Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
RECOMBINANT PROTEIN PRODUCTION SYSTEM
Document Type and Number:
WIPO Patent Application WO/2018/115429
Kind Code:
A1
Abstract:
The present disclosure relates generally to an improved gene expression system in the field of recombinant gene expression. The invention relates to a system showing an improved yield and quality of protein production and methods for increasing production of a protein produced by cultured cells, particularly cultured eukaryotic cells.

Inventors:
DE KEERSMAECKER KIM (BE)
SULIMA SERGEY (BE)
GIRARDI TIZIANA (BE)
KAMPEN KIM (BE)
Application Number:
PCT/EP2017/084359
Publication Date:
June 28, 2018
Filing Date:
December 22, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV LEUVEN KATH (BE)
International Classes:
C12N15/79
Foreign References:
US20080184384A12008-07-31
Other References:
HOFER ANNE ET AL: "Mutational analysis of the ribosomal protein Rpl10 from yeast", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 282, no. 45, November 2007 (2007-11-01), pages 32630 - 32639, XP002778706, ISSN: 0021-9258
DE KEERSMAECKER KIM ET AL: "Exome sequencing identifies mutation in CNOT3 and ribosomal genes RPL5 and RPL10 in T-cell acute lymphoblastic leukemia", NATURE GENETICS, vol. 45, no. 2, February 2013 (2013-02-01), pages 186 - 190, XP002778707
KLAUCK S M ET AL: "Mutations in the ribosomal protein gene RPL10 suggest a novel modulating disease mechanism for autism", MOLECULAR PSYCHIATRY, vol. 11, no. 12, December 2006 (2006-12-01), pages 1073 - 1084, XP002778708, ISSN: 1359-4184
SULIMA SERGEY O ET AL: "Bypass of the pre-60S ribosomal quality control as a pathway to oncogenesis", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 111, no. 15, April 2014 (2014-04-01), pages 5640 - 5645, XP002778709, ISSN: 0027-8424
DELABRE MARIE LAURE ET AL: "RPL29 codes for a non-essential protein of the 60S ribosomal subunit in Saccharomyces cerevisiae and exhibits synthetic lethality with mutations in genes for proteins required for subunit coupling", BIOCHIMICA ET BIOPHYSICA ACTA, vol. 1574, no. 3, 12 April 2002 (2002-04-12), pages 255 - 261, XP002778710, ISSN: 0006-3002
GIRARDI T ET AL: "The T-cell leukemia-associated ribosomal RPL10 R98S mutation enhances JAK-STAT signaling.", LEUKEMIA 24 JUL 2017, 24 July 2017 (2017-07-24), XP002778711, ISSN: 1476-5551
Download PDF:
Claims:
Claims

A method for producing a second recombinant protein said method comprising providing an eukaryotic host cell expressing a mutant RPL10 protein, wherein wild type RPL10 is not expressed or its expression is silenced within said eukaryotic cell, and introducing a nucleic acid encoding a recombinant protein under conditions that allow expression of the recombinant polypeptide.

The method according to claim 1, further comprising the step of recovering the expressed polypeptide from the host cell.

The method according to claim 1 or 2, wherein endogenous RPL10 DNA sequence has been deleted from the genome, wherein the expression of endogenous RPL10 has been silenced or wherein the endogenous RPL10 DNA sequence has been altered to encode the mutant RPL10.

The method according to any one of claims 1 to 3, wherein mutant RPL10 protein comprises one or more amino acids substitutions at positions 33, 66, 70, 98, 123 or an equivalent mutation at another position in the protein leading to increased expression of a recombinant protein.

The method according to any one of claims 1 to 4, wherein mutant RPL10 protein comprises one or more amino acids substitutions at positions 33, 66, 70, 98, and 123.

The method according to any one of claims 1 to 5, wherein mutant RPL10 protein comprises one or more amino acids substitutions at selected from the group consisting of 33Val, 66Gly, 70Met or 70Leu, 98Ser or 98Cys, and 123Pro.

The method according to any one of claims 1 to 6, wherein the mutant RPL10 protein contains the mutation 98Ser.

The method according to any one of claims 1 to 7, wherein the cells are grown in a medium comprising an antioxidant, such as NAC. The method according to any one of claims 1 to 8, wherein NOTCH 1 signalling is activated in the host cells.

The method according to claim 9, wherein NOTCH 1 signalling is activated by transduction with a retroviral vector harbouring MSCV plasmids encoding activated intracellular NOTCH 1 (NOTCH 1-ICN) [] .

The method according to any one of claims 1 to 10, wherein the recombinant eukaryotic cell host is a mammalian cell.

The method according to claim 11, wherein the mammalian cell is a cell line selected from the group consisting of CHO, COS, Vero, Hela, BHK, HEK293, Hek293T, HKB-11, MEF and Sp-2 cell lines.

Use of an eukaryotic cell expressing a mutant RPL10 protein, wherein wild type RPL10 is not expressed or its expression is silenced within said eukaryotic cell for the expression of recombinant proteins.

A eukaryotic cell expressing a mutant RPL10 protein, wherein wild type RPL10 is not expressed or its expression is silenced within said eukaryotic cell, characterized by activated NOTCH 1 signalling.

The cell according to claim 14, comprising intracellular NOTCH 1 (NOTCH 1- ICN).

A method for identifying RPL10 mutations which results in increased expression of a recombinant protein in a cell comprising such RPL10 compared to a cell comprising wild type RPL10,

introducing in a first cell a first nucleic acid encoding a mutated RPL10 protein under conditions that allow expression of the mutated, wherein wild type RPL10 is absent,

introducing in said first cell a second nucleic acid encoding a detectable protein under conditions that allow expression of the detectable protein, introducing in a second cell a third nucleic acid encoding a wild type RPL10 protein under conditions that allow expression of the mutated, introducing in said second cell the second nucleic acid encoding the detectable protein under conditions that allow expression of the detectable protein,

cultivating the first cell and the second cell under the same conditions and comparing the amount of detectable protein produced, wherein a cell with an RPL10 mutant wherein an increase of at least 2% (w/w) of detectable protein is obtained, is selected as host for the expression of recombinant proteins.

Use of a cell selected in the method of claim 16, in a method according to claim 1.

18. A non-human transgenic animal expressing a R98S RPL10 mutation. 19. The non-human transgenic animal according to claim 18, which is a conditional transgenic animal expressing the R98S RPL10 mutation via CRE-recombinase.

20. The non-human transgenic animal according to claim 18 or 19, wherein the R98S RPL10 mutation is tissue specific.

Description:
RECOMBINANT PROTEIN PRODUCTION SYSTEM FIELD OF THE INVENTION

The present disclosure relates generally to an improved protein expression system in the field of recombinant protein expression. The invention relates to a system showing an improved yield and quality of protein production and methods for increasing production of a protein produced by cultured cells, particularly cultured eukaryotic cells. BACKGROUND

Improved methodologies for maximizing protein production through recombinant gene expression is an on-going effort in the art. Of particular interest is the development of methodologies that maximize recombinant expression of biologically active proteins for producing commercially useful quantities of these proteins. While prokaryotic, typically bacterial, host cell systems have proven capable of generating large quantities of recombinant proteins, these hosts suffer from a number of disadvantages, including an inability to glycosylate proteins, inefficient cleavage of "pre" or "prepro" sequences from proteins (e.g., inefficient post translational modification), and a general inability to secrete proteins. Consequently the art has sought eukaryotic host systems, typically mammalian host cell systems, for mammalian protein production. One feature of such systems is that the protein produced has a structure most like that of the natural protein species, and, purification often is easier since the protein can be secreted into the culture medium in a biologically active form.

Proteins are commercially useful in a large variety of applications, including diagnostic, pharmacological, therapeutic, nutritional, and research applications. Large scale production of proteins for commercial use can be both laborious and expensive. Moreover, facilities that produce proteins for pharmacological use can incur significant cost to obtain building and regulatory approval. Thus, even small increases in the efficiency with which a protein can be produced are commercially valuable because of the limited number of facilities available for production and the expense of production.

Cultured mammalian cells have been used for production of some proteins, particularly recombinant proteins intended for pharmacological use, and numerous adjustments of culture conditions have been shown to affect the quantity and quality of protein produced. Chen et al. (2006) Appl Environ Microbiol. 72, 4001-4006 identified a tomato (Lycopersicon esculentum) gene encoding a QM-like protein (tQM) and found that stable expression of tQM conferred protection against oxidative damage from H202, paraquat, and heat. The same authors disclose in US20080184384 an eukaryote expression system with QM overexpression to increase the yield of a recombinant protein due to a reduced stress tolerance. The QM protein has homologues in yeast and vertebrates. The human homologue is known as RPL10. De Keersmaecker K. et al. (2013) Nat Genet. 45, 186-190 identify mutations in the ribosomal gene RPL10 in T-cell acute lymphoblastic leukaemia.

SUMMARY OF THE INVENTION

The present invention provides a new system that is designed to improve the yield and quality of protein production, particularly a recombinant protein, from cultured eukaryotic, in particular mammalian cells.

Although engineered RPL10 eukaryotic cells show a ribosome biogenesis defect, the present invention solves the problems of the related art by increasing translation efficiency and translation fidelity, thereby providing a system with an improved yield and quality of protein production.

The invention is summarized in the following statements:

1. A method for producing a second recombinant protein said method comprising providing an eukaryotic host cell expressing a mutant RPL10 protein, wherein wild type RPL10 is not expressed or its expression is silenced within said eukaryotic cell, and introducing a nucleic acid encoding a recombinant protein under conditions that allow expression of the recombinant polypeptide.

2. The method according to statement 1, further comprising the step of recovering the expressed polypeptide from the host cell.

3. The method according to statement 1 or 2, wherein endogenous RPL10 DNA sequence has been deleted from the genome, wherein the expression of endogenous RPL10 has been silenced or wherein the endogenous RPL10 DNA sequence has been altered to encode the mutant RPL10.

4. The method according to any one of statements 1 to 3, wherein mutant RPL10 protein comprises one or more amino acids substitutions at positions 33, 66, 70, 98, 123 or an equivalent mutation at another position in the protein leading to increased expression of a recombinant protein. 5. The method according to any one of statements 1 to 4, wherein mutant RPLIO protein comprises one or more amino acids substitutions at positions 33, 66, 70, 98, and 123.

6. The method according to any one of statements 1 to 5, wherein mutant RPLIO protein comprises one or more amino acids substitutions at selected from the group consisting of 33Val, 66Gly, 70Met or 70Leu, 98Ser or 98Cys, and 123Pro.

7. The method according to any one of statements 1 to 6, wherein the mutant RPLIO protein contains the mutation 98Ser.

8. The method according to any one of statements 1 to 7, wherein the cells are grown in a medium comprising an antioxidant.

9. The method according to any one of statements 1 to 8, wherein NOTCH 1 signalling is activated in the host cells.

10. The method according to statement 9, wherein NOTCH 1 signalling is activated by transduction with a retroviral vector harbouring MSCV plasmids encoding activated intracellular NOTCH 1 (NOTCH 1-ICN) for example as shown in SEQ ID NO: 5.

11. The method according to any one of statements 1 to 10, wherein the recombinant eukaryotic cell host is a mammalian cell.

12. The method according to statement 11, wherein the mammalian cell is a cell line selected from the group consisting of CHO, COS, Vero, Hela, BHK, HEK293, Hek293T, HKB-11, MEF and Sp-2 cell lines.

13. Use of an eukaryotic cell expressing a mutant RPL10 protein, wherein wild type RPL10 is not expressed or its expression is silenced within said eukaryotic cell for the expression of recombinant proteins.

14. A eukaryotic cell expressing a mutant RPL10 protein, wherein wild type RPL10 is not expressed or its expression is silenced within said eukaryotic cell, characterized by activated NOTCH 1 signalling.

15. The cell according to statement 14, comprising intracellular NOTCH 1 (NOTCH 1-ICN) for example as shown in SEQ ID NO: 5] .

16. A method for identifying RPL10 mutations which results in increased expression of a recombinant protein in a cell comprising such RPL10 compared to a cell comprising wild type RPL10,

introducing in a first cell a first nucleic acid encoding a mutated RPL10 protein under conditions that allow expression of the mutated, wherein wild type RPL10 is absent, introducing in said first cell a second nucleic acid encoding a detectable protein under conditions that allow expression of the detectable protein,

introducing in a second cell a third nucleic acid encoding a wild type RPL10 protein under conditions that allow expression of the mutated,

introducing in said second cell the second nucleic acid encoding the detectable protein under conditions that allow expression of the detectable protein.

Cultivating the first cell and the second cell under the same conditions and comparing the amount of detectable protein produced, wherein a cell with an RPL10 mutant wherein an increase of at least 2% (w/w) of detectable protein is obtained, is selected as host for the expression of recombinant proteins.

17. Use of a cell selected in the method of statement 16, in a method according to statement 1.

18. A non-human transgenic animal expressing a R98S RPL10 mutation.

19. The non-human transgenic animal according to statement 18, which is a conditional transgenic animal expressing the R98S RPL10 mutation via CRE- recombinase.

20. The non-human transgenic animal according to statement 18 or 19, wherein the R98S RPL10 mutation is tissue specific. Further scope of applicability of the present invention will become apparent from the detailed description given hereinafter. However, it should be understood that the detailed description and specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description. It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed. BRIEF DESCRIPTION OF DRAWINGS

The present invention will become more fully understood from the detailed description given herein below and the accompanying drawings which are given by way of illustration only, and thus are not limitative of the present invention, and wherein :

Figure 1 is a schematic overview of Programmed -1 ribosomal frameshifting (-1 PRF) assays. Wild type and mutant Ba/F3 cells were mock-electroporated or electroporated with plasmids harbouring the in frame control, the out of frame control, or a -1 PRF signal from the human IL7R gene between the upstream renilla and downstream firefly luciferase open reading frames.

Figure 2 is a schematic overview of nonsense and missense suppression analysis. Nonsense suppression analysis is carried out via a dual luciferase assay wherein a STOP codon is introduced between the renilla and firefly luciferase genes. In such a construct, the production of the firefly luciferase protein is dependent on a STOP codon read-through event. Missense suppression is assayed by employing a construct containing the R218S single base substitution in the firefly luciferase active site. In such a construct, the activity of firefly luciferase is dependent on the incorporation of a near-cognate arginine instead of a cognate serine.

Figure 3 shows increased translation efficiency in RPL10 R98S cells A) Representative western blot analysis of de novo protein labelling with AHA-Biotin detected with streptavidin-HRP antibody (* : p<0.05; T-test) on Ba/F3 WT versus R98S cells. B) Comparison of MFI (mean fluorescent intensity) of a GFP reporter expressed in RPL10 R98S versus WT expressing cells. Left: Ba/F3 cells; Right: Jurkat cells C) Representative western blot detection of phospo-eIF2a in RPL10 WT and R98S expressing Ba/F3 cells. D) Analysis of the distribution of indicated mRNAs in polysome profiles from RPL10 WT and R98S Ba/F3 expressing cells. Figure 4 shows that RPL10 R98S cells have a higher translation accuracy. Results from dual luciferase reporter assays testing -1 PRF levels (frameshifting), STOP codon read-trough and missense reading in Ba/F3 cells expressing WT or RPL10 R98S. ** p<0.01

Figure 5 illustrates that RPL10 R98S cells show reduced chymotrypsin-like and caspase-like protease activity. Chymotrypsin-Like, Trypsin-Like and Caspase- Like proteasomal activity of Ba/F3 (top) or Jurkat (bottom) cells expressing either WT or R98S RPL10. P-values were calculated using a T-test.

Figure 6 illustrates that NOTCH 1-ICN signalling rescues RPL10 R98S associated cell proliferation defects without affecting translation fidelity. A) Proliferation curves of Ba/F3 cells expressing indicated constructs. B) Results from dual luciferase reporter assays testing -1 PRF levels (frameshifting), STOP codon read-trough and missense reading in cells expressing indicated constructs.

Figure 7 shows a schematic representation of the conditional RpllO cKI R98S mouse model

Figure 8. shows that RPL10 R98S expressing mouse hematopoietic cells display increased expression and phosphorylation of Jak-Stat signalling components. Representative immunoblots of expression and phosphorylation of Jak-Stat proteins in hematopoietic cells derived from RpllOcKI R98S (labelled as WT in the figure) and MX-Cre Rpl lOcKI R98S (labelled as R98S in the figure) mice. Figure 9 shows a mass spectrometry screen for proteins and pathways showing differential expression between RPL10 WT and R98S cells. A) Hierarchical clustering analysis of the differential proteome results of the three analyzed RPL10 Wild type (WT#29, WT#36 and WT#28) and RPL10 R98S mutant (R98S# 13, R98S#35 and R98S# 11) cell clones. B) Volcano plot of the quantitative proteomics data comparing RPL10 WT and R98S samples. The dashed line illustrates the cut-off for significance (p<0.01; T-test). Red dots represent the 178 proteins that are significantly upregulated in the R98S samples, green dots correspond to the 68 proteins that are significantly downregulated. C) Pathways/processes that are significantly up- (left) or downright) regulated in R98S samples. The numbers in the pie chart represent the number of significant pathways corresponding to each process that are up or down in the cells.

Figure 10 illustrates that RPL10 R98S cells express altered levels of Jak-Stat signalling components. A) left: GSEA plot illustrating enrichment of JAK-STAT pathway proteins in the proteins upregulated in RPL10 R98S cells. FDR: false discovery rate, right: Volcano plot of the Jak-Stat signalling mediators identified by mass spectrometry. The dashed line illustrates the cut-off for significance (p<0.05; T-test). Red dots represent proteins that are significantly upregulated in the R98S samples, green dots correspond to proteins that are significantly downregulated in the R98S cells. B) Left: Immunoblot validation of differential expression of Jak-Stat pathway genes in three independent clones of RPL10 R98S versus WT expressing cells. The figure shows a representative blot of 3 independent experiments. Right: Quantification of the immunoblot validations. Signal of the JAK-STAT pathway proteins was normalized for loading. The quantification represents the average +/- standard deviation of a representative experiment comparing 3 independent RPL10 WT versus 3 independent R98S cell clones. P-values were calculated using a T-test. C) Quantification of JAK-STAT mRNA levels in RPL10 WT versus R98S expressing Ba/F3 cells. Results were obtained by mRNA sequencing on 3 RPL10 WT versus R98S Ba/F3 clones. The represented RNA levels are DESeq2-normalized counts relative to the WT. Significant changes (FDR<0.1) in RNA levels between R98S and WT were estimated using the DESeq R package. *p<0.05, **p<0.01, ***p<0.01.

Figure 11 shows that RPL10 R98S T-ALL patient samples have elevated expression of the JAK-STAT cascade, A) Immunoblot analysis of JAK-STAT pathway protein expression in 3 RPLIO WT and 3 RPLIO R98S mutant human T- ALL xenograft samples. Only those components of the JAK-STAT pathway for which significant changes were observed are represented. B) Quantification of the immunoblots shown in panel (A). The bars indicate the average +/- standard deviation of 3 independent RPLIO WT T-ALL patient samples versus 3 RPLIO R98S positive patient samples. P-values were calculated using a T-test. *p<0.05, **p<0.01.

Figure 12 shows that the Jak-Stat pathway genes contain functional -1 PRF signals, the frameshifting levels on some of which are influenced by the RPLIO R98S mutation. A) Results of enrichment analysis within all human genes containing predicted -1 PRF signals and extracted from the PRF database (Belew AT. et al. (2008) BMC Genomics 9, 339). Analysis was performed using the G : profiler (Reimand, J. et al. (2016) Nucleic Acids Res 44, W83-W89) software and KEGG databases, and statistical significance was calculated using Fisher's one-tailed test. B) Results from dual luciferase reporter assays (explained in Figure 18A) testing -1 PRF levels on computationally predicted -1 PRF signals in the indicated mouse genes. The out-of-frame (OOF) is a negative control. Assays were performed in Ba/F3 cells expressing RPL10 WT or R98S. The bars indicate the average +/- standard error of at least 5 biologically independent measurements. C) Percentages of -1 PRF on human IL7RA and JAK1 mRNAs as determined by dual luciferase reporter assays performed in WT versus R98S Ba/F3 cells. Plots show the average +/- standard error of at least 5 biologically independent measurements.

Figure 13 illustrates that RPL10 R98S expressing cells show altered proteasome expression and activity and enhanced stability of Jakl. A) Immunoblot analysis of Psmb9 and PsmblO expression in three independent clones of RPL10 R98S versus WT expressing cells. B) Quantification of the immunoblot validations shown in panel (A) C) Chymotrypsin-like, caspase-like and trypsin-like proteasomal activity of Ba/F3 cells expressing either WT or R98S RPL10. Plots show the average +/- standard deviation of three independent experiments comparing 3 biologically independent RPL10 WT versus 3 independent R98S cell clones. D) Relative proliferation of RPL10 WT and R98S cells treated with the indicated proteasome inhibitors measured using the ATPIite luminescence assay (Perking Elmer). The panel shows the average +/- standard deviation of a representative experiment comparing 3 biologically independent RPL10 WT versus 3 independent R98S cell clones. E) Immunoblots illustrating stability of Jakl and Csf2rb/2 proteins as assessed by cycloheximide chase assays. F) Quantification of the immunoblots shown in panel (E). CHX: cycloheximide. The quantification represents the average +/- standard deviation of three independent experiments comparing 3 biologically independent RPL10 WT versus 3 independent R98S cell clones. P-values were calculated using a T-test *p<0.05, **p<0.01, ***p<0.001.

Figure 14 shows Sanger sequences showing RpllO WT and R98S expression in Ba/F3 cells. Representative Sanger chromatogram of cDNA obtained from RPL10 WT (A) or R98S (B) expressing Ba/F3 cells.

Figure 15. Validation of -1 PRF signals by dual luciferase assays. Results from dual luciferase reporter assays in Hek293T cells testing -1 PRF levels on the indicated mouse and human PRF signals. Error bars denote standard errors.

**p<0.01 compared to the out of frame (OOF) control (T-test).

Figure 16 illustrates that RPL10 R98S mouse hematopoietic cells show upregulation of immunoproteasome subunits. Representative immunoblots showing protein expression levels of the specific subunits PsmblO and Psmb9 of the immunoproteasome in hematopoietic cells derived from Rpl lO cKI R98S (labelled as WT in the figure) and MX-Cre RpllO cKI R98S (labelled as R98S in the figure) mice.

Figure 17 shows thatRPLlO R98S mutant cells do not show altered poly- ubiquitination. A) Immunoblot analysis of poly-ubiquitinated proteins in RPL10 R98S versus WT expressing Ba/F3 cells. The figure shows a representative blot of 3 independent experiments. B) Quantification of the immunoblots of panel (A). The plot shows the average +/- st dev.

Figure 18 shows that the stability of Stat proteins remained unchanged within 150 minutes of CHX-chase. Immunoblots illustrating protein stability as assessed by cycloheximide (CHX) chase assays.

Figure 19 shows an alignment of human, mouse and yeast RPL10, with indication of the mutations discussed in the application.

Figure 20 illustrates that RPL10 R98S cells show enhanced survival under nutrient poor overgrowth conditions A) Viable cell counts of RPL10 R98S and WT expressing Ba/F3 cells that were cultured for 5 days without medium refreshment. In this overgrowth condition, Ba/F3 RPL10 R98S mutant cells displayed a survival benefit as compared to RPL10 WT cells. B) Ba/F3 clones were grown under normal conditions for 3 days, until their overgrowth condition started. At this moment, the culture media was replaced by media that had been exhausted by RPL10 WT Ba/F3 cells. After 2 days of culturing in the exhausted condition, the viable cell counts were determined comparing RPL10 WT clones versus R98S clones. C) Viable counts of RPL10 R98S and WT expressing JURKAT cells that were cultured for 5 days without medium refreshment. Averages +/- st dev are plotted.

Figure 21 shows a cell proliferation analysis of Ba/F3 RPL10 WT and R98S clones either untreated or treated with 5mM N-acetyl-L-cysteine (NAC). Cell proliferation is quantified using the MTS proliferation assay, in which colorimetric formazan production is measured by optical densitometry. The bar-graphs represent average +/- st dev of measured optimal density (OD) of the colorimetric MTS assay.

DETAILED DESCRIPTION

The present disclosure relates generally to an improved protein expression system in the field of recombinant protein expression. The invention relates to methods for increasing production of a protein produced by cultured cells, particularly cultured eukaryotic cells.

Several somatic ribosome defects have recently been discovered in cancer, yet their underlying oncogenic mechanisms remain poorly understood. Here we investigated the pathogenic role of the recurrent R98S mutation in the essential ribosomal protein L10 (RPL10) found in T-cell acute lymphoblastic leukaemia (T- ALL). In our T-ALL patient cohorts, the arginine to serine mutation at position 98 (R98S) in RPL10 (also known as uL16) was by far the most recurrent ribosomal defect, detected in 7.9% of paediatric patients. The R98 residue of RPL10 is centrally situated in the 60S subunit and closely approaches the ribosomal catalytic core. Yeast models have revealed that the RPL10 R98S mutation impairs ribosome assembly, translational fidelity, and cell proliferation. Although recombinant RPL10 eukaryotic cells show a ribosome biogenesis defect, they do exhibit enhanced survival properties in nutrient poor conditions and an improved yield and quality of protein production. Therefore the present invention solves the problems of the related art by increasing translation efficiency and translation fidelity, thereby providing a system showing an improved protein production.

The term "recombinant" as used herein refers to a product such as DNA or protein originating from combining genetic material from two or more different sources by means of genetic engineering.

"recombinant expression system" refers to any nucleic acid based approach or system for the expression of a gene product or gene products of interest, that has been artificially organized (man-made) of components directed toward the expression of the gene product or products. The components may be of naturally occurring genetic sources, synthetic or artificial, or some combination of natural and artificial genetic elements. Generally the gene product is a protein, polypeptide, or peptide.

A "recombinant protein" may be identical to a protein already expressed by the host cell, may be a mutated version and/or fragments of a protein already expressed by the host cell, or may be a protein (wild type of modified) origination from another organism. Recombinant proteins may further comprise additional elements such as tags or fusion parts for isolation and identification (e.g. fluorescent proteins) or other modifications.

Specific recombinant proteins in the context of the present invention are proteins with post-translational modifications such as glycosylated proteins, proteins with disulphide proteins, and the like.

Since the mutant RPL10 is a recombinant protein, the protein on the expression vector is referred to as a "further" or a second recombinant protein.

"Heterologous DNA sequence" refers to any DNA sequence that is foreign or not naturally associated with the other DNA sequences to which it is associated or linked (operably or otherwise), or a DNA sequence that is not naturally associated with the cell or organism into which it is introduced. An example of a heterologous DNA sequence is one that is used for the expression of a foreign or heterologous protein gene product in a host cell or organism. A heterologous DNA sequence can also be a part of a vector or expression construct having genetic material designed for directing the expression of a gene product, such as a protein, polypeptide, or peptide, in a host cell in vivo or in vitro, or in a cell free in vitro expression system.

Disclosed herein are:

Recombinant eukaryotic cell clones comprising a recombinant DNA sequence encoding mutant RPL10, wherein said cells expresses said mutant RPL10 and wherein the expression of the endogenous RPL10 DNA sequence has been deleted from the genome or wherein the expression of said endogenous RPL10 has been silenced or wherein the endogenous RPL10 DNA sequence has been altered to encode the mutant RPL10.

Typically said recombinant DNA sequence encodes a mutant RPL10 protein comprising one or more amino acid substitutions selected from the group consisting of I33V, E66G, I70M, I70L, R98S, R98C, H 123P and Q123P as compared to for instance the consensus RPL10 protein sequences SEQ ID NO: 2 (human), SEQ ID NO:3 (mouse), SEQ ID NO:4 (yeast) or homologues thereof. Specifically, in the recombinant DNA sequence encoding mutant RPL10 the nucleotide triplet encoding amino acid 98 has been replaced with a triplet encoding serine.

In these eukaryotic cell clones, NOTCH 1 signalling may be additionally activated by transduction with activated intracellular NOTCH 1. for example with a retroviral vector harbouring MSCV plasmids encoding activated intracellular NOTCH 1 (NOTCH 1-ICN) such as shown in SEQ ID NO: 5. Indeed, NOTCH 1 is a transmembrane protein whereby the released intracellular part can migrate to the nucleus where it functions as a transcription factor. Constructs such as the one depicted in SEQ ID NO: 6 are active as such since they are independent of NOTCH proteolytic processing.

The intracellular domain of NOTCH 1 is encoded by nucleotides 5260 to 7665 of NOTCH 1 as depicted in NCBI Reference Sequence: NM_017617.4 (version

11.12.2017). An embodiment of a protein sequence comprising the intracellular domain of NOTCH 1 is shown SEQ ID NO: 6, wherein the sequence of the intracellular domain is underlined. The double underlined sequence are the 22 N terminal amino acids of Notch 1.

The eukaryotic cell line is for example a mammalian cell line selected from the group consisting of CHO, COS, Vero, Hela, BHK, HEK293, Hek293T, Hek293S,

Hek293FT, HKB-11, MEF and Sp-2 cell lines

Herein disclosed are also methods of producing the recombinant eukaryotic cell clones described in the above paragraphs via a CRISPR-CAS9 system comprising stably or transiently transfecting eukaryotic cell lines with a Cas9 expression construct or Cas9 protein and transfecting with a gRNA targeting RpllO and with a donor oligo encoding the mutation selected from the group consisting of I33V, E66G, I70M, I70L, R98S, R98C, H 123P and Q123P as shown in SEQ ID NO: 2 for human, SEQ ID NO: 3 for mouse, and SEQ ID NO:4 for yeast.

Herein disclosed are also methods of producing the recombinant eukaryotic cell clones described in the above paragraphs via transduction with a retroviral vector encoding mutant RPL10 and knocking down endogenously expressed Rpl lO by transduction with an Rpl lO targeting shRNA construct as shown in SEQ ID NO: 6.

Herein disclosed are also methods of producing the recombinant eukaryotic cell clones described in the above paragraphs via a CRISPR-Cas9 system comprising stably or transiently transfecting eukaryotic cell lines with a Cas9 expression construct or Cas9 protein and transfecting with a gRNA targeting RpllO and with a donor oligo encoding the mutation selected from the group consisting of I33V, E66G, I70M, I70L, R98S, R98C, H 123P and Q123P wherein additionally NOTCH 1 signalling is activated by transduction with a retroviral vector harbouring MSCV plasmids encoding the active cleaved form of Notchl .

Herein disclosed are also methods of producing the recombinant eukaryotic cell clones described in the above paragraphs, via retroviral vector encoding the RPL10 mutant selected from the group consisting of I33V, E66G, I70M, I70L, R98S, R98C, H 123P and Q123P, wherein endogenously expressed RpllO is knocked down by transduction with an RpllO targeting shRNA construct as shown in SEQ ID NO:7 and wherein additionally NOTCH 1 signalling is activated by transduction with a retroviral vector harbouring MSCV plasmids encoding the active cleaved form of Notchl

In these methods of producing the recombinant eukaryotic cell clone the mutation is typically R98S.

Herein disclosed are methods for producing a second recombinant protein other than RPL10 said method comprising culturing a recombinant eukaryotic cell clone as described in the above paragraphs that has been transfected with a second recombinant DNA sequence encoding said second recombinant protein of interest and recovering the second protein so produced.

Herein disclosed are methods for increasing the protein production efficiency of an eukaryotic cell clone wherein said method comprises transforming said eukaryotic cell clone using any of the methods described above. The present invention relates to expression systems using a host cell comprising a mutant RPL10, typically mutated at one or more of positions 33, 66, 70, 98, and 123 with reference to the numbering of human RPL10 shown in figure 19. Mutations are for example mutations at position 33 e.g. into Val, Leu, Met, at position 66 into e.g. Gly, Ala, Ser, Thr, Cys, at position 70 e.g. into Val, Leu, Met, at position 98 into e.g. Gly, Ala, Ser, Thr, Cys .

More specific mutations are Ile33Val, Glu66Gly, Ile70Met or Ile70Leu, Arg98Ser, Arg98Cys, Gln/Hisl23Pro.

Equivalents are mutations of RPL10 other than Ile33Val, Glu66Gly, Ile70Met or Ile70Leu, Arg98Ser, Arg98Cys, Gln/Hisl23Pro, which lead to an increased overexpression of a recombinant protein. This can be tested by comparing the expression of a recombinant protein in a cell transfected with wild type RPL10 versus a cell transfected the mutant RPL10. Apart from point mutant, deletions of one more amino acids at the N terminus, C terminus and internally in the sequence are envisaged.

In host cells, the endogenous wild type RPL10 is mutated into one or more of the mutations Ile33Val, Glu66Gly, Ile70Met or Ile70Leu, Arg98Ser, Arg98Cys, 123Pro or an equivalent. Alternatively the wild type RPL10 is inactivated by silencing or by complete or partial deletion of the wild type RPL10 gene and an expression construct encoding RPL10 with mutations Ile33Val, Glu66Gly, Ile70Met or Ile70Leu, Arg98Ser, Arg98Cys, 123Pro or an equivalent is introduced. "Equivalents" are those mutations which lead to an increased expression as determined in the above described method.

The methods of recombinant expression can be performed in a variety of eukaryotic cells including yeast cells (e.g. saccharomyces, aspergillus, Pichia, insect cells, algae, plant cells and plants, mammalian cells (e.g. rodent, non- human primates, human). Specific embodiments are expression systems other than yeast. Commercial expression systems are available and the host cells are amenable for modifications such as transfection of a transgene RPL10 construct and the inactivation or mutation of endogenous RPL10, especially in view of the emerging Crisp/Cas technology.

The highly conserved ribosomal machinery and the high sequence similarity of RPL10 between unrelated organisms make it likely that e.g. expression of mouse or even yeast mutant RPL10 in human cells may be as effective as expression of human mutant RPL10 in human cells in order to increase overexpression of a transgene. Typically RPL10 will be from the same organism as the host cell.

The mutant RPL10 may as mentioned be a mutant cell line as isolated from a patient or a cell line used for protein expression (such as CHO) wherein the wild type RPL10 is mutated. When the mutant RPL10 is introduced in a cell with a silenced or deleted RPL10, the mutant RPL10 may be integrated in the genome or may occur on a plasmid, viral vector, or any suitable vector for protein expression. The mutant RPL10 may be under the control of an inducible promotor, or may be constitutively expressed.

The invention relates to the use of a recombinant eukaryotic cell clone expressing a recombinant DNA molecule comprising a nucleotide sequence encoding a mutant RPL10. Specific mutants of RPL10 are selected from any one of the group consisting of RPL10 I33V, RPL10 E66G, RPL10 I70M, RPLIO I70L, RPLIO R98C, and RPLIO R98S for mouse, yeast and human cells, RPLIO Q123P for mouse and human cells and RPLIO H 123P for yeast cells.

In a specific embodiment, the recombinant eukaryotic cell clone expresses a recombinant DNA molecule comprising a nucleotide sequence encoding RPLIO as shown in SEQ ID NO: l except that the nucleotide triplet encoding amino acid 98 has been replaced with a triplet encoding serine.

In specific embodiments of the invention, the recombinant RPLIO enhances survival properties in nutrient poor conditions and is combined with hyperactive NOTCH 1 signalling to rescue the RPLIO associated cell proliferation defects without affecting the translation fidelity, therefore making such recombinant eukaryotic cells much more amenable for routine recombinant protein expression. Furthermore, the RPLIO R98S associated proliferation defect can be rescued by addition of antioxidants such as e.g. 5 mM N-acetyl-L-cysteine (NAC). A suitable alternative for NAC is for example glutathione.

The invention provides the recombinant RPLIO R98S cell clone, wherein additionally NOTCH 1 signalling is activated by transduction with a retroviral vector harbouring MSCV plasmids encoding the active cleaved form of Notch 1 as shown in SEQ ID NO: 5.

The eukaryotic cell lines are conventional mammalian cell lines selected from the group consisting of CHO, COS, Vero, Hela, HEK 293, HEK 293T, BHK, HKB-11, MEF and Sp-2 cell lines. In another embodiment the eukaryotic cell line is derived from an animal model, expressing recombinant RPLIO, e.g. MEF derived according to Lei (2013) Methods Mol Biol 1031 : 59-64.

The invention provides methods for producing the recombinant eukaryotic cell clone via a CRISPR-CAS9 system comprising stably or transiently transfecting eukaryotic cell lines with a Cas9 expression construct or Cas9 protein and transfecting with a gRNA targeting RpllO and with a donor oligo encoding the R98S modification.

The invention provides methods for producing the recombinant eukaryotic cell clone via transduction with a retroviral vector encoding R98S mutant RPL10 and knocking down endogenously expressed RpllO by transduction with an RpllO targeting shRNA construct as shown in SEQ ID NO: 7.

The invention provides methods for producing the RPL10 R98S recombinant eukaryotic cell clone via a CRISPR-CAS9 system wherein additionally NOTCH 1 signalling is activated by transduction with a retroviral vector harbouring MSCV plasmids encoding the active cleaved form of Notchl

The invention provides methods for producing the recombinant eukaryotic cell clone via retroviral vector encoding R98S mutant RPL10 and knocking down endogenously expressed RpllO by transduction with an RpllO targeting shRNA construct as shown in SEQ ID NO: 7 and additionally activating NOTCH 1 signalling.

The invention provides methods for producing a recombinant product comprising growing the recombinant eukaryotic cell clone that has been transfected with a protein of interest and recovering the protein so produced.

Particular and preferred aspects of the invention are set out in the accompanying independent and dependent claims. Features from the dependent claims may be combined with features of the independent claims and with features of other dependent claims as appropriate and not merely as explicitly set out in the claims.

Thus, the claims following the detailed description are hereby expressly incorporated into this detailed description, with each claim standing on its own as a separate embodiment of this invention. EXAMPLES

Example 1: Generation of a stable cell model expressing the RPLIO R98S mutation

shRNA + overexpression model

Mouse lymphoid pro-B cells (Ba/F3) were transduced with retroviral vectors encoding WT and R98S mutant RPL10 according to standard methods. These retroviral constructs were described previously (De Keersmaecker et al. (2013) cited above). T-ALL samples containing the RPL10 R98S defect only express mutant RPL10 (De Keersmaecker et al. (2013) cited above). To mimic this, endogenously expressed RpllO was knocked down in the Ba/F3 cell lines by transduction with an RpllO targeting shRNA construct. To generate this shRNA construct, a short hairpin RNA sequence ( AACCG ACG ATCCTATTGTCATC : SEQ ID NO: 7) targeting mouse RpllO was cloned into a mir30 cassette that was introduced into the pMSCV-Neo vector. In order to obtain cells with efficient and stable knock down of endogenous RpllO, cultures were established from single cell colonies grown in Clonacell-TCS medium (Stemcell technologies). Only cultures with 90% or higher knock down of the endogenous Rpl lO as determined by qPCR were retained. Expression of RPL10 R98S and knock-down of endogenous RpllO were confirmed by Sanger sequencing of cDNA (Figure 14).

CRISPR-Cas9 model

In order to generate a stable CRISPR-Cas9 based model expressing the RPL10 R98S point mutation, Jurkat cells (DSMZ) were transduced with lentiCRISPR- Cas9, a lentiviral Cas9 encoding plasmid received from the laboratory of Prof. Jan Cools. These Jurkat cells were electroporated (6 square wave pulses, O. lms interval, 175V) with a pX321 vector (Van der Krogt et al. (2017) Haematologica, 102, 1605-1616) containing an RPL10 targeting gRNA ( 5 '-TCTTGTTG ATG - CGGATGACG-3' [SEQ ID NO:8]) and with a 127-nt donor oligo containing the R98S allele as well as 3 silent mutations to avoid re-recognition and cutting by the gRNA-Cas9 complex ( 5 '- CCTGTC AG CCCC AGC AC AGG AC AAC ATCTT- GTTAATGCTGATCACGTGAAAGGGGTGGAGCCGCACCCGGATATGGAAGCCATCTTTG CCACAAC I I I I I ACCATGTACTTATTGGCACAAATTCGGGCA-3' [SEQ ID NO:9] ; Integrated DNA Technologies). Following electroporation, cells were incubated for 48 hrs in the presence of 500 nM SCR7 (Sigma-Aldrich), followed by single cell sorting (BD FACSAria I) into 96 well plates. Outgrowing clones were expanded and screened for the desired modification using a PCR approach. Since Rpl lO is located on the X chromosome and our Jurkat cells only contained one X chromosome, a PCR screen was done using allele specific primers (Fw_WT: 5'- CTTCCACGTCATCCGCATC-3' [SEQ ID NO: 10] ; FW_R98S : 5'-CCTTTCACGT- G ATC AG C ATT- 3 ' [SEQ ID NO: 11]; Rev_WTandR98S: 5'-GCTCTGATAA- AATAATGCAAGCCTA- 3 ' [SEQ ID NO: 12]). Sanger sequencing was done to confirm the RPL10 status.

The exact same strategy is applicable to other human cell lines (e.g. Hek293 cells).

Subject to adaptions of the gRNA and donor oligo, mouse or hamster lines like Ba/F3, CHO can be generated.

Equally, depending on the Cas9 construct, transient Cas9 expression or transfection of Cas9 protein is possible.

Example 2: Metabolic labelling assay to determine levels of freshly synthesized proteins

Click-iT technology (Thermo Fisher Scientific) was employed to measure the levels of newly synthesized proteins in Ba/F3 cells. Briefly, 2 million cells were placed in methionine-free medium for 1 hour and then labelled for 2 hours with 35 μΜ AHA (L-azidohomoalanine). Cells were then lysed in lysis buffer (Cell Signalling Technology) and 100 μg of protein extract was used to perform the Click-iT biotin-conjugating reaction according to manufactures instruction. Subsequently, proteins were separated by gel electrophoresis and newly synthesized proteins were detected by immunoblot using a streptavidin-HRP antibody (Cell Signalling Technology).

Example 3: Analysis of GFP expression

The plasmid used for expressing RPL10 WT or R98S in the Ba/F3 shRNA/overexpression model contains an IRES-GFP expression cassette. Expression levels of this cassette were monitored by measuring the mean fluorescent intensity (MFI) on a Guava Easycyte HT flow cytometer (Millipore). For the Jurkat RPL10 WT or R98S cells, a plasmid encoding a GFP expression cassette was electroporated and expression levels of this cassette were determined by measuring the mean fluorescent intensity (MFI) on a Guava Easycyte HT flow cytometer (Millipore) at 24 hours after electroporation.

Example 4: Polysomal analysis

Ba/F3 cell pellets were lysed in ice-cold lOOmM KCI, 20 mM Hepes (Life technologies), 10 mM MgCI2, 1 mM DTT, 1% sodium deoxycholate, 1% NP-40 (Tergitol® solution, Sigma-Aldrich), 100 μg ml-1 cycloheximide, 1% Phosphatase Inhibitor Cocktail 2 (Sigma-Aldrich), 1% Phosphatase Inhibitor Cocktail 3 (Sigma-Aldrich), 1% Protease Inhibitor Cocktail (Sigma-Aldrich), 400 U ml-1 RNasin (Promega). After 10 minutes incubation on ice, lysates were centrifuged 5 minutes at 13,000 rpm and the resulting supernatant was loaded onto 10-60% sucrose density gradients (100 mM KCI, 20 mM Hepes, 10 mM MgCI2). Gradients were then centrifuged in a SW40Ti rotor (Beckman Coulter) at 37,000 rpm for 150 minutes and polysomal fractions were monitored through a live OD254 nm measurement on a BioLogic LP System (BIO-RAD) followed by collection of 12 fractions. RNA from each of these fractions was extracted and distribution of particular mRNAs over the fractions was analyzed by qPCR.

Example 5: Translation fidelity assays

Programmed -1 ribosomal fra mesh if ting (-1 PRF) assays

Dual luciferase assays (Promega), and statistical calculations were performed as previously described (Grentzmann et al. (1998) RNA 4, 479-486 ; Harger and Dinman (2003) RNA 9, 1019-1024; Jacobs and Dinman (2004) Nucleic acids Res. 32, el60). Briefly, wild type and mutant Ba/F3 cells were mock- electroporated or electroporated with plasmids harbouring the in frame control, the out of frame control, or a -1 PRF signal from the human IL7R gene between the upstream renilla and downstream firefly luciferase open reading frames (Figure 1) [Belew, A.T. et al. (2014) Nature 512, 265-269] . In the -1 PRF signal containing construct, the production of the firefly luciferase protein is dependent on a frameshifting event.

Three million cells were electroporated with 15 μg of each plasmid in 400 μΙ serum-free medium in 4 mm cuvettes using an exponential decay protocol (300V, 950 [ F) r and immediately transferred to 4 ml. of pre-warmed recovery medium (10% FBS, IL3, sodium pyruvate and MEM non-essential amino acids). Twenty four hours later, cells were lysed in 100 μΙ lysis buffer, and luciferase readings were collected in 96 well half-area white plates (10 μΙ of lysate per read) on an EnSpire plate reader with two injectors (PerkinElmer). The percent frameshifting was determined by dividing the firely/ renilla signal ratio in cells containing a -1 PRF construct or the out of frame control by the firefly/ renilla signal ratio in the corresponding cells containing the in frame control construct. Nonsense and missense suppression assays

Nonsense suppression analysis is carried out in the same way as described above, except a STOP codon is introduced between the renilla and firefly luciferase genes. In such a construct, the production of the firefly luciferase protein is dependent on a STOP codon read-through event (Figure 2).

Missense suppression is assayed by employing a construct containing the R218S single base substitution in the firefly luciferase active site. In such a construct, the activity of firefly luciferase is dependent on the incorporation of a near- cognate arginine instead of a cognate serine (Figure 2).

Example 6: Proteasome activity assay

Proteasome activity is tested on Ba/F3 and Jurkat cells expressing RPL10 WT or R98S using Proteasome-Glo Chymotrypsin-Like, Trypsin-Like and Caspase-Like Cell-Based Assays (Promega) according to the manual included in the kit (Figure 5).

Example 7: The RPLIO R98S mutation increases translation efficiency To characterize the effects of RPL10 R98S on ribosome function, we first looked at cellular levels of newly synthesized proteins using an assay that measures incorporation of a labelled amino acid in all cellular proteins. These experiments indicated that Ba/F3 cells expressing RPLIO R98S produce 25% more de novo proteins when compared to RPLIO WT cells (Figure 3A). In agreement with this, expression of the GFP reporter present in our RPLIO expression constructs was higher in the RPLIO R98S expressing cells (average MFI of 1358 in RPLIO WT cells versus 2154 in RPLIO R98S cells Figure 3B, left). Similarly, Jurkat cells electroporated with a GFP reporter plasmid displayed an elevated GFP MFI upon RPLIO R98S expression (average MFI of 2174 in RPLIO WT cells versus 3764 in RPLIO R98S cells; Figure 3B, right) This increase in detected proteins could be due to an upregulation in protein translation by the ribosome. In agreement with this, the mutant expressing Ba/F3 cells show downregulated phospho-eIF2a levels (Figure 3C). Upregulation of translation efficiency was further supported by the fact that all analyzed transcripts shifted towards higher polysomal fractions in polysome profiling experiments on Ba/F3 cells, indicating a general increase in translation efficiency (Figure 3D).

Example 8: RPLIO R98S Ba/F3 cells show increased translation fidelity

In order to verify if the RPLIO R98S mutation influences translation fidelity by the ribosome, we performed dual luciferase reporter assays in which firefly activity depends on the fidelity of amino acid incorporation, reading of a STOP codon, or programmed -1 ribosomal frameshifting (see figures in methods section above). RPLIO R98S expressing cells showed a 70-80% reduction of normalized firefly activity in all three assays, indicating higher translation accuracy (Figure 4). Example 9: RPLIO R98S cells show reduced protein degradation

In order to investigate whether the RPL10 R98S associated changes in ribosome function have implications on degradation of cellular proteins, the three major protease activities of the proteasome were analysed in RPL10 WT versus R98S cells. RPL10 R98S Ba/F3 cells displayed 28% and 23% decreases in chymotrypsin-like and caspase-like activity of the proteasome as compared to WT (Figure 5, top). Similarly, RPL10 R98S expressing Jurkat cells had a 17% and 25% decrease in proteasomal chymotrypsin-like and caspase-like activity as compared to WT (Figure 5, bottom). Example 10: Hyperactive NOTCH1 signalling rescues RPL10 R98S associated cell proliferation defects without affecting translation fidelity

RPL10 WT and R98S expressing Ba/F3 cells were transduced with retroviral vectors harbouring MSCV plasmids encoding either the active cleaved form of Notchl (intercellular Notch - ICN) (SEQ ID NO: 5) followed by an IRES sequence and an mcherry reporter or mCherry alone. After sorting for mCherry, cells were plated at 100,000 cells/ml, and cell numbers were counted every 24 hours on a on a Guava Easycyte HT flow cytometer (Millipore). Translational fidelity in RPL10 WT and R98S cells harbouring the ICN-IRES-mCherry or mCherry constructs was assayed as described above.

Mutant RPL10 R98S Ba/F3 cells display a clear growth defect as compared to wild type cells during the exponential growth phase (Figure 6A). The exogenous addition of NOTCH 1-ICN did not have any effect on the growth rate of wild type cells, but it rescued the growth defect of mutant cells (Figure 6A). However, while the growth defect of mutant cells was rescued with NOTCH 1-ICN, their translational alterations and increased ribosomal accuracy remained (Figure 6B).

Example 11: Treatment with N-acetyl-L-cyste ' i ' ne (NAC) rescues RPLIO R98S associated proliferation defects

RPL10 WT and R98S expressing Ba/F3 cells were plated at 0.25 x 10 6 cells/ ml and were grown for 48 hours under exponential growth conditions in the presence or absence of 5 mM N-acetyl-L-cysteine (NAC). The MTS cell proliferation assay in which colorimetric formazan production is measured by optical densitometry was used as read-out for cell numbers. R98S Ba/F3 cells displayed lower OD values indicating reduced cell numbers as compared to WT cells in the absence of NAC (due to the proliferation defect in exponential growth condition). These reduced cell numbers were entirely rescued by addition of 5 mM NAC (Figure 21).

Examples 10 and 11 support that the growth defect associated with expression of an RPL10 mutation construct, can be rescued by expression of the NOTCH 1- ICN allele or by addition of antioxidant agents such as NAC, suggesting that these manipulations will have a positive effect on protein expression.

Example 12: RPLIO R98S conditional knock-in mouse model

Generation of a conditional RpllO R98S knock-in mouse line (RPLlOcKI R98S) was performed by the company Polygene AG (Rumlang, Switzerland). In this model, the wild type genomic region of RpllO encompassing exons 5 up to 7 was flanked with loxP sites. Downstream of this cassette, a mutant version of exon 5 encoding the R98S mutation was placed (Figure 7, targeted allele RPL10 cKI R98S ). In this configuration, prior to Cre recombinase mediated recombination, the wild type gene containing its wild type introns including Snora70are expressed. After Cre recombination, the genomic configuration is identical to wild type with the exception of a remaining loxP site, and the introduced R98S point mutation (Figure 7, Cre RPL10 cKI R98S ).

To generate the targeting vector, the homology arms from a C57BI/6-derived BAC were subcloned, and a synthetic 1293 bp loxP-flanked cassette containing the RpllO genomic sequence encompassing exons 5-7 spiked with 3 silent changes was used. Upstream of the upstream loxP site, an FRT-flanked neomycin selection cassette was added for selection in cell culture. The homologous arms had sizes of 2.5 and 3.2 kbp. Targeting of C57BI/6N-derived ES cells (PolyGene AG) yielded 8 out of 388 clones with correct configuration verified by PCR and Southern hybridization. Blastocyst microinjection of these clones into C57BI/6grey-derived embryos (PolyGene AG) resulted in chimeric mice that transmitted to germ line after mating with C57BI/6grey Flp deleter mice. Neo-less correct heterozygous Fl genotypes were verified by PCR.

Rpl lO R98S conditional knock-in mice were crossed to MX-Cre C57BI/6 mice (B6.Cg-Tg(Mxl-cre)lCgn/J strain Jackson Laboratories). For the described experiments, lineage negative cells were isolated (EasySep Mouse Hematopoietic Progenitor Cell enrichment kit, Stemcell Technologies) from 6-8 weeks old male mice carrying the conditional RpllO R98S allele together with an MX-Cre allele (MX-Cre RpllO cKI R98S ) and from conditional RpllO R98S controls (Rpl lO cKI R98S ). Wells were plated at 2000 cells/ml in Methocult GF M3534 medium (Stemcell Technologies) containing 1250 units/ml of IFNP (R&D systems) to induce recombination of the conditional RPL10 R98S allele. After 10-15 days, cells were collected and lysed in lysis buffer (Cell Signalling Technology) with addition of 5 mM Na3V0 4 and protease inhibitors (Complete, Roche) and analyzed via immunoblotting. Expression of the R98S mutation upon Cre recombination was confirmed by Sanger sequencing of cDNA of the region encoding RpllO R98. In mouse hematopoietic cells derived from a conditional RpllO R98S knock-in mouse model, elevated protein expression and/or phosphorylation was confirmed for Csf2rb/2, Jakl, Stat3, Stat5 and Erk in (Figure 8). Example 13:RPL10 R98S cells show enhanced survival under nutrient poor overgrowth conditions

RPL10 R98S and WT expressing Ba/F3 were cultured for 5 days without medium refreshment. In this overgrowth condition, Ba/F3 RPL10 R98S mutant cells displayed a survival benefit as compared to RPL10 WT cells (Figure 20A). To ascertain that this survival benefit was independent of differences in residual nutrients due to the growth defect of RPL10 R98S cells in exponential phase (De Keersmaecker et al. (2013) cited above), the RPL10 WT and R98S Ba/F3 cells were cultured in nutrient deprived conditioned media from overgrown WT Ba/F3 cells, leading to an even stronger survival benefit for the RPL10 R98S cells (Figure 20B).

Similarly, RPL10 R98S expressing Jurkat cells in overgrowth condition (no medium exchange for 5 days) displayed an enhanced survival over RPL10 WT expressing JURKAT cells (Figure 20C).

The present example illustrates that cells with mutant RPL10 have an even better chance in survival under stress conditions. Use of cells with RPL10 mutations for protein expression is expected to give a higher protein yield, in view of the higher biomass that can be expected. It is expected that this advantage is even more pronounced under stress conditions such as the synthesis of proteins of an expression construct, which puts a significant metabolic burden on a cell.

Example 14: The T-cell leukaemia associated ribosomal RPLIO R98S mutation enhances JAK-STAT signalling

Quantitative proteomics

Cells derived from 3 monoclonal Ba/F3 cultures expressing WT or R98S RPL10 were lysed in lysis buffer (Cell Signalling Technology) with addition of 5mM Na3V04 and protease inhibitors (Complete, Roche). Twenty μg of protein as determined by the Bradford method were processed for quantitative proteomics as described in the supplementary methods. The entire list of identified proteins was ranked according to log2 fold changes and used as input for GSEA against the MSigDB C2 KEGG gene sets [Subramanian A. & Tamayo P. (2005) Proc Natl Acad Sci USA. 103, 155545-155550; Mootha VK. et al. (2003) Nat Genet. 34, 267-273] . Only GSEA results with a FDR q-value <0.25 were considered.

Programmed -1 ribosomal frameshifting (-1 PRF) assays

Dual luciferase assays (Promega), and statistical calculations were performed as follows. Briefly, Hek293T cells or Ba/F3 cells were transfected with plasmids harbouring the in frame control, the out of frame control, or a -1 PRF signal between the upstream renilla and downstream firefly luciferase open reading frames (Figure 1). Further details on the assay are described in example 5.

Quantitative proteomics

Cells derived from 3 monoclonal Ba/F3 cultures expressing either WT or R98S RPL10 were lysed in lysis buffer (Cell Signalling Technology) with addition of 5 mM Na3V04 and protease inhibitors (Complete, Roche). Twenty μg of protein as determined by the Bradford method was run through a 12% SDS-PAGE (Bio- Rad) and Coomassie stained using Simply Blue Safe Stain (Invitrogen). Entire SDS-PAGE gel lanes were sliced into pieces and proteins were reduced-alkylated before overnight digestion using Trypsin/LysC Mix (Promega). The resulting peptides were extracted and vacuum dried. Peptides were desalted on C18 StageTips and each sample was fractionated using SCX StageTips. All fractions were again vacuum dried prior to mass spectrometric analysis. For LC MS/MS analysis, peptides were resuspended and separated by reversed-phase chromatography on a Dionex Ultimate 3000 RSLC nanoUPLC system in-line connected with an Orbitrap Q Exactive Mass-Spectrometer (Thermo Fischer Scientific). Database searching was performed using Mascot 2.3 (Matrix Science), MS-Amanda and SEQUEST in Proteome Discoverer v.1.4 against a homemade database consisting of the human RL10 R98S protein (Uniprot Accession : P27635) and in the mouse reference proteome (UniProt release 2015_04; 45182 sequences). All searches were performed with trypsin cleavage specificity, up to 2 missed cleavages were allowed, and ion mass tolerance of 10 ppm for the precursor and 0.05 Da for the fragments. Carbamidomethylation was set as a fixed modification, whereas oxidation (M), acetylation (Protein N- term), phosphorylation (STY) were considered as variable modifications. Further processing of mass spectrometry data was performed in Scaffold 4 software (Proteome Software), using the quantitative value (normalized total spectra) setting. Unsupervised average-linkage hierarchical clustering was performed in IBM SPSS 23.0 with Euclidean distance as similarity metric. The entire list of identified proteins was ranked according to log2 fold changes and used as input for GSEA against the MSigDB C2 KEGG gene sets.1,2 Only GSEA results with a FDR q-value <0.25 were considered.

Polysomal and total mRNA sequencing

Up to three polysomal and total RNA sequencing libraries were generated for each of the 3 monoclonal Ba/F3 cultures expressing either WT or R98S RPL10. An amount of 15x106 cells were pelleted by centrifugation (5 min, 1500 rpm) and were lysed in ice-cold lOOmM KCI, 20 mM Hepes (Life technologies), 10 mM MgCI 2 , 1 mM DTT, 1% sodium deoxycholate, 1% NP-40 (Tergitol solution, Sigma-Aldrich), 100 μg ml-1 cycloheximide, 1% Phosphatase Inhibitor Cocktail 2 (Sigma-Aldrich), 1% Phosphatase Inhibitor Cocktail 3 (Sigma-Aldrich), 1% Protease Inhibitor Cocktail (Sigma-Aldrich), 100 U ml-1 RNasin (Promega). After 10 minutes incubation on ice, lysates were centrifuged 5 minutes at 13,000 rpm and the resulting supernatant was loaded onto 10-60% sucrose density gradients (100 mM KCI, 20 mM Hepes, 10 mM MgCI 2 ). Gradients were then centrifuged in a SW40Ti rotor (Beckman Coulter) at 37,000 rpm for 150 minutes and polysomal fractions were monitored through a live OD254 nm measurement on a BioLogic LP System (Bio-Rad). Polysomal fractions were collected on a fraction collector 2110 (Bio-Rad), followed by addition of proteinase K (50 μg/ml) and incubation for 30 min at 37°C. NaOAc 3M, pH5.5 (1/10 volume) was added followed by RNA extraction using the phenol/chloroform method with inclusion of an extra washing step with 70% ethanol. Next generation sequencing libraries were generated from total RNA and polysomal RNA using the TruSeq Stranded mRNA Sample Prep Kit (Illumina) and were sequenced on a NextSeq instrument (Illumina) using a 50-bp single read protocol. Ribosomal RNA and tRNA contamination were computationally removed and the remaining reads were aligned by Tophat v2.0.11 to the to the mmlO mouse reference genome (GRCm38) using the transcriptome defined by Mus_musculus.GRCm38.76.6 Only reads mapping uniquely and with high quality (mapqual> 10) were retained for further analyses. Gene expression was estimated from exon-mapped reads, which were counted using HTSeq-count in union mode.

The DESeq2 R package8 was applied on total RNA to identify significant differences in transcription between R98S and WT conditions (FDR<0.1).

Translational efficiency (TE) was estimated, for each gene and within each condition, as the ratio between polysome-associated mRNA counts and total mRNA counts. Differences in TE between R98S and WT conditions were calculated as a TE(R98S)-to-TE(WT) ratio. The Babel R package9 was used to estimate the statistical significance of detected TE differences between the R98S and WT condition. This method builds a regression model of polysome mRNA counts and total mRNA counts to identify genes whose polysome mRNA levels are not sufficiently explained by their total mRNA levels. RESULTS

Four percent of proteins show significantly altered expression levels in RPL10 R98S cells

To gain insights into the mechanisms by which the RPL10 R98S mutation contributes to T-ALL development, we screened for proteins that are differentially expressed between RPL10 wild type (WT) and RPL10 R98S (R98S) expressing cells. These experiments were conducted in the mouse pro-B Ba/F3 cell line, a well-established hematopoietic model for oncogenic studies. A label- free quantitative proteomics approach was used to compare the abundance of the 5557 most highly expressed proteins, and unsupervised hierarchical clustering showed that WT and R98S samples grouped into 2 independent clusters (Figure 9A). Whereas 96% (5311/5557) of proteins did not change significantly between WT and R98S samples, 3% (178) were significantly upregulated in the R98S samples, and 1% (68) were significantly downregulated (Figure 9B). Gene Set Enrichment Analysis (GSEA) revealed that many of the overexpressed proteins in the R98S cells fit into pathways of cell metabolism, signalling, and function of the immune system. In contrast, proteins related to essential cellular processes such as DNA replication and repair, transcription and translation, were downregulated in the mutant cells (Figure 9C). RPL10 R98S cells thus showed altered expression of 4% of proteins, with differential expression of several known cancer-associated processes.

RPLIO R98S cells express elevated levels of Jak-Stat signalling mediators

Analysis of the GSEA results of the proteomic data revealed an enrichment of JAK-STAT pathway members among the upregulated proteins in the RPL10 R98S cells (Figure 10A, left). Because of the importance of the JAK-STAT pathway in T-ALL pathogenesis and cancer in general, these results prompted a more detailed investigation. Three Jak kinases (Jakl, Jak3 and Tyk2) were detected in the mass spectrometry screen, of which Jakl showed a significant 1.9-fold upregulation in the R98S cells (Figure 10A, right). Of the five Stat proteins that were detectable, only Stat2 was unchanged. All others trended towards upregulation in the R98S cells, with Stat6 attaining a statistically significant increase. The mass spectrometric data also revealed a significant 2- to 3-fold upregulation of the Csf2rb and Csf2rb2 proteins. Csf2rb corresponds to the common beta chain of the receptor for IL3, IL5 and GM-CSF, whereas Csf2rb2 is an IL3-receptor specific beta subunit in mouse [Hara T, Miyajima A. (1992) EMBO J. 11, 1875-1884] . IL3, IL5 and GM-CSF signalling are all mediated via JAK-STATs. JAK-STAT signalling is tightly controlled via negative regulators including Pias proteins, Socs proteins and phosphatases such as Ptprc and Ptpn2 [Van Vlierberghe P. (2012) J Clin Invest 122, 3398-3406] . Only Ptprc and Ptpn2 were detectable among the mass spectrometry data, and Ptprc was significantly downregulated in the R98S cells (Figure 10A, right). Immunoblot analysis of Jak-Stat cascade proteins confirmed upregulation of Csf2rb and Csf2rb2, Jakl, Statl, Stat3, Stat5a, Stat5b, Stat6, downregulation of Ptprc and no change for Ptpn2 in R98S cells (Figure 10B). As no consistent changes were detected for Tyk2 and Jak3 by immunoblot, these were not considered in further analyses. Whereas the protein changes detected for Csf2rb/2, Stat5a and Ptprc were associated with corresponding changes on mRNA level, the transcripts of the other genes were unchanged in R98S cells (Figure IOC). RPLIO R98S T-ALL patient samples have elevated expression of the JAK- STAT cascade

Immunoblotting of protein lysates from xenografted T-ALL patient samples confirmed elevated expression of JAK1 (2.1-fold) and STAT5 (4-fold) (Figure 13A-B) in RPL10 R98S mutant T-ALL samples. None of these patient samples contained any IL7R-JAK-STAT mutations. The IL3 receptor is not expressed in T- ALL samples. Instead, the IL7 receptor is important for normal T-cell development and in T-ALL [Ribeiro D et al (2012). Adv. Boil. Reg. 53, 211-222] . Expression of the IL7RA chain was 3.7-fold upregulated in the RPL10 R98S T-ALL samples compared to RPL10 WT samples (Figure 11A-B).

RPLIO R98S decreases programmed -1 ribosomal frameshifting on several JAK-STAT pathway mRNAs.

We next investigated cellular mechanisms that could be contributing to the specific JAK-STAT overexpression. In this process, c/ ' s-acting mRNA elements (-1 PRF signals) direct translating ribosomes to slip on an mRNA by one base in the 5' direction, thus establishing a new reading frame. In mammalian cells, such -1 PRF events direct translating ribosomes towards premature termination codons, resulting in destabilization of the -1 PRF signal-containing mRNA (Klare N. et al. (2007) J Mol Biol. 373, 1-10; Advani VM & Dinman JD (2015) Bioessays. 38, 21-26). -1 PRF thus serves as a mechanism to fine-tune gene expression, and in silico algorithms predict that approximately 10% of human genes contain -1 PRF signals (Belew et al. (2014) cited above; Advani et al. (cited above). We have recently shown that several cytokine receptors, including the IL7RA chain, contain functional -1 PRF signals (Belew et al. (2014) cited above). To test whether the RPL10 R98S associated differences in JAK-STAT protein expression might be influenced by alterations in -1 PRF rates, the JAK-STAT pathway was first screened in silico for predicted -1 PRF signals. Enrichment analysis revealed that, within the 10% of human genes predicted to contain -1 PRF signals, the JAK-STAT pathway was enriched for such predicted signals using both GO (p=0.00056) and KEGG (p=0.0031) databases. Specifically, 30% of genes in the JAK-STAT pathway are predicted to contain -1 PRF signals compared to the 14.8% average in other pathways (Figure 12A). Several human and mouse JAK- STAT pathway members for which RPL10 R98S associated differential protein levels were observed harbored such predicted -1 PRF signals. We validated the frameshifting-promoting activity of these signals using dual luciferase reporter constructs (Figure 1) [Jacobs & Dinman et al. (2004) cited above] . Efficient rates of -1 PRF promoted by several mouse and human sequences were confirmed in human Hek293T cells (Figure 15), as well as in mouse Ba/F3 cells (Figure 12B- C). Interestingly, frameshifting levels induced by the signals in the mouse Stat genes were three to six times reduced in R98S cells (Figure 12B). In contrast, high levels (15%) of frameshifting induced by the mouse Jakl -1 PRF signal were unaffected by the R98S mutation in the Ba/F3 cells (Figure 12B). A 1.5-fold and 2-fold decrease in frameshifting rates induced by human -1 PRF signals in IL7RA and JAK1 mRNAs were observed in cells expressing the R98S mutation (Figure 12C). In summary, we have identified functional -1 PRF signals in several mouse and human JAK-STAT genes, the frameshifting levels on some of which were influenced by the RPL10 R98S mutation.

RPLIO R98S cells show altered proteasome activity

Altered frameshifting rates can only partially explain the upregulation of the Jak- Stat cascade, as there was one -1 PRF signal (Jakl) in which frameshifting levels were unaffected by RPL10 R98S, despite a change at the protein level. Additionally, our computational tools could not identify -1 PRF signals in the mRNAs encoding some of the other proteins in which changes at the protein level were observed . The altered levels of mouse Stat proteins are also likely not entirely explained by altered -1 PRF alone. The transcriptional changes detected for Csfr2b/2, Stat5a and Ptprc (Figure IOC) might partially explain the detected protein changes, and we investigated additional potential regulation at the translational or post-translational level. Polysomal RNA-seq analysis in the Ba/F3 cell model did not reveal any significantly altered translation efficiency of Csf2rb/2, Jak or Stat mRNAs (not shown). Interestingly, the mass spectrometry data revealed up- and downregulation of several proteasomal proteins in R98S cells (p<0.05), including upregulation of PsmblO, a catalytic subunit specific for the immunoproteasome (not shown). Upregulation of PsmblO and Psmb9, another immunoproteasome specific catalytic subunit, was confirmed on immunoblots from both available mouse hematopoietic cell models (Figure 13A-B and Figure 16). In light of this, proteasome activities were assayed . R98S Ba/F3 cells displayed 28% and 23% decreases in chymotrypsin-like and caspase-like activity of the proteasome (Figure 13C). Consistent with these observations, R98S cells were more sensitive to the proteasome inhibitors bortezomib, carfilzomib and MLN9708 (Figure 13D). Interestingly, this altered proteasomal activity was not accompanied by differences in total protein polyubiquitination (Figure 17). However, chase experiments after treatment with the translational inhibitor cycloheximide revealed increased stability of Jakl, but not Csf2rb/2, in R98S cells (Figure 13E-F). Stat protein stability remained unchanged within the 150 minutes of the CHX-chase (Figure 18). Altogether, our data indicate that RPL10 R98S cells display alterations in proteasome composition and activity, which may explain the increased stability of particular proteins such as Jakl in these cells.

DISCUSSION

We explored the molecular mechanism by which the RPL10 R98S mutation may drive selective upregulation of the JAK-STAT cascade. These effects may, at least partially, be mediated via reduced levels of -1 PRF on several JAK-STAT mRNAs. Initially described in viruses, it is becoming clear that this process is also relevant in mammalian cells (Belew et al. (2014) cited above). Only a limited set of predicted mammalian - 1 PRF signals have been experimentally validated, including signals in several cytokine receptors such as IL7RA (Belew et al. (2014) cited above). We show the functionality of several additional predicted -1 PRF sig nals in mammalian cells, and that the JAK-STAT signalling cascade, downstream of cytokine receptors, is enriched for such signals. Levels of - 1 PRF can be regulated by trans-acting proteins and miRNAs (Belew et al. (2014) cited above; Anzalone AV. et al . (2016) Nat Methods.13, 453-458; Li Y. et al. (2014) Proc Natl Acad Sci USA. Ill, E2172.) It is plausible that these - 1 PRF signals function in fine-tuning and controlling immune responses, opening the possibility for oncogenic factors such as RPL10 R98S to deregulate this control mechanism . Given the relatively low rates of -1 PRF promoted by Jak-Stat signals, it is clear that the altered frameshifting levels detected here cannot fully explain the changes observed at the protein level, suggesting contribution by additional mechanisms. We propose a model in which R98S associated decreases in -1 PRF levels, combined with changes in the degradation of particular proteins and potential other mechanisms such as transcriptional regulation, leads to an oncogenic program. The specificity of the frameshifting alterations can be explained by the presence of unique -1 PRF signals in Jak-Stat genes. However, the specificity of the transcriptional and degradation phenotype is less clear. We show altered levels of specific catalytic components of the immunoproteasome. This finding, together with the altered proteasome activity in R98S cells, may indicate expression of a distinct type of proteasome, and is consistent with previously described 'mixed type' proteasomes containing constitutive as well as immuno-subunits [Klare N. et al. (2007) J Mol Biol. 373, 1-10; Dahlmann B. et al. (2002) J Mol Biol. 303, 643-653]. Different proteasome varieties show quantitative differences in cleavage efficiency of particular epitopes, which might provide a certain degree of protein specificity [Mishto M. et al. (2014) Eur J Immunol. 44, 3508-3521; Huber EM. et al. (2012) Cell. 148, 727-738]. Moreover, the proteomics screen revealed several proteins with E3 ubiquitin ligase activity to be differentially expressed between RPL10 WT and R98S cells (not shown), which may further account for the specificity of the observed degradation phenotype.

SEQUENCES DEPICTED IN THE APPLICATION

Human RPL10 cDNA sequence (SEQ ID NO:l) and protein sequence (SEQ ID NO:2)

atgggccgccgccccgcccgttgttaccggtattgtaagaacaagccgtacccaaag tct

M G R R P A R C Y R Y C K N K P Y P K S

cgcttctgccgaggtgtccctgatgccaagattcgcatttttgacctggggcggaaa aag

R F C R G V P D A K I R I F D L G R K K

gcaaaagtggatgagtttccgctttgtggccacatggtgtcagatgaatatgagcag ctg

A K V D E F P L C G H M V S D E Y E Q L

tcctctgaagccctggaggctgcccgaatttgtgccaataagtacatggtaaaaagt tgt

S S E A L E A A R I C A N K Y M V K S C

ggcaaagatggcttccatatccgggtgcggctccaccccttccacgtcatccgcatc aac

G K D G F H I R V R L H P F H V I R I N

aagatgttgtcctgtgctggggctgacaggctccaaacaggcatgcgaggtgccttt gga

K M L S C A G A D R L Q T G M R G A F G aagccccagggcactgtggccagggttcacattggccaagttatcatgtccatccgcacc K P Q G T V A R V H I G Q V I M S I R T

aagctgcagaacaaggagcatgtgattgaggccctgcgcagggccaagttcaagttt cct K L Q N K E H V I E A L R R A K F K F P

ggccgccagaagatccacatctcaaagaagtggggcttcaccaagttcaatgctgat gaa G R Q K I H I S K K W G F T K F N A D E

tttgaagacatggtggctgaaaagcggctcatcccagatggctgtggggtcaagtac atc F E D M V A E K R L I P D G C G V K Y I

cccagtcgtggccctctggacaagtggcgggccctgcactcatga

P S R G P L D K W R A L H S

Mouse RPL10 (SEQ ID NO:3)

MGRRPARCYR YCKNKPYPKS RFCRGVPDAK IRIFDLGRKK AKVDEFPLCG 50

HMVSDEYEQL SSEALEAARI CANKYMVKSC GKDGFHIRVR LHPFHVIRIN 100 KMLSCAGADR LQTGMRGAFG KPQGTVARVH IGQVIMSIRT KLQNKEHVIE 150

ALRRAKFKFP GRQKIHISKK WGFTKFNADE FEDMVAEKRL IPDGCGVKYI 200

PNRGPLDKWA LHS 213

Yeast RPL10 (SEQ ID NO : 4 )

MARRPARCYR YQKNKPYPKS RYNRAVPDSK IRIYDLGKKK ATVDEFPLCV 50

HLVSNELEQL SSEALEAARI CANKYMTTVS GRDAFHLRVR VHPFHVLRIN 100

KMLSCAGADR LQQGMRGAWG KPHGLAARVD IGQIIFSVRT KDSNKDVWE 150

GLRRARYKFP GQQKI ILSKK WGFTNLDRPE YLKKREAGEV KDDGAFVKFL 200

SKKGSLENNI REFPEYFAAQ A 221

NOTCHl-ICN (SEQ ID NO:

atgccgccgc tcctggcgcc cctgctctgc ctggcgctgc tgcccgcgct cgccgcacga ggcccgcgag gatccatcgt ctacctggag attgacaacc ggcagtgtgt gcaggcctcc tcgcagtgct tccagagtgc caccgacgtg gccgcattcc tgggagcgct cgcctcgctg ggcagcctca acatccccta caagatcgag gccgtgcaga gtgagaccgt ggagccgccc ccgccggcgc agctgcactt catgtacgtg gcgcaggccg tggagccgcc cccgccggcg cagctgcact tcatgtacgt ggcggcggcc gcctttgtgc ttctgttctt cgtgggctgc ggggtgctgc tgtcccgcaa gcgccggcgg cagcatggcc agctctggtt ccctgagggc ttcaaagtgt ctgaggccag caagaagaag cggcgggagc ccctcggcga ggactccgtg ggcctcaagc ccctgaagaa cgcttcagac ggtgccctca tggacgacaa ccagaatgag tggggggacg aggacctgga gaccaagaag ttccggttcg aggagcccgt ggttctgcct gacctggacg accagacaga ccaccggcag tggactcagc agcacctgga tgccgctgac ctgcgcatgt ctgccatggc ccccacaccg ccccagggtg aggttgacgc cgactgcatg gacgtcaatg tccgcgggcc tgatggcttc accccgctca tgatcgcctc ctgcagcggg ggcggcctgg agacgggcaa cagcgaggaa gaggaggacg cgccggccgt catctccgac ttcatctacc agggcgccag cctgcacaac cagacagacc gcacgggcga gaccgccttg cacctggccg cccgctactc acgctctgat gccgccaagc gcctgctgga ggccagcgca gatgccaaca tccaggacaa catgggccgc accccgctgc atgcggctgt gtctgccgac gcacaaggtg tcttccagat cctgatccgg aaccgagcca cagacctgga tgcccgcatg catgatggca cgacgccact gatcctggct gcccgcctgg ccgtggaggg catgctggag gacctcatca actcacacgc cgacgtcaac gccgtagatg acctgggcaa gtccgccctg cactgggccg ccgccgtgaa caatgtggat gccgcagttg tgctcctgaa gaacggggct aacaaagata tgcagaacaa cagggaggag acacccctgt ttctggccgc ccgggagggc agctacgaga ccgccaaggt gctgctggac cactttgcca accgggacat cacggatcat atggaccgcc tgccgcgcga catcgcacag gagcgcatgc atcacgacat cgtgaggctg ctggacgagt acaacctggt gcgcagcccg cagctgcacg gagccccgct ggggggcacg cccaccctgt cgcccccgct ctgctcgccc aacggctacc tgggcagcct caagcccggc gtgcagggca agaaggtccg caagcccagc agcaaaggcc tggcctgtgg aagcaaggag gccaaggacc tcaaggcacg gaggaagaag tcccaggacg gcaagggctg cctgctggac agctccggca tgctctcgcc cgtggactcc ctggagtcac cccatggcta cctgtcagac gtggcctcgc cgccactgct gccctccccg ttccagcagt ctccgtccgt gcccctcaac cacctgcctg ggatgcccga cacccacctg ggcatcgggc acctgaacgt ggcggccaag cccgagatgg cggcgctggg tgggggcggc cggctggcct ttgagactgg cccacctcgt ctctcccacc tgcctgtggc ctctggcacc agcaccgtcc tgggctccag cagcggaggg gccctgaatt tcactgtggg cgggtccacc agtttgaatg gtcaatgcga gtggctgtcc cggctgcaga gcggcatggt gccgaaccaa tacaaccctc tgcgggggag tgtggcacca ggccccctga gcacacaggc cccctccctg cagcatggca tggtaggccc gctgcacagt agccttgctg ccagcgccct gtcccagatg atgagctacc agggcctgcc cagcacccgg ctggccaccc agcctcacct ggtgcagacc cagcaggtgc agccacaaaa cttacagatg cagcagcaga acctgcagcc agcaaacatc cagcagcagc aaagcctgca gccgccacca ccaccaccac agccgcacct tggcgtgagc tcagcagcca gcggccacct gggccggagc ttcctgagtg gagagccgag ccaggcagac gtgcagccac tgggccccag cagcctggcg gtgcacacta ttctgcccca ggagagcccc gccctgccca cgtcgctgcc atcctcgctg gtcccacccg tgaccgcagc ccagttcctg acgcccccct cgcagcacag stactcctyg cctgtggaca acacccccag ccaccagcta caggtgcctg agcacccctt cctcaccccg tcccctgagt cccctgacca gtggtccagc tygtccccgc attccaacgt ctccgactgg tccgagggcg tctccagccc tcccaccagc atgcagtccc agatcgcccg cattccggag gccttcaagt aa

NOTCH1-ICN (SEQ ID NO: 6)

[embodiment of SEQ ID NO : 6 wherein wherein AA2 is Pro, AA857 is Leu, AA860 is Ser and AA 891 is Ser]

MPPLLAPLLC LALLPALAAR GPRGSIVYLE IDNRQCVQAS SQCFQSATDV 50

AAFLGALASL GSLNIPYKIE AVQSETVEPP PPAQLHFMYV AQAVEPPPPA 100

QLHFMYVAAA AFVLLFFVGC GVLLSRKRRR QHGQLWFPEG FKVSEASKKK 150

RREPLGEDSV GLKPLKNASD GALMDDNQNE WGDEDLETKK FRFEEPWLP 200

DLDDQTDHRQ WTQQHLDAAD LRMSAMAPTP PQGEVDADCM DVNVRGPDGF 250

TPLMIASCSG GGLETGNSEE EEDAPAVISD FIYQGASLHN QTDRTGETAL 300

HLAARYSRSD AAKRLLEASA DANIQDNMGR TPLHAAVSAD AQGVFQILIR 350

NRATDLDARM HDGTTPLILA ARLAVEGMLE DLINSHADVN AVDDLGKSAL 400

HWAAAVNNVD AAWLLKNGA NKDMQNNREE TPLFLAAREG SYETAKVLLD 450

HFANRDI DH MDRLPRDIAQ ERMHHDIVRL LDEYNLVRSP QLHGAPLGGT 500

PTLSPPLCSP NGYLGSLKPG VQGKKVRKPS SKGLACGSKE AKDLKARRKK 550

SQDGKGCLLD SSGMLSPVDS LESPHGYLSD VASPPLLPSP FQQSPSVPLN 600

HLPGMPDTHL GIGHLNVAAK PEMAALGGGG RLAFETGPPR LSHLPVASGT 650

STVLGSSSGG ALNFTVGGST SLNGQCEWLS RLQSGMVPNQ YNPLRGSVAP 700

GPLSTQAPSL QHGMVGPLHS SLAASALSQM MSYQGLPSTR LATQPHLVQT 750

QQVQPQNLQM QQQNLQPANI QQQQSLQPPP PPPQPHLGVS SAASGHLGRS 800

FLSGEPSQAD VQPLGPSSLA VHTILPQESP ALPTSLPSSL VPPVTAAQFL 850

TPPSQHSYSS PVDNTPSHQL QVPEHPFLTP SPESPDQWSS SSPHSNVSDW 900

SEGVSSPPTS MQSQIARIPE AFK 923 shRNA construct targeting mouse rpllO (SEQ ID NO : 7 )

5' -aaccgacgatcctattgtcatc-3 '

gRNA targeting RPL10 (SEQ ID NO: 8)

5' -tcttgttgatgcggatgacg-3 '

RPL10 R98S donor oligo sequence (SEQ ID NO: 9)

5 ' -ctgtcagccc cagcacagga caacatcttg ttaatgctga tcacgtgaaa ggggtggagc cgcacccgga tatggaagcc atctttgcca caacttttta ccatgtactt attggcacaa attcgggca-3 '

PCR primers for distinguishing RPL10 WT and R98S in Jurkat cells Fw WT : 5' -cttccacgtcatccgcatc-3' [SEQ ID NO: 10] FW_R98S :

5' -cctttcacgtgatcagcatt-3' [SEQ ID NO: 11] Rev_WTandR98S :

5' -gctctgataaaataatgcaagccta-3' [SEQ ID NO