 184: 1165-79. 10.1534/genetics.109.112516.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Paddison PJ, Silva JM, Conklin DS, Schlabach M, Li M, Aruleba S, Balija V, O'Shaughnessy A, Gnoj L, Scobie K, Chang K, Westbrook T, Cleary M, Sachidanandam R, McCombie WR, Elledge SJ, Hannon GJ: A resource for large-scale RNA-interference-based screens in mammals. Nature. 2004, 428: 427-31. 10.1038/nature02370.

    Article  CAS  PubMed  Google Scholar 

  14. Boden D, Pusch O, Silbermann R, Lee F, Tucker L, Ramratnam B: Enhanced gene silencing of HIV-1 specific siRNA using microRNA designed hair pins. Nucleic Acids Res. 2004, 32: 1154-8. 10.1093/nar/gkh278.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. O'hare MJ, Bond J, Clarke C, Takeuchi Y, Atherton AJ, Berry C, Moody J, Silver ARJ, Davies DC, Alsop AE, Neville AM, Jat PS: Conditional immortalization of freshly isolated human mammary fibroblasts and endothelial cells. Proc Natl Acad Sci USA. 2001, 98: 646-651. 10.1073/pnas.98.2.646.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Rovillain E, Mansfield L, Caetano C, Alvarez-Fernandez M, Caballero OL, Medema RH, Hummerich H, Jat PS: Activation of Nuclear Factor-kappa B signaling promotes cellular senescence. Oncogene. 2011, 30: 2356-66. 10.1038/onc.2010.611.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Mansfield LV: Dissecting the telomere-independent pathways underlying human cellular senescence. 2006, PhD thesis, University of London

    Google Scholar 

  18. Matsuda A, Suzuki Y, Honda G, Muramatsu S, Matsuzaki O, Nagano Y, Doi T, Shimotohno K, Harada T, Nishida E, Hayashi H, Sugano S: Large-scale identification and characterization of human genes that activate NF-kappaB and MAPK signaling pathways. Oncogene. 2003, 22: 3307-18. 10.1038/sj.onc.1206406.

    Article  CAS  PubMed  Google Scholar 

  19. Dodeller F, Gottar M, Huesken D, Iourgenko V, Cenni B: The lysosomal transmembrane protein 9B regulates the activity of inflammatory signalling pathways. J Biol Chem. 2008, 283: 21487-94. 10.1074/jbc.M801908200.

    Article  CAS  PubMed  Google Scholar 

  20. Westbrook TF, Martin ES, Schlabach MR, Leng Y, Liang AC, Feng B, Zhao JJ, Roberts TM, Mandel G, Hannon GJ, Depinho RA, Chin L, Elledge SJ: A genetic screen for candidate tumor suppressors identifies REST. Cell. 2005, 121: 837-48. 10.1016/j.cell.2005.03.033.

    Article  CAS  PubMed  Google Scholar 

  21. Oklü R, Hesketh R: The latent transforming growth factor beta binding protein (LTBP) family. Biochem J. 2000, 352: 601-10. 10.1042/0264-6021:3520601.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Penttinen C, Saharinen J, Weikkolainen K, Hyytiäinen M, Keski-Oja J: Secretion of human latent TGF-beta-binding protein-3 (LTBP-3) is dependent on co-expression of TGF-beta. J Cell Sci. 2002, 115: 3457-68.

    CAS  PubMed  Google Scholar 

  23. Katakura Y: Molecular basis for the cellular senescence program and its application to anticancer therapy. Biosci Biotechnol Biochem. 2006, 70: 1076-81. 10.1271/bbb.70.1076.

    Article  CAS  PubMed  Google Scholar 

  24. Frippiat C, Chen QM, Zdanov S, Magalhaes JP, Remacle J, Toussaint O: Subcytotoxic H2O2 stress triggers a release of transforming growth factor-beta 1, which induces biomarkers of cellular senescence of human diploid fibroblasts. J Biol Chem. 2001, 276: 2531-7. 10.1074/jbc.M006809200.

    Article  CAS  PubMed  Google Scholar 

  25. Frippiat C, Dewelle J, Remacle J, Toussaint O: Signal transduction in H2O2-induced senescence-like phenotype in human diploid fibroblasts. Free Radic Biol Med. 2002, 33: 1334-46. 10.1016/S0891-5849(02)01044-4.

    Article  CAS  PubMed  Google Scholar 

  26. Debacq C, Heraud JM, Asquith B, Bangham C, Merien F, Moules V, Mortreux F, Wattel E, Burny A, Kettmann R, Kazanji M, Willems L: Reduced cell turnover in lymphocytic monkeys infected by human T-lymphotropic virus type 1. Oncogene. 2005, 24: 7514-23. 10.1038/sj.onc.1208896.

    Article  CAS  PubMed  Google Scholar 

  27. Marz P, Probst A, Lang S, Schwager M, Rose-John S, Otten U, Ozbek S: Ataxin-10, the spinocerebellar ataxia type 10 neurodegenerative disorder protein, is essential for survival of cerebellar neurons. J Biol Chem. 2004, 279: 35542-50. 10.1074/jbc.M405865200.

    Article  PubMed  Google Scholar 

  28. Waragai M, Nagamitsu S, Xu W, Li YJ, Lin X, Ashizawa T: Ataxin 10 induces neuritogenesis via interaction with G-protein beta2 subunit. J Neurosci Res. 2006, 83: 1170-8. 10.1002/jnr.20807.

    Article  CAS  PubMed  Google Scholar 

  29. Chen Z, Zhuo W, Wang Y, Ao X, An J: Down-regulation of layilin, a novel hyaluronan receptor, via RNA interference, inhibits invasion and lymphatic metastasis of human lung A549 cells. Biotechnol Appl Biochem. 2008, 50: 89-96. 10.1042/BA20070138.

    Article  CAS  PubMed  Google Scholar 

  30. Draviam VM, Stegmeier F, Nalepa G, Sowa ME, Chen J, Liang A, Hannon GJ, Sorger PK, Harper JW, Elledge SJ: A functional genomic screen identifies a role for TAO1 kinase in spindle-checkpoint signalling. Nat Cell Biol. 2007, 9: 556-564. 10.1038/ncb1569.

    Article  CAS  PubMed  Google Scholar 

  31. Han J, Sun P: The pathways to tumor suppression via route p38. Trends Biochem Sci. 2007, 32: 364-71. 10.1016/j.tibs.2007.06.007.

    Article  CAS  PubMed  Google Scholar 

  32. Lockyer PJ, Kupzig S, Cullen PJ: CAPRI regulates Ca2+-dependent inactivation of the Ras-MAPK pathway. Curr Biol. 2001, 11: 981-986. 10.1016/S0960-9822(01)00261-5.

    Article  CAS  PubMed  Google Scholar 

  33. Kurochkin IV, Yonemitsu N, Funahashi SI, Nomura H: ALEX1, a Novel Human Armadillo Repeat Protein That Is Expressed Differentially in Normal Tissues and Carcinomas. Biochem Biophys Res Commun. 2001, 280: 340-347. 10.1006/bbrc.2000.4125.

    Article  CAS  PubMed  Google Scholar 

  34. Besnier C, Ylinen L, Strange B, Lister A, Takeuchi Y, Goff SP, Towers GJ: Characterization of murine leukemia virus restriction in mammals. J Virol. 2003, 77: 13403-6. 10.1128/JVI.77.24.13403-13406.2003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are indebted to Catherine King (the UCL shRNA library core facility) for providing constructs, Gary Adamson for DNA sequencing and to Ray Young for graphics. PSJ gratefully acknowledges financial support from the Wellcome Trust (078305) and an equipment grant from the Brain Research Trust.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Parmjit S Jat.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

ER and LM-carried out the screen. ER and PSJ rescued the inserts. CJL and AA amplified and provided the shRNAmir library. PSJ wrote the manuscript. All authors have read and approved the final manuscript.

Electronic supplementary material

12864_2010_3526_MOESM1_ESM.XLS

Additional file 1:Volume of virus supernatants used for the screen. The table shows the volume of virus supernatants used for each of the pools. (XLS 20 KB)

12864_2010_3526_MOESM2_ESM.XLS

Additional file 2:Primary screen. This table shows the pool number, the number of cells obtained after puromycin selection, the number of T-75 and T-25cm2 flasks reseeded and the number of growing colonies observed after 3 weeks at 38°C. Numbers indicated in red correspond to flasks that were reseeded for extracting genomic DNA. (XLS 40 KB)

12864_2010_3526_MOESM3_ESM.XLS

Additional file 3:Unidentified inserts. This table shows the hair pin sequences that were not 100% homologous to a gene by BLASTN analysis of the NCBI human genome data base (http://blast.ncbi.nlm.nih.gov/Blast.cgi) or could not be linked back to a hair pin sequence within the Open Biosystems SM2 library; the number of times the insert was isolated is also indicated. (XLS 32 KB)

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Rovillain, E., Mansfield, L., Lord, C.J. et al. An RNA interference screen for identifying downstream effectors of the p53 and pRB tumour suppressor pathways involved in senescence. BMC Genomics 12, 355 (2011). https://doi.org/10.1186/1471-2164-12-355

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2164-12-355

Keywords