M6A-COUPLED EFFECTOR PROTEIN EXPRESSION SYSTEM AND METHODS OF MAKING AND USING SAME

Abstract
The present disclosure provides systems and methods for m6A-dependent delivery and m6A-dependent delivery targeted of a polypeptide to a cell. In certain embodiments, compositions, systems, and methods are provided that provide for m6A-dependent delivery of effector proteins, for example, effector proteins, such as a tumor suppression proteins and m6a regulation systems, mediated by CRISPRi in embodiments.
Description
REFERENCE TO A SEQUENCE LISTING SUBMITTED AS A XML FILE VIA EFS-WEB

The official copy of the sequence listing named “1412057 (DU7940US) Sequence Listing.xml”, created on Jan. 16, 2024, and having a size of 172 KB is submitted electronically via Patent Center in .xml format and is hereby incorporated by reference in its entirety.


BACKGROUND

The N6-methyladenosine (m6A) modification is found in thousands of cellular mRNAs and is a critical regulator of gene expression and cellular physiology. In pathological instances, m6A modifications may be dysregulated, contributing to several human diseases. For example, m6A dysregulation can lead to hypermethylation of oncogenic mRNAs and, in turn, leads to increased translation and cancer progression. The m6A methyltransferase machinery therefore has emerged as a promising therapeutic target.


Ongoing study of m6A modifications enabled by new tools provides clues as to strategies for overcoming hypermethylation. Current strategies for overcoming hypermethylation are focused on developing drugs that inhibit methyltransferase such as, for example, m(6)A methyltransferase (METTL3). However, as they can impact the methylation of all mRNAs, these approaches can have unwanted effects. Thus, targeted approaches for decreasing m6A hypermethylation and expression of upregulated oncogenes caused by m6A dysregulation are necessary avoid to globally affecting m6A modifications in unwanted areas, such as non-pathological tissue or cells.


SUMMARY

The Summary is provided to introduce a selection of concepts that are further described below in the Detailed Description. This Summary is not intended to identify key or essential features of the claimed subject matter, nor is it intended to be used as an aid in limiting the scope of the claimed subject matter.


Provided herein is a N6-methyladenosine (m6A)-coupled effector protein expression system and methods of introducing same into a cell, tissue, and/or animal model to achieve m6A-dependent protein expression.


In some embodiments, the m6A-coupled effector protein expression system comprises (a) a nucleic acid sequence encoding a fusion protein, wherein the fusion protein comprises an m6A binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase, and (b) a nucleic acid sequence encoding an effector protein (e.g., a protein that modulates expression of one or more proteins in the cell) and dihydrofolate reductase (DHFR). In some embodiments, the m6A-coupled effector protein expression system comprises (a) a nucleic acid sequence encoding a fusion protein, wherein the fusion protein comprises an m6A binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase, and (b) a nucleic acid sequence encoding an effector protein (e.g., a protein that modulates expression of one or more proteins in the cell), a M6A sensing domain, and dihydrofolate reductase (DHFR).


In some embodiments, the expression system is a vector system wherein a first plasmid comprises the nucleic acid sequence encoding the fusion protein comprising an N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase, and a second plasmid comprises the nucleic acid sequence encoding an effector protein (e.g., a protein that modulates expression of one or more proteins in the cell or otherwise targets a component of an expression system to a cell or within a cell) and dihydrofolate reductase (DHFR).


In some embodiments, the catalytic domain of the cytosine deaminase is the catalytic domain of apolipoprotein B mRNA editing enzyme (APOBEC-1). In some embodiments, the effector protein is a tumor suppressor protein, for example, METTL3. In some embodiments, the effector protein is an RNA-guided endonuclease. In some embodiments, the RNA-guided endonuclease is a dead RNA-guided endonuclease, for example, dead Cas9 (dCas9). In some embodiments, the effector protein comprises dCas9 linked or fused to a transcriptional regulator, for example, a transcriptional repressor (e.g., KRAB).


In further embodiments, the expression system can comprise: a first DNA construct comprising a nucleic acid sequence encoding a fusion protein, wherein the fusion protein comprises (i) a catalytically-dead RNA-targeting CRISPR-Cas system enzyme fused to (ii) a catalytic domain of a cytidine deaminase fused to (iii) an N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domain-containing protein; a second DNA construct comprising a nucleic acid sequence encoding: an effector protein; a m6A sensor sequence; and a polypeptide encoding dihydrofolate reductase (DHFR); and a guide RNA configured to bind to the nucleic acid of the second DNA construct. In embodiments, the cytidine deaminase can be APOBEC-1. In embodiments, the effector protein can be a tumor suppressor protein. In some embodiments, the effector protein can be a p53 or a SOCS2. In some embodiments, the dead RNA-guided endonuclease can be a dead type VI dCas13. In some embodiments, the fusion protein can further comprise a nuclear localization sequence (NLS).


In embodiments, described herein is an expression system comprising: (a) a first DNA construct comprising a polynucleotide encoding a fusion protein, wherein the fusion protein comprises an N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase or a catalytic domain of an adenosine deaminase; and a second DNA construct comprising a polynucleotide encoding a heterologous polypeptide, the polynucleotide encoding a heterologous polypeptide comprising: polynucleotide encoding an effector protein; polynucleotide encoding a m6A sensor sequence; and a polynucleotide encoding a dihydrofolate reductase (DHFR).


In embodiments, the m6A binding domain comprises a sequence having at least 90% or greater sequence identity to SEQ ID Nos: 66 or 108-116. In embodiments, the m6A binding domain is fused to the catalytic domain via a peptide linker. In embodiments, the catalytic domain comprises a polypeptide having at least 95% identity to SEQ ID NO 78 or a catalytic fragment thereof, SEQ ID NO: 79 or a catalytic fragment thereof, SEQ ID NO: 80 or a catalytic fragment thereof; or SEQ ID NO: 81. In embodiments, a vector comprises the first DNA construct, a second DNA construct, or both. In embodiments, the nucleic acid sequence encoding a fusion protein, the nucleic acid sequence encoding a heterologous polypeptide and a polypeptide encoding dihydrofolate reductase (DHFR), or both, are operably linked to a first promoter. In embodiments, the system further comprises a nucleic acid sequence encoding a selectable marker operably linked to a second promoter. In embodiments, the first promoter is a constitutive or an inducible promoter. In embodiments, the first promoter is a constitutive or an inducible promoter. In embodiments, the cytidine deaminase is APOBEC-1. In embodiments, the effector protein is a tumor suppressor protein or a catalytically dead RNA-guided endonuclease. In embodiments, the tumor suppressor protein is suppressor of cytokine signaling 2 (SOC2) or p53 or one of the proteins listed in Table 1. In embodiments, the catalytically dead RNA-guided endonuclease is a dCas9 or a dCas13.


In embodiments, described herein is a polynucleotide comprising a nucleic acid sequence encoding an effector protein polypeptide, a m6A sensor sequence, and a polypeptide encoding dihydrofolate reductase (DHFR). Also described herein are vectors and host cells comprising one or more components of expression systems as described herein, as well as non-human transgenic animals comprising one or more components of expression vectors as described herein. Described herein additionally are kits comprising any one or more components of expression systems as described herein.


Further described herein are methods. In embodiments, described herein are methods of increasing expression of a tumor suppressor protein in one or more cells, comprising introducing the expression system of claim 1 into the one or more cells, for example, hepatocellular carcinoma (HCC) cells. In embodiments, the tumor suppression protein is SOCS2 or p53 or one of the proteins listed in Table 1.


Described herein are methods of reducing m6A effector regulator expression in a sample or a subject. In embodiments, described herein is a method of reducing M6A effector regulator expression, comprising: introducing an expression system into a subject having or suspected of having a cancer, wherein the expression system comprises: a first DNA construct comprising a polynucleotide encoding a fusion protein, wherein the fusion protein comprises an N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase or a catalytic domain of an adenosine deaminase; and a second DNA construct comprising a polynucleotide encoding a heterologous polypeptide, the polynucleotide encoding a heterologous polypeptide comprising: a polynucleotide encoding a catalytically-dead RNA-guided endonuclease; a polynucleotide encoding a m6A sensor sequence; and a polynucleotide encoding a dihydrofolate reductase (DHFR); an sgRNA configured to bind to an m6A regulator. In embodiments, the sgRNA is configured to bind to a m6A regulator listed in Table 2. In embodiments, the cancer comprises at least one of acute myeloid leukemia (AML), glioblastoma (GBM), lung cancer, endometrial cancer, cervical cancer, ovarian cancer, breast cancer, colorectal cancer (CRC), a hepatocellular carcinoma (HCC), pancreatic cancer, gastric cancer, prostate cancer, or renal cell carcinoma. In embodiments, the cancer is a cancer listed in Table 1 or Table 2. In embodiments, the catalytically-dead RNA-guided endonuclease is a dCas9 or dCas13.


Described herein are methods of reducing m6A hypermethylation in a subject or sample. In embodiments, methods comprising: introducing an expression system into a subject having or suspected of having a cancer, wherein the expression system comprises: a first DNA construct comprising a polynucleotide encoding a fusion protein, wherein the fusion protein comprises an N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase or a catalytic domain of an adenosine deaminase; and a second DNA construct comprising a polynucleotide encoding a heterologous polypeptide, the polynucleotide encoding a heterologous polypeptide comprising: a polynucleotide encoding a catalytically-dead RNA-guided endonuclease; a polynucleotide encoding a m6A sensor sequence; and a polynucleotide encoding a dihydrofolate reductase (DHFR); an sgRNA configured to bind to an m6A regulator. The sgRNA is configured to bind to a m6A regulator listed in Table 2. The cancer comprises at least one of acute myeloid leukemia (AML), glioblastoma (GBM), lung cancer, endometrial cancer, cervical cancer, ovarian cancer, breast cancer, colorectal cancer (CRC), a hepatocellular carcinoma (HCC), pancreatic cancer, gastric cancer, prostate cancer, or renal cell carcinoma. The cancer is a cancer listed in Table 1 or Table 2. The catalytically-dead RNA-guided endonuclease is a dCas9 or dCas13.


In embodiments, described herein are methods of inhibiting cancer cells. In an embodiment, a method of inhibiting a cancer cell, the method comprising: introducing the expression system as described herein into the cancer cell, wherein the cancer cell comprises m6A RNA hypermethylation, and wherein the second DNA construct comprising a polynucleotide encoding an effector protein, the effector protein comprising a tumor suppressor protein.


The cancer cell can comprise an acute myeloid leukemia (AML) cell, a glioblastoma (GBM) cell, a lung cancer cell, an endometrial cancer, a cervical cancer cell, an ovarian cancer cell, a breast cancer cell, a colorectal cancer (CRC) cell, a hepatocellular carcinoma (HCC) cell, a pancreatic cancer cell, a gastric cancer cell, a prostate cancer cell, or a renal cell carcinoma cell. In an embodiment, the lung cancer cell is a non-small cell lung carcinoma cell. In an embodiment, the cancer cell is a hepatocellular carcinoma cell. In an embodiment, the tumor suppressor protein comprises at least one of the tumor suppressor proteins listed in Table 1. In an embodiment, expression of the tumor suppressor protein upregulates downstream signaling targets. In an embodiment, the tumor suppressor protein comprises p53. In an embodiment, expression of p53 upregulates at least one of CDKN1A or GADD45A. In an embodiment, the tumor suppressor protein comprises suppressor of cytokine signaling 2 (SOCS2). In an embodiment, the expression system is introduced into the cancer cell by transfection, viral infection, or electroporation. In an embodiment of methods as described herein, inhibiting the cancer cell comprises decreasing at least one of cell proliferation, cell migration, or metastasis.


Described herein are methods of treating a subject having a cancer. In embodiments, methods of treating a subject having a cancer characterized by m6A RNA hypermethylation, the methods comprise inhibiting a cancer cell according to the methods as described above. In embodiments, the cancer comprises at least one of acute myeloid leukemia (AML), glioblastoma (GBM), lung cancer, endometrial cancer, cervical cancer, ovarian cancer, breast cancer, colorectal cancer (CRC), a hepatocellular carcinoma (HCC), pancreatic cancer, gastric cancer, prostate cancer, or renal cell carcinoma. In embodiments, the cancer comprises hepatocellular carcinoma. In embodiments, expression of the tumor suppressor protein results in decreasing at least one of cell proliferation, cell migration, or metastasis of the cancer. In embodiments, the expression system is introduced into the subject by viral infection or electroporation.





BRIEF DESCRIPTION OF THE DRAWINGS

The present application includes the following figures. The figures are intended to illustrate certain embodiments and/or features of the compositions and methods, and to supplement any description(s) of the compositions and methods. The figures do not limit the scope of the compositions and methods, unless the written description expressly indicates that such is the case.



FIG. 1 is a schematic of the m6A sensor system according to certain embodiments of this disclosure. The m6A reporter mRNA is shown with the m6A sensor sequence expanded. When this sequence is unmethylated, APO1-YTH does not bind to the sensor sequence and no editing takes place. As a result, GFP-DHFR is produced and rapidly degraded (left panel). Methylation of either adenosine (red) in the sensor sequence results in recruitment of APO1-YTH and C-to-U editing of either or both convertible stop codons. Translation leads to GFP production and cell fluorescence (right panel).



FIGS. 2A-2M show that GEMS depends on m6A recognition. FIG. 2A. HEK293T cells were transfected with the GEMS reporter mRNA alone or the reporter mRNA together with APO1-YTH and imaged 24 h later. Cells expressing the reporter mRNA with APO1-YTH exhibit robust EGFP fluorescence, whereas cells expressing the reporter mRNA alone are dark. Scale bar: 100 μm. FIG. 2B. RT-PCR and Sanger sequencing of the m6A sensor sequence from cells in (FIG. 2A) shows C-to-U editing (C-to-T in cDNA) of the convertible stop codon cytidines (marked with an asterisk) only in the presence of APO1-YTH. Quantification of % C2U at the indicated cytidines is shown below the sequencing traces. ***p<0.001. n=3 biological replicates. FIG. 2C. Western blot from cells in (FIG. 2A) indicates EGFP production from the GEMS reporter mRNA only in cells expressing APO1-YTH. FIG. 2D. HEK293T cells were transfected with the GEMS system containing APO1-YTH or m6A binding-deficient APO1-YTHmut and imaged after 24 h. Cells expressing APO1-YTH contain EGFP fluorescence whereas cells expressing APO1-YTHmut are dark. Scale bar: 100 μm. FIG. 2E. Western blot from cells in (FIG. 2D) indicates loss of EGFP production from the GEMS reporter mRNA when cells co-express APO1-YTHmut. FIG. 2F. RT-PCR and Sanger sequencing from cells in (FIG. 2D) shows C-to-U editing of the m6A sensor sequence only in cells co-expressing APO1-YTH. Quantification of % C2U at the indicated cytidines is shown below the sequencing traces. ***p<0.001. n=2 biological replicates. FIGS. 2G-2L show that GEMS mRNA methylation mirrors endogenous mRNA methylation. FIG. 2G. HEK293T cells were co-transfected with the m6A reporter mRNA and APO1-YTH or APO1-YTHmut, then subjected to flow cytometry. Robust EGFP fluorescence is detected only in cells expressing the reporter mRNA and APO1-YTH. FIG. 2H. HEK293T cells were transfected with the GEMS system, and flow cytometry was used to sort cells into three populations based on EGFP fluorescence intensity. RT-qPCR-based m6A quantification of the sensor sequence shows an increase in m6A with increasing EGFP fluorescence. **p<0.01, n=2 biological replicates. FIG. 2I. RNA was isolated from sorted cell populations in (FIG. 2H) and analyzed by RT-PCR/Sanger sequencing. C-to-U editing of the m6A sensor sequence is increased in cells with higher levels of EGFP fluorescence. FIG. 2J. HEK293T cells were transfected with GEMS containing the full length m6A sensor sequence or a version with RAC motifs mutated (GEMS ARAC). EGFP fluorescence is abolished in cells expressing GEMS ARAC. Scale bar: 100 μm. FIG. 2K. Top schematic shows the m6A reporter mRNA with a portion of the m6A sensor sequence expanded. This sequence is based off of a sequence within the human ACTB 3′UTR (bottom schematic), which contains two m6A sites at positions A1216 and A1222 that have been shown to be methylated in several m6A mapping studies. FIG. 2L. RNA was isolated from HEK293T cells expressing the GEMS system for 24 h, followed by RT-PCR and Sanger sequencing. Sequencing traces show C-to-U editing of site A1222 in endogenous ACTB and the second consensus A in the m6A sensor sequence. N.s.=no significant difference, n=3 biological replicates. FIG. 2M. Relative m6A quantification using an RT-qPCR m6A detection approach shows similar levels of methylation of the m6A sensor sequence (GEMS GAC) and endogenous ACTB site A1222 on which the sensor sequence is based. M6A is not detected at non-consensus adenosines in the m6A sensor sequence (UAC and CAG). Dotted line at 0.5 represents the minimum cutoff value indicating the presence of m6A. Schematic below shows the presence of the indicated adenosines within the m6A sensor sequence. ***p<0.001. n=3 biological replicates.



FIGS. 3A-3T show that GEMS is METTL3-dependent and responds to changes in METTL-3 levels. FIG. 3A. Mass spectrometry analysis of purified mRNA from METTL3 degron cells shows a decrease in m6A in auxin-treated cells. ***p<0.001, n=2 biological replicates. FIG. 3B. The GEMS system was transfected into HEK293T cells containing an auxin-inducible degron tag fused to endogenous METTL3. Addition of auxin leads to reduced EGFP fluorescence. Scale bar: 100 μm. FIG. 3C. Western blot confirms loss of METTL3 and EGFP following auxin treatment of cells in (FIG. 3B). FIG. 3D. Quantification of EGFP/EGFP-DHFR ratio from western blot samples in (FIG. 3C) shows a decrease in EGFP/EGFP-DHFR in response to auxin treatment. ***p<0.001, n=5 biological replicates. FIG. 3E. Sanger sequencing traces show C-to-U editing of the m6A sensor sequence in RNA extracts from cells in (FIG. 3B). Editing is reduced in auxin-treated cells. ***p<0.001, n=3 biological replicates FIG. 3F. RT-qPCR quantification of m6A sensor sequence methylation decreased m6A after auxin-mediated METTL3 depletion. ***p<0.001, n=3 biological replicates. FIG. 3G. RT-qPCR quantification of m6A reporter mRNA expression was performed on RNA samples extracted from METTL3 degron cells with and without auxin treatment. N.s.=no significant difference, n=2 biological replicates. FIG. 3H. Densitometry analysis of western blot data was used to quantify total reporter mRNA protein production (EGFP+EGFP-DHFR) relative to cyclophilin A in METTL3 degron cells expressing the GEMS system. n.s.=no significant difference, n=3 biological replicates. FIG. 3I. HEK293T cells were transfected with GEMS for 24 hours and then treated with cycloheximide (CHX) and collected after 0, 20, 40, 60, 90, and 120 minutes of treatment. RNA was extracted at each time point and m6A reporter mRNA abundance was measured by RT-qPCR. N.s.=no significant difference, n=3 biological replicates. FIG. 3J. The GEMS system was transfected into HEK293T cells in the presence or absence of METTL3 overexpression. EGFP fluorescence is increased in METTL3-overexpressing cells. Scale bar: 100 μm. FIG. 3K. Western blot analysis shows an increase in EGFP protein expression in cells overexpressing METTL3. FIG. 3L. Quantification of EGFP/EGFP-DHFR ratio from western blot data indicates increased EGFP/EGFP-DHFR in METTL3-overexpressing cells. *p<0.05, n=3 biological replicates. FIG. 3M. Sanger sequencing analysis of RNA extracts from cells in (FIG. 3J) shows increased C-to-U editing of the m6A sensor sequence in response to METTL3 overexpression. **p<0.01, n=3 biological replicates. FIG. 3N. RNA was extracted from cells transfected with the GEMS system with or without simultaneous METTL3 overexpression and subjected to RT-qPCR to measure the expression of the m6A reporter mRNA. N.s.=no significant difference, n=3 biological replicates. FIG. 3O. RNA was extracted from HEK293T cells transfected with GEMS with or without simultaneous overexpression of METTL3, followed by RT-qPCR-based m6A quantification of the m6A sensor sequence. ***p<0.001, n=3 biological replicates. FIG. 3P Correlation between the change in m6A level in the reporter mRNA and EGFP protein production (EGFP/EGFP-DHFR) in response to METTL3 overexpression. Line represents best-fit and shaded area represents confidence interval around the fit. R=0.971; n=4 biological replicates. FIG. 3Q. Schematic showing the main components of the GEMS plasmid with the addition of DsRed under the control of a separate promoter. FIG. 3R. HEK293T cells infected with Cas9 and either METTL3 sgRNA or AAVS1 sgRNA (control) were transfected with the GEMS system and subjected to flow cytometry based on EGFP and DsRed fluorescence. The proportion of cells in the indicated flow-sorted populations that contain METTL3 indels is shown. FIG. 3S. RNA samples were prepared from cell populations sorted in (i) and subjected to RT-PCR/Sanger sequencing of the m6A sensor sequence. C-to-U editing is only detected in the EGFP+ population. FIG. 3T. RNA was isolated from sorted cell populations and analyzed by RT-qPCR for METTL3 expression. The DsRed+/EGFP− population shows significantly lower METTL3 expression. ***p<0.001, n=2 biological replicates.



FIGS. 4A-4G show that GEMS detects differences in methylation across cell types. FIG. 4A. The GEMS system containing an internal m6A-independent DsRed reporter was transfected into HEK293T, HeLa, and Huh-7 cells followed by fluorescence microscopy 24 h later. M6A-coupled EGFP fluorescence is reduced in Huh-7 cells compared to HEK293T and HeLa cells. Scale bar: 100 μm. FIG. 4B. Western blot analysis of cells in (FIG. 4A) shows decreased EGFP expression in Huh-7 cells compared to HEK293T and HeLa cells. FIG. 4C. Quantification of EGFP/EGFP-DHFR ratio relative to DsRed expression in HEK293T, HeLa, and Huh-7 cells. N.s.=no significant difference, 915 ***p<0.001; n=3 biological replicates. FIG. 4D. Sanger sequencing shows C-to-U editing of the m6A sensor sequence in RNA samples from cells in (FIG. 4A). Huh-7 cells have reduced C-to-U editing compared to HEK293T and HeLa cells. Quantification of % C-to-U is shown on the right. ***p<0.001; n=3 biological replicates. FIG. 4E. The GEMS system expressing APO1-YTH or APO1-YTHmut was transfected into the indicated cell types followed by fluorescence microscopy 24 h later. In all cell types, EGFP fluorescence was detected in the presence of APO1-YTH but not APO1-YTHmut, indicating m6A-dependent activity of the GEMS system. Scale bar: 10 μm. FIG. 4F. Cell lysates were prepared from cells transfected as in (a) and analyzed by western blot. All cell types show decreased EGFP protein production in the presence of APO1-YTHmut. FIG. 4G. RT-PCR/Sanger sequencing analysis of the m6A sensor sequence shows C-to-U editing in all cell types tested which is absent in the presence of APO1-YTHmut. Quantification of editing is shown below. N.s.=not statistically significant. N=3 biological replicates. FIG. 4H. RT-qPCR-based m6A quantification shows reduced m6A in the m6A sensor sequence in Huh-7 cells compared to HEK293T and HeLa cells. Dotted line indicates minimum m6A detection threshold. N.s.=no significant difference, **p<0.01, n=3 biological replicates.



FIG. 4I. Bioanalyzer traces are shown for purified mRNA samples from HEK293T, HeLa, and Huh-7 cells that were subsequently analyzed by mass spectrometry to quantify cellular m6A. The traces confirm removal of rRNA in each sample. FIG. 4J. Mass spectrometry was used to quantify m6A in purified mRNA from HEK29T, HeLa, and Huh-7 cells.



FIGS. 5A-5F show that GEMS senses changes in m6A caused by small molecule inhibition of METTL3. FIG. 5A. EGFP fluorescence from the GEMS system is reduced in HEK293T cells treated with the METTL3 inhibitor STM2457. GEMS-expressing cells were treated with 30 μM STM2457 for 24 h. Scale bar: 100 μm. FIG. 5B. Quantitative microscopy was performed on HEK293T cells expressing the GEMS system and treated with 30 μM STM2457. Treatment with STM2457 shows a significant reduction in EGFP fluorescence intensity. ***p<0.001; n>400 cells per condition. EGFP signal in each cell was normalized to DsRed. FIG. 5C. Western blot shows decreased EGFP protein in STM2457-treated cells. FIG. 5D. Densitometry analysis indicates reduced EGFP/EGFP-DHFR ratio in cells treated with 30 μM STM2457 for 24 h. ***p<0.001; n=2 biological replicates. FIG. 5E. HEK293T cells were treated with 10 or 30 μM of STM2457 for 24 hours followed by transfection with the GEMS system containing DsRed as an m6A-uncoupled internal control. Increasing amounts of STM2457 lead to increased depletion of m6A-coupled EGFP fluorescence. Scale bar: 100 μm. FIG. 5F. Western blot from cells in (FIG. 5E) shows decreased production of EGFP protein with increasing doses of STM2457. FIG. 5G. Sanger sequencing traces (top) and quantification (bottom) of the m6A sensor sequence from cells in (FIG. 5E) indicates decreased editing with increasing amounts of STM2457. N=3 biological replicates. FIG. 5H. Mass spectrometry analysis of purified mRNA from cells in (FIG. 5E) indicates depletion of m6A following STM2457 treatment. ***p<0.001, n=2 biological replicates. FIG. 5I. RNA was extracted from cells in FIG. 5E that were treated with 30 μM STM2457 or DMSO and subjected to RT-qPCR to measure abundance of the m6A reporter mRNA. N.s.=no significant difference; n=2 biological replicates. FIG. 5J. Sanger sequencing of the m6A sensor sequence in RNA samples from cells in FIG. 5A shows reduced C-to-U editing in cells treated with STM2457. Quantification of % C-to-U is shown on the right. ***p<0.001; n=3 biological replicates. FIG. 5K. RT-qPCR-based m6A detection was used to quantify relative m6A levels of endogenous ACTB A1222 and the m6A sensor sequence. STM2457 treatment leads to similar reductions in m6A in endogenous ACTB and the m6A sensor sequence. ***p<0.001; n=3 biological replicates. FIG. 5L. HEK293T cells were treated as in (FIG. 5E) but transfected with a version of GEMS containing EGFP fused to a PEST destabilization domain. The EGFP signal shows greater depletion at lower doses of STM2457 compared to (FIG. 5E), indicating improved sensitivity of GEMS-EGFP-PEST as a readout for changes in m6A compared to GEMS-EGFP. FIG. 5M. Western blot from cells in (FIG. 5I) shows decreased production of EGFP protein with increasing doses of STM2457. FIG. 5N. Sanger sequencing traces of the m6A sensor sequence from cells in (FIG. 5I) indicates decreased editing with increasing amounts of STM2457. N=3 biological replicates. FIG. 5O. Quantification of EGFP/EGFP-DHFR ratio following STM2457 treatment of HEK293T cells expressing GEMS with EGFP or EGFP-PEST. The EGFP-PEST version shows an improved response at low doses of STM2457 compared to EGFP. ***p<0.001, **p<0.01, n=3 biological replicates. FIG. 5P is a cartoon depicting an example of an alternative FP that could be utilized in the GEMS system in place of EGFP. This schematic shows primary neurons that are infected with a lentivirus expressing a photoconvertible FP such as Dendra2, which emits green fluorescence that is converted to red fluorescence upon exposure to UV light. New Dendra2 protein can the subsequently be identified by green fluorescence.



FIG. 6 is a schematic of the m6A feedback system according to certain embodiments of this disclosure. METTL3 transcription leads to methylation of the sensor sequence and translation of dCas9-KRAB. Constitutive expression of METTL3 sgRNA targets dCas9-KRAB to the METTL3 locus to inhibit transcription. This results in decreased methylation of the sensor sequence and dCas9-KRAB depletion, allowing METTL3 transcription to resume.



FIG. 7 shows that dCas9-KRAB can be expressed in place of GFP in the m6A reporter mRNA. HEK293T cells were transfected with the m6A sensor system using a plasmid in which GFP was replaced with the coding sequence for dCas9-KRAB. Western blot shows expression of dCas9-KRAB and APO1-YTH. Cyclophilin A is shown as a loading control.



FIGS. 8A-8F show that dCas13-tethered APO1-YTH enables targeted m6A sensor sequence editing. FIG. 8A. Schematic showing the main components of the GEMS system with dCas13-APO1-YTH (dCas13-GEMS). Location of regions in the m6A reporter mRNA targeted by the indicated gRNAs is shown. FIG. 8B. HEK293T cells were co-transfected with dCas13-GEMS and the indicated gRNA. Only gRNAs targeting within the m6A sensor sequence enable GEMS activity (EGFP fluorescence). gRNA CTL=scrambled gRNA control. Scale bar: 100 μm. FIG. 8C1. RNA from cells in (FIG. 8B) was subjected to RT-PCR/Sanger sequencing targeting the m6A sensor sequence and known m6A sites in four cellular mRNAs (ACTB A1222, HERC2 A14782, NIPA1 A6089, and SMUG1 A1303). In cells expressing dCas13-GEMS and a gRNA targeting the sensor sequence, C-to-U editing is only detected in the m6A sensor sequence and not in cellular mRNAs. In contrast, cells expressing the APO1-YTH version of GEMS have editing of both the sensor sequence and cellular mRNAs. Asterisks denote m6A sites. FIG. 8C2. Top row shows RT-PCR/Sanger sequencing-based quantification of C-to-U editing of the m6A sensor sequence after expression of the GEMS system in HEK293T cells. This version of the GEMS system contains the APO1-YTH protein. Bottom row shows the same, but for cells expressing a version of the GEMS system with dCas13-APO1-YTH and a m6A reporter mRNA-targeting gRNA. FIG. 8D. Quantification of editing at the 2 convertible stop codons within the m6A sensor sequence from I. n.s.=no significant difference. ***p<0.001. n=2 biological replicates. FIG. 8E. RNA was extracted from cells in (b) and RT-qPCR-based m6A quantification was used to measure m6A in the m6A sensor sequence. n.s.=no significant difference, n=3 biological replicates. FIG. 8F. HEK293T cells were treated with STM2457 for 16 hours and then co-transfected with dCas13-GEMS and the indicated gRNAs. EGFP fluorescence activated by dCas13-GEMS is decreased in response to STM2457 treatment. Scale bar: 100 μm.



FIGS. 9A-9R show that m6A-coupled effector protein delivery counteracts the effects of m6A hypermethylation in cancer cells. FIG. 9A. Schematic showing m6A-coupled expression of a tumor suppressor protein to counteract the effects of m6A hypermethylation in cancer cells. FIG. 9B. Left: schematic shows the GEMS system Middle: Schematic showing the results of previously published studies (paper above) which found that hypermethylation of the SOCS2 mRNA leads to its degradation and reduced SOCS2 protein expression in liver cancer cells. SOCS2 is an inhibitor of the JAK/STAT pathway and acts as a tumor suppressor in hepatocellular carcinoma Right: Schematic of the GEMS system in which EGFP has been replaced with SOCS2. FIG. 9C. Huh-7 cells treated with increasing amounts of STM2457 show a dose-dependent increase in SOCS2 mRNA expression as measured by RT-qPCR. These data are consistent with m6A-mediated inhibition of SOCS2 abundance. ***p<0.001, **p<0.01. n=3 biological replicates. FIG. 9D. GEMS was used to deliver either EGFP (GEMS-EGFP) or SOCS2 (GEMS-SOCS2) into Huh-7 cells. Western blot indicates robust expression of SOCS2 in cells expressing GEMS-SOCS2. FIG. 9E. RT-qPCR shows SOCS2 coding sequence expression in Huh-7 cells transfected with GEMS-SOCS2. FIG. 9F. Sanger sequencing of the m6A sensor sequence from cells in (FIG. 9D) indicates similar C-to-U editing rates of the GEMS-EGFP and GEMS-SOCS2 mRNAs. n.s.=no significant difference; n=3 biological replicates FIG. 9G. Quantification of EGFP/EGFP-DHFR ratio and SOCS2/SOCS2-DHFR ratio from western blot data from cells expressing GEMS-EGFP or GEMS-SOCS2 indicates similar ratios. FIG. 9H. Western blot analysis of downstream SOCS2 targets shows a decrease in STAT5 and JAK2 phosphorylation in Huh-7 cells expressing GEMS-SOCS2. FIG. 9I. RT-qPCR shows reduced expression of SOCS2 target mRNAs IGF1 and CyclinD1 in Huh-7 cells expressing GEMS-SOCS2. ***p<0.001, n=3 biological replicates. FIG. 9J. Cell growth assays show reduced growth of Huh-7 cells transfected with 952 GEMS-SOCS2 compared to non-transfected cells (Control). Growth curves for both were normalized to cells expressing GEMS-EGFp. n=3 biological replicates. FIG. 9K. Huh-7 cell migration is diminished following expression of GEMS-SOCS2 compared to GEMS-EGFP. FIG. 9L. Western blot shows elevated p53 levels in Huh-7 cells expressing GEMS-p53 compared to GEMS-EGFP. Top panel shows brightfield images of cells migration; bottom panel shows quantification of the total number of cells migrated. ***p<0.001; n=3 biological replicates. FIGS. 9M-90 shows that GEMS achieves m6A-coupled p53 expression in cancer cells. FIG. 9M. Huh-7 cells expressing GEMS-EGFP or GEMS-p53 were subjected to RT-PCR and Sanger sequencing of the m6A sensor sequence. Similar C-to-U editing of target cytidines is achieved with GEMS-EGFP and GEMS-p53. FIG. 9N. RT-qPCR shows TP53 coding sequence expression in Huh-7 cells transfected with GEMS-p53. FIG. 9O. RT-qPCR shows reduced expression of p53 target mRNAs CDKN1A and GADD45A in Huh-7 cells expressing GEMS-p53. ***p<0.001, n=3 biological replicates. FIG. 9P. Cell growth is reduced in Huh-7 cells transfected with GEMS-p53 compared to non-transfected cells (Control). Growth curves for both were normalized to cells expressing GEMS-EGFP. ***p<0.001; n=3 biological replicates. FIG. 9Q. Brightfield images (top) and quantification (bottom) of Huh-7 cell migration measured 24 h after transfection with GEMS-EGFP or GEMS-p53. ***p<0.001; n=3 biological replicates. FIG. 9R. Comparison of the effects of GEMS-p53 delivery into Huh-7 cells (which express mutant p53) and HepG2 cells (which express wild type p53). The number of cells following GEMS-p53 transfection relative to GEMS-EGFP transfection for each cell type across 5 days is shown. ***p<0.001, n=3 biological replicates



FIGS. 10A-10D show that GEMS enables tunable protein expression with m6A levels. FIG. 10A. Huh-7 and HepG2 cells were transfected with either GEMS-p53 or GEMS-SOCS2. Western blot shows increased production of SOCS2 and p53 proteins in HepG2 cells. FIG. 10B. Quantification of p53/p53-DHFR and SOCS2/SOCS2-DHFR ratios in Huh-7 and HepG2 cells shows increased ratios in HepG2 cells compared to Huh-7 cells. *p<0.05, n=2 biological replicates. FIG. 10C. Top: Sanger sequencing traces of the m6A sensor sequence from cells in (FIG. 10A) indicates increased editing of convertible stop codons in HepG2 cells compared to Huh-7 cells. Bottom: quantification of C-to-U editing. ***p<0.001; n=3 biological replicates. FIG. 10D. RT-qPCR-based m6A quantification shows increased m6A in the m6A sensor sequence in HepG2 cells compared to Huh-7 cells. Dotted line indicates minimum m6A detection threshold. **p<0.01, *p<0.05; n=3 biological replicates.



FIG. 11: Neurons can be isolated from transgenic mice expressing the APOBEC1-YTH enzyme and then the m6A reporter mRNA could be introduced with viral infection or other means to examine m6A dynamics. This could also be done using mice that express the GEMS system.



FIG. 12: the GEMS system is compatible with HTS, so it could be used for HTS studies such as those seeking to identify cellular proteins/pathways that control m6A abundance or drugs/small molecules that inhibit METTL3 or m6A demethylases.





DETAILED DESCRIPTION

The following description recites various aspects and embodiments of the present compositions and methods. No particular embodiment is intended to define the scope of the compositions and methods. Rather, the embodiments merely provide non-limiting examples of various compositions and methods that are at least included within the scope of the disclosed compositions and methods. The description is to be read from the perspective of one of ordinary skill in the art; therefore, information well known to the skilled artisan is not necessarily included.


Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. All patents, patent applications and publications referred to throughout the disclosure herein are incorporated by reference in their entirety.


Articles “a” and “an” are used herein to refer to one or to more than one (i.e. at least one) of the grammatical object of the article. By way of example, “an element” means at least one element and can include more than one element.


The use of any and all examples or exemplary language (e.g., “such as”) provided herein, is intended merely to better illustrate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed.


The terms “may,” “may be,” “can,” and “can be,” and related terms are intended to convey that the subject matter involved is optional (that is, the subject matter is present in some examples and is not present in other examples), not a reference to a capability of the subject matter or to a probability, unless the context clearly indicates otherwise.


“About” is used to provide flexibility to a numerical range endpoint by providing that a given value may be “slightly above” or “slightly below” the endpoint without affecting the desired result.


The use herein of the terms “including,” “comprising,” or “having” and variations thereof, is meant to encompass the elements listed thereafter and equivalents thereof as well as additional elements. Embodiments recited as “including,” “comprising,” or “having” certain elements are also contemplated as “consisting essentially of” and “consisting of” those certain elements. As used herein, “and/or” refers to and encompasses any and all possible combinations of one or more of the associated listed items, as well as the lack of combinations where interpreted in the alternative (“or”).


As used herein, the transitional phrase “consisting essentially of” (and grammatical variants) is to be interpreted as encompassing the recited materials or steps and those that do not materially affect the basic and novel characteristic(s)” of the claimed invention. See, In re Herz, 537 F.2d 549, 551-52, 190 U.S.P.Q. 461, 463 (CCPA 1976) (emphasis in the original); see also MPEP § 2111.03. Thus, the term “consisting essentially of” as used herein should not be interpreted as equivalent to “comprising.”


As used throughout, the term “nucleic acid” or “nucleotide” refers to deoxyribonucleic acids (DNA) or ribonucleic acids (RNA) and polymers thereof in either single- or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides that have similar properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. A nucleic acid sequence can comprise combinations of deoxyribonucleic acids and ribonucleic acids. Such deoxyribonucleic acids and ribonucleic acids include both naturally occurring molecules and synthetic analogues. The polynucleotides of the invention also encompass all forms of sequences including, but not limited to, single-stranded forms, double-stranded forms, hairpins, stem-and-loop structures, and the like.


Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, and complementary sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 (1985); and Rossolini et al., Mol. Cell. Probes 8:91-98 (1994)).


As used throughout the term “mRNA” or “mRNA transcript” refers to a single-stranded RNA having at least one open reading frame that can be translated by a cell to express a protein, The cell can be an in vitro cell or an in vivo cell.


“Polypeptide,” “peptide,” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. As used herein, the terms encompass amino acid chains of any length, including full-length proteins, wherein the amino acid residues are linked by covalent peptide bond”.


“Contacting” as used herein, e.g., as in “contacting a cell” refers to contacting a cell directly or indirectly in vitro, ex vivo, or in vivo (i.e., within a subject as defined herein). Contacting a cell may include addition of a compound (e.g., a genetically encoded m6A-coupled effector protein delivery system) to a cell, or administration to a subject. Contacting encompasses administration to a solution, cell, tissue, mammal, subject, patient, or human. Further, contacting a cell includes adding an agent to a cell culture.


As used herein, the terms “subject” and “patient” are used interchangeably herein and refer to both human and nonhuman animals. The term “nonhuman animals” of the disclosure includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dog, cat, horse, cow, chickens, amphibians, reptiles, and the like, as well as animal models, such as transgenic animals, and the like. The methods and compositions disclosed herein can be used on a sample either in vitro (for example, on isolated cells or tissues) or in vivo in a subject (i.e., living organism, such as a patient or animal model). In embodiments of methods as described herein, the sample comprises a plurality of cells.


As used throughout, a catalytic domain of a cytidine deaminase is a polypeptide comprising a cytidine deaminase, for example, Apolipoprotein B mRNA Editing Enzyme Catalytic Subunit (APOBEC1), activation induced cytidine deaminase (AICDA), Apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3A (APOBEC3A), or a catalytic fragment of any thereof, that catalyzes deamination of cytidine (“C”) to uridine (“U”) in RNA molecules. As used throughout, a catalytic domain of an adenosine deaminase, is a polypeptide comprising an adenosine deaminase, for example, double-stranded RNA-specific adenosine deaminase (ADAR1), or a catalytic fragment thereof, that catalyzes deamination of adenosine (“A”) to inosine (“I”) in RNA molecules. In some embodiments, the catalytic domain retains at least about 75%, 80%, 90%, 95%, or 99% of the enzymatic activity of the wildtype deaminase from which the domain is derived.


As used throughout, the term “Cas9 polypeptide” means a Cas9 protein or a fragment thereof present in any bacterial species that encodes a Type II CRISPR/Cas9 system. See, for example, Makarova et al. Nature Reviews, Microbiology, 9: 467-477 (2011), including supplemental information, hereby incorporated by reference in its entirety. For example, the Cas9 protein or a fragment thereof can be from Streptococcus pyogenes. Full-length Cas9 is an endonuclease comprising a recognition domain and two nuclease domains (HNH and RuvC, respectively) that creates double-stranded breaks in DNA sequences. In the amino acid sequence of Cas9, HNH is linearly continuous, whereas RuvC is separated into three regions, one left of the recognition domain, and the other two right of the recognition domain flanking the HNH domain. Cas9 from Streptococcus pyogenes is targeted to a genomic site in a cell by interacting with a guide RNA that hybridizes to a 20-nucleotide DNA sequence that immediately precedes an NGG motif recognized by Cas9. This results in a double-strand break in the genomic DNA of the cell.


As used throughout, a dCas9 polypeptide is a deactivated or nuclease-dead Cas9 (dCas9) that has been modified to inactivate Cas9 nuclease activity. Modifications include, but are not limited to, altering one or more amino acids to inactivate the nuclease activity or the nuclease domain. For example, and not to be limiting, D10A and H840A mutations can be made in Cas9 from Streptococcus pyogenes to inactivate Cas9 nuclease activity. Other modifications include removing all or a portion of the nuclease domain of Cas9, such that the sequences exhibiting nuclease activity are absent from Cas9. Accordingly, a dCas9 may include polypeptide sequences modified to inactivate nuclease activity or removal of a polypeptide sequence or sequences to inactivate nuclease activity. The dCas9 retains the ability to bind to DNA even though the nuclease activity has been inactivated. Accordingly, dCas9 includes the polypeptide sequence or sequences required for DNA binding but includes modified nuclease sequences or lacks nuclease sequences responsible for nuclease activity. It is understood that similar modifications can be made to inactivate nuclease activity in other site-directed nucleases, for example in Cpf1 or C2c2.


In some examples, the dCas9 protein is a full-length Cas9 sequence from S. pyogenes lacking the polypeptide sequence of the RuvC nuclease domain and/or the HNH nuclease domain and retaining the DNA binding function. In other examples, the dCas9 protein sequences have at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or 99% identity to Cas9 polypeptide sequences lacking the RuvC nuclease domain and/or the HNH nuclease domain and retains DNA binding function. In other examples, the dCas9 protein sequence is encoded by a polynucleotide that has at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or 99% to SEQ ID NO: 59.


As used throughout, the term “Cas13 polypeptide” means a Cas13 protein or a fragment thereof present in any bacterial species that encodes a Type VI CRISPR/Cas13 system. Exemplary Cas13 polypeptides include dPspCas13b, dLwaCas13a, and dRfxCas13d. Additional Cas13 polypeptides are described, for example, in Abudayyeh et al., Science. 2016 August 5; 353(6299): aaf5573. doi:10.1126/science.aaf5573, including supplemental information, hereby incorporated by reference in its entirety; Cox et al., Science 358, 1019-1027 (2017) including supplemental information, hereby incorporated by reference in its entirety; and Tang et al., Front. Cell Dev. Biol., 27 Jul. 2021 Sec. Epigenomics and Epigenetics Volume 9-2021; doi: 10.3389/fcell.2021.677587. For example, the Cas13 protein or a fragment thereof with ssRNA targeting activity can be from Leptotrichia wadei, Leptotrichia shahii, Prevotella sp. P5-125 (PspCas13b), or Ruminococcus flavefaciens. Generally, Cas13 enzymes have two higher eukaryotes and prokaryotes nucleotide-binding (HEPN) endoRNase domains that mediate precise RNA cleavage with a preference for targets with protospacer flanking sites (PFSs) observed biochemically and in bacteria.


As used throughout, a dCas13 polypeptide is a deactivated or nuclease-dead Cas13 (dCas13) that has been modified to inactivate Cas13 nuclease activity. Modifications include, but are not limited to, altering one or more amino acids to inactivate the nuclease activity or the nuclease domain. For example, and not to be limiting, H133A and H1058A mutations can be made in Cas13 HEPN domains from Prevotella sp. P5-125 (PspCas13b) to inactivate Cas13 nuclease activity (see, for example, Cox et al., Science 358, 1019-1027 (2017) including supplemental information, hereby incorporated by reference in its entirety, and International Patent Publication WO 2019/005884, also incorporated by reference in its entirety). Other modifications include removing all or a portion of the nuclease domain of Cas13 (for example, A984-1090 H133A of Cas13b is from Prevotella sp. P5-125; see, for example, Programmable m(6)A modification of cellular RNAs with a Cas13-directed methyltransferase. Wilson C, Chen P J, Miao Z, Liu D R. Nat Biotechnol. 2020 Jun. 29. pii: 10.1038/s41587-020-0572-6. doi: 10.1038/s41587-020-0572-6. 10.1038/s41587-020-0572-6 PubMed 32601430), such that the sequences exhibiting nuclease activity are absent from Cas13. Exemplary dCas13 polypeptide mutations include R474A/R1046A in dCas13 from L. wadei and mutations R239R/H244A/ and R858A/H863A from Ruminococcus flavefaciens strain XPD3002. Accordingly, a dCas13 may include polypeptide sequences modified to inactivate nuclease activity or removal of a polypeptide sequence or sequences to inactivate nuclease activity. The dCas13 retains the ability to target ssRNA even though the nuclease activity has been inactivated. Accordingly, dCas13 includes the polypeptide sequence or sequences required for ssRNA targeting but includes modified nuclease sequences or lacks nuclease sequences responsible for nuclease activity.


In some examples, the dCas13 protein is a full-length Cas13 sequence from L. wadei, L. shahii, Prevotella sp. P5-125 (PspCas13b), or R. flavefaciens having one or more mutations in one or more HEPN domains and retaining the ssRNA targeting function. In other examples, the dCas13 protein sequences have at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or 99% identity to Cas13 polypeptide sequences with HEPN mutations and retains RNA binding function. In other examples, the dCas13 protein sequence is encoded by a dCas13 polynucleotide coding fragment that has at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or 99% to the corresponding dCas13 polynucleotide coding fragment present in SEQ ID NO: 60.


I. Introduction

N6-methyladenosine (m6A) is the most abundant internal mRNA modification and influences several steps of the RNA life cycle, including splicing, stability, and translation 1, 2. The majority of m6A sites in cells are deposited co-transcriptionally by a single methyltransferase, METTL3, which interacts with additional accessory proteins to target RNAs for methylation. In mammals, m6A occurs in a unique consensus sequence which at its core consists of RAC (R=G or A), and It is enricled in proximal 3′UTRs and in the vicinity of the stop codon3, 4. m6A carries out its diverse RNA regulatory functions by recruiting m6A binding proteins, which mediate the ability of m6A to impact the expression of thousands of cellular mRNAs.


Consistent with the broad roles for m6A in gene expression control, m6A has emerged as an important regulator of cellular function. m6A is necessary for several physiological processes, including stem cell maintenance, development, innate immunity, and learning and memory5-7. Additionally, dynamic regulation of m6A provides a mechanism for cells to fine-tune gene expression in response to changing cellular conditions. For instance, some forms of cellular stress can lead to hyper- or hypomethylated states which impact the expression of stress response genes 8-10 and synaptic activity alters mRNA methylation in the brain to control the expression of synaptic plasticity genes5, 11-13 In addition, abnormal regulation of m6A levels in cells contributes to a variety of human diseases, including cardiovascular disease, the response to viral infection, and several cancers14-16 METTL3 and other methyltransferase complex proteins are often upregulated in cancer, leading to elevated levels of m6A that promote the expression of genes that support cancer cell proliferation and migration. Thus, detecting changes in m6A levels across cell types or under certain cellular conditions is important for understanding how m6A contributes to cellular function in both healthy and disease states.


Much of the progress that has been made in understanding m6A regulation in cells has been through the development of new tools that have enabled m6A detection. Strategies for detecting global changes in cellular m6A levels have primarily used three approaches: m6A antibodies, thin-layer chromatography, or mass spectrometry. However, these methods suffer from several limitations, including high cost, the need for large amounts of RNA, and multiple sample processing steps. Moreover, antibody-based methods suffer from non-specificity, mass spectrometry requires specialized equipment, and TLC depends on radioactivity. More recently, alternatives to antibody-based global m6A mapping have been developed17-21, but these methods often require substantial amounts of input RNA. Importantly, all current strategies involve isolation of RNA from cells and therefore do not enable real-time monitoring of m6A methylation in living cells. These limitations have been a major barrier for understanding how cellular m6A is dynamically regulated. In addition, no method exists for providing a specific readout of cellular m6A methylation in a manner compatible with high-throughput screening (HTS). This has substantially limited drug discovery efforts aimed at identifying inhibitors of METTL3, and it has prevented other high-throughput studies designed to identify factors that regulate m6A in cells.


Based on the aforementioned deficiencies, there existed a great need to develop a simple, low-cost method for detecting adenosine methylation in living cells which is also compatible with HTS. As described at least in International Patent Application PCT/US2022/079709 (which is incorporated by reference as if fully set forth herein), progress has been made in this area. Genetically Encoded m6A Sensor technology (also referred to herein as “GEMS”) is described at least in International Patent Application PCT/US2022/079709, which can couple protein expression, such as a fluorescent signal, with cellular mRNA methylation. Sensors and methods as described therein can detect changes in m6A levels caused by pharmacological inhibition of the m6A methyltransferase, giving it potential utility for drug discovery efforts.


However, prior methods for studying m6A required RNA isolation and did not provide a real-time readout of mRNA methylation in living cells, leading to the development of technology such as the Genetically Encoded m6A Sensor technology (also referred to herein as “GEMS”). Other aspects of GEMS system components are additional described for example, at least in U.S. Pat. No. 11,680,109, which is incorporated by reference as if fully set forth herein.


Some of these prior approaches to date, however, may risk editing of off-target endogenous RNAs when fusion proteins comprising N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domains are utilized. Furthermore, while drug discovery efforts have been made aimed at METTL3 inhibition, targeted delivery of therapeutics (such as tumor suppressors and cell cycle proteins, for example), an area of research which can see improvements.


Described herein are constructs, expression systems, methods, kits, animals, and cells relating to programmable sensors and methods which can be programmed for targeted delivery of cells to achieve m6A-dependent delivery of custom protein payloads in cells. Thus, constructs, expression systems, and methods as described herein can provide a versatile platform based on m6A sensing, allowing for (at least): (1) a simple readout for m6A methylation; (2) a system for m6A-coupled protein expression; and (3) a system for targeted m6A-coupled protein expression. Furthermore, the GEMS systems as described can be modified for effector protein expression (e.g., expression of proteins related to tumor suppression or cell cycle regulation, such as p53 or suppressor of tumor signaling 2 (SOCS2)) or an RNA-guided endonuclease that has been modified to remove cleavage activity (e.g., a “dead” CAS protein). Systems as described herein additionally can be employed in transgenic or knock-in animals or cells derived from animal models as described herein.


Disclosed herein are compositions, systems, and methods related to overcoming the aforementioned limitations.


Disclosed herein are genetically encoded sensors for m6A which can provide a fluorescent readout when m6A is deposited on mRNA. The sensor may be used for detecting mRNA methylation in a variety of cell types (without intending to be limiting in immortalized or primary tumor cells in vitro, for example), and for responding to small molecule inhibition of the m6A methyltransferase, METTL3, as discussed. In addition, as disclosed herein, the m6A sensor platform can be utilized to express effector proteins of interest instead of a reporter protein (i.e., eGFP), such as anti-tumor therapeutics or tumor suppression proteins. For example, sensors as described herein can achieve m6A-coupled delivery of anti-tumor therapeutics (for example, tumor suppressor proteins to slow the growth of cancer cells through the expression of p53 or other tumor suppressor proteins) in cancer cells that have elevated m6A levels.


Additionally, components of the compositions, systems, and methods as described herein can be targeted to prevent off-target effects (such as unwanted editing of off-target RNAs in physiologically normal or otherwise healthy cells) utilizing catalytically-dead CRISPR-associated (Cas) enzymes, for example, of RNA-targeting (also referred to herein as “RNA-guided”) type III (i.e., Csm/Csr), type VI (i.e. Cas13), or type II (i.e., Cas9) CRISPR-Cas systems. Altogether, the system provides a simple, highly versatile approach that can be used for sensing m6A in living cells and coupling mRNA methylation to effector protein expression.


II. Expression Systems

Provided herein is an expression system comprising: (a) a first DNA construct comprising a nucleic acid sequence encoding a fusion protein, wherein the fusion protein comprises an N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase or a catalytic domain of an adenosine deaminase (e.g., APOBEC1); and (b) a second DNA construct comprising (i) a nucleic acid sequence encoding an effector polypeptide; (ii) a m6A sensor sequence; and (iii) a polypeptide encoding dihydrofolate reductase (DHFR). The nucleic acid sequence encoding an effector; (ii) a m6A sensor sequence; and (iii) a polypeptide encoding dihydrofolate reductase (DHFR) is also referred to as the mRNA reporter sequence or effector sequence. Also provided is a nucleic acid sequence comprising a nucleic acid sequence encoding an effector protein, a m6A sensor sequence, and, a polypeptide encoding dihydrofolate reductase (DHFR).


The m6A methylation sensor system previously discovered by the inventors, as described in PCT/US2022/079709, U.S. Pat. No. 11,680,109, and Meyer, K. D., “DART-seq: an antibody-free method for global m(6)A detection,” Nat Methods. 2019 December, 16(12):1275-1280 (published online Sep. 23, 2019); doi: 10.1038/s41592-019-0570-0, the entire contents of all of which (including sequence information and any supplemental information) are incorporated by reference in their entirety as fully set forth herein includes at least two components: 1) expression of APO1-YTH, and 2) expression of a protein in the presence of m6A (FIG. 1; the reporter protein eGFP is shown, which can be interchanged for an effector protein as described herein). The mRNA of the effector protein comprises the coding sequence for an effector protein (for example, a dCas or a tumor suppression protein), followed by a short m6A “sensor sequence” (for example, 5′GACUUACGACAG3′), which contains two m6A consensus motifs (GAC) and two tandem “convertible” stop codon sequences that are in-frame with EGFP (FIG. 1). The m6A sensor sequence can be modified from a similar sequence in the human ACTB mRNA 3′UTR, which contains two methylated GAC sequences that have been reported in many different cell types. When unedited, the convertible stop codons encode arginine and glutamine (CGA and CAG, respectively). However, C-to-U editing produces two stop codons (UGA and UAG) (FIG. 1). Downstream of the m6A sensor sequence and in-frame with EGFP is the coding sequence for a destabilization domain modified from the Escherichia coli dihydrofolate reductase gene (DHFR). This DHFR destabilization domain induces rapid, proteasome-mediated degradation of proteins to which it is tethered. Thus, when the GFP-DHFR m6A reporter mRNA is introduced into cells together with APO1-YTH, if the reporter mRNA is not methylated, there will be no editing of the m6A sensor sequence by APO1-YTH and the full-length GFP-DHFR protein will be translated. The result is rapid degradation of GFP-DHFR and no fluorescence (FIG. 1, left panel). However, if either of the GAC sequences within the m6A sensor sequence is methylated, APO1-YTH will bind to the m6A and deaminate one or both cytidine residues within the two convertible stop codons of the sensor sequence. The result is translation of GFP followed by translation termination before the ribosome encounters the DHFR sequence. The GFP protein will not be degraded since it will not be fused to DHFR, resulting in GFP fluorescence (FIG. 1, right panel). Thus, this system provides a simple fluorescent readout for the presence of m6A (i.e., no m6A=no GFP fluorescence; m6A=GFP fluorescence). Again, although eGFP is shown in FIG. 1, it would be understood that the eGFP polypeptide can be interchanged for an effector protein polypeptide.


Although the m6A sensor system uses m6A-coupled GFP expression as a readout, any gene of interest can be cloned in place of GFP to achieve m6A-dependent protein expression. Such an m6A-coupled effector protein delivery system has several potential applications (e.g., in cancer therapy). Additional aspects of expression systems are provided in Sections I and II above.


The recombinant nucleic acids provided herein can be included in expression cassettes for expression in a host cell or an organism of interest. The cassette will include 5′ and 3′ regulatory sequences operably linked to a recombinant nucleic acid provided herein that allows for expression of the modified polypeptide. The cassette may additionally contain at least one additional gene or genetic element to be co-transformed into the organism. Where additional genes or elements are included, the components are operably linked. Alternatively, the additional gene(s) or element(s) can be provided on multiple expression cassettes. Such an expression cassette is provided with a plurality of restriction sites and/or recombination sites for insertion of the polynucleotides to be under the transcriptional regulation of the regulatory regions. The expression cassette may additionally contain a selectable marker gene. The expression cassette will include in the 5′ to 3′ direction of transcription: a transcriptional and translational initiation region (i.e., a promoter), a polynucleotide of the invention, and a transcriptional and translational termination region (i.e., termination region) functional in the cell or organism of interest. The promoters of the invention are capable of directing or driving expression of a coding sequence in a host cell. The regulatory regions (i.e., promoters, transcriptional regulatory regions, and translational termination regions) may be endogenous or heterologous to the host cell or to each other. As used herein, “heterologous” in reference to a sequence is a sequence that originates from a foreign species, or, if from the same species, is substantially modified from its native form in composition and/or genomic locus by deliberate human intervention.


Additional regulatory signals include, but are not limited to, transcriptional initiation start sites, operators, activators, enhancers, other regulatory elements, ribosomal binding sites, an initiation codon, termination signals, and the like. See Sambrook et al. (1992) Molecular Cloning: A Laboratory Manual, ed. Maniatis et al. (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.) (hereinafter “Sambrook 11”); Davis et al., eds. (1980) Advanced Bacterial Genetics (Cold Spring Harbor Laboratory Press), Cold Spring Harbor, N.Y., and the references cited therein.


Further provided is a vector comprising a nucleic acid or expression cassette set forth herein. The vector is contemplated to have the necessary functional elements that direct and regulate transcription of the inserted nucleic acid. These functional elements include, but are not limited to, a promoter, regions upstream or downstream of the promoter, such as enhancers that may regulate the transcriptional activity of the promoter, an origin of replication, appropriate restriction sites to facilitate cloning of inserts adjacent to the promoter, antibiotic resistance genes or other markers which can serve to select for cells containing the vector or the vector containing the insert, RNA splice junctions, a transcription termination region, or any other region which may serve to facilitate the expression of the inserted gene or hybrid gene (See generally, Sambrook et al. Molecular Cloning: A Laboratory Manual, 4th ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 2012). The vector, for example, can be a plasmid.


The expression vectors described herein can also include the nucleic acids as described herein under the control of an inducible promoter such as the tetracycline inducible promoter or a glucocorticoid inducible promoter. The nucleic acids of the present invention can also be under the control of a tissue-specific promoter to promote expression of the nucleic acid in specific cells, tissues or organs. Any regulatable promoter, such as a metallothionein promoter, a heat-shock promoter, and other regulatable promoters, of which many examples are well known in the art are also contemplated. Furthermore, a Cre-loxP inducible system can also be used, as well as a Flp recombinase inducible promoter system, both of which are known in the art.


Provided herein is a m6A-coupled effector protein expression system and methods of introducing same into a cell, tissue, and/or animal model to achieve m6A-dependent protein expression. In some embodiments, the m6A-coupled effector protein expression system comprises (a) a nucleic acid sequence encoding a fusion protein, wherein the fusion protein comprises an N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase, and (b) a nucleic acid sequence encoding an effector protein and dihydrofolate reductase (DHFR). In some embodiments, the catalytic domain of the cytosine deaminase is the catalytic domain of apolipoprotein B mRNA editing enzyme (APOBEC-1). Also provided is a vector comprising any of the nucleic acid sequences described herein.


In some embodiments, the effector protein is a tumor suppressor protein, for example, METTL3. In some embodiments, the effector protein is an RNA-guided endonuclease. In some embodiments, the RNA-guided endonuclease is a dead RNA-guided endonuclease, for example, dead Cas9 (dCas9). In some embodiments, the effector protein comprises dCas9 linked or fused to a transcriptional regulator, for example, a transcriptional repressor (e.g., KRAB). In some embodiments, the effector protein comprises dCas9 linked or fused to a transcriptional activator. In any of the methods described herein, one or more guide RNAs can be introduced into the cell to guide the dCas9 to a specific site in the genome of the cell.


Also provided is a DNA construct comprising a promoter operably linked to a recombinant nucleic acid described herein. A nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence. Numerous promoters can be used in the constructs described herein. A promoter is a region or a sequence located upstream and/or downstream from the start of transcription that is involved in recognition and binding of RNA polymerase and other proteins to initiate transcription. The promoter can be a eukaryotic or a prokaryotic promoter. In some embodiments the promoter is an inducible promoter. In some embodiments, the promoter is a constitutive promoter.


Any of the nucleic acid sequences provided herein can be included in expression cassettes for expression in a host cell or an organism of interest. The cassette will include 5′ and 3′ regulatory sequences operably linked to a recombinant nucleic acid provided herein that allows for expression of the modified polypeptide. A nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence. Numerous promoters can be used in the constructs described herein. A promoter is a region or a sequence located upstream and/or downstream from the start of transcription that is involved in recognition and binding of RNA polymerase and other proteins to initiate transcription. The promoter can be a eukaryotic or a prokaryotic promoter. In some embodiments the promoter is an inducible promoter. In some embodiments, the promoter is a constitutive promoter.


In some embodiments, the nucleic acid sequence encoding a fusion protein comprising an N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase or a catalytic domain of an adenosine deaminase is operably linked to an inducible promoter, e.g., a tetracycline inducible promoter; and the nucleic acid construct encoding the mRNA reporter sequence is operably linked to a constitutive promoter (e.g., a CMV promoter)”


A “constitutive” promoter is a promoter that is active under most environmental and developmental conditions. Examples of constitutive promoters include, but are not limited to, a CMV promoter, a U6 promoter, a PGK promoter, a EF-1α promoter and a SV40 promoter.


An “inducible” promoter is a promoter that is active under environmental or developmental regulation, for example, regulated by the presence or absence of a drug. Examples of inducible promoters include, but are not limited to, the pL promoter (induced by an increase in temperature), the pBAD promoter, (induced by the addition of arabinose to the growth medium). the tetracycline-controlled transcriptional activation system (Tet-On/Tet-Off, Bujard and Gossen, PNAS, 89(12):5547-5551 (1992)), the Lac switch inducible system (Wyborski et al., Environ Mol Mutagen, 28(4):447-58 (1996)), the ecdysone-inducible gene expression system (No et al., PNAS, 93(8):3346-3351 (1996)), the cumate gene-switch system (Mullick et al., BMC Biotechnology, 6:43 (2006)), and the tamoxifen-inducible gene expression (Zhang et al., Nucleic Acids Research, 24:543-548 (1996)). Furthermore, a Cre-loxP inducible system can also be used, as well as a Flp recombinase inducible promoter system, both of which are known in the art.


In some embodiments, the promoter is a cell-specific or tissue-specific promoter. When using a cell- or tissue-specific promoter, expression occurs primarily, but not exclusively, in a particular cell or tissue. For example, expression can occur in at least 90%, 95%, or 99% of the targeted cell or tissue. It will be understood, however, that tissue-specific promoters may have a detectable amount of background or base activity in those tissues where they are mostly silent.


Examples of tissue-specific promoters include, but are not limited to, liver-specific promoters (e.g., APOA2, SERPINA1, CYP3A4, MIR122), pancreatic-specific promoters (e.g., insulin, insulin receptor substrate 2, pancreatic and duodenal homeobox 1, Aristaless-like homeobox 3, and pancreatic polypeptide), cardiac-specific promoters (e.g., myosin, heavy chain 6, myosin, light chain 2, troponin I type 3, natriuretic peptide precursor A, solute carrier family 8), central nervous system promoters (e.g., glial fibrillary acidic protein, internexin neuronal intermediate filament protein, Nestin, myelin-associated oligodendrocyte basic protein, myelin basic protein, tyrosin hydroxylase, and Forkhead box A2), skin-specific promoters (e.g., Filaggrin, Keratin 14 and transglutaminase 3), pluripotent and embryonic germ layer promoters (e.g., POU class 5 homeobox 1, Nanog homeobox, Nestin, and MicroRNA 122).


The cassette may additionally contain at least one additional gene or genetic element to be co-transformed into the organism (i.e., a cell, plurality of cells, tissue, or animal). Where additional genes or elements are included, the components are operably linked. Alternatively, the additional gene(s) or element(s) can be provided on multiple expression cassettes. Such an expression cassette is provided with a plurality of restriction sites and/or recombination sites for insertion of the polynucleotides to be under the transcriptional regulation of the regulatory regions. The expression cassette may additionally contain a selectable marker gene. The expression cassette will include in the 5′ to 3′ direction of transcription: a transcriptional and translational initiation region (i.e., a promoter), a polynucleotide of the invention, and a transcriptional and translational termination region (i.e., termination region) functional in the cell or organism of interest. The promoters of the invention are capable of directing or driving expression of a coding sequence in a host cell. The regulatory regions (i.e., promoters, transcriptional regulatory regions, and translational termination regions) may be endogenous or heterologous to the host cell or to each other. As used herein the term “heterologous” refers to a nucleotide sequence or polypeptide not normally found in a given cell in nature. As such, a heterologous nucleotide sequence or heterologous polypeptide may be: (a) foreign to its host cell (i.e., is exogenous to the cell); (b) naturally found in the host cell (i.e., endogenous) but present at an unnatural quantity in the cell (i.e., greater or lesser quantity than naturally found in the host cell); or (c) be naturally found in the host cell but positioned outside of its natural locus.


Additional regulatory signals include, but are not limited to, transcriptional initiation start sites, operators, activators, enhancers, other regulatory elements, ribosomal binding sites, an initiation codon, termination signals, and the like. See Sambrook et al. (1992) Molecular Cloning: A Laboratory Manual, ed. Maniatis et al. (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.) (hereinafter “Sambrook 11”); Davis et al., eds. (1980) Advanced Bacterial Genetics (Cold Spring Harbor Laboratory Press), Cold Spring Harbor, N.Y., and the references cited therein.


In preparing the expression cassette, the various DNA fragments may be manipulated, to provide for the DNA sequences in the proper orientation and, as appropriate, in the proper reading frame. Toward this end, adapters or linkers may be employed to join the DNA fragments or other manipulations may be involved to provide for convenient restriction sites, removal of superfluous DNA, removal of restriction sites, or the like. For this purpose, in vitro mutagenesis, primer repair, restriction, annealing, resubstitutions, e.g., transitions and transversions, may be involved.


Further provided is a vector comprising a nucleic acid or expression cassette set forth herein. The vector is contemplated to have the necessary functional elements that direct and regulate transcription of the inserted nucleic acid. These functional elements include, but are not limited to, a promoter, regions upstream or downstream of the promoter, such as enhancers that may regulate the transcriptional activity of the promoter, an origin of replication, appropriate restriction sites to facilitate cloning of inserts adjacent to the promoter, antibiotic resistance genes or other markers which can serve to select for cells containing the vector or the vector containing the insert, RNA splice junctions, a transcription termination region, or any other region which may serve to facilitate the expression of the inserted gene or hybrid gene (See generally, Sambrook et al. Molecular Cloning: A Laboratory Manual, 4th ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 2012). The vector, for example, can be a plasmid.


In some embodiments, a vector comprises the first DNA construct. In some embodiments, a vector comprises the second DNA construct. In some embodiments, a vector comprises the first and second DNA construct. In some embodiments, the vector is a plasmid. In some embodiments, a vector comprises the first DNA construct, the second DNA construct and a nucleic acid encoding a selectable marker. In some embodiments, the first DNA construct and the second DNA construct are operably linked to a first promoter, and the nucleic acid sequence encoding a selectable marker is operably linked to a second promoter (i.e., a promoter that is different from the first promoter). In some embodiments, the selectable marker is a fluorescent protein, that is different from the effector protein or the fluorescent protein encoded by second DNA construct, for example, dsRed. An exemplary dual-promoter construct that can be modified to express effector proteins as described herein, for example, but exchanging the nucleic acid sequence encoding a fluorescent report for an effector protein comprises: (1) a nucleic acid sequence encoding an effector protein, a m6A reporter sequence and DHFR; (2) a nucleic acid sequence encoding a fusion protein (APOBEC1-YTH); and (3) a nucleic acid sequence encoding dsRed (provided herein as SEQ ID NO: 107). In certain embodiments, the first DNA construct and second DNA construct do not contain nucleic acid sequences encoding a fluorescent protein.


There are numerous E. coli expression vectors known to one of ordinary skill in the art, which are useful for the expression of any of the nucleic acid sequences described herein (e.g., any of the fusion proteins described herein). Other microbial hosts suitable for use include bacilli, such as Bacillus subtilis, and other enterobacteriaceae, such as Salmonella, Senatia, and various Pseudomonas species. In these prokaryotic hosts, one can also make expression vectors, which will typically contain expression control sequences compatible with the host cell (e.g., an origin of replication). In addition, any number of a variety of well-known promoters will be present, such as the lactose promoter system, a tryptophan (Trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda. Additionally, yeast expression can be used.


“Polypeptide,” “peptide,” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. As used herein, the terms encompass amino acid chains of any length, including full-length proteins, wherein the amino acid residues are linked by covalent peptide bonds.


As used throughout, a “fusion protein” is a protein comprising two different polypeptide sequences, i.e. a binding domain and a catalytic domain, that are joined or linked to form a single polypeptide. The two amino acid sequences are encoded by separate nucleic acid sequences that have been joined so that they are transcribed and translated to produce a single polypeptide. In some embodiments, the fusion protein comprises, in the following order, a m6A binding domain, and a catalytic domain of a cytidine deaminase or an adenosine deaminase.


As used throughout, “m6A” refers to posttranscriptional methylation of an adenosine residue in the RNA of prokaryotes and eukaryotes (e.g., mammals, insects, plants and yeast).


As used throughout an “m6A sensor sequence” is a sequence comprising one or more m6A methylation consensus motifs (GAC). The m6A sensor sequence can also comprise at least one sequence that can be converted to a stop codon when the m6A sensor sequence is methylated in the cell. In the constructs described herein, the m6A sensor sequence is in-frame with the nucleic acid encoding the heterologous protein, e.g. a reporter protein. The m6A sensor sequence is flanked by the nucleic acid sequence encoding the heterologous protein (e.g., reporter protein) and the nucleic acid sequence encoding a destabilization domain, e.g., DHFR. When the construct is methylated in the cell, a C to U modification generates a stop codon in the m6A sensor sequence. The stop codon prevents expression of the destabilization domain, thus preventing degradation of the heterologous protein. Exemplary m6A sensor sequences include, but are not limited to, a nucleic acid sequence comprising, consisting of, or consisting essentially of, SEQ ID NOs: 66 and 108-116. Nucleic acid sequences having at least 90, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity with a nucleic acid sequence comprising, consisting essentially of, or consisting of SEQ ID NOs: 66 and 108-116 are also provided. One of skill in the art would understand that these sequences are merely exemplary because any m6A sensor sequence comprising at least one m6A methylation consensus motif (GAC) (e.g., one, two, three, four etc.) can be used as a sensor sequence.


As used throughout, a m6A binding domain of a YT521-B homology (YTH) domain-containing protein is a polypeptide fragment of a YTH domain-containing protein that binds to m6A-containing sequence (e.g., a RNA, such as a mRNA or a m6A sensor sequence). The m6A binding domain derived from a YT521-B homology (YTH) domain-containing protein can be of any size as long as it retains binding activity and is not the full-length YTH domain-containing protein. In some embodiments, the binding domain retains at least about 75%, 80%, 90%, 95%, or 99% of the binding activity of the wildtype YTH domain-containing protein from which the binding domain is derived.


In some embodiments, the DNA construct encodes a m6A binding domain comprising a polypeptide having at least 95% identity, for example, at least about 95%, 96%, 97%, 98% or 99% identity, to SEQ ID NO: 67 (amino acid sequence of YTHDF2-YTH, a m6A binding domain of YTHDF2), SEQ ID NO: 68 (amino acid sequence of YTHDF2-YTH_W432A_W486A, a mutated m6A binding domain of YTHDF2), SEQ ID NO: 69 (amino acid sequence of YTHDF2-YTHmut, an amino acid sequence that includes the YTH domain of YTHDF2, and does not include the m6A-binding domain), SEQ ID NO: 70 (amino acid sequence of YTHDF2-YTHmut, an amino acid sequence comprising SEQ ID NO: 69, with a W432A mutation and a W486a mutation), SEQ ID NO: 71 (amino acid sequence of YTHDF2-YTH D422N, a mutated m6A binding domain of YTHDF2), SEQ ID NO: 72 (amino acid sequence of a m6A binding domain of YTHDF1), SEQ ID NO: 73 (amino acid sequence of YTHDF1mut, an amino acid sequence that includes the YTH domain of YTHDF2, and does not include the m6A-binding domain), SEQ ID NO: 74 (amino acid sequence of YTHDF1 D401N, a mutated m6A binding domain of YTHDF1), SEQ ID NO: 75 (amino acid sequence of a m6A binding domain of YTHDF3); SEQ ID NO: 76 (amino acid sequence of a m6A binding domain of YTHDC1) or SEQ ID NO: 77 (amino acid sequence of a m6A binding domain of YTHDC2).


As used throughout, a catalytic domain of a cytidine deaminase is a polypeptide comprising a cytidine deaminase, for example, Apolipoprotein B MRNA Editing Enzyme Catalytic Subunit (APOBEC1 or APO1), activation induced cytidine deaminase (AICDA) or Apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like 3A (APOBEC3A), or a catalytic fragment thereof, that catalyzes deamination of cytidine (“C”) to uridine (“U”) in RNA molecules. As used throughout, a catalytic domain of an adenosine deaminase, is a polypeptide comprising an adenosine deaminase, for example, double-stranded RNA-specific adenosine deaminase (ADAR1), or a catalytic fragment thereof, that catalyzes deamination of adenosine (“A”) to inosine (“I”) in RNA molecules. In some embodiments, the catalytic domain retains at least about 75%, 80%, 90%, 95%, or 99% of the enzymatic activity of the wildtype deaminase from which the domain is derived.


In some embodiments, the catalytic domain comprises a polypeptide having at least 95% identity, for example, at least about 95%, 96%, 97%, 98% or 99% identity, to SEQ ID NO: 78 (amino acid sequence of rAPOBEC1) or its catalytic domain (SEQ ID NO: 120), SEQ ID NO: 13 (amino acid sequence of hAICDA) or its catalytic domain (SEQ ID NO: 79); SEQ ID NO: 80 (amino acid sequence of hAPOBEC3A) or its catalytic domain (SEQ ID NO: 128); SEQ ID NO: 81 (amino acid sequence of ADAR2) or its catalytic domain (SEQ ID NO: 119); or SEQ ID NO: 121 (amino acid sequence of ADAR1) or its catalytic domain (SEQ ID NO: 122).


The catalytic domain can also comprise a polypeptide having at least 95% identity to SEQ ID NO: 119 (amino acid sequence of catalytic domain of ADAR2), as set forth in U.S. Patent Application Publication No. 20190010478.


In some embodiments, the DNA construct encodes a m6A binding domain fused to the catalytic domain via a peptide linker. The peptide linker can be about 2 to about 150 amino acids in length. For example, the linker can be a linker of from about 5 to about 20 amino acids in length, from about 5 to about 25 amino acids in length, from about 10 to about 30 amino acids in length, 5 to about 35 amino acids in length, from about 5 to about 40 amino acids in length, from about 5 to about 45 amino acids in length, from about 5 to about 50 amino acids in length, from about 5 to about 55 amino acids in length, from about 5 to about 60 amino acids in length, from about 5 to about 65 amino acids in length, from about 5 to about 70 amino acids in length, from about 5 to about 75 amino acids in length, from about 5 to about 80 amino acids in length, from about 5 to about 85 amino acids in length, from about 5 to about 90 amino acids in length, from about 5 to about 95 amino acids in length, from about 5 to about 100 amino acids in length, from about 5 to about 105 amino acids in length, from about 5 to about 110 amino acids in length, from about 5 to about 115 amino acids in length, from about 5 to about 120 amino acids in length, from about 5 to about 125 amino acids in length, from about 5 to about 130 amino acids in length, from about 5 to about 135 amino acids in length, from about 5 to about 140 amino acids in length, from about 5 to about 145 amino acids in length, or from about 5 to about 150 amino acids in length.


Exemplary peptide linkers include, but are not limited to, peptide linkers comprising SEQ ID NO: 82 (SGSETPGTSESATPE), SEQ ID NO: 83 (SGSETPGTSESATPES), SEQ ID NO: 84 ((GGGGS)3), SEQ ID NO: 85 ((GGGGS)10), SEQ ID NO: 117 ((GGGGS)20), SEQ ID NO: 86 (A(EAAAK)3A), SEQ ID NO: 123 (A(EAAAK)10A), or SEQ ID NO: 124 (A(EAAAK)2MA).


In some embodiments, the fusion protein further comprises a localization element. In some embodiments, the localization element is fused to the N-terminus or the C-terminus of the fusion protein. As used herein, a localization element targets or localizes the fusion protein to one or more subcellular compartments. Subcellular compartments include but are not limited to, the nucleus, the endoplasmic reticulum, the mitochondria, chromatin, the cellular membrane, and RNA granules (for example, P-bodies, stress granules and transport granules). In some embodiments, the fusion protein can be targeted to the nuclear lamina, nuclear speckles nuclear paraspeckles in the nucleus of a cell. In some embodiments, the protein can be targeted to the outer mitochondrial membrane or the inner mitochondrial membrane.


Exemplary localization elements include, but are not limited to, a peptide comprising a nuclear localization signal, for example, SEQ ID NO: 89 (PKKKRKV), a peptide comprising a nuclear export signal, for example, SEQ ID NO: 90 (LPPLERLTL), a peptide comprising an endoplasmic reticulum targeting sequence, for example, SEQ ID NO: 91 (MDPVVVLGLCLSCLLLLSLWKQSYGGG), or SEQ ID NO: 92 (METDTLLLWVLLLWVPGSTGD), a peptide comprising a Myc tag, for example, SEQ ID NO: 93 (EQKLISEEDL), a peptide comprising a V5 tag, for example, SEQ ID NO:94 (GKPIPNPLLGLDST) or SEQ ID NO: 95 (IPNPLLGLD), a peptide comprising a FLAG tag, for example, SEQ ID NO: 96 (DYKDDDDK), a peptide comprising a 3×FLAG tag, for example, SEQ ID NO: 97 (DYKDHDGDYKDHDIDYKDDDDK) and a peptide comprising a DHFR destabilization domain, for example, SEQ ID NO: 98 (ISLIAALAVDHVIGMETVMPWNLPADLAWFKRNTLNKPVIMGRHTWESIGRPLPGRKNI ILSSQPSTDDRVTWVKSVDEAIAACGDVPEIMVIGGGRVYEQFLPKAQKLYLTHIDAEVE GDTHFPDYEPDDWESVFSEFHDADAQNSHSYCFEILERR). HA tags and NLS tags can also be utilized as known in the art.


Exemplary targeting effector proteins, such as catalytically-inactive RNA-guided endonucleases are provided above in the definitions above (for example, dCas9 and dCas13).


Exemplary effector proteins being tumor suppression proteins include p53 and SOCS2. In some embodiments, p53 comprises a polypeptide (or a polynucleotide encoding a polypeptide) having at least 95% identity, for example, at least about 95%, 96%, 97%, 98% or 99% identity, to SEQ ID NO: 125. In some embodiments, human SOCS2 comprises a polypeptide (or a polynucleotide encoding a polypeptide) having at least 95% identity, for example, at least about 95%, 96%, 97%, 98% or 99% identity, to SEQ ID NO: 126. Other tumor suppression proteins may be utilized, for example, those that affect the cell cycle or other proteins that are upstream or downstream of the JAK/STAT signaling pathway.


III. Polypeptides

Provided herein are polypeptides that relate to methyladenosine (m6A) sensors and systems for detecting m6A modifications, in addition to effector protein expression systems and systems for targeting sensing and/or effector expression. Polypeptides as described herein can comprise more than one coding sequence for a protein of interest that are translationally fused so as to create a fusion protein. Provided herein are polypeptides encoded by any of the polynucleotides as described herein.


Modifications to any of the polypeptides or proteins provided herein are made by known methods. By way of example, modifications are made by site specific mutagenesis of nucleotides in a nucleic acid encoding the polypeptide, thereby producing a DNA encoding the modification, and thereafter expressing the DNA in recombinant cell culture to produce the encoded polypeptide. Techniques for making substitution mutations at predetermined sites in DNA having a known sequence are well known. For example, M13 primer mutagenesis and PCR-based mutagenesis methods can be used to make one or more substitution mutations. Any of the nucleic acid sequences provided herein can be codon-optimized to alter, for example, maximize expression, in a host cell or organism.


The amino acids in the polypeptides described herein can be any of the 20 naturally occurring amino acids, D-stereoisomers of the naturally occurring amino acids, unnatural amino acids, and chemically modified amino acids. Unnatural amino acids (that is, those that are not naturally found in proteins) are also known in the art, as set forth in, for example, Zhang et al. “Protein engineering with unnatural amino acids,” Curr. Opin. Struct. Biol. 23(4): 581-587 (2013); Xie et al. “Adding amino acids to the genetic repertoire,” 9(6): 548-54 (2005)); and all references cited therein. B and γ amino acids are known in the art and are also contemplated herein as unnatural amino acids.


As used herein, a chemically modified amino acid refers to an amino acid whose side chain has been chemically modified. For example, a side chain can be modified to comprise a signaling moiety, such as a fluorophore or a radiolabel. A side chain can also be modified to comprise a new functional group, such as a thiol, carboxylic acid, or amino group. Post-translationally modified amino acids are also included in the definition of chemically modified amino acids.


Also contemplated are conservative amino acid substitutions. By way of example, conservative amino acid substitutions can be made in one or more of the amino acid residues, for example, in one or more lysine residues of any of the polypeptides provided herein. One of skill in the art would know that a conservative substitution is the replacement of one amino acid residue with another that is biologically and/or chemically similar. The following eight groups each contain amino acids that are conservative substitutions for one another:

    • 1) Alanine (A), Glycine (G);
    • 2) Aspartic acid (D), Glutamic acid (E);
    • 3) Asparagine (N), Glutamine (Q);
    • 4) Arginine (R), Lysine (K);
    • 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V);
    • 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W);
    • 7) Serine (S), Threonine (T); and
    • 8) Cysteine (C), Methionine (M).


By way of example, when an arginine to serine is mentioned, also contemplated is a conservative substitution for the serine (e.g., threonine). Nonconservative substitutions, for example, substituting a lysine with an asparagine, are also contemplated.


IV. Polynucleotides

Recombinant nucleic acids encoding any of the polypeptides described herein are also provided.


As used throughout, the term “nucleic acid” or “nucleotide” refers to deoxyribonucleic acids (DNA) or ribonucleic acids (RNA) and polymers thereof in either single- or double-stranded form. It is understood that when an RNA is described, its corresponding cDNA is also described, wherein uridine is represented as thymidine. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides that have similar properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. A nucleic acid sequence (i.e., a polynucleotide) can comprise combinations of deoxyribonucleic acids and ribonucleic acids. Such deoxyribonucleic acids and ribonucleic acids include both naturally occurring molecules and synthetic analogues. The polynucleotides of the invention also encompass all forms of sequences including, but not limited to, single-stranded forms, double-stranded forms, hairpins, stem-and-loop structures, and the like.


Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, and complementary sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues. See Batzer et al., Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 (1985); and Rossolini et al., Mol. Cell. Probes 8:91-98 (1994).


The term “identity” or “substantial identity,” as used in the context of a polynucleotide or polypeptide sequence described herein, refers to a sequence that has at least 60% sequence identity to a reference sequence. Alternatively, percent identity can be any integer from 60% to 100%. Exemplary embodiments include at least: 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%, as compared to a reference sequence using the programs described herein; preferably BLAST using standard parameters, as described below. One of skill will recognize that these values can be appropriately adjusted to determine corresponding identity of proteins encoded by two nucleotide sequences by taking into account codon degeneracy, amino acid similarity, reading frame positioning, and the like.


For sequence comparison, typically one sequence acts as a reference sequence to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated. The sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.


A “comparison window,” as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Methods of alignment of sequences for comparison are well-known in the art. Optimal alignment of sequences for comparison may be conducted by the local homology algorithm of Smith and Waterman Add. APL. Math. 2:482 (1981), by the homology alignment algorithm of Needleman and Wunsch J. Mol. Biol. 48:443 (1970), by the search for similarity method of Pearson and Lipman Proc. Natl. Acad. Sci. (U.S.A.) 85: 2444 (1988), by computerized implementations of these algorithms (e.g., BLAST), or by manual alignment and visual inspection.


Algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1990) J. Mol. Biol. 215: 403-410 and Altschul et al. (1977) Nucleic Acids Res. 25: 3389-3402, respectively. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (NCBI) web site. The algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al, supra). These initial neighborhood word hits acts as seeds for initiating searches to find longer HSPs containing them. The word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always <0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a word size (W) of 28, an Iectation (E) of 10, M=1, N=−2, and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a word size (W) of 3, Iexpectation (E) of 10, and the BLOSUM62 scoring matrix. See Henikoff & Henikoff, Proc. Natl. Acad. Sci. USA 89:10915 (1989).


The BLAST algorithm also performs a statistical analysis of the similarity between two sequences. See, e.g., Karlin & Altshcul, Proc. Nat'l. Acad. Sci. USA 90:5873-5787 (1993). One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.01, more preferably less than about 10−5, and most preferably less than about 10−20.


A target-specific guide RNA (gRNA) can comprise a nucleotide sequence that is complementary to a polynucleotide or RNA target sequence as described herein (for example one encoding a GEMS as described herein), and thereby mediates binding of the Cas-gRNA complex by hybridization at the target site. A target-specific guide RNA (gRNA) can comprise a nucleotide sequence that is complementary to a polynucleotide or RNA target sequence as described herein (for example METLL3, or other methylation target or therapeutic target in the cell, for example, a regulator of the cell cycle or protein involved in the JAK/STAT signaling pathway), and thereby mediates binding of the Cas-gRNA complex by hybridization at the target site. In certain embodiments, the gRNA is 5-50 nucleotides, 10-30 nucleotides, 15-25 nucleotides, 18-22 nucleotides, or 19-21 nucleotides in length, or any length between the stated ranges, including, for example, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 nucleotides in length.


V. Constructs, Vectors, Host Cells, and Animal Models
A. Constructs

Provided herein are DNA constructs comprising aspects of expression systems as described herein, for example, components as described in Section I and II above.


The recombinant nucleic acids provided herein can be included in expression cassettes for expression in a host cell or an organism of interest. The cassette will include 5′ and 3′ regulatory sequences operably linked to a recombinant nucleic acid provided herein that allows for expression of the modified polypeptide. The cassette may additionally contain at least one additional gene or genetic element to be co-transformed into the cell or organism. Where additional genes or elements are included, the components are operably linked. Alternatively, the additional gene(s) or element(s) can be provided on multiple expression cassettes. Such an expression cassette is provided with a plurality of restriction sites and/or recombination sites for insertion of the polynucleotides to be under the transcriptional regulation of the regulatory regions. The expression cassette may additionally contain a selectable marker gene. The expression cassette will include in the 5′ to 3′ direction of transcription: a transcriptional and translational initiation region (i.e., a promoter), a polynucleotide of the invention, and a transcriptional and translational termination region (i.e., termination region) functional in the cell or organism of interest. The promoters of the invention are capable of directing or driving expression of a coding sequence in a host cell. The regulatory regions (i.e., promoters, transcriptional regulatory regions, and translational termination regions) may be endogenous or heterologous to the host cell or to each other. As used herein, “heterologous” in reference to a sequence is a sequence that originates from a foreign species, or, if from the same species, is substantially modified from its native form in composition and/or genomic locus by deliberate human intervention.


Additional regulatory signals include, but are not limited to, transcriptional initiation start sites, operators, activators, enhancers, other regulatory elements, ribosomal binding sites, an initiation codon, termination signals, and the like. See Sambrook et al. (1992) Molecular Cloning: A Laboratory Manual, ed. Maniatis et al. (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.); Davis et al., eds. (1980) Advanced Bacterial Genetics (Cold Spring Harbor Laboratory Press), Cold Spring Harbor, N.Y., and the references cited therein.


The expression cassette can also comprise a selectable marker gene for the selection of transformed cells. Marker genes include genes conferring antibiotic resistance, such as those conferring hygromycin resistance, ampicillin resistance, gentamicin resistance, neomycin resistance, to name a few. Additional selectable markers are known and any can be used.


In preparing the expression cassette, the various DNA fragments may be manipulated, so as to provide for the DNA sequences in the proper orientation and, as appropriate, in the proper reading frame. Toward this end, adapters or linkers may be employed to join the DNA fragments or other manipulations may be involved to provide for convenient restriction sites, removal of superfluous DNA, removal of restriction sites, or the like. For this purpose, in vitro mutagenesis, primer repair, restriction, annealing, resubstitutions, e.g., transitions and transversions, may be involved.


In preparing the expression cassette, the various DNA fragments may be manipulated, so as to provide for the DNA sequences in the proper orientation and, as appropriate, in the proper reading frame. Toward this end, adapters or linkers may be employed to join the DNA fragments or other manipulations may be involved to provide for convenient restriction sites, removal of superfluous DNA, removal of restriction sites, or the like. For this purpose, in vitro mutagenesis, primer repair, restriction, annealing, resubstitutions, e.g., transitions and transversions, may be used.


B. Vectors

Further provided is a vector comprising a nucleic acid or expression cassette set forth herein. The vector is contemplated to have the necessary functional elements that direct and regulate transcription of the inserted nucleic acid. These functional elements include, but are not limited to, a promoter, regions upstream or downstream of the promoter, such as enhancers that may regulate the transcriptional activity of the promoter, an origin of replication, appropriate restriction sites to facilitate cloning of inserts adjacent to the promoter, antibiotic resistance genes or other markers which can serve to select for cells containing the vector or the vector containing the insert, RNA splice junctions, a transcription termination region, or any other region which may serve to facilitate the expression of the inserted gene or hybrid gene. See generally, Sambrook et al. Molecular Cloning: A Laboratory Manual, 4th ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 2012. The vector, for example, can be a plasmid.


There are numerous E. coli expression vectors known to one of ordinary skill in the art, which are useful for the expression of a nucleic acid. Other microbial hosts suitable for use include bacilli, such as Bacillus subtilis, and other enterobacteriaceae, such as Salmonella, Senatia, and various Pseudomonas species. In these prokaryotic hosts, one can also make expression vectors, which will typically contain expression control sequences compatible with the host cell (e.g., an origin of replication). In addition, any number of a variety of well-known promoters will be present, such as the lactose promoter system, a tryptophan (Trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda. Additionally, yeast expression can be used. Provided herein is a nucleic acid encoding a polypeptide of the present invention, wherein the nucleic acid can be expressed by a yeast cell. More specifically, the nucleic acid can be expressed by Pichia pastoris or S. cerevisiae.


Mammalian cells also permit the expression of proteins in an environment that favors important post-translational modifications such as folding and cysteine pairing, addition of complex carbohydrate structures, and secretion of active protein. Vectors useful for the expression of active proteins in mammalian cells are known in the art and can contain genes conferring hygromycin resistance, geneticin or G418 resistance, or other genes or phenotypes suitable for use as selectable markers, or methotrexate resistance for gene amplification. A number of suitable host cell lines capable of secreting intact human proteins have been developed in the art, and include CHO cells, HeLa cells, HEK-293 cells, HEK-293T cells, U2OS cells, or any other primary or transformed cell line. Other suitable host cell lines include COS-7 cells, myeloma cell lines, Jurkat cells, etc. Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, an enhancer, and necessary information processing sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences. Preferred expression control sequences are promoters derived from immunoglobulin genes, SV40, Adenovirus, Bovine Papilloma Virus, etc.


The expression vectors described herein can also include the nucleic acids as described herein under the control of an inducible promoter such as the tetracycline inducible promoter or a glucocorticoid inducible promoter. The nucleic acids of the present invention can also be under the control of a tissue-specific promoter to promote expression of the nucleic acid in specific cells, tissues or organs. Any regulatable promoter, such as a metallothionein promoter, a heat-shock promoter, and other regulatable promoters, of which many examples are well known in the art are also contemplated. Furthermore, a Cre-loxP inducible system can also be used, as well as a Flp recombinase inducible promoter system, both of which are known in the art.


Insect cells also permit the expression of the polypeptides. Recombinant proteins produced in insect cells with baculovirus vectors undergo post-translational modifications similar to that of wild-type mammalian proteins.


Also provided herein is a vector comprising the polynucleotides as described herein. The vector may be a DNA vector or a RNA vector. In some embodiments, the vector is a non-viral vector (e.g., a plasmid or naked DNA) or a viral vector. In some embodiments, the vector is a viral vector. Examples of viral vectors include, but are not limited to, an adeno-associated virus (AAV) vector, a retroviral vector, a lentiviral vector, a herpes simplex viral vector, or an adenoviral vector. It is understood that any of the viral vectors described herein can be packaged into viral particles or virions for administration to the subject.


In some aspects, the disclosure provides a virus comprising the nucleic acid comprising a nucleotide sequence encoding a polypeptide as described herein or the viral vector as described herein. The virus may be a AAV, a lentivirus, or a retrovirus.


Non-viral vectors can also be used to deliver the polynucleotides described herein. Accordingly, in some embodiments, the vector is a non-viral vector. For example, non-viral systems, such as naked DNA formulated as a microparticle, may be used. In some embodiments, delivery may include using virus-like particles (VLPs), cationic liposomes, nanoparticles, cell-derived nanovesicles, direct nucleic acid injection, hydrodynamic injection, use of nucleic acid condensing peptides and non-peptides. In one approach, virus-like particles (VLP's) are used to deliver the polypeptide(s). The VLP comprises an engineered version of a viral vector, where nucleic acids are packaged into VLPs through alternative mechanisms (e.g., mRNA recruitment, protein fusions, protein-protein binding). See Itaka and Kataoka, 2009, “Recent development of nonviral gene delivery systems with virus-like structures and mechanisms,” Eur J Pharma and Biopharma 71:475-483; and Keeler et al., 2017, “Gene Therapy 2017: Progress and Future Directions” Clin. Transl. Sci. (2017) 10, 242-248, incorporated by reference.


C. Host Cells and Animal Models

Aspects of this disclosure include host cells and transgenic animals comprising the nucleic acid sequences or constructs described herein as well as methods of making such cells and transgenic animals.


a. Host Cells


A host cell comprising a nucleic acid or a vector or an expression as described herein is provided. The host cell can be an in vitro, ex vivo, or in vivo host cell. Populations of any of the host cells described herein are also provided. A cell culture comprising one or more host cells described herein is also provided. Methods for the culture and production of many cells, including cells of bacterial (for example E. coli and other bacterial strains), animal (especially mammalian), and archebacterial origin are available in the art. See e.g., Sambrook, Ausubel, and Berger (all supra), as well as Freshney (1994) Culture of Animal Cells, a Manual of Basic Technique, 3rd Ed., Wiley-Liss, New York and the references cited therein; Doyle and Griffiths (1997) Mammalian Cell Culture: Essential Techniques John Wiley and Sons, NY; Humason (1979) Animal Tissue Techniques, 4th Ed. W.H. Freeman and Company; and Ricciardelli, et al., (1989) In vitro Cell Dev. Biol. 25:1016-1024.


The host cell can be a prokaryotic cell, including, for example, a bacterial cell. Alternatively, the cell can be a eukaryotic cell, for example, a mammalian cell. In some embodiments, the cell can be an HEK293T cell, a Chinese hamster ovary (CHO) cell, a COS-7 cell, a HELA cell, an avian cell, a myeloma cell, a Pichia cell, an insect cell or a plant cell. A number of other suitable host cell lines have been developed and include myeloma cell lines, fibroblast cell lines, and a variety of tumor cell lines such as melanoma cell lines. The vectors containing the nucleic acid segments of interest can be transferred or introduced into the host cell by well-known methods, which vary depending on the type of cellular host. Host cells can be derived from any of the animals models discussed in (b) below.


In some embodiments, the provided cells express the protein stably or transiently by introducing an expression system (or any component thereof) into the cell. Stable expression of the protein in a cell refers to integration of any of the nucleic acids, DNA constructs, or vectors described herein into the genome of the cell, thereby allowing the cell to express the protein. Transient expression refers to expression of the protein directly from any of the nucleic acids, DNA constructs, and/or vectors following introduction into the cell (i.e., the gene encoding the protein is not integrated into the genome of the cell).


As used herein, the phrase “introducing” in the context of introducing a nucleic acid into a cell refers to the translocation of the nucleic acid sequence from outside a cell to inside the cell. In some cases, introducing refers to translocation of the nucleic acid from outside the cell to inside the nucleus of the cell. Various methods of such translocation are contemplated, including but not limited to, electroporation, nanoparticle delivery, viral delivery, contact with nanowires or nanotubes, receptor mediated internalization, translocation via cell penetrating peptides, liposome mediated translocation, DEAE dextran, lipofectamine, calcium phosphate or any method now known or identified in the future for introduction of nucleic acids into prokaryotic or eukaryotic cellular hosts. A targeted nuclease system (e.g., an RNA-guided nuclease, a transcription activator-like effector nuclease (TALEN), a zinc finger nuclease (ZFN), or a megaTAL (MT) (Li et al. Signal Transduction and Targeted Therapy 5, Article No. 1 (2020)) can also be used to introduce a nucleic acid, for example, a nucleic acid encoding a fusion protein and/or mRNA transcript (e.g, mRNA reporter mRNA) described herein, into a host cell.


In some embodiments, the provided cells express the protein constitutively or inducibly. Constitutive expression refers to ongoing, continuous expression of a gene (i.e., of a protein), whereas inducible expression refers to gene (protein) expression that is responsive to a stimulus. Inducible expression is generally regulated via an inducible promoter, a description of which is included above.


The CRISPR/Cas9 system, an RNA-guided nuclease system that employs a Cas9 endonuclease, can be used to edit the genome of a host cell or organism. Other RNA-guided CAS effector proteins can be used as well, for example, Cas13. The “CRISPR/Cas” system refers to a widespread class of bacterial systems for defense against foreign nucleic acid. CRISPR/Cas systems are found in a wide range of eubacterial and archaeal organisms. CRISPR/Cas systems include type I, II, and III sub-types. Wild-type type II CRISPR/Cas systems utilize an RNA-mediated nuclease, for example, Cas9, in complex with guide and activating RNA to recognize and cleave foreign nucleic acid. Guide RNAs having the activity of both a guide RNA and an activating RNA are also known in the art. In some cases, such dual activity guide RNAs are referred to as a single guide RNA (sgRNA).


Any of the components encoded by the nucleic acid constructs described herein, for example, fusion proteins or a m6A/effector protein fusion protein, can be purified or isolated from a host cell or population of host cells. For example, a recombinant nucleic acid encoding any of the fusion proteins described herein can be introduced into a host cell under conditions that allow expression of the fusion protein. In some embodiments, the recombinant nucleic acid is codon-optimized for expression. After expression in the host cell, the fusion protein can be isolated or purified. Similarly, any of the nucleic acids encoding a m6A reporter mRNA described herein can be introduced into a host cell under conditions that allow transcription of the m6A reporter mRNA. After expression in the host cell, the m6A reporter mRNA can be isolated or purified.


b. Animal Models


Also provided is a non-human transgenic animal comprising a mammalian host cell that comprises any of the nucleic acid sequences or constructs described herein. Methods for making transgenic animals, include, but are not limited to, oocyte pronuclear DNA microinjection, intracytoplasmic sperm injection, embryonic stem cell manipulation, somatic nuclear transfer, recombinase systems (for example, Cre-LoxP systems, Flp-FRT systems and others), zinc finger nucleases (ZNFs), transcriptional activator-like effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeat/CRISPR-associated protein 9 (CRISPR/Cas9). See, for example, Volobueva et al. Braz. J. Med. Biol. Res. 52(5): e8108 (2019)).


The term “transgenic animal” as used herein means an animal into which a genetic modification has been introduced by a genetic engineering procedure and in particular an animal into which has been introduced an exogenous nucleic acid, and may loosely also encompass “knock in” animals. That is the animal comprises a nucleic acid sequence which is not normally present in the animal. Such animals can be created by a one-for-one substitution of DNA sequence information in a predetermined genetic locus or the insertion of sequence information not found within the locus.


A transgenic animal may be developed, for example, from embryonic cells into which the genetic modification (e.g. exogenous nucleic acid sequence) has been directly introduced or from the progeny of such cells. The exogenous nucleic acid is introduced artificially into the animal (e.g. into a founder animal). Animals that are produced by transfer of an exogenous nucleic acid through breeding of the animal comprising the nucleic acid (into whom the nucleic acid was artificially introduced), which are progeny animals, are also included. Representative examples of non-human mammals include, but are not limited to non-human primates, mice, rats, rabbits, pigs, goats, sheep, horses, zebrafish and cows. A cell or a population of cells from any of the non-human transgenic animals provided herein is also provided.


The exogenous nucleic acid may be integrated into the genome of the animal or it may be present in an non-integrated form, e.g. as an autonomously-replicating unit, for example, an artificial chromosome which does not integrate into the genome, but which is maintained and inherited substantially stably in the animal. In some embodiments, the exogenous nucleic acid is under the control of a cell-specific or tissue-specific promoter. For example, transgenic animals that express a fusion protein and a mRNA reporter sequence in specific cells or tissues can be produced by introducing one or more nucleic acids into fertilized eggs, embryonic stem cells or the germline of the animal, wherein the one or more nucleic acids are under the control of a specific promoter which allows expression of the nucleic acid fusion protein and mRNA reporter sequence in specific types of cells or tissues. As used herein, a protein or mRNA is expressed predominantly in a given tissue, cell type, cell lineage or cell, when 90% or greater of the observed expression occurs in the given tissue cell type, cell lineage or cell.


In some embodiments, the exogenous nucleic acid in the animal is under the control of a constitutive or an inducible promoter, as described above. Inducible systems can also be used to allow expression of the fusion and/or mRNA reporter sequence at designated times during development, expanding the temporal specificity of fusion protein and/or mRNA reporter expression in the transgenic animal.


Included are both progenitor and progeny animals. Progeny animals include animals which are descended from the progenitor as a result of sexual reproduction or cloning and which have inherited genetic material from the progenitor. Thus, the progeny animals comprise the genetic modification introduced into the parent. A transgenic animal may be developed, for example, from embryonic cells into which the genetic modification (e.g. exogenous nucleic acid sequence) has been directly introduced or from the progeny of such cells. The exogenous nucleic acid is introduced artificially into the animal (e.g. into a founder animal). Animals that are produced by transfer of an exogenous nucleic acid through breeding of the animal comprising the nucleic acid (into whom the nucleic acid was artificially introduced), which are progeny animals, are also included.


Although the present disclosure is described primarily in a mouse, one of ordinary skill in the art would understand that other non-human mammals, for example, rodent, rabbit, bovine, ovine, canine, feline, equine, porcine, camelid, non-human primate, and other mammals, can also be engineered to express aspects of the present disclosure in a similar fashion, and these transgenic animals can also be used for applications as disclosed herein. A cell or a population of cells from any of the non-human transgenic animals provided herein is also provided.


A. Pharmaceutical Compositions

Also provided herein are pharmaceutical compositions of the nucleic acids, the vectors, the viruses, or the cells described herein. The pharmaceutical compositions described herein are for delivery to subjects in need thereof by any suitable route or a combination of different routes. The pharmaceutical compositions can be delivered to a subject, so as to allow expression of the polypeptide in cells of the subject and produce an effective amount of the polypeptide that treats a condition in the subject. In some embodiments, the pharmaceutical composition comprising the nucleic acid, the vector, the virus, or the cell as described herein further comprises a pharmaceutically acceptable excipient or carrier.


The terms “pharmaceutically acceptable carrier” and “pharmaceutically acceptable excipient” are used interchangeably and refer to a substance or compound that aids or facilitates preparation, storage, administration, delivery, effectiveness, absorption by a subject, or any other feature of the composition for its intended use or purpose. Such pharmaceutically acceptable carrier is not biologically or otherwise undesirable and can be included in the compositions of the present invention without causing a significant adverse toxicological effect on the subject or interacting in a deleterious manner with the other components of the pharmaceutical composition.


In some approaches, sterile injectable solutions can be prepared with the vectors in the required amount and an excipient suitable for injection into a human patient. In some embodiments, the pharmaceutically and/or physiologically acceptable excipient is particularly suitable for administration to the cardiac muscle. For example, a suitable carrier may be buffered saline or other buffers, e.g., HEPES, to maintain pH at appropriate physiological levels, stabilizing agents, adjuvants, diluents, or surfactants. In some embodiments, the pharmaceutically acceptable excipient comprises a non-ionic detergent, such as, for example, Pluronic F-681. For injection, the excipient will typically be a liquid. Exemplary pharmaceutically acceptable excipients include sterile, pyrogen-free water and sterile, pyrogen-free, phosphate buffered saline. Pharmaceutically acceptable salts can be included therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. The preparation of pharmaceutically acceptable carriers, excipients and formulations is described in, e.g., Remington: The Science and Practice of Pharmacy, 22nd edition, Loyd V. Allen et al, editors, Pharmaceutical Press (2012). See also Bennicelli et al., “Reversal of blindness in animal models of leber congenital amaurosis using optimized AAV2-mediated gene transfer,” Mol Ther. (2008); 16(3):458-65. A variety of known carriers are also provided in U.S. Pat. Nos. 7,629,322, and 6,764,845, incorporated herein by reference.


VI. Methods

Provided herein are methods for inducing m6A methylation-dependent expression of a heterologous polypeptide (comprising an effector protein) in one or more cells, a biological samples (for example a group of cells or tissue biopsy from a mammalian subject), or a subject having or suspected of having a cancer as described herein (or otherwise derived from a cancer in the case of cells in vitro), comprising introducing or administering any of the expression systems described herein into one or more cells, the sample, or the subject. As set forth above, when any of the expression systems described herein is introduced into a cell, sample, or subject, if m6A methylation occurs in the cell, the effector protein expressed by the expression system, i.e., a mRNA comprising a heterologous protein, a m6A sensor sequence and a destabilization domain (e.g., DHFR), will be methylated (at the m6A sensor sequence). Upon methylation, C to U editing results in a stop codon in the m6A sensor sequence that inhibits expression of DHFR, thus allowing the heterologous protein to be expressed without degradation.


In embodiments, the cell can be an in vitro, ex vivo or in vivo cell. The cell may be a mammalian cell or a rodent cell.


Provided herein are also methods for administering or introducing any of the expression systems described herein to a cell, sample, or subject as described herein. The administering or introducing to one or more cells, a sample, or a subject, can be by mechanisms known in the art to introduce exogenous nucleic acids into cells, for example, lipofection, nucleofection, or electroporation. Alternatively, the skilled artisan would understand that aspects of expression systems as described herein can be cloned into viral expression vectors and packaged into an adenoviral (AAV) or lentiviral (LV) vector, and subsequently used to transduce the exogenous genetic material into the cell, sample, or subject.


Also provided is a virus (e.g., an AAV, a lentivirus, or a retrovirus) comprising any of the nucleic acids or vectors described in this disclosure.


Also provided is a cell comprising any of the nucleic acids, vectors, or viruses described in this disclosure.


Provided herein is also a pharmaceutical composition comprising any of the nucleic acids, vectors, viruses, or cells described herein, and a pharmaceutically acceptable excipient.


A. Tumor Suppression

One aspect provided in this disclosure is a method of inhibiting a cancer cell, the method comprising introducing into the cancer cell the expression system as provided in this disclosure. In some embodiments, inhibiting the cancer cell by methods as described herein results in decreasing at least one of cell proliferation, cell migration, or metastasis.


In some embodiments of this method, the cancer cell can comprise m6A RNA hypermethylation. In some embodiments, the cancer cell comprises an acute myeloid leukemia (AML) cell, a glioblastoma (GBM) cell, a lung cancer cell, an endometrial cancer, a cervical cancer cell, an ovarian cancer cell, a breast cancer cell, a colorectal cancer (CRC) cell, a hepatocellular carcinoma (HCC) cell, a pancreatic cancer cell, a gastric cancer cell, a prostate cancer cell, or a renal cell carcinoma cell. In certain embodiments, the lung cancer cell is a non-small cell lung carcinoma cell. In certain embodiments, the cancer cell is a hepatocellular carcinoma cell.


In some embodiments, the second DNA construct comprises a polynucleotide encoding an effector protein, wherein the effector protein comprises a tumor suppressor protein. In some embodiments, expression of the tumor suppressor protein upregulates downstream signaling targets. The tumor suppressor protein may comprise at least one of the tumor suppressor proteins listed in Table 1. In certain embodiments, the tumor suppressor protein can comprise p53. In some embodiments, expression of p53 upregulates at least one of CDKN1A or GADD45A. In certain embodiments, the tumor suppressor protein comprises suppressor of cytokine signaling 2 (SOCS2).


The expression system may be introduced into the cancer cell by viral infection (in particular, adenoviral, lentiviral, or AAV infection).


In another aspect of this disclosure, provided herein is a method of treating a subject having a cancer characterized by m6A RNA hypermethylation, the method comprising introducing into a cancer cell in the subject the expression system as provided in this disclosure. In some embodiments, the method comprises inhibiting a cancer cell of the subject's cancer in the subject. In some embodiments, expression of the tumor suppressor protein results in decreasing at least one of cell proliferation, cell migration, or metastasis of the cancer.


In some embodiments, the cancer can comprise acute myeloid leukemia (AML), glioblastoma (GBM), lung cancer, endometrial cancer, cervical cancer, ovarian cancer, breast cancer, colorectal cancer (CRC), a hepatocellular carcinoma (HCC), pancreatic cancer, gastric cancer, prostate cancer, and/or renal cell carcinoma. In certain embodiments, the cancer comprises hepatocellular carcinoma.


In some embodiments, the expression system can be introduced into the subject by viral infection (adenoviral, lentiviral, AAV).


While SOCS2 and p53 are provided as examples, the skilled artisan would recognize that other tumor suppression proteins can be expressed depending on the type of cancer by cloning a coding sequence of any of the gene products from Table 1 into the expression system as the effector protein.


Effects of expression of tumor suppression proteins according to the present disclosure include: inhibition of mitogenic signaling pathways; inhibition of cell cycle progression; inhibition of “pro-growth” programs of metabolism and angiogenesis; inhibition of invasion and metastasis; stabilization of the genome; DNA repair factors; and induction of apoptosis.


Additional examples are provided in Table 1 below:









TABLE 1







Embodiments of Tumor Suppression Proteins and Cancer


Types According to the Present Disclosure:










Gene
Type of Cancer







APC
Colon/rectum carcinoma



BRCA1
Breast and ovarian carcinomas



BRCA2
Breast carcinoma



CDH1
Autosomal dominant familial gastric




carcinoma



CDKN2A
Melanomas, Leukemias, and Carcinomas



DPC4
Pancreatic carcinoma



INK4
Melanoma, lung carcinoma, brain tumors,




leukemias, lymphomas



MADR2
Colon/rectum carcinoma



NF1
Neurofibrosarcoma



NF2
Meningioma



p53 (i.e., TP53)
Brain tumors; breast, colon/rectum,




esophageal, liver, and lung carcinomas;




sarcomas; leukemias and lymphomas



PARP-1
Breast carcinoma



PTC
Basal cell carcinoma



PTEN
Brain tumors; melanoma; prostate,




endometrial, kidney, and lung carcinomas



Rb
Retinoblastoma; sarcomas; bladder, breast,




and lung carcinomas



SOCS2
hepatocellular carcinoma



VHL
Renal cell carcinoma



WT1
Wilms' tumor










B. m6A Regulator Targeting

Described herein are methods of reducing m6A effector regulator expression in a sample or a subject. In particular, CRISPRi can be utilized to knock-down expression of m6A “writers”, which are proteins that are responsible for m6A dysregulation (in particular hypermethylation) observed in cancer cells.


In embodiments, described herein is a method of reducing m6A effector regulator expression, comprising: introducing an expression system into a subject having or suspected of having a cancer, wherein the expression system comprises: a first DNA construct comprising a polynucleotide encoding a fusion protein, wherein the fusion protein comprises an N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase or a catalytic domain of an adenosine deaminase; and a second DNA construct comprising a polynucleotide encoding a heterologous polypeptide, the polynucleotide encoding a heterologous polypeptide comprising: a polynucleotide encoding a catalytically-dead RNA-guided endonuclease; a polynucleotide encoding a m6A sensor sequence; and a polynucleotide encoding a dihydrofolate reductase (DHFR); an sgRNA configured to bind to an m6A regulator. In embodiments, the sgRNA is configured to bind to a m6A regulator listed in Table 2. In embodiments, the cancer comprises at least one of acute myeloid leukemia (AML), glioblastoma (GBM), lung cancer, endometrial cancer, cervical cancer, ovarian cancer, breast cancer, colorectal cancer (CRC), a hepatocellular carcinoma (HCC), pancreatic cancer, gastric cancer, prostate cancer, or renal cell carcinoma. In embodiments, the cancer is a cancer listed in Table 1 or Table 2. In embodiments, the catalytically-dead RNA-guided endonuclease is a dCas9 or dCas13.


Described herein are methods of reducing m6A hypermethylation in a subject or sample. In embodiments, methods comprising: introducing an expression system into a subject having or suspected of having a cancer, wherein the expression system comprises: a first DNA construct comprising a polynucleotide encoding a fusion protein, wherein the fusion protein comprises an N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase or a catalytic domain of an adenosine deaminase; and a second DNA construct comprising a polynucleotide encoding a heterologous polypeptide, the polynucleotide encoding a heterologous polypeptide comprising: a polynucleotide encoding a catalytically-dead RNA-guided endonuclease; a polynucleotide encoding a m6A sensor sequence; and a polynucleotide encoding a dihydrofolate reductase (DHFR); an sgRNA configured to bind to an m6A regulator. The sgRNA is configured to bind to a m6A regulator listed in Table 2. The cancer comprises at least one of acute myeloid leukemia (AML), glioblastoma (GBM), lung cancer, endometrial cancer, cervical cancer, ovarian cancer, breast cancer, colorectal cancer (CRC), a hepatocellular carcinoma (HCC), pancreatic cancer, gastric cancer, prostate cancer, or renal cell carcinoma. The cancer is a cancer listed in Table 1 or Table 2. The catalytically-dead RNA-guided endonuclease is a dCas9 or dCas13.


A subject can be a subject having or suspected of having a cancer as described herein. Introducing to the subject can comprise viral infection or electroporation. Modulating m6A levels by affecting an m6A regulator can decreasing at least one of cell proliferation, cell migration, or metastasis.


While METTL3 is provided as an example, the skilled artisan would recognize that other tumor suppression proteins can be expressed depending on the type of cancer by cloning a coding sequence of any of the gene products in Table 2 below into the expression system as the effector protein in order to reduce hypermethylation (or the effects thereof). It would be recognized that m6A “washers” may be also be expressed as tumor suppression proteins, while m6A “writers” can be targeted by the catalytically-dead RNA-guided endonuclease to block protein expression of the writers.


Additional aspects of CRISPi generally, can be found, for example in Carroll & Giacca, CRISPR activation and interference as investigative tools in the cardiovascular system, Int. J. of Biochem. &Cell Bio., Volume 155, February 2023, 106348, the contents of which regarding CRISPRi are incorporated by reference as if fully set forth herein.









TABLE 2







Embodiments of M6A Regulators and Cancer Types According to the Present Disclosure:










Pathway
m6A Regulator
Cancer
Function





C-myc pathway
METTL3
Lung cancer
promote growth and migration



METTL3
Bladder cancer
promote cell proliferation, invasion and survival



METTL3
Oral squamous
promote growth, invasion, migration and progression




cell carcinoma,




Colorectal




cancer, Prostate




carcinoma



METTL3
Gastric cancer
promote proliferation and metastasis



METTL3
Acute myeloid
inhibit diferentiation and increase proliferation




leukemia



FTO
Colorectal cancer
inhibit apoptosis and improve cell proliferation, migration, and





invasion



IGF2BP2
Thyroid cancer
promote proliferation, invasion, migration and anti-apoptosis



YTHDF2
Glioblastoma
support glioblastoma stem cells viability


PI3K/AKT/mTOR pathway
YTHDF1
Gastric cancer



FTO
Endometrial
promote invasion and metastasis




cancer



YTHDF1
Colorectal
promote tumorigenicity and cell cycle




carcinoma



METTL3
Hepatocellular
accelerate development




carcinoma





Hepatocellular carcinoma



METTL3
Colorectal
promote the stemness and chemoresistance




carcinoma



METTL3
Nasopharyngeal
promote cisplatin resistance




carcinoma (NPC)



METTL14
Breast cancer
promote stemness and progression


p53 pathway
METTL3
Colorectal
promote multidrug resistance



YTHDF1 and
Melanoma
promote the development



HNRNPA2B1



METTL14
Pancreatic cancer
promote the growth and metastasis



METTL3
Breast cancer
promote the proliferation


EMT signaling pathway
METTL14
Colorectal
mediate EMT process




carcinoma



METTL3
Gastric cancer
accelerate the EMT



METTL3
Hepatocellular
promote the EMT




carcinoma



YTHDF3
Hepatocellular
facilitate migration, invasion, and EMT




carcinoma



METTL3
Lung cancer,
accelerate the EMT and promote the development




Ovarian cancer,




Colorectal




carcinoma


MAPK signaling pathway.
METTL3
Colorectal
promote metastasis




carcinoma


P38/ERK
METTL3
Colorectal
suppress proliferation, migration and invasion




carcinoma


ERK1/2 and STAT3
HNRNPA2B1
Breast cancer
promote the tumorigenicity, and decrease apoptosis


pathways


BCL-2
METTL3
Breast cancer
accelerate proliferation, decrease the apoptosis










(Adapted from Pan J, Huang T, Deng Z, Zou C. Roles and therapeutic implications of m6A modification in cancer immunotherapy. Front Immunol. 2023 Mar. 7; 14:1132601. doi: 10.3389/fimmu.2023.1132601. PMID: 36960074; PMCID: PMC10028070). Additional m6a regulators can be found, for example, in Gu. et al. RNA m6A Modification in Cancers: Molecular Mechanisms and Potential Clinical Applications, Cell, The Innovation 1, 100066, Nov. 25, 2020, as well as Chen, X Y., Zhang, J. & Zhu, J S. The role of m6A RNA methylation in human cancer. Mol Cancer 18, 103 (2019). Doi: 10.1186/s12943-019-1033-z; Chang, G., et al., RNa m6A Modification in Cancers: Molecular Mechanisms and Potential Clinical Applications. The Innovation, Vol. 1, Issue 3, Article 100066, Nov. 25, 2020. doi: 10.1016/j.xinn.2020.100066; and Chen, X. Y., et al. The role of m6A RNA methylation in human cancer. Molecular Cancer, Vol. 18, Article 103 (2019). doi: 10.1186/s12943-019-1033-z, molecular-cancer.biomedcentral.com/articles/10.1186/s12943-019-1033-z the contents of all of which are incorporated by reference regarding m6A regulators and cancers and effectors of regulators the m6A regulators.


Also provided are methods of treating a disease or disorder in a subject in need thereof, wherein the method comprises administering any of the expressions systems described herein to the subject. In some methods, the subject has cancer. In some methods, the subject is diagnosed with a disease or disorder (e.g., cancer).


As used herein, the term “administering” “administration”, or “administer” means delivering the pharmaceutical composition as described herein to a target cell or a subject. Administration refers to the act of introducing, injecting or otherwise physically delivering a substance as it exists outside the body (e.g., one or more nucleic acids, vectors, viruses, cells, or pharmaceutical compositions described herein) into a subject. The compositions described herein can be delivered to subjects in need thereof by any suitable route or a combination of different routes. Any suitable route of administration or combination of different routes can be used, including systemic administration (e.g., intravenous, intravascular, or intra-arterial injection), local injection into the heart muscle, local injection into the CNS (e.g., intracranial injection, intracerebral injection, intracerebroventricular, or injection into the Cerebrospinal fluid (CSF) via the cerebral ventricular system, cisterna magna, or intrathecal space), or local injection at other bodily sites (e.g. intraocular, intramuscular, subcutaneous, intradermal, or transdermal injection). In some embodiments, the compositions described herein are administered into the coronary arteries. In some embodiments, the compositions described herein are administered into the coronary sinus.


As used herein the terms “treatment”, “treat”, or “treating” refers to a clinical intervention made in response to a disease, disorder or physiological condition manifested by a patient or to which a patient may be susceptible. The aim of treatment includes the reduction, alleviation, slowing, or stopping the progression or worsening of a disease, disorder, or condition including reducing or preventing one or more of the effects or symptoms of the disease, disorder, or condition and/or the remission of the disease, disorder or condition, for example, a cardiac condition, in the subject. Thus, in the disclosed methods, treatment can refer to a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% reduction in the severity of a cardiac condition. For example, a method for treating a cardiac condition is considered to be a treatment if there is a 10% reduction in one or more symptoms of a cardiac condition in a subject as compared to a control. Thus the reduction can be a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or any percent reduction in between 10% and 100% as compared to native or control levels. It is understood that treatment does not necessarily refer to a cure or complete ablation of the disease or symptoms of the disease.


Administration can be performed by injection, by use of an osmotic pump, by electroporation, or by other means. In some approaches, administration of the compositions of the present disclosure can be performed before, after, or simultaneously with surgical treatment.


Dosage values may depend on the nature of the product and the severity of the condition. It is to be understood that for any particular subject, specific dosage regimens can be adjusted over time and in course of the treatment according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions. Accordingly, dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition.


A therapeutically effective amount of such a composition may vary according to factors such as the disease state, age, sex, weight of the individual, and whether it is used concomitantly with other therapeutic agents. Dosage regimens may be adjusted to provide the optimum response. A suitable dose can also depend on the particular viral vector used, or the ability of the viral vector to elicit a desired response in the individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the viral vector are outweighed by the therapeutically beneficial effects. Other factors determining a dose can include, e.g., other medical disorders concurrently or previously affecting the subject, the general health of the subject, the genetic disposition of the subject, diet, time of administration, and any other additional therapeutics that are administered to the subject. It should also be understood that a specific dosage and treatment regimen for any particular subject also depends upon the judgment of the treating medical practitioner.


The effective amount of the compositions described herein can be determined by one of ordinary skill in the art. One of skill in the art will appreciate that an effective amount of a composition, for example, comprising an AAV or a lentivirus, can be empirically determined. An effective amount of any of the compositions described herein will vary and can be determined by one of skill in the art through experimentation and/or clinical trials. For example, quantification of genome copies (GC), vector genomes (VG), virus particles (VP), or infectious viral titer may be used as a measure of the dose contained in a formulation or suspension. Any method known in the art can be used to determine the GC, VG, VP or infectious viral titer of the virus compositions of the invention, including as measured by qPCR, digital droplet PCR (ddPCR), UV spectrophotometry, ELISA, next-generation sequencing, or fluorimetry as described in, e.g., in Dobkin et al., “Accurate Quantification and Characterization of Adeno-Associated Viral Vectors.” Front Microbiol 10: 1570-1583 (2019); Lock et al., “Absolute determination of single-stranded and self-complementary adeno-associated viral vector genome titers by droplet digital PCR.” Hum Gene Ther Methods 25: 115-125 (2014); Sommer, et al., “Quantification of adeno-associated virus particles and empty capsids by optical density measurement.” Mol Ther 7: 122-128 (2003); Grimm, et al. “Titration of AAV-2 particles via a novel capsid ELISA: packaging of genomes can limit production of recombinant AAV-2.” Gene Ther 6: 1322-1330 (1999); Maynard et al., “Fast-Seq: A Simple Method for Rapid and Inexpensive Validation of Packaged Single-Stranded Adeno-Associated Viral Genomes in Academic Settings.” Hum Gene Ther 30(6): 195-205 (2019); Piedra, et al., “Development of a rapid, robust, and universal picogreen-based method to titer adeno-associated vectors.” Hum Gene Ther Methods 26: 35-42 (2015); which are incorporated herein by reference. For intravenous injection, an exemplary human dosage range in vector particles (vp) may be between 5×10e13-10×10e14 vp per kilogram bodyweight (vp/kg) in a volume of 1-100,000 μl. In one embodiment, an exemplary human dose for intramuscular (cardiac muscle injection) or intracoronary delivery may be 1×10e14-5×10e14 vp per injection into the heart in a volume of 1-1000 μl.


In one approach, the composition is administered in a single dosage selected from those above listed. In another embodiment, the method involves administering the compositions in two or more dosages (e.g., split dosages). In another embodiment, multiple injections are made at different locations. In another embodiment, a second administration of the composition is performed at a later time point. Such time point may be weeks, months or years following the first administration. In some embodiments, multiple treatments may be required in any given subject over a lifetime.


EXAMPLES

As mentioned above, a targeted m6A-coupled effector protein delivery system can be used in cancer therapy. For example, METTL3 is elevated in many cancers, and hypermethylation of oncogenic mRNAs leads to increased translation and cancer progression (Vu et al. “The N(6)-methyladenosine (m(6)A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells,” Nat Med. 2017; 23(11):1369-76). Current strategies for overcoming this have focused on developing drugs that inhibit METTL3. However, this approach can have unwanted effects since it can impact the methylation of all mRNAs. Thus, using the m6A sensor system to express a tumor suppressor protein or to deliver CRISPR systems targeting upregulated oncogenes offers a more targeted approach.


Additionally, the m6A sensor system can be used to develop an m6A-coupled effector protein expression system. To demonstrate the utility and versatility of this technology, m6A sensor systems can be engineered and utilized to deliver a tumor suppressor protein to counteract the effects of hypermethylation in cancer cells, and (in embodiments) to express METTL3-targeting CRISPRi tools to maintain cellular m6A levels through a METTL3 feedback mechanism or express other tumor suppression proteins (such as cycle proteins like p53 for example). The utility of the system, to influence physiological outcomes can also be studied.


Example 1: Materials and Methods

Cell culture. All cell types used in this study were cultured at 37° C. and 5% CO2 using the recommended cell type-specific growth medium. HEK293T cells (ATCC, CRL-3216), HeLa cells (ATCC, CRM-CCL-2), and NIH/3T3 cells (ATCC, CRL-1658) were cultured in Dulbecco's Modified 430 Eagle's Medium (DMEM, Corning). A549 cells (ATCC, CCL-185) and CHO-K1 cells (ATCC, CCL61) were cultured in Ham's F-12K (Kaighn's) Medium (Gibco). Huh-7 cells (obtained through the Duke University Cell Culture Facility) were cultured in Dulbecco's Modified Eagle's Medium (DMEM, Corning) with the addition of 12.5 mL of 1M HEPES (Fisher Scientific). HepG2 cells (ATCC, HB-8065) were cultured in Gibco Minimum Essential Media (MEM, Gibco) with the addition of 1% Sodium Pyruvate (Fisher Scientific) and 1% NEAA (Fisher Scientific). METTL3 degron cells were cultured as for HEK293T cells. All cell lines were cultured with the addition of 10% fetal bovine serum (Avantor) and 10 units/mL Penicillin/10 μg/mL Streptomycin (Gibco) to their respective growth media. HEK293T cells were tested for mycoplasma infection by the Duke University Cell Culture Facility and were confirmed to be mycoplasma-free.


Plasmids and cloning. The sequence for the EGFP-DHFR reporter mRNA was synthesized using custom gene synthesis (IDT gblock). All RAC consensus motifs within the EGFP sequence were mutated to avoid m6A methylation and potential editing of the EGFP coding sequence. Synonymous/codon-445 optimized mutations were used when possible. The EGFP and DHFR coding sequences are separated by a linker region which contains the first 81 nt of the human ACTB 3′UTR with some modifications (Table 2). The “m6A sensor sequence” consists of 5′-GCGGACUUACGACAG-3′ and contains the m6A sites at positions 1216 and 1222 of ACTB, with mutations of some nearby residues to enable C-to-U editing sites that produce in-frame stop codons (Table 3). The DHFR sequence contains the E. coli DHFR gene as previously described25. This EGFP-DHFR gblock sequence was cloned into the pCMV-APOBEC1-YTH plasmid20 at Not1/XhoI sites. The resulting plasmid, pCMV-EGFP-DHFR, was used for experiments involving expression of EGFP-DHFR alone or co-transfected with APOBEC1-YTH or APOBEC1-YTHmut. For all other experiments, the pGEMS plasmid was used, which contains CMV-EGFP-DHFR and EF1a-APOBEC1-YTH. pGEMSmut is the same plasmid but contains EF1a-APOBEC1-YTHmut. To generate pGEMS and pGEMSmut, we first generated iDuet101A-APOBEC1-YTH and iDuet101A-APOBEC1-YTHmut by cloning APOBEC1-YTH/YTHmut from pCMV-APOBEC1-YTH/YTHmut into iDuet101A (a gift from Linzhao Cheng, Addgene plasmid #17629) using XbaI/ClaI sites. EGFP-DHFR was then cloned into iDuet101A-APOBEC1-YTH and -YTHmut at NruI and SanDI sites to generate pGEMS and pGEMSmut, both of which contain the EGFP-DHFR sequence under control of the CMV promoter and APOBEC1-YTH or APOBEC1-YTHmut under control of the EF1a promoter. The puromycin-P2A-rtTA from TLCV2 (a gift from Adam Karpf, Addgene #87360) was also inserted into pGEMS and pGEMSmut using Gibson assembly.


The hPGK-DsRed-Express2 construct was cloned out of LVDP-CArG-RE-GPR (a gift from Stelios Andreadis, Addgene plasmid #89762) and subcloned into pGEMS and pGEMSmut by Gibson assembly to generate pGEMS-II and pGEMSmut-II. For experiments using GEMS-EGFP-PEST, the PEST destabilization domain was subcloned from the pCAG-GFP-PEST plasmid (a gift from Debra Silver) and inserted at the c-terminus of EGFP to produce pGEMS-II-PEST. To generate pGEMS-SOCS2 and pGEMS-p53, human SOCS2 and p53 CDSs were amplified from a cDNA library prepared from HeLa cells and subcloned into pGEMS-II in place of EGFP.


The GEMS-dCas13 system (dCas13-NLS-APO1-YTH) was adapted from pCMV-dCas13-M3nls (a gift from David Liu, plasmid #155366), by subcloning dCas13-NLS upstream of APO1-YTH in the pGEMS-II-PEST plasmid. dCas13 gRNA sequences (listed in Table 3) were subcloned into the pC016 plasmid (a gift from Feng Zhang, plasmid #91906).


Plasmid Transfection. Transfections were performed using Fugene HD (Promega) according to the manufacturer's instructions. For METTL3 inhibition experiments, cells were treated with 10 μM or 30 μM of STM2457 (MedChemExpress) for 16 hours prior to transfection. Cells were treated with 0.1% DMSO (VWR Life Science) as a control. For experiments using METTL3 degron cells, 0.1 mg/mL of auxin or equivalent volume of H2O (control) was added to the cells for 24 hours prior to plasmid transfection.


Microscopy. All images were obtained using a Leica DMi8 inverted fluorescence microscope. Images were processed using the Leica LAS X software. 4-5 fields of view were obtained per sample, and representative images were selected for each experiment.


Quantitative Microscopy. HEK293T cells were plated in 10 cm cell culture-treated dishes at a density of 2.2×106 cells per plate and allowed to grow overnight. Cells were then treated with 30 μM of STM2457 or DMSO control for 16 hours. Cells were then transfected with the GEMS-PEST-DsRed plasmid and allowed to grow for an additional 16 hours. Prior to imaging, media was replaced with 1×PBS with 1 μg/mL Hoechst nuclear fluorescent stain (ThermoFisher Scientific) for 30 minutes. Cells were imaged using a Leica DMi8 fluorescence microscope. Images were analyzed by ImageJ image analysis software. First, fluorescence channels were separated and RGB fluorescence channels were converted to grayscale (16-bit). Binary image thresholds were set in relation to DsRed fluorescence signal and background noise was subtracted. Individual cells were selected, and pixel intensities were generated of each individual cell within the field of view. Similar binary image thresholding, cell selection, and intensity datasets were generated for the EGFP image channel. Data were analyzed by dividing EGFP signal intensity for each cell by average DsRed signal intensity. Data were plotted in JMP software (IMP 17.0, SAS Institute Inc.) by normalized EGFP signal intensity for each treatment. Boxplots represent interquartile range of the data and whiskers represent minimum and maximum data points excluding outliers. Statistical significance was calculated using a two-way t-test assuming unequal variance.


Western blotting. Cells were dissociated in culture plates using TrypLE (Gibco) and collected by centrifugation (6,000 rpm, 10 minutes, 4° C.). Cell pellets were resuspended in 150 μL chilled standard RIPA buffer (1% Triton X-100, 0.1% Sodium Deoxycholate, 0.1% SDS) prepared with the addition of Complete Mini protease inhibitor (Sigma Aldrich) immediately before use. Cells were resuspended and incubated on ice in RIPA buffer for 30 minutes. Cell lysates were cleared by centrifugation (13,000 rpm, 10 minutes, 4° C.) and mixed in a 1:1 ratio with NuPage LDS sample buffer (Invitrogen) with 5% Beta-Mercaptoethanol (Sigma). Samples were separated by gel electrophoresis on NuPage 4-12% Bis-Tris SDS-PAGE gels (Invitrogen), then transferred onto PVDF membranes (Amersham) using semi-dry transfer (Trans-Blot Turbo, Biorad). Membranes were blocked with 4% milk powder in 0.1% PBST and incubated with the appropriate primary antibody overnight at 4° C. Membranes were then washed with 0.1% PBST and incubated for 1 hour at room temperature in secondary antibody. Blots were washed and incubated in chemiluminescent ECL reagent solution (Amersham) then imaged in a ChemiDoc MP imaging system (BioRad) under chemiluminescent and colorimetric light. In western blot images, cyclophilin A is shown as a loading control and all western blots are representative of a minimum of 3 independent biological replicates. The following antibodies were used in this study: GFP tag Polyclonal antibody (Proteintech, 50430-2-AP). Anti-HA rabbit monoclonal antibody (Cell Signaling, 3724). Anti-Cyclophilin A antibody (Cell Signaling, 2175S). Anti-DsRed-Express2 Monoclonal Antibody (Fisher Scientific, CF180014). Anti-METTL3 antibody (abcam, ab195352). Anti-SOCS2 antibody (abcam, ab109245). Anti-p53 (7F5) Rabbit monoclonal antibody (Cell Signaling, 2527S). Recombinant Anti-JAK2 Antibody (abcam, ab108596). Recombinant Anti-STAT5 (phosphorylated Y694) (abcam, ab32364). Anti-STAT5 antibody (Cell Signaling, 25656S). Secondary antibodies used in this study: Goat Anti-Rabbit IgG HRP (Abcam, ab6721), Goat anti-mouse IgG HRP (Fisher Scientific, 62-6520).


For densitometry analysis, western blot images were quantified using ImageJ software 58. EGFP band intensity was normalized to either EGFP-DHFR or (EGFP+EGFP-DHFR) as indicated in each experiment. Similar densitometry analysis was used to measure total protein production (EGFP+EGFP-DHFR), normalized to the Cyclophilin A loading control. At least two replicates were used for each western blot quantification analysis. Bars are plotted as mean intensity ratio and error bars represent standard deviation.


Sanger sequencing and RT-qPCR. Cells were dissociated in culture using TrypLE (Gibco) treatment for 2 minutes and collected by centrifugation (6,000 rpm, 10 minutes, 4° C.). RNA was extracted using the RNA easy Plus Mini kit (Qiagen) according to the manufacturer's protocol. Extracted RNA was purified for genomic and plasmid DNA contamination by incubation with 1 μL DNase I at 37° C. for 30 minutes, and purified RNA was precipitated in 2.5 volumes of isopropanol overnight. RNA quantification was performed using the Qubit 4.0 fluorometer following the Qubit RNA broad range assay kit (Invitrogen). 500 ng of RNA was used for reverse transcription with the iScript reverse transcription supermix (Biorad) following the manufacturer's protocol. PCR was then used to 550 amplify the region of interest in the m6A sensor mRNA sequence (see Table 2). PCR products were column purified before Sanger sequencing using QiaQuick spin columns (Qiagen) and 10 μL reactions were submitted for standard amplicon sequencing (Azenta Life Sciences). C-to-U editing percentage was calculated using the EditR web server 59. To measure gene expression using gene-specific oligos, qPCR was performed using iTaq universal SYBR green Supermix (Biorad). 20 μL reactions were set up with 1 μL of cDNA for each sample, in 3 technical replicates. qPCR was performed on a Biorad CFX Duet real-time PCR instrument, and results were analyzed by normalizing threshold cycle of each target gene to 18S rRNA according to established methods60. At least 2 biological replicates were used for each sample, and results are plotted as mean relative fold expression comparing treatment to the control group as indicated. Error bars represent standard deviation, and statistical significance was calculated using a two-way t-test assuming unequal variance.


m6A detection using RT-qPCR. RNA was extracted and treated with DNase I as described above, and a relative quantification of m6A by RT-qPCR was adapted from28. 4 reverse transcription reactions were set up using 150 ng of RNA for each: 2 reactions using BstI polymerase (NEB), and 2 reactions using Superscript reverse transcriptase enzyme (Fisher Scientific). For each reverse transcriptase, one reaction included a primer adjacent (+) to the site being tested (reverse compliment reverse oligo immediately downstream of the site), and the other reaction included a primer that is non-adjacent (−) to the site. The BstI reaction consisted of 10 U BstI polymerase, 50 mM dNTPs, 500 nM oligos (adjacent (+) or non-adjacent (−)), in 1× ThermoPol Buffer. Reactions were incubated in a thermal cycler with the following cycling settings: 3 minutes at 25° C., 30 minutes at 50° C., and 3 minutes at 85° C. Superscript III (SSIII) reactions consisted of 200 U Superscript III, 1M DTT, 25 mM MgCl2+, 10 mM dNTPs, 500 nM oligos (adjacent or non-adjacent), 2 μL 10× FS Buffer, and water up to 20 μL. SSIII thermal cycler settings were set according to manufacturer's protocol. All 4 reactions were used as a template in a qPCR reaction, in 3 technical replicates, using primers that flank the m6A site being tested. Threshold cycle values were obtained and relative m6A was calculated using the formula: 2−(CT Bst(−)−Ct SSIII(−)/Ct Bst(+)−Ct SSIII(+)). At least 2 biological replicates were used for each sample. Error bars represent standard deviation, and statistical significance was calculated using a two-way t-test assuming unequal variance.


Flow cytometry. After 24 hours, the culture media was replaced with media containing 2 μM puromycin for an additional 72 hours to select against the non-infected cells. Cells were then cultured in puromycin-free media for 48 hours, followed by 589 transfection with the GEMS-EGFP system. After 24 hours, cells were dissociated by TrypLE (Gibco) treatment for 10 minutes at 37° C. and 5% CO2. Trypsinized cells were resuspended in 5 mL of growth media containing 1% FBS and passed through a 4 μm cell filter to further separate the culture into single cells. Flow cytometry analysis was performed on a Sony MA900 cell sorter. Cell suspensions were first sorted by size and forward scatter to gate on live cells (BSC-A vs. FSC-A) and to eliminate doublets (FSC-H vs. FSC-A). 2 lasers were used to sort EGFP-positive cells (laser excitation 488 nm) and DsRed-positive cells (laser excitation 561 nm). Cells were sorted in a 4-way channel and collected in 5 mL conical tubes containing 1 mL of 1×PBS. Thresholds for EGFP and DsRed negative fluorescence were pre-calibrated using non-transfected HEK293T cells. Collection stopped when 500,000 cells were collected in the target populations (DsRed+/EGFP− and DsRed+/EGFP+). Sorted cells were collected by centrifugation (6,000 rpm, 10 minutes, 4° C.), and genomic DNA was extracted and analyzed by sequencing using METTL3 locus-specific primers. Indels at the METTL3 locus were identified by aligning the obtained sequences with the genomic sequence of endogenous METTL3 (GRCh38; chr14:21503198-21503835). Results were reported as the percentage of cells containing METTL3 indels out of the total number of cells obtained in each sorted cell population.


For APO1-YTH vs. APO1-YTHmut analysis, HEK293T cells were co-transfected with EGFP-DHFR and either the APO1-YTH or APO1-YTHmut plasmid. Cells were collected 24 hours later and samples were prepared for flow cytometry analysis as described above. Samples were analyzed using a Sony MA900 cell sorter and 1 million cells were recorded to measure EGFP fluorescence (last excitation 488 nm). FCS files were analyzed using Floreada.io software and plotted on a density plot as the frequency of events vs. EGFP fluorescence for each sample.


Mass spectrometry analysis. Total RNA was extracted as described above, and mRNA was purified using two rounds of oligo(dT) purification with Dynabeads oligo-(dT) mRNA purification kit (Invitrogen), followed by 2 rounds of rRNA depletion using NEBNext rRNA Depletion Kit (V2.0, NEB), and an additional two rounds of oligo(dT) purification with Dynabeads oligo-(dT) mRNA purification kit (Invitrogen). All mRNA purification steps were performed following the manufacturer's instructions. Purified mRNA quality was checked using a Bioanalyzer high sensitivity RNA analysis 6000 pico kit (Agilent). For mass spectrometry analysis, 100-200 ng of purified mRNA was incubated with 2 U of Nuclease P1 (Sigma) with 2.5 mM ZnCl and 25 mM NaCl at 37° C. for 2 hours. mRNA samples were treated with 5 U of antarctic phosphatase (NEB) for 2 h at 37° C. Samples were then processed using the Xevo TQ-S mass spectrometry system. All nucleosides were quantified by retention time and ion mass transitions of 268.2 to 133.2 (A) and 282.2 to 150.1 (m6A). Data were plotted as a percentage of m6A relative to A. At least 2 biological replicates were performed for each sample.


Cell growth assays. Huh-7 and HepG2 cells were plated in 6-well culture plates and transiently transfected with the indicated plasmids. 12 hours after transfection, cells were dissociated using TrypLE (Gibco) treatment for 2 minutes at 37° C. and 5% CO2. Trypsinized cells were resuspended in 5 mL of cell type-specific growth media, and an aliquot was used to count the number of cells in the culture using a hemocytometer. 10,000 cells for each sample were plated in one well of a 6-well culture plate for a total of 6 wells per condition. A hemocytometer was used to count the number of cells in each well every 24 hours for 5 days. Counts were performed in 3 technical replicates, and the average number of cells was used to calculate the ratio as indicated in each experiment.


Migration assays. Huh-7 cells were plated in 6-well culture plates and transiently transfected with the indicated plasmids. 12 hours after transfection, cells were dissociated using TrypLE (Gibco) treatment for 2 minutes at 37° C. and 5% CO2. 500 cells from each sample were plated on the top of a 6.5 mm transwell membrane with 8 μm pores (Corning). Culture medium inside the transwell chamber was formulated without the addition of FBS, while the culture medium at the bottom of the well included 10% FBS as a chemoattractant. 24 hours after plating the cells in the transwells, transwells were washed twice with 1×PBS, and the non-migrated cells were cleared using a cotton swab on the top of the transwell membrane. The membrane was fixed with methanol for 30 minutes, then washed with 1×PBS. Membranes were then stained with 5% Crystal Violet (VWR) for 30 minutes, then washed 3 times with 1×PBS. Transwell membranes were placed on a microscope glass slide and imaged under a brightfield 20× objective. At least 4 images were obtained for each condition and representative images were selected.


Example 2: m6A Sensor Design and Validation

A system for sensing m6A in cellular mRNAs was envisioned. That system has three main features: 1) it is genetically encoded to enable m6A sensing in living cells, 2) it is versatile and capable of being used in a variety of cell and tissue types, and 3) it provides a simple readout compatible with high-throughput studies. To achieve these goals, a system was designed that uses a reporter mRNA which produces a fluorescent protein (EGFP) only when the mRNA is methylated. This simple system was referred to as GEMS (genetically encoded m6A sensor), and, therefore couples cellular fluorescence with m6A methylation.


To achieve m6A-dependent production of EGFP in the GEMS system, the DART-seq was used. DART-seq is a method that previously developed for m6A detection20. DART-seq identifies m6A residues in cells by using a fusion protein consisting of the YTH domain, which directly binds to m6A sites, tethered to the cytidine deaminase APOBEC1. When the APOBEC1-YTH fusion protein is expressed in cells, it binds to m6A and catalyzes C-to-U editing of nearby cytidine residues (FIG. 1). This property of the APOBEC1-YTH fusion protein (hereafter APO1-YTH) could be harnessed to develop a system in which m6A-dependent C-to-U editing produces a stop codon that alters the expression of EGFP and provides a readout for m6A.


The GEMS system contains two components: APO1-YTH and an m6A reporter mRNA (FIG. 1). The reporter mRNA contains the coding sequence for EGFP followed by an m6A “sensor sequence”, which contains two m6A consensus motifs (GAC) and two tandem convertible codons in-frame with EGFP. When unedited, these codons encode arginine and glutamine (CGA and CAG, respectively). However, C-to-U editing produces two stop codons (UGA and UAG). The surrounding m6A sensor sequence is modified from a similar sequence in the human ACTB mRNA 3′UTR which contains two methylated GAC sequences that have been reported in many different cell types3, 4, 20, 22-24. Downstream of the m6A sensor sequence and in-frame with EGFP is the coding sequence for a destabilization domain modified from the Escherichia coli dihydrofolate reductase gene (ecDHFR). This destabilization domain was previously engineered to induce rapid, proteasome-mediated degradation of proteins to which it is tethered25. Thus, when the EGFP-DHFR m6A reporter mRNA is introduced into cells together with APO1-YTH, if the reporter mRNA is not methylated, there will be no editing of the m6A sensor sequence by APO1-YTH and the full-length EGFP-DHFR protein will be translated. The result will be rapid degradation of EGFP-DHFR and no fluorescence (FIG. 1). However, if either of the GAC sequences within the m6A sensor sequence is methylated, APO1-YTH will bind to the m6A and deaminate one or both cytidine residues within the two convertible stop codons of the sensor sequence. The result will be translation of EGFP followed by translation termination before the ribosome encounters the DHFR sequence. The EGFP protein will not be degraded since it will not be fused to DHFR, resulting in EGFP fluorescence (FIG. 1). Thus, this system provides a simple fluorescent readout for the presence of m6A (no m6A=no EGFP fluorescence; m6A=EGFP fluorescence).


To determine whether the GEMS system can sense cellular mRNA methylation, the system was transfected into HEK293T cells and assessed cellular fluorescence 24 hours later. Cells expressing APO1-YTH together with the m6A reporter mRNA exhibit robust EGFP fluorescence, whereas cells only expressing the m6A reporter mRNA are dark (FIG. 2A). In addition, Sanger sequencing of the reporter mRNA indicates C-to-U editing of the convertible stop codon sequences only in cells expressing APO1-YTH (FIG. 2B). This is further confirmed by western blot, which shows that cells expressing the reporter mRNA together with APO1-YTH produce both EGFP and EGFP-DHFR, whereas cells lacking APO1-YTH only produce EGFP-DHFR (FIG. 2C).


As an additional control to demonstrate that EGFP production and cellular fluorescence are due to recognition of m6A by APO1-YTH, cells were transfected with the m6A reporter mRNA and APO1-YTHmut, a mutant version of the APO1-YTH fusion protein which lacks the full m6A binding region of the YTH domain and exhibits greatly reduced m6A-binding activity20. This resulted in loss of EGFP fluorescence and EGFP protein production as well as decreased editing of the m6A sensor sequence (FIGS. 2D-2G), indicating that EGFP fluorescence and m6A sensor sequence editing are due to m6A recognition by APO1-YTH. Consistent with this, when the cells expressing the GEMS system were subjected to flow cytometry and sorted cells based on EGFP fluorescence, methylation and C-to-U editing of the m6A sensor sequence were found to be increased in cells with higher EGFP fluorescence (FIGS. 2H-2I). Additionally, when the RAC motifs within the m6A sensor sequence were mutated to preclude methylation, GEMS activity was abolished (FIG. 2J), further demonstrating that GEMS activity depends on m6A modification of the reporter mRNA. FIG. 2K. Top schematic shows the m6A reporter mRNA with a portion of the m6A sensor sequence expanded. This sequence is based off of a sequence within the human ACTB 3′UTR (bottom schematic), which contains two m6A sites at positions A1216 and A1222 that have been shown to be methylated in several m6A mapping studies.


Both the EGFP and EGFP-DHFR protein products are detected in cells expressing the GEMS system (FIGS. 2C, 2E). This likely reflects the presence of both methylated and unmethylated copies of the reporter mRNA, which is consistent with the known sub-stoichiometric abundance of m6A in cellular mRNAs17, 19, 22, 26, 27. Therefore, the level of methylation in the m6A sensor sequence was analyzed to determine if it is comparable to that of endogenous mRNAs. Previously, the proportion of APO1-YTH-mediated C-to-U editing (% C2U) near m6A sites is correlated with m6A stoichiometry20, 26. The stoichiometry of m6A in cellular mRNAs is generally low, with most sites estimated to be at levels of 40% or less17, 19, 22, 26, 27 Therefore, if the m6A sensor is a faithful representation of cellular mRNA methylation, one may expect C-to-U editing levels of the m6A sensor sequence to be comparable to endogenous mRNA. Indeed, when comparing the editing rate of the m6A sensor sequence to the editing seen in the region of the endogenous ACTB 3′UTR on which the sensor sequence is modeled, highly similar values were observed (FIG. 2L). Additionally, an orthogonal, RT-qPCR-based method for m6A detection and quantification28 was used to target the m6A sensor sequence as well as endogenous ACTB. This validated that m6A consensus adenosines within the sensor sequence are methylated in cells at a similar level as the corresponding region in endogenous ACTB, whereas non-consensus adenosines are unmethylated (FIG. 2M).


Altogether, these data demonstrate that the GEMS system produces robust EGFP fluorescence that depends both on the m6A-binding ability of APO1-YTH and on methylation and C-to-U editing of the m6A sensor sequence. Furthermore, methylation of the GEMS reporter mRNA mirrors the m6A level seen in a similar region of the ACTB mRNA, indicating that GEMS is an accurate representation of endogenous cellular mRNA methylation.


Example 3: GEMS Responds to Changes in METTL3 Expression Levels

This example discusses that the m6A sensor is METTL3-dependent. The GEMS system was expressed in HEK293T cells that contain an auxin-inducible degradation tag at the endogenous METTL3 locus and which exhibit decreased levels of m6A in the presence of auxin (FIG. 3A). Substantially reduced EGFP fluorescence was observed in auxin-treated cells compared to DMSO-treated cells (FIG. 3B). This was accompanied by a reduced EGFP:EGFP-DHFR ratio as assessed by western blot and decreased C-to-U editing of the m6A sensor sequence (FIGS. 3C-3E). Importantly, it was confirmed that these effects are due to loss of m6A, as mass spectrometry- and RT-qPCR-based m6A quantification showed reduced methylation of cellular mRNAs and the m6A sensor sequence, respectively, in auxin-treated cells (FIG. 3F, FIG. 3A). Also, m6A reporter mRNA abundance was quantified in METTL3-depleted cells after 24 hours; similar levels of abundance was found as in cells without METTL3 depletion, indicating that methylation of the reporter mRNA does not alter its stability (FIG. 3G). Furthermore, loss of m6A does not alter total protein production from the reporter mRNA (FIG. 3H). Lastly, the premature termination codons introduced by C-to-U editing of the reporter mRNA do not trigger nonsense-mediated decay (NMD), as treatment of cells with cycloheximide to limit NMD has no effect on reporter mRNA levels (FIG. 3I). Consistent with this, the reporter mRNA lacks introns and is therefore not expected to be susceptible to exon junction complex-dependent NMD.


To determine whether the GEMS system can also detect elevated methylation caused by increased levels of METTL3, GEMS was introduced into HEK293T cells together with exogenous expression of METTL3. This led to increased EGFP fluorescence, a higher EGFP:EGFP-DHFR ratio, and increased C-to-U editing of the sensor sequence (FIGS. 3J-3M). Overexpression of METTL3 also led to an increase in methylation of the sensor sequence without affecting reporter mRNA stability (FIGS. 3N-3O)—as little as a 1.29-fold increase in reporter mRNA methylation was sufficient to produce a significant increase in EGFP (FIG. 3P). Thus, the GEMS system is sensitive to both increased and decreased cellular m6A methylation caused by changes in METTL3 expression.


Since GEMS uses EGFP fluorescence as a readout for m6A, factors that inhibit general transcription, translation, or fluorescent protein (FP) production could potentially lead to a false readout and limit the utility of GEMS for some applications. To address this, the GEMS system was modified to include DsRed under the control of a separate promoter to control for transcription and general FP production (FIG. 3Q). Then, tests were performed to see whether selective reduction of EGFP (m6A-coupled) compared to DsRed (m6A-uncoupled) fluorescent signal could be used to detect genetic disruption of METTL3.


HEK293T cells were infected with a Cas9-expressing lentivirus and sgRNAs targeting either METTL3 or the AAVS1 safe harbor gene locus followed by transfection with the GEMS system and flow cytometry to isolate cells based on red/green fluorescence. Cells were then subjected to targeted sequencing of the METTL3 locus to determine whether CRISPR-induced indels are enriched in DsRed+/EGFP− cells, which would be expected if selective reduction of EGFP fluorescence reflects METTL3 disruption. Indeed, METTL3 indels are substantially higher in DsRed+/EGFP− cells compared to DsRed+/EGFP+ cells (FIG. 3R). This is accompanied by a decrease in C-to-U editing of the m6A sensor sequence, with nearly undetectable editing in the DsRed+/EGFP− pool of cells (FIG. 3S). Consistent with this, METTL3 expression is significantly decreased in the DsRed+/EGFP− pool of cells (FIG. 3T). Altogether, these data show that GEMS can be used to detect genetic perturbation of METTL3 and demonstrate its sensitivity to both increased and decreased METTL3 expression through m6A-coupled FP production.


Example 4: GEMS Detects Differential Methylation Across Cell Types

This example discusses the utility of GEMS for sensing m6A across diverse cell types by expressing the system in a variety of mouse and human cell lines. For each cell type, EGFP protein production and fluorescence were observed, as well as editing of the m6A sensor sequence, indicating that the GEMS system is active (FIGS. 4A-4D, FIGS. 4E-4G). It was confirmed that this is due to m6A recognition, since cells were dark and had greatly reduced C-to-U editing of the sensor sequence when APO1-YTH was replaced with APO1-YTHmut (FIGS. 4E-4G). Thus, GEMS can be used in a variety of different cell types to detect m6A.


Interestingly, some cell types were observed to have higher levels of EGFP fluorescence and sensor sequence editing than others. This could reflect different levels of m6A and perhaps GEMS can be used to report differential mRNA methylation across distinct cell types. To test this, the system was expressed in three commonly used human cell lines (HEK293T, HeLa, and Huh-7) with DsRed as an internal control. It was found that EGFP fluorescence, EGFP:EGFP-DHFR ratio, and m6A sensor sequence editing are highly similar in HEK293T and HeLa cells but substantially reduced in Huh-7 cells (FIGS. 4A-4D). This is not caused by differences in expression of APO1-YTH, as similar levels were observed across the three cell types (FIG. 4B). Furthermore, quantification of sensor sequence methylation in each cell type revealed similar m6A levels in HEK293T and HeLa cells but reduced levels in Huh-7 cells (FIG. 4H), indicating that EGFP fluorescence can be used to detect cell type-dependent differences in methylation of the GEMS reporter mRNA.


GEMS uses a single mRNA to sense m6A. To validate that GEMS activity reflects mRNA methylation levels globally, mRNA was purified from HEK293T, HeLa, and Huh-7 cells and performed mass spectrometry to quantify m6A levels (FIG. 4J). Consistent with the relative GEMS activity across the three cell lines, mRNA from HEK293T and HeLa cells have similar levels of m6A, whereas the amount of m6A in Huh-7 cellular mRNA is reduced (FIG. 4E, FIG. 4I). Thus, these data demonstrate that the GEMS system can be used to sense differences in m6A methylation of mRNAs across different cell types.


Example 5: GEMS Detects Pharmacological Inhibition of METTL3

The m6A methyltransferase machinery has recently emerged as a promising therapeutic target for the potential treatment of cancer and other diseases29-32. However, efforts to identify METTL3 inhibitors have been hampered by the lack of methods that provide a simple readout for m6A methyltransferase activity in living cells on a scale that is compatible with HTS. Since GEMS couples m6A methylation with cellular fluorescence, it has potential utility as a HTS-compatible technology for determining the effects of drugs or small molecules on m6A levels in cells.


To explore whether the GEMS system can detect pharmacological inhibition of METTL3, HEK293T cells expressing GEMS were subjected to STM2457, a small molecule inhibitor of METTL329, and performed quantitative microscopy. A significant decrease in EGFP fluorescence was observed following STM2457 treatment (FIGS. 5A-5D), an effect that is exacerbated with increasing doses of STM2457 (FIGS. 5E-5I). This is accompanied by reduced C-to-U editing of the sensor sequence and reduced m6A in the sensor sequence (FIG. 5J, FIG. 5K).


The ability of the GEMS system to report m6A reduction depends in part on the half-life of EGFP: if cellular mRNA methylation decreases, this can potentially be difficult to detect due to the presence of pre-existing EGFP protein. It may be that an improved GEMS system could be developed by tagging EGFP with a destabilizing domain to reduce its half-life in cells. A PEST degradation sequence was therefore added to the EGFP coding sequence in the GEMS reporter mRNA; this modified system was tested for its ability to respond to METTL3 inhibition with STM2457. Indeed, the EGFP-PEST reporter enabled improved detection of m6A depletion compared to the original EGFP version (FIGS. 5L-5O). Altogether, these data show that the GEMS system effectively detects m6A depletion mediated by pharmacological inhibition of METTL3 and demonstrate that the use of more short-lived FPs in the GEMS system can improve the detection of m6A dynamics in cells. FIG. 5P is a cartoon depicting an example of an alternative FP that could be utilized in the GEMS system in place of EGFP. This schematic shows primary neurons that are infected with a lentivirus expressing a photoconvertible FP such as Dendra2, which emits green fluorescence that is converted to red fluorescence upon exposure to UV light. New Dendra2 protein can the subsequently be identified by green fluorescence.


Example 6: m6A-Coupled CRISPRi Targeting

Because the APO1-YTH protein edits cellular methylated mRNAs in addition to the GEMS reporter mRNA, it could potentially lead to unwanted effects in cells. Therefore, an alternative approach was developed to target APO1-YTH specifically to the reporter mRNA and reduce editing of endogenous cellular transcripts. An additional application of the m6A-coupled effector protein delivery system is driving expression of CRISPR tools that target METTL3. This can provide an m6A-dependent feedback mechanism which reduces METTL3 expression when m6A levels become too high and could therefore serve as a way to maintain m6A homeostasis in cells. This can be tested by developing a system that expresses m6A-coupled CRISPRi tools to inhibit METTL3 transcription (FIG. 6). Embodiments employing dCas9 and dCas13 are described below, but other catalytically-inactive RNA-guided endonucleases may be employed.


dCas9


In the present embodiment, the GFP sequence of the m6A sensor system can be replaced with dCas9-KRAB, which is a fusion protein consisting of inactive Cas9 tethered to the Kruppel-associated box (KRAB) transcriptional repressor (Alerasool et al., An efficient KRAB domain for CRISPRi applications in human cells. Nat Methods. 2020; 17(11):1093-6). Then, a U6-METTL3 sgRNA cassette can be introduced into this plasmid. The result will be constitutive expression of the METTL3 sgRNA but only m6A-dependent dCas9-KRAB expression in the presence of doxycyclin, which induces APO1-YTH (FIG. 6). The efficacy of METTL3 sgRNA targeting with CRISPRi can be tested separately in HEK293T cells before choosing which sgRNA sequence to use.


Then, a lentivirus expressing this “m6A feedback system” can be packaged and infect HEK293T cells. RNA and protein can be isolated at various timepoints over the course of 72 hours (this can be expanded to longer times as needed). Sensor sequence methylation can be measured using SELECT. Sensor sequence editing can be evaluated with Sanger sequencing. Western blot can be used to assess METTL3, APO1-YTH, and dCas9-KRAB/dCas9-KRAB-DHFR protein levels. Global m6A levels in cellular mRNA can also be measured using UPLC-MS/MS. Collectively, these readouts can provide important quantitative metrics of how the m6A feedback system responds to gain/loss of m6A and how effective the feedback system is at maintaining m6A levels as the cell cycles between high and low levels of METTL3. As an additional approach, the cycling of m6A levels can be assessed using the m6A sensor system. Cells infected with the m6A feedback system can be transfected with the GFP-encoding m6A reporter mRNA. Live-cell imaging will be used to monitor GFP fluorescence over the course of 72 hours (or longer, as needed).


To determine whether a new protein of interest can be produced in place of GFP in the m6A sensor system, dCas9-KRAB was cloned in place of GFP. Robust dCas9-KRAB expression was detected using western blot (FIG. 7), demonstrating that other polypeptides can be expressed from the m6A reporter mRNA in place of GFP.


The dCas9-KRAB effector protein delivery system can be expressed in METTL3 degron cells to show that auxin treatment (which leads to METTL3 degradation) reduces dCas9-KRAB expression. The system can also be used to show that STM2457, a METTL3 inhibitor, reduces dCas9-KRAB expression when the system is expressed in wildtype cells. Finally, experiments can be done to show that METTL3 overexpression increases dCas9-KRAB expression. [0218] gRNAs that target other genes of interest (e.g., oncogenes) can be evaluated using this system. It is understood that any gene in the genome of a cell can be targeted by the dCas9-KRAB effector protein, as long as one or more gRNA guide the dCas9-KRAB to the gene of interest.


Any polypeptide can be expressed in a cell by using the m6A-coupled effector protein expression system. Other proteins of interest include, but are not limited to SOCS2 and other tumor suppressors, which can be expressed in cancer cells to determine if expression of a tumor suppressor can reduce cancer cell proliferation, migration, and colony formation.


dCas13


Previous studies have shown that dCas13 can be tethered to the m6A methyltransferase machinery and coupled with guide RNA (gRNA)-mediated targeting to achieve methylation of cellular mRNAs of interest33. A similar approach of fusing dCas13 to APO1-YTH might enable targeted m6A recognition and C-to-U editing of the sensor sequence in the GEMS reporter mRNA. Thus, APO1-YTH in the GEMS system was replaced with dCas13-APO1-YTH and co-expressed this in cells together with a gRNA targeting the m6A sensor sequence (FIG. 8A). This led to EGFP expression and m6A sensor sequence editing which was strongest for gRNAs binding closest to the sensor sequence adenosines (FIGS. 8B-8D). In contrast, cells co-expressing a non-targeting gRNA were dark and had no sensor sequence editing despite unchanged sensor sequence methylation (FIGS. 8B-8E). Additionally, treatment with STM2457 abolished GEMS activity, indicating that dCas13-APO1-YTH retains its requirement for recognizing m6A-modification of the reporter mRNA (FIG. 8F). Importantly, replacing APO1-YTH with dCas13-tethered APO1-YTH did not cause C-to-U editing of methylated cellular mRNAs (FIGS. 8C1-8C2). Altogether, these data show that dCas13-fused APO1-YTH can be used in the GEMS system together with a reporter mRNA-targeting gRNA to achieve m6A-dependent sensor sequence editing and GEMS activation while reducing editing of endogenous cellular mRNAs.


Example 7: m6A-Coupled Effector Protein Delivery Slows Cancer Cell Growth

The m6A-coupled payload delivery system can be used to influence cellular function. METTL3 expression is elevated in several cancers, and m6A hypermethylation has been shown to promote cancer cell proliferation and tumorigenesis4. Although pharmacological inhibition of METTL3 is a promising strategy for counteracting the effects of hypermethylation of transcripts associated with cancer progression, such approaches may have unwanted consequences because they also influence methylation of other RNAs in the cell. Since the GEMS system can couple protein expression with m6A methylation, it could replace EGFP with effector proteins of interest to overcome the oncogenic effects of mRNA hypermethylation in cancer cells.


A protein expression system described herein can be tested by infecting Huh-7 cells with a lentivirus expressing the system (or other means of introducing exogenous polynucleotides into a cell, for example, lipofection, nucleofection, or electroporation). The GFP-expressing sensor system can be used as a control, with both systems including APO1-YTH under an inducible promoter. Cells can be treated with doxycycline, and cell proliferation and colony formation will be tested over the course of 72 hours using established protocols (Chen et al.). METTL3 inhibition with STM2457 will be used in wildtype cells as a control to confirm METTL3-dependent effects on proliferation and colony formation.


The m6A feedback system can be tested using a similar approach with Huh-7 cells as well as MOLM-13 cells, an AML cell line with high levels of METTL3 that exhibits reduced proliferation and colony formation in response to STM2457. Thus, in both cell types, it is expected that expression of the m6A feedback system can lead to high levels of m6A sensor sequence methylation and editing. Effector protein or other therapeutic delivery can lead to METTL3 transcription inhibition, reduced cell proliferation and colony formation as the cell cycles from high to low m6A.


Huh-7 cells are a hepatocyte-derived carcinoma cell line frequently used to model hepatocellular carcinoma (HCC). Previous studies have shown that METTL3 and other methyltransferase complex components are upregulated in HCC and associated with increased disease severity and cancer progression38,39. One mechanism for this is through hypermethylation of the SOCS2 mRNA, which acts as a tumor suppressor in HCC40, 41 Elevated m6A methylation of SOCS2 promotes its degradation and reduces SOCS2 protein levels to accelerate cancer cell growth36, 42. This system was chosen because METTL3-induced hypermethylation of SOCS2 mRNA leads to m6A-dependent transcript degradation and a reduction in SOCS2 protein, which in turn promotes HCC cell proliferation, migration, and colony formation (Chen et al. RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2,” Hepatology. 2018; 67(6):2254-70.) One could use the GEMS system to couple cellular m6A levels with SOCS2 protein expression and effectively rescue the loss of SOCS2 protein that is caused by hypermethylation of the SOCS2 mRNA (FIGS. 9A-9B).


To test this, the EGFP sequence in the GEMS reporter mRNA was replaced with the coding sequence for SOCS2. Expression of GEMS-SOCS2 in Huh-7 cells led to robust expression of SOCS2 protein and m6A sensor sequence editing (FIGS. 9C-9F). Importantly, expression of the GEMS-SOCS2 system in Huh-7 cells led to similar levels of sensor sequence editing and stop codon usage as GEMS-EGFP, indicating that the EGFP coding sequence can be effectively replaced with other sequences without compromising the ability of the system to sense m6A (FIGS. 9F-9G).


To confirm activity of the GEMS-delivered SOCS2 protein, the JAK-STAT signaling pathway, which is inhibited by SOCS family proteins43-45, was examined. Reduced levels of phosphorylated JAK2 and STAT5 were observed; both JAK2 and STAT5 are known targets of SOCS2-mediated inhibition 45 (FIG. 9H). Additionally, significant downregulation of IGF1 and CyclinD1 genes was observed in GEMS-SOCS2-expressing cells, consistent with the role of SOCS2 in negatively regulating IGF1 and CyclinD1 transcription46-48 (FIG. 9I).


Previous studies have shown that m6A-mediated SOCS2 depletion promotes cancer cell proliferation and migration40, 41 To determine whether GEMS-SOCS2 expression can reverse these effects, Huh-7 cells were transfected with GEMS-SOCS2 and measured cell growth over the course of 5 days. Expression of GEMS-SOCS2 significantly reduced Huh-7 cell growth, indicating that SOCS2 delivery with the GEMS system can counteract the effects of m6A hypermethylation on cancer cell proliferation (FIG. 9J). Additionally, Huh-7 cells transfected with GEMS-SOCS2 exhibit reduced migration capacity compared to cells expressing GEMS-EGFP (FIG. 9K). Thus, m6A-coupled delivery of SOCS2 overcomes the effects of SOCS2 hypermethylation and slows cancer cell growth and migration.


In other embodiments, The SOCS2 coding sequence can be cloned in place of GFP in the m6A sensor system described above using a lentiviral backbone. Huh-7 cells can then be infected with the system. APO1-YTH expression can be induced with doxycycline treatment. RNA and protein will be collected at various timepoints over the course of 72 hours. Sensor sequence methylation and editing can be measured with SELECT and Sanger sequencing, respectively, and SOCS2/SOCS2-DIFR and APO1-YTH levels can be assessed by Western blot. These studies can establish the timing and amount of SOCS2 protein expression that can be achieved by the system. To confirm that SOCS2 expression is m6A-dependent, the experiments can be repeated in cells treated with STM2457 to inhibit METTL3. Expression of the GFP-encoding m6A sensor system can be used in parallel as a control.


Next, the GEMS system was analyzed as a possible general strategy to deliver tumor suppressors to inhibit cancer cell growth. The tumor suppressor protein p53 regulates transcriptional programs involved in cell cycle arrest, apoptosis, and DNA repair and plays a critical role in the prevention of cancer progression49, 50. Consistent with this, the TP53 gene is mutated in nearly half of human cancers51, 52 Huh-7 cells express mutated p53 (Y220C) which is stable but has impaired DNA binding and transcriptional activity53, 54. Therefore, the wild type TP53 coding sequence was cloned into the GEMS system and introduced it into Huh-7 cells. This led to robust p53 expression and upregulation of downstream p53 transcription targets, including CDKN1A and GADD45A (FIGS. 9L-90). Expression of GEMS-p53 also slowed Huh-7 cell growth and migration (FIGS. 9P-9Q). Additionally, when the effect of GEMS-p53 expression on the growth of HepG2 cells (which do not harbor TP53 mutations55), was tested, it was found that the growth reduction was specific to Huh-7 cells (FIG. 9R). Overall, these data demonstrate that the GEMS system can be used to couple m6A methylation with the expression of tumor suppressor proteins to slow cancer cell growth and migration.


A major advantage of the GEMS platform is that it enables protein output to be tuned to m6A levels. Indeed, the amounts of SOCS2 and p53 protein delivered by the GEMS system were compared in HepG2 and Huh-7 cells—elevated levels of both proteins were found in HepG2 cells, which have higher m6A41 (FIGS. 10A-10D). In theory, the system can be programmed to deliver any effector protein of interest in an m6A-dependent manner, making it an attractive strategy for tuning gene expression in a highly targeted manner in response to mRNA methylation levels.


Discussion

Disclosed herein is a genetically encoded m6A sensor system which provides a fluorescent readout in cells when m6A is deposited on mRNA. This disclosure offers a simple, low-cost method for cellular m6A sensing which can be implemented in virtually any cell or tissue type and easily carried out by a standard molecular biology lab. The ability of GEMS to sense changes in m6A methylation in living cells makes it an attractive system for monitoring m6A dynamics in a variety of cell types and conditions. Indeed, as disclosed herein, GEMS can be used as a readout for m6A in a variety of mouse and human cell lines and that relative differences in EGFP reporter fluorescence can be used to identify differences in methylation levels between cell types. GEMS may also have wide utility for studies of m6A dynamics in cells. Since the sensitivity of GEMS for reporting changes in mRNA methylation depends in part on the half-life of EGFP, using a reporter protein with a short half-life will improve the sensitivity of GEMS for sensing dynamic regulation of m6A. Consistent with this, as discussed above, adding a PEST sequence to EGFP substantially reduces sensor protein longevity and improves the ability to detect changes in m6A caused by pharmacological inhibition of METTL3. Depending on the application, photoconvertible proteins or other reporter proteins could also be substituted for EGFP to further improve detection of m6A dynamics.


Also, GEMS may be utilized for in vivo monitoring of m6A. This could be achieved either through the generation of transgenic animals expressing the two main components of the GEMS system or by introducing GEMS into a desired tissue of interest using viral-mediated or other delivery methods. Such studies might be useful for monitoring the in vivo effects of m6A methylation inhibitors, for examining how certain conditions or stresses alter m6A, or for understanding tissue-specific differences in methylation.


Due to its simple design and ability to sense m6A in living cells, the GEMS system may be useful for a variety of HTS-based approaches. For instance, the factors that control m6A methylation in cells are still not completely understood, so GEMS may be useful for global knockout screens designed to identify cellular proteins that influence m6A. Additionally, GEMS will be highly enabling for drug discovery efforts, as it provides a simple method for screening drug or small molecule libraries to identify novel inhibitors of METTL3. Other methyltransferase complex proteins such as METTL14 and WTAP have also been implicated in human disease and are upregulated in several cancers56, 57 so such screens have the potential to uncover inhibitors of these proteins as well.


Although GEMS opens up several new avenues for both low- and high-throughput studies of m6A, there are some important considerations when using the system. For instance, factors that influence proteasomal degradation could impact EGFP-DHFR stability and therefore cellular fluorescence. Additionally, changes in transcription or translation rates could influence FP production, although the use of m6A-uncoupled internal reporters such as DsRed can help mitigate this. Lastly, since GEMS requires APO1-YTH expression, factors that influence the fusion protein's activity or m6A recognition could impact the system. APO1-YTH also edits cellular methylated RNAs in addition to the GEMS reporter mRNA, which could influence other processes in the cell. Importantly, as discussed above, tethering APO1-YTH to dCas13 enables targeted editing of the GEMS reporter mRNA and reduces editing of cellular mRNAs. Additional refining of the GEMS system based on this approach may further improve its functionality by limiting unwanted effects of APO1-YTH-mediated editing of endogenous methylated RNAs.


In addition to its utility as an m6A-coupled fluorescent reporter, the GEMS system can be programmed to deliver protein payloads of interest in an m6A-dependent manner. As discussed above, GEMS may be used to express SOCS2 and p53 in liver cancer cells, leading to slowed cell growth and reduced migration capacity. Thus, the GEMS system can be used both to rescue the expression of proteins whose production is decreased by mRNA hypermethylation, as in the case of SOCS2, or as a general strategy for tumor suppressor protein expression in cells with elevated m6A, as with p53. In theory, any protein of interest can be expressed using the GEMS system, opening up numerous possibilities for m6A-coupled effector protein expression as a means of achieving desired cellular outcomes or counteracting the effects of high or low levels of m6A. For instance, GEMS could be used to deliver CRISPR/Cas9 tools targeting METTL3 itself, which could be used to activate or inhibit METTL3 expression in response to changing levels of m6A and therefore maintain m6A homeostasis. Given the numerous associations between m6A dysregulation and human disease, the GEMS system has potential utility as a novel therapeutic strategy.


Example 8: System-Expressing Cells from Transgenic/Knock-In Animals


FIG. 11 shows an embodiment of host cells as described herein and methods of use. Briefly, primary neurons can be isolated from transgenic mice expressing the APOBEC1-YTH enzyme. The cells can be cultured and then the m6A reporter mRNA could be introduced with viral infection or other means to examine m6A dynamics. Alternatively, mice that express the GEMS system can be created, and neurons (or other cells) isolated and cultured utilizing methods known in the art.


Example 9: GEMS Utilization for HTS Applications


FIG. 12: the GEMS system is compatible with HTS, so it could be used for HTS studies such as those seeking to identify cellular proteins/pathways that control m6A abundance or drugs/small molecules that inhibit METTL3 or m6A demethylases. As shown in FIG. 12, the expression system could be introduced to cell of choice, plated, and cultured m6A abundance or drugs/small molecules that inhibit METTL3 or m6A demethylases could then be studied using reporters, such as fluorescent reporters and measured using plate readers as known in the art. Below is a list of references relevant to this disclosure.

  • 1. Zaccara, S., Ries, R. J., and Jaffrey, S. R. (2019). Reading, writing and erasing mRNA methylation. Nat. Rev. Mol. Cell Biol. 20. 10.1038/s41580-019-0168-5.
  • 2. Shi, H., Wei, J., and He, C. (2019). Where, When, and How: Context-Dependent Functions of RNA Methylation Writers, Readers, and Erasers. Mol. Cell 74. 10.1016/j.molcel.2019.04.025.
  • 3. Meyer, K. D., Saletore, Y., Zumbo, P., Elemento, O., Mason, C. E., and Jaffrey, S. R. (2012). Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell. 10.1016/j.cell.2012.05.003.
  • 4. Dominissini, D., Moshitch-Moshkovitz, S., Schwartz, S., Salmon-Divon, M., Ungar, L., Osenberg, S., Cesarkas, K., Jacob-Hirsch, J., Amariglio, N., Kupiec, M., et al. (2012). Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 10.1038/naturel 1112.
  • 5. Flamand, M. N., and Meyer, K. D. (2019). The epitranscriptome and synaptic plasticity. Curr. Opin. Neurobiol. 10.1016/j.conb.2019.04.007.
  • 6. Roundtree, I. A., Evans, M. E., Pan, T., and He, C. (2017). Dynamic RNA Modifications in Gene Expression Regulation. Cell 169. 10.1016/j.cell.2017.05.045.
  • 7. Meyer, K. D., and Jaffrey, S. R. (2017). Rethinking m6A readers, writers, and erasers. Annu. Rev. Cell Dev. Biol. 33. 10.1146/annurev-cellbio-100616-060758.
  • 8. Meyer, K. D., Patil, D. P., Zhou, J., Zinoviev, A., Skabkin, M. A., Elemento, O., Pestova, T. V., Qian, S. B., and Jaffrey, S. R. (2015). 5′ UTR m6A Promotes Cap-Independent Translation. Cell. 10.1016/j.cell.2015.10.012.
  • 9. Ries, R. J., Zaccara, S., Klein, P., Olarerin-George, A., Namkoong, S., Pickering, B. F., Patil, D. P., Kwak, H., Lee, J. H., and Jaffrey, S. R. (2019). m6A enhances the phase separation potential of mRNA. Nature 571. 10.1038/s41586-019-1374-1.
  • 10. Zhou, J., Wan, J., Shu, X. E., Mao, Y., Liu, X. M., Yuan, X., Zhang, X., Hess, M. E., Brüning, J. C., and Qian, S. B. (2018). N6-Methyladenosine Guides mRNA Alternative Translation during Integrated Stress Response. Mol. Cell 69. 10.1016/j.molcel.2018.01.019.
  • 11. Engel, M., Eggert, C., Kaplick, P. M., Eder, M., Röh, S., Tietze, L., Namendorf, C., Arloth, J., Weber, P., Rex-Haffner, M., et al. (2018). The Role of m6A/m-RNA Methylation in Stress Response Regulation. Neuron 99. 10.1016/j.neuron.2018.07.009.
  • 12. Widagdo, J., Zhao, Q. Y., Kempen, M. J., Tan, M. C., Ratnu, V. S., Wei, W., Leighton, L., Spadaro, P. A., Edson, J., Anggono, V., et al. (2016). Experience-dependent accumulation of N6-methyladenosine in the prefrontal cortex is associated with memory processes in mice. J. Neurosci. 36. 10.1523/JNEUROSCI.4053-15.2016.
  • 13. Zhang, Z., Wang, M., Xie, D., Huang, Z., Zhang, L., Yang, Y., Ma, D., Li, W., Zhou, Q., Yang, Y. G., et al. (2018). METTL3-mediated N 6-methyladenosine mRNA modification enhances long-term memory consolidation. Cell Res. 28. 10.1038/s41422-018-0092-9.
  • 14. Dai, D., Wang, H., Zhu, L., Jin, H., and Wang, X. (2018). N6-methyladenosine links RNA metabolism to cancer progression review-article. Cell Death Dis. 9. 10.1038/s41419-017-0129-x.
  • 15. Jiang, X., Liu, B., Nie, Z., Duan, L., Xiong, Q., Jin, Z., Yang, C., and Chen, Y. (2021). The role of m6A modification in the biological functions and diseases. Signal Transduct. Target. Ther. 6. 10.1038/s41392-020-00450-x.
  • 16. Williams, G. D., Gokhale, N. S., and Homer, S. M. (2019). Regulation of Viral Infection by the RNA Modification N6-Methyladenosine. Annu. Rev. Virol. 6. 10.1146/annurev-virology-092818-015559.
  • 17. Garcia-Campos, M. A., Edelheit, S., Toth, U., Safra, M., Shachar, R., Viukov, S., Winkler, R., Nir, R., Lasman, L., Brandis, A., et al. (2019). Deciphering the “m6A Code” via Antibody-Independent Quantitative Profiling. Cell 178. 10.1016/j.cell.2019.06.013.
  • 18. Zhang, Z., Chen, L. Q., Zhao, Y. L., Yang, C. G., Roundtree, I. A., Zhang, Z., Ren, J., Xie, W., He, C., and Luo, G. Z. (2019). Single-base mapping of m6A by an antibody-independent method. Sci. Adv. 5. 10.1126/sciadv.aax0250.
  • 19. Liu, C., Sun, H., Yi, Y., Shen, W., Li, K., Xiao, Y., Li, F., Li, Y., Hou, Y., and Lu, B. (2022). Absolute quantification of single-base m6A methylation in the mammalian transcriptome using GLORI. Nat. Biotechnol., 1-12.
  • 20. Meyer, K. D. (2019). DART-seq: an antibody-free method for global m6A detection. Nat. Methods. 10.1038/s41592-019-0570-0.
  • 21. Hu, L., Liu, S., Peng, Y., Ge, R., Su, R., Senevirathne, C., Harada, B. T., Dai, Q., Wei, J., Zhang, L., et al. (2022). m6A RNA modifications are measured at single-base resolution across the mammalian transcriptome. Nat. Biotechnol. 40. 10.1038/s41587-022-01243-z.
  • 22. Liu, N., Parisien, M., Dai, Q., Zheng, G., He, C., and Pan, T. (2013). Probing N6-methyladenosine RNA modification status at single nucleotide resolution in mRNA and long noncoding RNA. RNA 19. 10.1261/rna.041178.113.
  • 23. Linder, B., Grozhik, A. V., Olarerin-George, A. O., Meydan, C., Mason, C. E., and Jaffrey, S. R. (2015). Single-nucleotide-resolution mapping of m6A and m6Am throughout the transcriptome. Nat. Methods. 10.1038/nmeth.3453.
  • 24. Ke, S., Alemu, E. A., Mertens, C., Gantman, E. C., Fak, J. J., Mele, A., Haripa, B., Zucker-Scharff, I., Moore, M. J., Park, C. Y., et al. (2015). A majority of m6A residues are in the last exons, allowing the potential for 3′ UTR regulation. Genes Dev. 29.
  • 25. Iwamoto, M., Bjorklund, T., Lundberg, C., Kirik, D., and Wandless, T. J. (2010). A general chemical method to regulate protein stability in the mammalian central nervous system. Chem. Biol. 10.1016/j.chembiol.2010.07.009.
  • 26. Tegowski, M., Flamand, M. N., and Meyer, K. D. (2022). scDART-seq reveals distinct m6A signatures and mRNA methylation heterogeneity in single cells. Mol. Cell 82. 10.1016/j.molcel.2021.12.038.
  • 27. Xiao, Y.-L., Liu, S., Ge, R., Wu, Y., He, C., Chen, M., and Tang, W. (2023). Transcriptome-wide profiling and quantification of N6-methyladenosine by enzyme-assisted adenosine deamination. Nat. Biotechnol., 1-11.
  • 28. Castellanos-Rubio, A., Santin, I., Olazagoitia-Garmendia, A., Romero-Garmendia, I., Jauregi-Miguel, A., Legarda, M., and Bilbao, J. R. (2019). A novel RT-QPCR-based assay for the relative quantification of residue specific m6A RNA methylation. Sci. Rep. 9. 10.1038/s41598-019-40018-6.
  • 29. Yankova, E., Blackaby, W., Albertella, M., Rak, J., De Braekeleer, E., Tsagkogeorga, G., Pilka, E. S., Aspris, D., Leggate, D., Hendrick, A. G., et al. (2021). Small-molecule inhibition of METTL3 as a strategy against myeloid leukaemia. Nature. 10.1038/s41586-021-03536-w.
  • 30. Cui, Q., Shi, H., Ye, P., Li, L., Qu, Q., Sun, G., Sun, G., Lu, Z., Huang, Y., Yang, C. G., et al. (2017). m6A RNA Methylation Regulates the Self-Renewal and Tumorigenesis of Glioblastoma Stem Cells. Cell Rep. 18. 10.1016/j.celrep.2017.02.059.
  • 31. Tzelepis, K., De Braekeleer, E., Yankova, E., Rak, J., Aspris, D., Domingues, A. F., Fosbeary, R., Hendrick, A., Leggate, D., Ofir-Rosenfeld, Y., et al. (2019). Pharmacological Inhibition of the RNA m6a Writer METTL3 As a Novel Therapeutic Strategy for Acute Myeloid Leukemia. Blood 134. 10.1182/blood-2019-127962.
  • 32. Huang, H., Weng, H., and Chen, J. (2020). m6A Modification in Coding and Non-coding RNAs: Roles and Therapeutic Implications in Cancer. Cancer Cell 37. 10.1016/j.ccell.2020.02.004.
  • 33. Wilson, C., Chen, P. J., Miao, Z., and Liu, D. R. (2020). Programmable m6A modification of cellular RNAs with a Cas13-directed methyltransferase. Nat. Biotechnol. 10.1038/s41587-020-0572-6.
  • 34. Lan, Q., Liu, P. Y., Haase, J., Bell, J. L., Hüttelmaier, S., and Liu, T. (2019). The critical role of RNA m6A methylation in cancer. Cancer Res. 79, 1285-1292.
  • 35. Liu, J., Eckert, M. A., Harada, B. T., Liu, S.-M., Lu, Z., Yu, K., Tienda, S. M., Chryplewicz, A., Zhu, A. C., and Yang, Y. (2018). m6A mRNA methylation regulates AKT activity to promote the proliferation and tumorigenicity of endometrial cancer. Nat. Cell Biol. 20, 1074-1083.
  • 36. Xu, J., Chen, Q., Tian, K., Liang, R., Chen, T., Gong, A., Mathy, N. W., Yu, T., and Chen, X. (2020). m6A methyltransferase METTL3 maintains colon cancer tumorigenicity by suppressing SOCS2 to promote cell proliferation. Oncol. Rep. 44, 973-986.
  • 37. Pinello, N., Sun, S., and Wong, J. J.-L. (2018). Aberrant expression of enzymes regulating m6A mRNA methylation: implication in cancer. Cancer Biol. Med. 15, 323.
  • 38. Lin, Y., Wei, X., Jian, Z., and Zhang, X. (2020). METTL3 expression is associated with glycolysis metabolism and sensitivity to glycolytic stress in hepatocellular carcinoma. Cancer Med. 9. 10.1002/cam4.2918.
  • 39. Liu, X., Qin, J., Gao, T., Li, C., Chen, X., Zeng, K., Xu, M., He, B., Pan, B., Xu, X., et al. (2020). Analysis of METTL3 and METTL14 in hepatocellular carcinoma. Aging (Albany. NY). 12. 10.18632/aging.103959.
  • 40. Liu, J., Liu, Z., Li, W., and Zhang, S. (2021). SOCS2 is a potential prognostic marker that suppresses the viability of hepatocellular carcinoma cells. Oncol. Lett. 21. 10.3892/OL.2021.12660.
  • 41. Cui, M., Sun, J., Hou, J., Fang, T., Wang, X., Ge, C., Zhao, F., Chen, T., Xie, H., Cui, Y., et al. (2016). The suppressor of cytokine signaling 2 (SOCS2) inhibits tumor metastasis in hepatocellular carcinoma. Tumor Biol. 37. 10.1007/s13277-016-5215-7.
  • 42. Chen, M., Wei, L., Law, C. T., Tsang, F. H. C., Shen, J., Cheng, C. L. H., Tsang, L. H., Ho, D. W. H., Chiu, D. K. C., Lee, J. M. F., et al. (2018). RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology 67. 10.1002/hep.29683.
  • 43. O'Shea, J. J., and Murray, P. J. (2008). Cytokine Signaling Modules in Inflammatory Responses. Immunity 28. 10.1016/j.immuni.2008.03.002.
  • 44. O'Sullivan, L. A., Liongue, C., Lewis, R. S., Stephenson, S. E. M., and Ward, A. C. (2007). Cytokine receptor signaling through the Jak-Stat-Socs pathway in disease. Mol. Immunol. 801 44. 10.1016/j.molimm.2006.11.025.
  • 45. Durham, G. A., Williams, J. J. L., Nasim, M. T., and Palmer, T. M. (2019). Targeting SOCS Proteins to Control JAK-STAT Signalling in Disease. Trends Pharmacol. Sci. 40. 10.1016/j.tips.2019.03.001.
  • 46. Zhou, Y., Zhang, Z., Wang, N., Chen, J., Zhang, X., Guo, M., John Zhong, L., and Wang, Q. (2018). Suppressor of cytokine signalling-2 limits IGF1R-mediated regulation of epithelial-mesenchymal transition in lung adenocarcinoma article. Cell Death Dis. 9. 10.1038/s41419-018-0457-5.
  • 47. Dobie, R., MacRae, V. E., Huesa, C., van′t Hof, R., Ahmed, S. F., and Farquharson, C. (2014). Direct stimulation of bone mass by increased GH signalling in the osteoblasts of Socs2−/− mice. J. Endocrinol. 223. 10.1530/JOE-14-0292.
  • 48. Zhang, Y., Ma, Y., Wu, G., Xie, M., Luo, C., Huang, X., Tian, F., Chen, J., and Li, X. (2021). SENP1 promotes MCL pathogenesis through regulating JAK-STAT5 pathway and SOCS2 expression. Cell Death Discov. 7. 10.1038/s41420-021-00578-x.
  • 49. Hu, J., Cao, J., Topatana, W., Juengpanich, S., Li, S., Zhang, B., Shen, J., Cai, L., Cai, X., and Chen, M. (2021). Targeting mutant p53 for cancer therapy: direct and indirect strategies. J. Hematol. Oncol. 14. 10.1186/s13045-021-01169-0.
  • 50. Zhang, C., Liu, J., Xu, D., Zhang, T., Hu, W., and Feng, Z. (2020). Gain-of-function mutant p53 in cancer progression and therapy. J. Mol. Cell Biol. 12. 10.1093/jmcb/mjaa040.
  • 51. Ozaki, T., and Nakagawara, A. (2011). Role of p53 in cell death and human cancers. Cancers (Basel). 3, 994-1013.
  • 52. Hollstein, M., Sidransky, D., Vogelstein, B., and Harris, C. C. (1991). p53 mutations in human cancers. Science (80-.). 253, 49-53.
  • 53. Bauer, M. R., Jones, R. N., Tareque, R. K., Springett, B., Dingler, F. A., Verduci, L., Patel, K. J., Fersht, A. R., Joerger, A. C., and Spencer, J. (2019). A structure-guided molecular chaperone approach for restoring the transcriptional activity of the p53 cancer mutant Y220C. Future Med. Chem. 11, 2491-2504.
  • 54. Liu, X., Wilcken, R., Joerger, A. C., Chuckowree, I. S., Amin, J., Spencer, J., and Fersht, A. R. (2013). Small molecule induced reactivation of mutant p53 in cancer cells. Nucleic Acids Res. 41, 6034-6044.
  • 55. Courtois, S., Verhaegh, G., North, S., Luciani, M.-G., Lassus, P., Hibner, U., Oren, M., and Hainaut, P. (2002). ΔN-p53, a natural isoform of p53 lacking the first transactivation domain, counteracts growth suppression by wild-type p53. Oncogene 21, 6722-6728. 10.1038/sj.onc.1205874.
  • 56. Chen, X. Y., Zhang, J., and Zhu, J. S. (2019). The role of m6A RNA methylation in human cancer. Mol. Cancer 18. 10.1186/s12943-019-1033-z.
  • 57. Guan, Q., Lin, H., Miao, L., Guo, H., Chen, Y., Zhuo, Z., and He, J. (2022). Functions, mechanisms, and therapeutic implications of METTL14 in human cancer. J. Hematol. Oncol. 15. 10.1186/s13045-022-01231-5.
  • 58. Abràmoff, M. D., Magalhães, P. J., and Ram, S. J. (2004). Image processing with imageJ. Biophotonics Int. 11. 10.1201/9781420005615.ax4.
  • 59. Kluesner, M. G., Nedveck, D. A., Lahr, W. S., Garbe, J. R., Abrahante, J. E., Webber, B. R., and Moriarity, B. S. (2018). EditR: a method to quantify base editing from Sanger sequencing. Cris. J. 1, 239-250.
  • 60. Kuchipudi, S. V., Tellabati, M., Nelli, R. K., White, G. A., Perez, B. B., Sebastian, S., Slomka, M. J., Brookes, S. M., Brown, I. H., Dunham, S. P., et al. (2012). 18S rRNA is a reliable normalisation gene for real time PCR based on influenza virus infected cells. Virol. J. 9. 10.1186/1743-422X-9-230.


Additional aspects of sequences relating to polynucleotides and polypeptides as described herein and in Table 3 below can be found, for example, in PCT/US2022/079709, U.S. Pat. No. 11,680,109, and Meyer, K. D., “DART-seq: an antibody-free method for global m(6)A detection,” Nat Methods. 2019 December, 16(12):1275-1280 (published online Sep. 23, 2019); doi: 10.1038/s41592-019-0570-0, the entire contents of all of which (including sequence information and any supplemental information) are incorporated by reference in their entirety as fully set forth herein.









TABLE 3







INFORMAL SEQUENCE LISTING











Oligonucleotide,




SEQ
polypeptide,




ID
plasmid, or

Description and


NO
sequence name
Sequence (5′ to 3′)
Notes













1
FM36_CMV5′UpseqFW
GAAGAATCTGCTTAGGGTTAGGCG
sequencing oligo at





CMV promoter





upstream of eGFP





2
FM37_DHFR3′seqFW
GAATTCCACGATGCTGATG
sequencing oligo at





DHFR domain





downstream of





EGFP





3
FM38_APOBEC5′seqFW
CGAGCCCCATGAGTTTGAGG
sequencing oligo at





5′ end of APO1





4
FM39_APOBEC3′seqFW
CCACAGCTGACATTCTTTACC
sequencing oligo at





3′ end of APO1





5
FM40_YTH3′seqFW
CTACAAGCACACCACTTCC
sequencing oligo at





3′ end of YTH





domain





downstream of





APO1





6
FM65_TLCV2tempR2
CAACTTACTTCTGACAACGATCGGAGG
sequencing oligo in




ACCGA
the middle of AMP





resistance cassette





7
FM66_TLCV2tempF1
TAGCTCCTTCGGTCCTCCGATCGTTGT
sequencing oligo in




CAGAA
the middle of AMP





resistance cassette





8
FM87_ODCmid1
AGAATAGACCGAGATAGGGTTGAGTG
sequencing oligo in





the middle of F1





origin of





replication cassette





9
FM88_ODCmid2
CACTCAACCCTATCTCGGTCTATTCT
sequencing oligo in





the middle of F1





origin of





replication cassette





10
FM149_dsREDN
GTACTGGAACTGGGGGGACAG
sequencing oligo at





5′ end of dsRED-





Express2





11
FM150_hPGK_F
GTGTTCCGCATTCTGCAAG
sequencing oligo in





hPGK promoter





upstream of





dsRED-Express2





12
FM255_SOCS2ampFW
CCTCTAGAGCCGCCATGACC
sanger GEMS-





SOCS2 (use with





FM2-actm6aRV)





13
FM256_P53ampFW
CCCTCTAGAGCCGCCATG
sanger GEMS-p53





(use with FM2-





actm6aRV)





14
FM1-actm6aFW
CAAGATCCGCCACAATATCGAGG
sanger GEMS-





EGFP (use with





FM2-actm6aRV)





15
FM2-actm6aRV
GTACTCGAGAGAATGCACAATGAGGCT
Used for amplicon




TATC
generation for





sanger in GEMS-





EGFP/SOCS2/p53





16
ActinFwd
CAACACAGTGCTGTCTGGC
sanger ACTB





A1222; Tegowski





and Meyer (2022)





17
ActinRev
CAAGATGAGATTGGCATGGC
sanger ACTB





A1222; Tegowski





and Meyer (2022)





18
FM186_595FragFW
CCCTCTAGAGCCGCCATGACCCTGCGG
amplify SOCS2




TGC
from cDNA for





GEMS-SOCS2





cloning





19
FM187_595FragRV
CTGTCGTAAGTCCGCTACCTGGAATTT
amplify SOCS2




ATATTCTTCCAAGTAATCTTTTAGT
from cDNA for





GEMS-SOCS2





cloning





20
FM202_600FragFW
CCCTCTAGAGCCGCCATGGAGGAGCCG
amplify p53 from




C
cDNA for GEMS-





SOCS2 cloning





21
FM203_600FragRV
CTGTCGTAAGTCCGCGTCTGAGTCAGG
amplify p53 from




C
cDNA for GEMS-





SOCS2 cloning





22
FM59_repadj2RV
GAAAGGGTGTAACGCAACTG
m6A sensor





sequence adjacent





RAC site 2 for





SELECT





23
FM60_repadj1RV
GAAAGGGTGTAACGCAACTGTCG
m6A sensor





sequence adjacent





RAC site 1 for





SELECT





24
FM61_repnonadjRV
CTTCTCATTGGGATCTTTGCTC
nonadjacent for





both m6A sensor





sequence RAC





sites for SELECT





25
FM62_qPCRFW
CCACAATATCGAGGATGGC
qPCR for both





m6A sensor





sequence RAC





sites for SELECT





26
FM126_adjRep1625
CGAGAAAGGGTGTAACGCAAC
m6A sensor





sequence adjacent





non-RAC site for





SELECT





27
FM127_adjRep1620
GGGTGTAACGCAACTGTCG
m6A sensor





sequence adjacent





non-RAC site for





SELECT





28
ACTBqPCRFwd
CAGCAAGCAGGAGTATGACGAGTC
qPCR for ACTB





A1222 for





SELECT





29
ACTB_nonAdjRev
CATGCCAATCTCATCTTG
nonadjacent for





ACTB A1222 for





SELECT





30
ActinA1222_AdjRev
TTGTCAAGAAAGGGTGTAACGCAACTA
ACTB A1222




AG
sequence adjacent





RAC site for





SELECT;





Tegowski and





Meyer (2022)





(reference 26)





31
FM201_SOCS2FW
CTGTTAATGAAGCCAAAGAG
qPCR of SOCS2





expression





32
FM201_SOCS2RV
GCACATCTGAACATAGTAG
qPCR of SOCS2





expression





33
FM214_p53FW
CAGATGAAGCTCCCAGAATG
qPCR of p53





expression





34
FM215_p53RV
GTCCCAGAATGCAAGAAG
qPCR of p53





expression





35
FM233_CCND1FW
GCTGCGAAGTGGAAACCATC
qPCR of CyclinD1





expression





36
FM234_CCNDIRV
CCTCCTTCTGCACACATTTGAA
qPCR of CyclinD1





expression





37
FM239_IGF1FW
GCTCTTCAGTTCGTGTGTGGA
qPCR of IGF1





expression








38
FM240_IGF1RV
GCCTCCTTAGATCACAGCTCC
qPCR of IGF1





expression





39
FM259_P21FW
TGTCCGTCAGAACCCATGC
qPCR of p21





expression





40
FM260_P21RV
AAAGTCGAAGTTCCATCGCTC
qPCR of p21





expression





41
FM263_RRM2AFW
AGAGGCTCGCTGTTTCTATGG
qPCR of RRM2A





expression





42
FM263_RRM2ARV
GCAAGGCCCAATCTGCTTTTT
qPCR of RRM2A





expression





43
AK_22_M3F
tggatctgttgtgatatccgctacc
qPCR of METTL3





expression





44
AK_23_M3R
ctgcctgtgacccagaggaagag
qPCR of METTL3





expression





45
FM220_dsREDNfw
CTTCAAGGTGCACATGGAG
qPCR of dsRed-





Express2





expression (use





with





FM149_dsREDN)





46
HZ_SMUG1_F
TTGACACTACTCCTGTTTGCCC
sanger sequencing





of SMUG1 m6A





site





47
HZ_SMUG1_R
AAAAGTTCCAAGTTTCAAAGCTGGG
sanger sequencing





of SMUG1 m6A





site





48
HZ_HERC2_F
TGTCGACTCCTTTGCTTCGGA
sanger sequencing





of HERC2 m6A





site





49
HZ_HERC2_R
AAATCGATTGCACCCACAAGTA
sanger sequencing





of HERC2 m6A





site





50
HZ_NIPA1_F
AGCCGCACAGAAAGATCACC
sanger sequencing





of NIPA1 m6A site





51
HZ_NIPA1_R
AAGTGCAAAAATACTAAGGTGGG
sanger sequencing





of NIPA1 m6A site





52
dCas13_GEMS-gRNA1
AGCGCGATCACATGGTCCTGCTGGAGT
Guide RNA co-




TCG
transfected with





GEMS-dCas13





targeted system.





Targeting as





marked in





FIGS. 8A-8F: end





of EGFP





53
dCas13_GEMS-gRNA2
CCGCCGCATCTAACACATTGATCCTAG
Guide RNA co-




CAG
transfected with





GEMS-dCas13





targeted system.





54
dCas13_GEMS-gRNA3
GCGGACTTACGACAGTTGCGTTACACC
Guide RNA co-




CTT
transfected with





GEMS-dCas13





targeted system. r





55
dCas13_GEMS-gRNA4
TTGCGGCGTTAGCGGTAGATCACGTTA
Guide RNA co-




TCG
transfected with





GEMS-dCas13





targeted system.





Targeting as





marked in





FIGS. 8A-8F: start





of DHFR





56
pCMV-APOBEC1-YTH
atgagctcagagactggcccagtggct
APOBEC1-YTH-




gtggaccccacattgagacggcggatc
HA to clone in




gagccccatgagtttgaggtattcttc
GEMS:




gatccgagagagctccgcaaggagacc
Addgene #131636,




tgcctgctttacgaaattaattggggg
the sequence of




ggccggcactccatttggcgacataca
which is accessible




tcacagaacactaacaagcacgtcgaa
at addgene.org/




gtcaacttcatcgagaagttcacgaca
browse/sequence/




gaaagatatttctgtccgaacacaagg
260783/ and




tgcagcattacctggtttctcagctgg
incorporated




agcccatgcggcgaatgtagtagggcc
by reference




atcactgaattcctgtcaaggtatccc





cacgtcactctgtttatttacatcgca





aggctgtaccaccacgctgacccccgc





aatcgacaaggcctgcgggatttgatc





tcttcaggtgtgactatccaaattatg





actgagcaggagtcaggatactgctgg





agaaactttgtgaattatagcccgagt





aatgaagcccactggcctaggtatccc





catctgtgggtacgactgtacgttctt





gaactgtactgcatcatactgggcctg





cctccttgtctcaacattctgagaagg





aagcagccacagctgacattctttacc





atcgctcttcagtcttgtcattaccag





cgactgcccccacacattctctgggcc





accgggttgaaaagcggcagcgagact





cccgggacctcagagtccgccacacca





gaaccccacccagtgttggagaagctt





cggtccattaataactataaccccaaa





gattttgactggaatctgaaacatggc





cgggttttcatcattaagagctactct





gaggacgatattcaccgttccattaag





tataatatttggtgcagcacagagcat





ggtaacaagagactggatgctgcttat





cgttccatgaacgggaaaggccccgtt





tacttacttttcagtgtcaacggcagt





ggacacttctgtggcgtggcagaaatg





aaatctgctgtggactacaacacatgt





gcaggtgtgtggtcccaggacaaatgg





aagggtcgttttgatgtcaggtggatt





tttgtgaaggacgttcccaatagccaa





ctgcgacacattcgcctagagaacaac





gagaataaaccagtgaccaactctagg





gacactcaggaagtgcctctggaaaag





gctaagcaggtgttgaaaattatagcc





agctacaagcacaccacttccattttt





gatgacttctcacactatgagaaacgc





caagaggaagaagaaagtgttaaaaag





gaacgtcaaggtcgtgggaaactcgag






57
pCMV-APOBEC1-YTHmut
ATGAGCTCAGAGACTGGCCCAGTGGCT
APOBEC1-




GTGGACCCCACATTGAGACGGCGGATC
YTHmutant-HA to




GAGCCCCATGAGTTTGAGGTATTCTTC
clone in




GATCCGAGAGAGCTCCGCAAGGAGACC
GEMSmut:




TGCCTGCTTTACGAAATTAATTGGGGG
Addgene #131637,




GGCCGGCACTCCATTTGGCGACATACA
the sequence of




TCACAGAACACTAACAAGCACGTCGAA
which is accessible




GTCAACTTCATCGAGAAGTTCACGACA
at addgene.org/




GAAAGATATTTCTGTCCGAACACAAGG
131637/ and




TGCAGCATTACCTGGTTTCTCAGCTGG
incorporated




AGCCCATGCGGCGAATGTAGTAGGGCC
by reference




ATCACTGAATTCCTGTCAAGGTATCCC





CACGTCACTCTGTTTATTTACATCGCA





AGGCTGTACCACCACGCTGACCCCCGC





AATCGACAAGGCCTGCGGGATTTGATC





TCTTCAGGTGTGACTATCCAAATTATG





ACTGAGCAGGAGTCAGGATACTGCTGG





AGAAACTTTGTGAATTATAGCCCGAGT





AATGAAGCCCACTGGCCTAGGTATCCC





CATCTGTGGGTACGACTGTACGTTCTT





GAACTGTACTGCATCATACTGGGCCTG





CCTCCTTGTCTCAACATTCTGAGAAGG





AAGCAGCCACAGCTGACATTCTTTACC





ATCGCTCTTCAGTCTTGTCATTACCAG





CGACTGCCCCCACACATTCTCTGGGCC





ACCGGGTTGAAAAGCGGCAGCGAGACT





CCCGGGACCTCAGAGTCCGCCACACCA





GAAGGCCGGGTTTTCATCATTAAGAGC





TACTCTGAGGACGATATTCACCGTTCC





ATTAAGTATAATATTTGGTGCAGCACA





GAGCATGGTAACAAGAGACTGGATGCT





GCTTATCGTTCCATGAACGGGAAAGGC





CCCGTTTACTTACTTTTCAGTGTCAAC





GGCAGTGGACACTTCTGTGGCGTGGCA





GAAATGAAATCTGCTGTGGACTACAAC





ACATGTGCAGGTGTGTGGTCCCAGGAC





AAATGGAAGGGTCGTTTTGATGTCAGG





TGGATTTTTGTGAAGGACGTTCCCAAT





AGCCAACTGCGACACATTCGCCTAGAG





AACAACGAGAATAAACCAGTGACCAAC





TCTAGGGACACTCAGGAAGTGCCTCTG





GAAAAGGCTAAGCAGGTGTTGAAAATT





ATAGCCAGCTACAAGCACACCACTTCC





ATTTTTGATGACTTCTCACACTATGAG





AAACGCCAAGAGGAAGAAGAAAGTGTT





AAAAAGGAACGTCAAGGTCGTGGGAAA





CTCGAG






58
iDuet101a
GTCGACGGATCGGGAGATCTCCCGATC
GEMS backbone




CCCTATGGTGCACTCTCAGTACAATCT
plasmid:




GCTCTGATGCCGCATAGTTAAGCCAGT
Addgene #17629,




ATCTGCTCCCTGCTTGTGTGTTGGAGG
the sequence of




TCGCTGAGTAGTGCGCGAGCAAAATTT
which is accessible




AAGCTACAACAAGGCAAGGCTTGACCG
at addgene.org/




ACAATTGCATGAAGAATCTGCTTAGGG
17629/ and




TTAGGCGTTTTGCGCTGCTTCGCGATG
incorporated




TACGGGCCAGATATACGCGTTGACATT
by reference




GATTATTGACTAGTTATTAATAGTAAT





CAATTACGGGGTCATTAGTTCATAGCC





CATATATGGAGTTCCGCGTTACATAAC





TTACGGTAAATGGCCCGCCTGGCTGAC





CGCCCAACGACCCCCGCCCATTGACGT





CAATAATGACGTATGTTCCCATAGTAA





CGCCAATAGGGACTTTCCATTGACGTC





AATGGGTGGAGTATTTACGGTAAACTG





CCCACTTGGCAGTACATCAAGTGTATC





ATATGCCAAGTACGCCCCCTATTGACG





TCAATGACGGTAAATGGCCCGCCTGGC





ATTATGCCCAGTACATGACCTTATGGG





ACTTTCCTACTTGGCAGTACATCTACG





TATTAGTCATCGCTATTACCATGGTGA





TGCGGTTTTGGCAGTACATCAATGGGC





GTGGATAGCGGTTTGACTCACGGGGAT





TTCCAAGTCTCCACCCCATTGACGTCA





ATGGGAGTTTGTTTTGGCACCAAAATC





AACGGGACTTTCCAAAATGTCGTAACA





ACTCCGCCCCATTGACGCAAATGGGCG





GTAGGCGTGTACGGTGGGAGGTCTATA





TAAGCAGCGCGTTTTGCCTGTACTGGG





TCTCTCTGGTTAGACCAGATCTGAGCC





TGGGAGCTCTCTGGCTAACTAGGGAAC





CCACTGCTTAAGCCTCAATAAAGCTTG





CCTTGAGTGCTTCAAGTAGTGTGTGCC





CGTCTGTTGTGTGACTCTGGTAACTAG





AGATCCCTCAGACCCTTTTAGTCAGTG





TGGAAAATCTCTAGCAGTGGCGCCCGA





ACAGGGACTTGAAAGCGAAAGGGAAAC





CAGAGGAGCTCTCTCGACGCAGGACTC





GGCTTGCTGAAGCGCGCACGGCAAGAG





GCGAGGGGCGGCGACTGGTGAGTACGC





CAAAAATTTTGACTAGCGGAGGCTAGA





AGGAGAGAGATGGGTGCGAGAGCGTCA





GTATTAAGCGGGGGAGAATTAGATCGC





GATGGGAAAAAATTCGGTTAAGGCCAG





GGGGAAAGAAAAAATATAAATTAAAAC





ATATAGTATGGGCAAGCAGGGAGCTAG





AACGATTCGCAGTTAATCCTGGCCTGT





TAGAAACATCAGAAGGCTGTAGACAAA





TACTGGGACAGCTACAACCATCCCTTC





AGACAGGATCAGAAGAACTTAGATCAT





TATATAATACAGTAGCAACCCTCTATT





GTGTGCATCAAAGGATAGAGATAAAAG





ACACCAAGGAAGCTTTAGACAAGATAG





AGGAAGAGCAAAACAAAAGTAAGACCA





CCGCACAGCAAGCGGCCGCTGATCTTC





AGACCTGGAGGAGGAGATATGAGGGAC





AATTGGAGAAGTGAATTATATAAATAT





AAAGTAGTAAAAATTGAACCATTAGGA





GTAGCACCCACCAAGGCAAAGAGAAGA





GTGGTGCAGAGAGAAAAAAGAGCAGTG





GGAATAGGAGCTTTGTTCCTTGGGTTC





TTGGGAGCAGCAGGAAGCACTATGGGC





GCAGCGTCAATGACGCTGACGGTACAG





GCCAGACAATTATTGTCTGGTATAGTG





CAGCAGCAGAACAATTTGCTGAGGGCT





ATTGAGGCGCAACAGCATCTGTTGCAA





CTCACAGTCTGGGGCATCAAGCAGCTC





CAGGCAAGAATCCTGGCTGTGGAAAGA





TACCTAAAGGATCAACAGCTCCTGGGG





ATTTGGGGTTGCTCTGGAAAACTCATT





TGCACCACTGCTGTGCCTTGGAATGCT





AGTTGGAGTAATAAATCTCTGGAACAG





ATTTGGAATCACACGACCTGGATGGAG





TGGGACAGAGAAATTAACAATTACACA





AGCTTAATACACTCCTTAATTGAAGAA





TCGCAAAACCAGCAAGAAAAGAATGAA





CAAGAATTATTGGAATTAGATAAATGG





GCAAGTTTGTGGAATTGGTTTAACATA





ACAAATTGGCTGTGGTATATAAAATTA





TTCATAATGATAGTAGGAGGCTTGGTA





GGTTTAAGAATAGTTTTTGCTGTACTT





TCTATAGTGAATAGAGTTAGGCAGGGA





TATTCACCATTATCGTTTCAGACCCAC





CTCCCAACCCCGAGGGGACCCGACAGG





CCCGAAGGAATAGAAGAAGAAGGTGGA





GAGAGAGACAGAGACAGATCCATTCGA





TTAGTGAACGGATCGGCACTGCGTGCG





CCAATTCTGCAGACAAATGGCAGTATT





CATCCACAATTTTAAAAGAAAAGGGGG





GATTGGGGGGTACAGTGCAGGGGAAAG





AATAGTAGACATAATAGCAACAGACAT





ACAAACTAAAGAATTACAAAAACAAAT





TACAAAAATTCAAAATTTTCGGGTTTA





TTACAGGGACAGCAGAGATCCAGTTTG





GTTAccagTGTGATGGATATCTGCAGA





ATTCGCCCTTGGATCCGAATTCCTGCA





GCCCCGACTTTCACTTTTCTCTATCAC





TGATAGGGAGTGGTAAACTCGACTTTC





ACTTTTCTCTATCACTGATAGGGAGTG





GTAAACTCGACTTTCACTTTTCTCTAT





CACTGATAGGGAGTGGTAAACTCGACT





TTCACTTTTCTCTATCACTGATAGGGA





GTGGTAAACTCGACTTTCACTTTTCTC





TATCACTGATAGGGAGTGGTAAACTCG





ACTTTCACTTTTCTCTATCACTGATAG





GGAGTGGTAAACTCGACTTTCACTTTT





CTCTATCACTGATAGGGAGTGGTAAAC





TCGAGGGGGATCCACTAGCATGAAGGG





CGAATTCCAGCACActggTAAcccgtg





tcggctccagatctGGCCTCCGCGCCG





GGTTTTGGCGcctcccgcgggcgcccc





cctcctcacggcgagccgcgTTGACAT





TGATTATTGACTAGGCTTTTGCAAAAA





GCTTTGCAAAGATGGATAAAGTTTTAA





ACAGAGAGGAATCTTTGCAGCTAATGG





ACCTTCTAGGTCTTGAAAGGAGTGGGA





ATTGGCTCCGGTGCCCGTCAGTGGGCA





GAGCGCACATCGCCCACAGTCCCCGAG





AAGTTGGGGGGAGGGGTCGGCAATTGA





ACCGGTGCCTAGAGAAGGTGGCGCGGG





GTAAACTGGGAAAGTGATGTCGTGTAC





TGGCTCCGCCTTTTTCCCGAGGGTGGG





GGAGAACCGTATATAAGTGCAGTAGTC





GCCGTGAACGTTCTTTTTCGCAACGGG





TTTGCCGCCAGAACACAGGTAAGTGCC





GTGTGTGGTTCCCGCGGGCCTGGCCTC





TTTACGGGTTATGGCCCTTGCGTGCCT





TGAATTACTTCCACCTGGCTGCAGTAC





GTGATTCTTGATCCCGAGCTTCGGGTT





GGAAGTGGGTGGGAGAGTTCGAGGCCT





TGCGCTTAAGGAGCCCCTTCGCCTCGT





GCTTGAGTTGAGGCCTGGCCTGGGCGC





TGGGGCCGCCGCGTGCGAATCTGGTGG





CACCTTCGCGCCTGTCTCGCTGCTTTC





GATAAGTCTCTAGCCATTTAAAATTTT





TGATGACCTGCTGCGACGCTTTTTTTC





TGGCAAGATAGTCTTGTAAATGCGGGC





CAAGATCTGCACACTGGTATTTCGGTT





TTTGGGGCCGCGGGCGGCGACGGGGCC





CGTGCGTCCCAGCGCACATGTTCGGCG





AGGCGGGGCCTGCGAGCGCGGCCACCG





AGAATCGGACGGGGGTAGTCTCAAGCT





GGCCGGCCTGCTCTGGTGCCTGGCCTC





GCGCCGCCGTGTATCGCCCCGCCCTGG





GCGGCAAGGCTGGCCCGGTCGGCACCA





GTTGCGTGAGCGGAAAGATGGCCGCTT





CCCGGCCCTGCTGCAGGGAGCTCAAAA





TGGAGGACGCGGCGCTCGGGAGAGCGG





GCGGGTGAGTCACCCACACAAAGGAAA





AGGGCCTTTCCGTCCTCAGCCGTCGCT





TCATGTGACTCCACGGAGTACCGGGCG





CCGTCCAGGCACCTCGATTAGTTCTCG





AGCTTTTGGAGTACGTCGTCTTTAGGT





TGGGGGGAGGGGTTTTATGCGATGGAG





TTTCCCCACACTGAGTGGGTGGAGACT





GAAGTTAGGCCAGCTTGGCACTTGATG





TAATTCTCCTTGGAATTTGCCCTTTTT





GAGTTTGGATCTTGGTTCATTCTCAAG





CCTCAGACAGTGGTTCAAAGTTTTTTT





CTTCCATTTCAGGTGTCGTGAGGAATT





AGCTTGGTACTAATACGACTCACTATA





GGGAGACCCAAGCTGGCTAGGTAAGCT





TGGTACCGAGCTCGGATCCACTAGTCC





AGTGTGGTGGAATTCTGCAGATATCCA





GCACAGTGGCGGCCGCTCGAGtctaga





ggatccccgggtaccggtcgccaccAT





GGTGAGCAAGGGCGAGGAGCTGTTCAC





CGGGGTGGTGCCCATCCTGGTCGAGCT





GGACGGCGACGTAAACGGCCACAAGTT





CAGCGTGTCCGGCGAGGGCGAGGGCGA





TGCCACCTACGGCAAGCTGACCCTGAA





GTTCATCTGCACCACCGGCAAGCTGCC





CGTGCCCTGGCCCACCCTCGTGACCAC





CCTGACCTACGGCGTGCAGTGCTTCAG





CCGCTACCCCGACCACATGAAGCAGCA





CGACTTCTTCAAGTCCGCCATGCCCGA





AGGCTACGTCCAGGAGCGCACCATCTT





CTTCAAGGACGACGGCAACTACAAGAC





CCGCGCCGAGGTGAAGTTCGAGGGCGA





CACCCTGGTGAACCGCATCGAGCTGAA





GGGCATCGACTTCAAGGAGGACGGCAA





CATCCTGGGGCACAAGCTGGAGTACAA





CTACAACAGCCACAACGTCTATATCAT





GGCCGACAAGCAGAAGAACGGCATCAA





GGTGAACTTCAAGATCCGCCACAACAT





CGAGGACGGCAGCGTGCAGCTCGCCGA





CCACTACCAGCAGAACACCCCCATCGG





CGACGGCCCCGTGCTGCTGCCCGACAA





CCACTACCTGAGCACCCAGTCCGCCCT





GAGCAAAGACCCCAACGAGAAGCGCGA





TCACATGGTCCTGCTGGAGTTCGTGAC





CGCCGCCGGGATCACTCTCGGCATGGA





CGAGCtgtacactcgaggttaacgaat





tctaccgggtaggggaggcgcttttcc





caaggcagtctggagcatgcgctttag





cagccccgctgggcacttggcgctaca





caagtggcctctggcctcgcacacatt





ccacatccaccggtaggcgccaaccgg





ctccgttctttggtggccccttcgcgc





caccttctactcctcccctagtcagga





agttcccccccgccccgcagctcgcgt





cgtgcaggacgtgacaaatggaagtag





cacgtctcactagtctcgtgcagatgg





acagcaccgctgagcaatggaagcggg





taggcctttggggcagcggccaatagc





agctttgctccttcgctttctgggctc





agaggctgggaaggggtgggtccgggg





ggggctcaggggcgggctcaggggggg





gggggcccgaaggtcctccggaggccc





ggcattctgcacgcttcaaaagcgcac





gtctgccgcgctgttctcctcttcctc





atctccgggcctttcgacctgcatccc





gccaccATGAAAAAGCCTGAACTCACC





GCGACGTCTGTCGAGAAGTTTCTGATC





GAAAAGTTCGACAGCGTCTCCGACCTG





ATGCAGCTCTCGGAGGGCGAAGAATCT





CGTGCTTTCAGCTTCGATGTAGGAGGG





CGTGGATATGTCCTGCGGGTAAATAGC





TGCGCCGATGGTTTCTACAAAGATCGT





TATGTTTATCGGCACTTTGCATCGGCC





GCGCTCCCGATTCCGGAAGTGCTTGAC





ATTGGGGAATTCAGCGAGAGCCTGACC





TATTGCATCTCCCGCCGTGCACAGGGT





GTCACGTTGCAAGACCTGCCTGAAACC





GAACTGCCCGCTGTTCTGCAGCCGGTC





GCGGAGGCCATGGATGCGATCGCTGCG





GCCGATCTTAGCCAGACGAGCGGGTTC





GGCCCATTCGGACCGCAAGGAATCGGT





CAATACACTACATGGCGTGATTTCATA





TGCGCGATTGCTGATCCCCATGTGTAT





CACTGGCAAACTGTGATGGACGACACC





GTCAGTGCGTCCGTCGCGCAGGCTCTC





GATGAGCTGATGCTTTGGGCCGAGGAC





TGCCCCGAAGTCCGGCACCTCGTGCAC





GCGGATTTCGGCTCCAACAATGTCCTG





ACGGACAATGGCCGCATAACAGCGGTC





ATTGACTGGAGCGAGGCGATGTTCGGG





GATTCCCAATACGAGGTCGCCAACATC





TTCTTCTGGAGGCCGTGGTTGGCTTGT





ATGGAGCAGCAGACGCGCTACTTCGAG





CGGAGGCATCCGGAGCTTGCAGGATCG





CCGCGGCTCCGGGGCGTATATGCTCCG





CATTGGTCTTGACCAACTCTATCAGAG





CTTGGTTGACGGCAATTTCGATGATGC





AGCTTGGGCGCAGGGTCGATGCGACGC





AATCGTCCGATCCGGAGCCGGGACTGT





CGGGCGTACACAAATCGCCCGCAGAAG





CGCGGCCGTCTGGACCGATGGCTGTGT





AGAAGTACTCGCCGATAGTGGAAACCG





ACGCCCCAGCACTCGTCCGAGGGCAAA





GGAATAGAGTAGATGCCGACCGAACAA





GAGCTGATTTCGAGAACGCCTCAGCCA





GCAACTCGCGCGAGCCTAGCAAGGCAA





ATGCGAGAGAACGGCCTTACGCTTGGT





GGCACAGTTCTCGTCCACAGTTCGCTA





AGCTCGCTCGGCTGGGTCGCGGGAGGG





CCGGTCGCAGTGATTCAGGCCCTTCTG





GATTGTGTTGGTCCCCAGGGCACGATT





GTCATGCCCACGCACTCGGGTGATCTG





ACTGATCCCGCAGATTGGAGATCGCCG





CCCGTGCCTGCCGATTGGGTGCAGATC





CGTCGAGttaacAAAAGAAAAGGGGGG





ACTGGAAGGGCTAATTCACTCCCAACG





AAGACAAGATatcataacttcgtatag





catacattatacgaagttatcggctag





ctggtccggaCTGTACTGGGTCTCTCT





GGTTAGACCAGATCTGAGCCTGGGAGC





TCTCTGGCTAACTAGGGAACCCACTGC





TTAAGCCTCAATAAAGCTTGCCTTGAG





TGCTTCAAGTAGTGTGTGCCCGTCTGT





TGTGTGACTCTGGTAACTAGAGATCCC





TCAGACCCTTTTAGTCAGTGTGGAAAA





TCTCTAGCAGGGCCCGTTTAAACCCGC





TGATCAGCCTCGACTGTGCCTTCTAGT





TGCCAGCCATCTGTTGTTTGCCCCTCC





CCCGTGCCTTCCTTGACCCTGGAAGGT





GCCACTCCCACTGTCCTTTCCTAATAA





AATGAGGAAATTGCATCGCATTGTCTG





AGTAGGTGTCATTCTATTCTGGGGGGT





GGGGTGGGGCAGGACAGCAAGGGGGAG





GATTGGGAAGACAATAGCAGGCATGCT





GGGGATGCGGTGGGCTCTATGGCTTCT





GAGGCGGAAAGAACCAGCTGGGGCTCT





AGGGGGTATCCCCACGCGCCCTGTAGC





GGCGCATTAAGCGCGGCGGGTGTGGTG





GTTACGCGCAGCGTGACCGCTACACTT





GCCAGCGCCCTAGCGCCCGCTCCTTTC





GCTTTCTTCCCTTCCTTTCTCGCCACG





TTCGCCGGCTTTCCCCGTCAAGCTCTA





AATCGGGGGCTCCCTTTAGGGTTCCGA





TTTAGTGCTTTACGGCACCTCGACCCC





AAAAAACTTGATTAGGGTGATGGTTCA





CGTAGTGGGCCATCGCCCTGATAGACG





GTTTTTCGCCCTTTGACGTTGGAGTCC





ACGTTCTTTAATAGTGGACTCTTGTTC





CAAACTGGAACAACACTCAACCCTATC





TCGGTCTATTCTTTTGATTTATAAGGG





ATTTTGCCGATTTCGGCCTATTGGTTA





AAAAATGAGCTGATTTAACAAAAATTT





AACGCGAATTAATTCTGTGGAATGTGT





GTCAGTTAGGGTGTGGAAAGTCCCCAG





GCTCCCCAGCAGGCAGAAGTATGCAAA





GCATGCATCTCAATTAGTCAGCAACCA





GGTGTGGAAAGTCCCCAGGCTCCCCAG





CAGGCAGAAGTATGCAAAGCATGCATC





TCAATTAGTCAGCAACCATAGTCCCGC





CCCTAACTCCGCCCATCCCGCCCCTAA





CTCCGCCCAGTTCCGCCCATTCTCCGC





CCCATGGCTGACTAATTTTTTTTATTT





ATGCAGAGGCCGAGGCCGCCTCTGCCT





CTGAGCTATTCCAGAAGTAGTGAGGAG





GCTTTTTTGGAGGCCTAGGCTTTTGCA





AAAAGCTCCCGGGAGCTTGTATATCCA





TTTTCGGATCTGATCAGCACGTGTTGA





CAATTAATCATCGGCATAGTATATCGG





CATAGTATAATACGACAAGGTGAGGAA





CTAAACCATGGCCAAGTTGACCAGTGC





CGTTCCGGTGCTCACCGCGCGCGACGT





CGCCGGAGCGGTCGAGTTCTGGACCGA





CCGGCTCGGGTTCTCCCGGGACTTCGT





GGAGGACGACTTCGCCGGTGTGGTCCG





GGACGACGTGACCCTGTTCATCAGCGC





GGTCCAGGACCAGGTGGTGCCGGACAA





CACCCTGGCCTGGGTGTGGGTGCGCGG





CCTGGACGAGCTGTACGCCGAGTGGTC





GGAGGTCGTGTCCACGAACTTCCGGGA





CGCCTCCGGGCCGGCCATGACCGAGAT





CGGCGAGCAGCCGTGGGGGCGGGAGTT





CGCCCTGCGCGACCCGGCCGGCAACTG





CGTGCACTTCGTGGCCGAGGAGCAGGA





CTGACACGTGCTACGAGATTTCGATTC





CACCGCCGCCTTCTATGAAAGGTTGGG





CTTCGGAATCGTTTTCCGGGACGCCGG





CTGGATGATCCTCCAGCGCGGGGATCT





CATGCTGGAGTTCTTCGCCCACCCCAA





CTTGTTTATTGCAGCTTATAATGGTTA





CAAATAAAGCAATAGCATCACAAATTT





CACAAATAAAGCATTTTTTTCACTGCA





TTCTAGTTGTGGTTTGTCCAAACTCAT





CAATGTATCTTATCATGTCTGTATACC





GTCGACCTCTAGCTAGAGCTTGGCGTA





ATCATGGTCATAGCTGTTTCCTGTGTG





AAATTGTTATCCGCTCACAATTCCACA





CAACATACGAGCCGGAAGCATAAAGTG





TAAAGCCTGGGGTGCCTAATGAGTGAG





CTAACTCACATTAATTGCGTTGCGCTC





ACTGCCCGCTTTCCAGTCGGGAAACCT





GTCGTGCCAGCTGCATTAATGAATCGG





CCAACGCGCGGGGAGAGGCGGTTTGCG





TATTGGGCGCTCTTCCGCTTCCTCGCT





CACTGACTCGCTGCGCTCGGTCGTTCG





GCTGCGGCGAGCGGTATCAGCTCACTC





AAAGGCGGTAATACGGTTATCCACAGA





ATCAGGGGATAACGCAGGAAAGAACAT





GTGAGCAAAAGGCCAGCAAAAGGCCAG





GAACCGTAAAAAGGCCGCGTTGCTGGC





GTTTTTCCATAGGCTCCGCCCCCCTGA





CGAGCATCACAAAAATCGACGCTCAAG





TCAGAGGTGGCGAAACCCGACAGGACT





ATAAAGATACCAGGCGTTTCCCCCTGG





AAGCTCCCTCGTGCGCTCTCCTGTTCC





GACCCTGCCGCTTACCGGATACCTGTC





CGCCTTTCTCCCTTCGGGAAGCGTGGC





GCTTTCTCATAGCTCACGCTGTAGGTA





TCTCAGTTCGGTGTAGGTCGTTCGCTC





CAAGCTGGGCTGTGTGCACGAACCCCC





CGTTCAGCCCGACCGCTGCGCCTTATC





CGGTAACTATCGTCTTGAGTCCAACCC





GGTAAGACACGACTTATCGCCACTGGC





AGCAGCCACTGGTAACAGGATTAGCAG





AGCGAGGTATGTAGGCGGTGCTACAGA





GTTCTTGAAGTGGTGGCCTAACTACGG





CTACACTAGAAGAACAGTATTTGGTAT





CTGCGCTCTGCTGAAGCCAGTTACCTT





CGGAAAAAGAGTTGGTAGCTCTTGATC





CGGCAAACAAACCACCGCTGGTAGCGG





TGGTTTTTTTGTTTGCAAGCAGCAGAT





TACGCGCAGAAAAAAAGGATCTCAAGA





AGATCCTTTGATCTTTTCTACGGGGTC





TGACGCTCAGTGGAACGAAAACTCACG





TTAAGGGATTTTGGTCATGAGATTATC





AAAAAGGATCTTCACCTAGATCCTTTT





AAATTAAAAATGAAGTTTTAAATCAAT





CTAAAGTATATATGAGTAAACTTGGTC





TGACAGTTACCAATGCTTAATCAGTGA





GGCACCTATCTCAGCGATCTGTCTATT





TCGTTCATCCATAGTTGCCTGACTCCC





CGTCGTGTAGATAACTACGATACGGGA





GGGCTTACCATCTGGCCCCAGTGCTGC





AATGATACCGCGAGACCCACGCTCACC





GGCTCCAGATTTATCAGCAATAAACCA





GCCAGCCGGAAGGGCCGAGCGCAGAAG





TGGTCCTGCAACTTTATCCGCCTCCAT





CCAGTCTATTAATTGTTGCCGGGAAGC





TAGAGTAAGTAGTTCGCCAGTTAATAG





TTTGCGCAACGTTGTTGCCATTGCTAC





AGGCATCGTGGTGTCACGCTCGTCGTT





TGGTATGGCTTCATTCAGCTCCGGTTC





CCAACGATCAAGGCGAGTTACATGATC





CCCCATGTTGTGCAAAAAAGCGGTTAG





CTCCTTCGGTCCTCCGATCGTTGTCAG





AAGTAAGTTGGCCGCAGTGTTATCACT





CATGGTTATGGCAGCACTGCATAATTC





TCTTACTGTCATGCCATCCGTAAGATG





CTTTTCTGTGACTGGTGAGTACTCAAC





CAAGTCATTCTGAGAATAGTGTATGCG





GCGACCGAGTTGCTCTTGCCCGGCGTC





AATACGGGATAATACCGCGCCACATAG





CAGAACTTTAAAAGTGCTCATCATTGG





AAAACGTTCTTCGGGGCGAAAACTCTC





AAGGATCTTACCGCTGTTGAGATCCAG





TTCGATGTAACCCACTCGTGCACCCAA





CTGATCTTCAGCATCTTTTACTTTCAC





CAGCGTTTCTGGGTGAGCAAAAACAGG





AAGGCAAAATGCCGCAAAAAAGGGAAT





AAGGGCGACACGGAAATGTTGAATACT





CATACTCTTCCTTTTTCAATATTATTG





AAGCATTTATCAGGGTTATTGTCTCAT





GAGCGGATACATATTTGAATGTATTTA





GAAAAATAAACAAATAGGGGTTCCGCG





CACATTTCCCCGAAAAGTGCCACCTGA





C






59
TLCV2
ATGGACAAGAAGTACAGCATCGGCCTG
CAS9 fragment




GACATCGGCACCAACTCTGTGGGCTGG
source to clone in




GCCGTGATCACCGACGAGTACAAGGTG
GEMS:




CCCAGCAAGAAATTCAAGGTGCTGGGC
Addgene #87360,




AACACCGACCGGCACAGCATCAAGAAG
the sequence of




AACCTGATCGGAGCCCTGCTGTTCGAC
which is accessible




AGCGGCGAAACAGCCGAGGCCACCCGG
at addgene.org/




CTGAAGAGAACCGCCAGAAGAAGATAC
87360/ and




ACCAGACGGAAGAACCGGATCTGCTAT
incorporated




CTGCAAGAGATCTTCAGCAACGAGATG
by reference




GCCAAGGTGGACGACAGCTTCTTCCAC





AGACTGGAAGAGTCCTTCCTGGTGGAA





GAGGATAAGAAGCACGAGCGGCACCCC





ATCTTCGGCAACATCGTGGACGAGGTG





GCCTACCACGAGAAGTACCCCACCATC





TACCACCTGAGAAAGAAACTGGTGGAC





AGCACCGACAAGGCCGACCTGCGGCTG





ATCTATCTGGCCCTGGCCCACATGATC





AAGTTCCGGGGCCACTTCCTGATCGAG





GGCGACCTGAACCCCGACAACAGCGAC





GTGGACAAGCTGTTCATCCAGCTGGTG





CAGACCTACAACCAGCTGTTCGAGGAA





AACCCCATCAACGCCAGCGGCGTGGAC





GCCAAGGCCATCCTGTCTGCCAGACTG





AGCAAGAGCAGACGGCTGGAAAATCTG





ATCGCCCAGCTGCCCGGCGAGAAGAAG





AATGGCCTGTTCGGAAACCTGATTGCC





CTGAGCCTGGGCCTGACCCCCAACTTC





AAGAGCAACTTCGACCTGGCCGAGGAT





GCCAAACTGCAGCTGAGCAAGGACACC





TACGACGACGACCTGGACAACCTGCTG





GCCCAGATCGGCGACCAGTACGCCGAC





CTGTTTCTGGCCGCCAAGAACCTGTCC





GACGCCATCCTGCTGAGCGACATCCTG





AGAGTGAACACCGAGATCACCAAGGCC





CCCCTGAGCGCCTCTATGATCAAGAGA





TACGACGAGCACCACCAGGACCTGACC





CTGCTGAAAGCTCTCGTGCGGCAGCAG





CTGCCTGAGAAGTACAAAGAGATTTTC





TTCGACCAGAGCAAGAACGGCTACGCC





GGCTACATTGACGGCGGAGCCAGCCAG





GAAGAGTTCTACAAGTTCATCAAGCCC





ATCCTGGAAAAGATGGACGGCACCGAG





GAACTGCTCGTGAAGCTGAACAGAGAG





GACCTGCTGCGGAAGCAGCGGACCTTC





GACAACGGCAGCATCCCCCACCAGATC





CACCTGGGAGAGCTGCACGCCATTCTG





CGGCGGCAGGAAGATTTTTACCCATTC





CTGAAGGACAACCGGGAAAAGATCGAG





AAGATCCTGACCTTCCGCATCCCCTAC





TACGTGGGCCCTCTGGCCAGGGGAAAC





AGCAGATTCGCCTGGATGACCAGAAAG





AGCGAGGAAACCATCACCCCCTGGAAC





TTCGAGGAAGTGGTGGACAAGGGCGCT





TCCGCCCAGAGCTTCATCGAGCGGATG





ACCAACTTCGATAAGAACCTGCCCAAC





GAGAAGGTGCTGCCCAAGCACAGCCTG





CTGTACGAGTACTTCACCGTGTATAAC





GAGCTGACCAAAGTGAAATACGTGACC





GAGGGAATGAGAAAGCCCGCCTTCCTG





AGCGGCGAGCAGAAAAAGGCCATCGTG





GACCTGCTGTTCAAGACCAACCGGAAA





GTGACCGTGAAGCAGCTGAAAGAGGAC





TACTTCAAGAAAATCGAGTGCTTCGAC





TCCGTGGAAATCTCCGGCGTGGAAGAT





CGGTTCAACGCCTCCCTGGGCACATAC





CACGATCTGCTGAAAATTATCAAGGAC





AAGGACTTCCTGGACAATGAGGAAAAC





GAGGACATTCTGGAAGATATCGTGCTG





ACCCTGACACTGTTTGAGGACAGAGAG





ATGATCGAGGAACGGCTGAAAACCTAT





GCCCACCTGTTCGACGACAAAGTGATG





AAGCAGCTGAAGCGGCGGAGATACACC





GGCTGGGGCAGGCTGAGCCGGAAGCTG





ATCAACGGCATCCGGGACAAGCAGTCC





GGCAAGACAATCCTGGATTTCCTGAAG





TCCGACGGCTTCGCCAACAGAAACTTC





ATGCAGCTGATCCACGACGACAGCCTG





ACCTTTAAAGAGGACATCCAGAAAGCC





CAGGTGTCCGGCCAGGGCGATAGCCTG





CACGAGCACATTGCCAATCTGGCCGGC





AGCCCCGCCATTAAGAAGGGCATCCTG





CAGACAGTGAAGGTGGTGGACGAGCTC





GTGAAAGTGATGGGCCGGCACAAGCCC





GAGAACATCGTGATCGAAATGGCCAGA





GAGAACCAGACCACCCAGAAGGGACAG





AAGAACAGCCGCGAGAGAATGAAGCGG





ATCGAAGAGGGCATCAAAGAGCTGGGC





AGCCAGATCCTGAAAGAACACCCCGTG





GAAAACACCCAGCTGCAGAACGAGAAG





CTGTACCTGTACTACCTGCAGAATGGG





CGGGATATGTACGTGGACCAGGAACTG





GACATCAACCGGCTGTCCGACTACGAT





GTGGACCATATCGTGCCTCAGAGCTTT





CTGAAGGACGACTCCATCGACAACAAG





GTGCTGACCAGAAGCGACAAGAACCGG





GGCAAGAGCGACAACGTGCCCTCCGAA





GAGGTCGTGAAGAAGATGAAGAACTAC





TGGCGGCAGCTGCTGAACGCCAAGCTG





ATTACCCAGAGAAAGTTCGACAATCTG





ACCAAGGCCGAGAGAGGCGGCCTGAGC





GAACTGGATAAGGCCGGCTTCATCAAG





AGACAGCTGGTGGAAACCCGGCAGATC





ACAAAGCACGTGGCACAGATCCTGGAC





TCCCGGATGAACACTAAGTACGACGAG





AATGACAAGCTGATCCGGGAAGTGAAA





GTGATCACCCTGAAGTCCAAGCTGGTG





TCCGATTTCCGGAAGGATTTCCAGTTT





TACAAAGTGCGCGAGATCAACAACTAC





CACCACGCCCACGACGCCTACCTGAAC





GCCGTCGTGGGAACCGCCCTGATCAAA





AAGTACCCTAAGCTGGAAAGCGAGTTC





GTGTACGGCGACTACAAGGTGTACGAC





GTGCGGAAGATGATCGCCAAGAGCGAG





CAGGAAATCGGCAAGGCTACCGCCAAG





TACTTCTTCTACAGCAACATCATGAAC





TTTTTCAAGACCGAGATTACCCTGGCC





AACGGCGAGATCCGGAAGCGGCCTCTG





ATCGAGACAAACGGCGAAACCGGGGAG





ATCGTGTGGGATAAGGGCCGGGATTTT





GCCACCGTGCGGAAAGTGCTGAGCATG





CCCCAAGTGAATATCGTGAAAAAGACC





GAGGTGCAGACAGGCGGCTTCAGCAAA





GAGTCTATCCTGCCCAAGAGGAACAGC





GATAAGCTGATCGCCAGAAAGAAGGAC





TGGGACCCTAAGAAGTACGGCGGCTTC





GACAGCCCCACCGTGGCCTATTCTGTG





CTGGTGGTGGCCAAAGTGGAAAAGGGC





AAGTCCAAGAAACTGAAGAGTGTGAAA





GAGCTGCTGGGGATCACCATCATGGAA





AGAAGCAGCTTCGAGAAGAATCCCATC





GACTTTCTGGAAGCCAAGGGCTACAAA





GAAGTGAAAAAGGACCTGATCATCAAG





CTGCCTAAGTACTCCCTGTTCGAGCTG





GAAAACGGCCGGAAGAGAATGCTGGCC





TCTGCCGGCGAACTGCAGAAGGGAAAC





GAACTGGCCCTGCCCTCCAAATATGTG





AACTTCCTGTACCTGGCCAGCCACTAT





GAGAAGCTGAAGGGCTCCCCCGAGGAT





AATGAGCAGAAACAGCTGTTTGTGGAA





CAGCACAAGCACTACCTGGACGAGATC





ATCGAGCAGATCAGCGAGTTCTCCAAG





AGAGTGATCCTGGCCGACGCTAATCTG





GACAAAGTGCTGTCCGCCTACAACAAG





CACCGGGATAAGCCCATCAGAGAGCAG





GCCGAGAATATCATCCACCTGTTTACC





CTGACCAATCTGGGAGCCCCTGCCGCC





TTCAAGTACTTTGACACCACCATCGAC





CGGAAGAGGTACACCAGCACCAAAGAG





GTGCTGGACGCCACCCTGATCCACCAG





AGCATCACCGGCCTGTACGAGACACGG





ATCGACCTGTCTCAGCTGGGAGGCGAC






60
pCMV-dCas13-M3nls
GTGATGCGGTTTTGGCAGTACATCAAT
Targeted m6A




GGGCGTGGATAGCGGTTTGACTCACGG
RNA methylation




GGATTTCCAAGTCTCCACCCCATTGAC
in mammalian cells




GTCAATGGGAGTTTGTTTTGGCACCAA
Addgene #155366,




AATCAACGGGACTTTCCAAAATGTCGT
the sequence of




AACAACTCCGCCCCATTGACGCAAATG
which is accessible




GGCGGTAGGCGTGTACGGTGGGAGGTC
at addgene.org/




TATATAAGCAGAGCTGGTTTAGTGAAC
155366/ and




CGTCAGATCCGCTAGAGATCCGCGGCC
incorporated




GCTAATACGACTCACTATAGGGAGAGC
by reference




CGCCACCATGAAACGGACAGCCGACGG





AAGCGAGTTCGAGTCACCAAAGAAGAA





GCGGAAAGTCAACATCCCCGCTCTGGT





GGAAAACCAGAAGAAGTACTTTGGCAC





CTACAGCGTGATGGCCATGCTGAACGC





TCAGACCGTGCTGGACCACATCCAGAA





GGTGGCCGATATTGAGGGCGAGCAGAA





CGAGAACAACGAGAATCTGTGGTTTCA





CCCCGTGATGAGCCACCTGTACAACGC





CAAGAACGGCTACGACAAGCAGCCCGA





GAAAACCATGTTCATCATCGAGCGGCT





GCAGAGCTACTTCCCATTCCTGAAGAT





CATGGCCGAGAACCAGAGAGAGTACAG





CAACGGCAAGTACAAGCAGAACCGCGT





GGAAGTGAACAGCAACGACATCTTCGA





GGTGCTGAAGCGCGCCTTCGGCGTGCT





GAAGATGTACAGGGACCTGACCAACGC





ATACAAGACCTACGAGGAAAAGCTGAA





CGACGGCTGCGAGTTCCTGACCAGCAC





AGAGCAACCTCTGAGCGGCATGATCAA





CAACTACTACACAGTGGCCCTGCGGAA





CATGAACGAGAGATACGGCTACAAGAC





AGAGGACCTGGCCTTCATCCAGGACAA





GCGGTTCAAGTTCGTGAAGGACGCCTA





CGGCAAGAAAAAGTCCCAAGTGAATAC





CGGATTCTTCCTGAGCCTGCAGGACTA





CAACGGCGACACACAGAAGAAGCTGCA





CCTGAGCGGAGTGGGAATCGCCCTGCT





GATCTGCCTGTTCCTGGACAAGCAGTA





CATCAACATCTTTCTGAGCAGGCTGCC





CATCTTCTCCAGCTACAATGCCCAGAG





CGAGGAACGGCGGATCATCATCAGATC





CTTCGGCATCAACAGCATCAAGCTGCC





CAAGGACCGGATCCACAGCGAGAAGTC





CAACAAGAGCGTGGCCATGGATATGCT





CAACGAAGTGAAGCGGTGCCCCGACGA





GCTGTTCACAACACTGTCTGCCGAGAA





GCAGTCCCGGTTCAGAATCATCAGCGA





CGACCACAATGAAGTGCTGATGAAGCG





GAGCAGCGACAGATTCGTGCCTCTGCT





GCTGCAGTATATCGATTACGGCAAGCT





GTTCGACCACATCAGGTTCCACGTGAA





CATGGGCAAGCTGAGATACCTGCTGAA





GGCCGACAAGACCTGCATCGACGGCCA





GACCAGAGTCAGAGTGATCGAGCAGCC





CCTGAACGGCTTCGGCAGACTGGAAGA





GGCCGAGACAATGCGGAAGCAAGAGAA





CGGCACCTTCGGCAACAGCGGCATCCG





GATCAGAGACTTCGAGAACATGAAGCG





GGACGACGCCAATCCTGCCAACTATCC





CTACATCGTGGACACCTACACACACTA





CATCCTGGAAAACAACAAGGTCGAGAT





GTTTATCAACGACAAAGAGGACAGCGC





CCCACTGCTGCCCGTGATCGAGGATGA





TAGATACGTGGTCAAGACAATCCCCAG





CTGCCGGATGAGCACCCTGGAAATTCC





AGCCATGGCCTTCCACATGTTTCTGTT





CGGCAGCAAGAAAACCGAGAAGCTGAT





CGTGGACGTGCACAACCGGTACAAGAG





ACTGTTCCAGGCCATGCAGAAAGAAGA





AGTGACCGCCGAGAATATCGCCAGCTT





CGGAATCGCCGAGAGCGACCTGCCTCA





GAAGATCCTGGATCTGATCAGCGGCAA





TGCCCACGGCAAGGATGTGGACGCCTT





CATCAGACTGACCGTGGACGACATGCT





GACCGACACCGAGCGGAGAATCAAGAG





ATTCAAGGACGACCGGAAGTCCATTCG





GAGCGCCGACAACAAGATGGGAAAGAG





AGGCTTCAAGCAGATCTCCACAGGCAA





GCTGGCCGACTTCCTGGCCAAGGACAT





CGTGCTGTTTCAGCCCAGCGTGAACGA





TGGCGAGAACAAGATCACCGGCCTGAA





CTACCGGATCATGCAGAGCGCCATTGC





CGTGTACGATAGCGGCGACGATTACGA





GGCCAAGCAGCAGTTCAAGCTGATGTT





CGAGAAGGCCCGGCTGATCGGCAAGGG





CACAACAGAGCCTCATCCATTTCTGTA





CAAGGTGTTCGCCCGCAGCATCCCCGC





CAATGCCGTCGAGTTCTACGAGCGCTA





CCTGATCGAGCGGAAGTTCTACCTGAC





CGGCCTGTCCAACGAGATCAAGAAAGG





CAACAGAGTGGATGTGCCCTTCATCCG





GCGGGACCAGAACAAGTGGAAAACACC





CGCCATGAAGACCCTGGGCAGAATCTA





CAGCGAGGATCTGCCCGTGGAACTGCC





CAGACAGATGTTCGACAATGAGATCAA





GTCCCACCTGAAGTCCCTGCCACAGAT





GGAAGGCATCGACTTCAACAATGCCAA





CGTGACCTATCTGATCGCCGAGTACAT





GAAGAGAGTGCTGGACGACGACTTCCA





GACCTTCTACCAGTGGAACCGCAACTA





CCGGTACATGGACATGCTTAAGGGCGA





GTACGACAGAAAGGGCTCCCTGCAGCA





CTGCTTCACCAGCGTGGAAGAGAGAGA





AGGCCTCTGGAAAGAGCGGGCCTCCAG





AACAGAGCGGTACAGAAAGCAGGCCAG





CAACAAGATCCGCAGCAACCGGCAGAT





GAGAAACGCCAGCAGCGAAGAGATCGA





GACAATCCTGGATAAGCGGCTGAGCAA





CAGCCGGAACGAGTACCAGAAAAGCGA





GAAAGTGATCCGGCGCTACAGAGTGCA





GGATGCCCTGCTGTTTCTGCTGGCCAA





AAAGACCCTGACCGAACTGGCCGATTT





CGACGGCGAGAGGTTCAAACTGAAAGA





AATCATGCCCGACGCCGAGAAGGGAAT





CCTGAGCGAGATCATGCCCATGAGCTT





CACCTTCGAGAAAGGCGGCAAGAAGTA





CACCATCACCAGCGAGGGCATGAAGCT





GAAGAACTACGGCGACTTCTTTGTGCT





GGCTAGCGACAAGAGGATCGGCAACCT





GCTGGAACTCGTGGGCAGCGACATCGT





GTCCAAAGAGGATGGATCCAAAAGAAC





CGCCGACGGCAGCGAATTCGAGCCCAA





GAAGAAGAGGAAAGTCTCTGGCAGCGA





GACACCAGGAACAAGCGAGTCAGCAAC





ACCAGAGAGCCAAGAGTTCTGTGACTA





TGGCACCAAAGAGGAGTGCATGAAGGC





TAGCGACGCTGATCGTCCATGCCGTAA





GCTGCACTTCCGTCGCATCATTAACAA





ACACACCGACGAGAGCCTGGGCGACTG





CAGCTTCCTGAACACCTGCTTTCACAT





GGACACCTGCAAGTACGTGCACTATGA





GATCGACGCGTGCATGGATAGCGAAGC





TCCGGGCAGCAAAGATCACACCCCGAG





CCAGGAACTGGCCCTGACCCAGAGCGT





GGGTGGTGACAGCAGCGCGGATCGTCT





GTTCCCACCACAGTGGATCTGCTGCGA





CATTCGTTACCTGGATGTGAGCATCCT





GGGCAAGTTTGCTGTTGTGATGGCCGA





CCCGCCGTGGGATATTCACATGGAGCT





GCCGTATGGTACCCTGACCGACGATGA





AATGCGTCGCCTGAACATCCCGGTGCT





GCAGGACGATGGCTTCCTGTTTCTGTG





GGTGACGGGTCGTGCTATGGAGCTGGG





TCGTGAATGCCTGAACCTGTGGGGTTA





CGAGCGTGTGGACGAAATCATTTGGGT





GAAAACCAACCAGCTGCAGCGTATCAT





TCGCACCGGCCGTACCGGTCACTGGCT





GAACCACGGCAAGGAGCACTGCCTGGT





GGGCGTGAAAGGCAACCCGCAGGGCTT





TAACCAGGGTCTGGACTGCGATGTGAT





CGTGGCTGAAGTGCGCAGCACCAGCCA





CAAGCCGGACGAGATCTACGGCATGAT





TGAACGCCTGAGCCCGGGTACCCGTAA





AATTGAGCTGTTCGGCCGTCCGCACAA





CGTGCAGCCGAACTGGATCACCCTGGG





CAACCAGCTGGACGGTATTCACCTGCT





GGACCCAGATGTGGTGGCTCGCTTTAA





ACAGCGTTATCCGGATGGCATCATTAG





CAAACCGAAGAATCTGTAACCGGTCAT





CATCACCATCACCATTGAGTTTAAACC





CGCTGATCAGCCTCGACTGTGCCTTCT





AGTTGCCAGCCATCTGTTGTTTGCCCC





TCCCCCGTGCCTTCCTTGACCCTGGAA





GGTGCCACTCCCACTGTCCTTTCCTAA





TAAAATGAGGAAATTGCATCGCATTGT





CTGAGTAGGTGTCATTCTATTCTGGGG





GGTGGGGTGGGGCAGGACAGCAAGGGG





GAGGATTGGGAAGACAATAGCAGGCAT





GCTGGGGATGCGGTGGGCTCTATGGCT





TCTGAGGCGGAAAGAACCAGCTGGGGC





TCGATACCGTCGACCTCTAGCTAGAGC





TTGGCGTAATCATGGTCATAGCTGTTT





CCTGTGTGAAATTGTTATCCGCTCACA





ATTCCACACAACATACGAGCCGGAAGC





ATAAAGTGTAAAGCCTAGGGTGCCTAA





TGAGTGAGCTAACTCACATTAATTGCG





TTGCGCTCACTGCCCGCTTTCCAGTCG





GGAAACCTGTCGTGCCAGCTGCATTAA





TGAATCGGCCAACGCGCGGGGAGAGGC





GGTTTGCGTATTGGGCGCTCTTCCGCT





TCCTCGCTCACTGACTCGCTGCGCTCG





GTCGTTCGGCTGCGGCGAGCGGTATCA





GCTCACTCAAAGGCGGTAATACGGTTA





TCCACAGAATCAGGGGATAACGCAGGA





AAGAACATGTGAGCAAAAGGCCAGCAA





AAGGCCAGGAACCGTAAAAAGGCCGCG





TTGCTGGCGTTTTTCCATAGGCTCCGC





CCCCCTGACGAGCATCACAAAAATCGA





CGCTCAAGTCAGAGGTGGCGAAACCCG





ACAGGACTATAAAGATACCAGGCGTTT





CCCCCTGGAAGCTCCCTCGTGCGCTCT





CCTGTTCCGACCCTGCCGCTTACCGGA





TACCTGTCCGCCTTTCTCCCTTCGGGA





AGCGTGGCGCTTTCTCATAGCTCACGC





TGTAGGTATCTCAGTTCGGTGTAGGTC





GTTCGCTCCAAGCTGGGCTGTGTGCAC





GAACCCCCCGTTCAGCCCGACCGCTGC





GCCTTATCCGGTAACTATCGTCTTGAG





TCCAACCCGGTAAGACACGACTTATCG





CCACTGGCAGCAGCCACTGGTAACAGG





ATTAGCAGAGCGAGGTATGTAGGCGGT





GCTACAGAGTTCTTGAAGTGGTGGCCT





AACTACGGCTACACTAGAAGAACAGTA





TTTGGTATCTGCGCTCTGCTGAAGCCA





GTTACCTTCGGAAAAAGAGTTGGTAGC





TCTTGATCCGGCAAACAAACCACCGCT





GGTAGCGGTGGTTTTTTTGTTTGCAAG





CAGCAGATTACGCGCAGAAAAAAAGGA





TCTCAAGAAGATCCTTTGATCTTTTCT





ACGGGGTCTGACACTCAGTGGAACGAA





AACTCACGTTAAGGGATTTTGGTCATG





AGATTATCAAAAAGGATCTTCACCTAG





ATCCTTTTAAATTAAAAATGAAGTTTT





AAATCAATCTAAAGTATATATGAGTAA





ACTTGGTCTGACAGTTACCAATGCTTA





ATCAGTGAGGCACCTATCTCAGCGATC





TGTCTATTTCGTTCATCCATAGTTGCC





TGACTCCCCGTCGTGTAGATAACTACG





ATACGGGAGGGCTTACCATCTGGCCCC





AGTGCTGCAATGATACCGCGAGACCCA





CGCTCACCGGCTCCAGATTTATCAGCA





ATAAACCAGCCAGCCGGAAGGGCCGAG





CGCAGAAGTGGTCCTGCAACTTTATCC





GCCTCCATCCAGTCTATTAATTGTTGC





CGGGAAGCTAGAGTAAGTAGTTCGCCA





GTTAATAGTTTGCGCAACGTTGTTGCC





ATTGCTACAGGCATCGTGGTGTCACGC





TCGTCGTTTGGTATGGCTTCATTCAGC





TCCGGTTCCCAACGATCAAGGCGAGTT





ACATGATCCCCCATGTTGTGCAAAAAA





GCGGTTAGCTCCTTCGGTCCTCCGATC





GTTGTCAGAAGTAAGTTGGCCGCAGTG





TTATCACTCATGGTTATGGCAGCACTG





CATAATTCTCTTACTGTCATGCCATCC





GTAAGATGCTTTTCTGTGACTGGTGAG





TACTCAACCAAGTCATTCTGAGAATAG





TGTATGCGGCGACCGAGTTGCTCTTGC





CCGGCGTCAATACGGGATAATACCGCG





CCACATAGCAGAACTTTAAAAGTGCTC





ATCATTGGAAAACGTTCTTCGGGGCGA





AAACTCTCAAGGATCTTACCGCTGTTG





AGATCCAGTTCGATGTAACCCACTCGT





GCACCCAACTGATCTTCAGCATCTTTT





ACTTTCACCAGCGTTTCTGGGTGAGCA





AAAACAGGAAGGCAAAATGCCGCAAAA





AAGGGAATAAGGGCGACACGGAAATGT





TGAATACTCATACTCTTCCTTTTTCAA





TATTATTGAAGCATTTATCAGGGTTAT





TGTCTCATGAGCGGATACATATTTGAA





TGTATTTAGAAAAATAAACAAATAGGG





GTTCCGCGCACATTTCCCCGAAAAGTG





CCACCTGACGTCGACGGATCGGGAGAT





CGATCTCCCGATCCCCTAGGGTCGACT





CTCAGTACAATCTGCTCTGATGCCGCA





TAGTTAAGCCAGTATCTGCTCCCTGCT





TGTGTGTTGGAGGTCGCTGAGTAGTGC





GCGAGCAAAATTTAAGCTACAACAAGG





CAAGGCTTGACCGACAATTGCATGAAG





AATCTGCTTAGGGTTAGGCGTTTTGCG





CTGCTTCGCGATGTACGGGCCAGATAT





ACGCGTTGACATTGATTATTGACTAGT





TATTAATAGTAATCAATTACGGGGTCA





TTAGTTCATAGCCCATATATGGAGTTC





CGCGTTACATAACTTACGGTAAATGGC





CCGCCTGGCTGACCGCCCAACGACCCC





CGCCCATTGACGTCAATAATGACGTAT





GTTCCCATAGTAACGCCAATAGGGACT





TTCCATTGACGTCAATGGGTGGAGTAT





TTACGGTAAACTGCCCACTTGGCAGTA





CATCAAGTGTATCATATGCCAAGTACG





CCCCCTATTGACGTCAATGACGGTAAA





TGGCCCGCCTGGCATTATGCCCAGTAC





ATGACCTTATGGGACTTTCCTACTTGG





CAGTACATCTACGTATTAGTCATCGCT





ATTACCATG






61
pC016
ATTGATTTAAAACTTCATTTTTAATTT
Backbone for




AAAAGGATCTAGGTGAAGATCCTTTTT
cloning LwCas13a




GATAATCTCATGACCAAAATCCCTTAA
guides under U6




CGTGAGTTTTCGTTCCACTGAGCGTCA
promoter




GACCCCGTAGAAAAGATCAAAGGATCT
Addgene #91906,




TCTTGAGATCCTTTTTTTCTGCGCGTA
the sequence of




ATCTGCTGCTTGCAAACAAAAAAACCA
which is accessible




CCGCTACCAGCGGTGGTTTGTTTGCCG
at addgene.org/




GATCAAGAGCTACCAACTCTTTTTCCG
91906/ and




AAGGTAACTGGCTTCAGCAGAGCGCAG
incorporated




ATACCAAATACTGTTCTTCTAGTGTAG
by reference




CCGTAGTTAGGCCACCACTTCAAGAAC





TCTGTAGCACCGCCTACATACCTCGCT





CTGCTAATCCTGTTACCAGTGGCTGCT





GCCAGTGGCGATAAGTCGTGTCTTACC





GGGTTGGACTCAAGACGATAGTTACCG





GATAAGGCGCAGCGGTCGGGCTGAACG





GGGGGTTCGTGCACACAGCCCAGCTTG





GAGCGAACGACCTACACCGAACTGAGA





TACCTACAGCGTGAGCTATGAGAAAGC





GCCACGCTTCCCGAAGGGAGAAAGGCG





GACAGGTATCCGGTAAGCGGCAGGGTC





GGAACAGGAGAGCGCACGAGGGAGCTT





CCAGGGGGAAACGCCTGGTATCTTTAT





AGTCCTGTCGGGTTTCGCCACCTCTGA





CTTGAGCGTCGATTTTTGTGATGCTCG





TCAGGGGGGCGGAGCCTATGGAAAAAC





GCCAGCAACGCGGCCTTTTTACGGTTC





CTGGCCTTTTGCTGGCCTTTTGCTCAG





CTAGCGAGGGCCTATTTCCCATGATTC





CTTCATATTTGCATATACGATACAAGG





CTGTTAGAGAGATAATTGGAATTAATT





TGACTGTAAACACAAAGATATTAGTAC





AAAATACGTGACGTAGAAAGTAATAAT





TTCTTGGGTAGTTTGCAGTTTTAAAAT





TATGTTTTAAAATGGACTATCATATGC





TTACCGTAACTTGAAAGTATTTCGATT





TCTTGGCTTTATATATCTTGTGGAAAG





GACGAAACACCGATTTAGACTACCCCA





AAAACGAAGGGGACTAAAACGGGTCTT





CGAGAAGACCTTTTTTTTGAATTCTGA





TGCGGTATTTTCTCCTTACGCATCTGT





GCGGTATTTCACACCGCATACGTCAAA





GCAACCATAGTACGCGCCCTGTAGCGG





CGCATTAAGCGCGGCGGGTGTGGTGGT





TACGCGCAGCGTGACCGCTACACTTGC





CAGCGCCTTAGCGCCCGCTCCTTTCGC





TTTCTTCCCTTCCTTTCTCGCCACGTT





CGCCGGCTTTCCCCGTCAAGCTCTAAA





TCGGGGGCTCCCTTTAGGGTTCCGATT





TAGTGCTTTACGGCACCTCGACCCCAA





AAAACTTGATTTGGGTGATGGTTCACG





TAGTGGGCCATCGCCCTGATAGACGGT





TTTTCGCCCTTTGACGTTGGAGTCCAC





GTTCTTTAATAGTGGACTCTTGTTCCA





AACTGGAACAACACTCAACTCTATCTC





GGGCTATTCTTTTGATTTATAAGGGAT





TTTGCCGATTTCGGTCTATTGGTTAAA





AAATGAGCTGATTTAACAAAAATTTAA





CGCGAATTTTAACAAAATATTAACGTT





TACAATTTTATGGTGCACTCTCAGTAC





AATCTGCTCTGATGCCGCATAGTTAAG





CCAGCCCCGACACCCGCCAACACCCGC





TGACGCGCCCTGACGGGCTTGTCTGCT





CCCGGCATCCGCTTACAGACAAGCTGT





GACCGTCTCCGGGAGCTGCATGTGTCA





GAGGTTTTCACCGTCATCACCGAAACG





CGCGAGACGAAAGGGCCTCGTGATACG





CCTATTTTTATAGGTTAATGTCATGAT





AATAATGGTTTCTTAGACGTCAGGTGG





CACTTTTCGGGGAAATGTGCGCGGAAC





CCCTATTTGTTTATTTTTCTAAATACA





TTCAAATATGTATCCGCTCATGAGACA





ATAACCCTGATAAATGCTTCAATAATA





TTGAAAAAGGAAGAGTATGAGTATTCA





ACATTTCCGTGTCGCCCTTATTCCCTT





TTTTGCGGCATTTTGCCTTCCTGTTTT





TGCTCACCCAGAAACGCTGGTGAAAGT





AAAAGATGCTGAAGATCAGTTGGGTGC





ACGAGTGGGTTACATCGAACTGGATCT





CAACAGCGGTAAGATCCTTGAGAGTTT





TCGCCCCGAAGAACGTTTTCCAATGAT





GAGCACTTTTAAAGTTCTGCTATGTGG





CGCGGTATTATCCCGTATTGACGCCGG





GCAAGAGCAACTCGGTCGCCGCATACA





CTATTCTCAGAATGACTTGGTTGAGTA





CTCACCAGTCACAGAAAAGCATCTTAC





GGATGGCATGACAGTAAGAGAATTATG





CAGTGCTGCCATAACCATGAGTGATAA





CACTGCGGCCAACTTACTTCTGACAAC





GATCGGAGGACCGAAGGAGCTAACCGC





TTTTTTGCACAACATGGGGGATCATGT





AACTCGCCTTGATCGTTGGGAACCGGA





GCTGAATGAAGCCATACCAAACGACGA





GCGTGACACCACGATGCCTGTAGCAAT





GGCAACAACGTTGCGCAAACTATTAAC





TGGCGAACTACTTACTCTAGCTTCCCG





GCAACAATTAATAGACTGGATGGAGGC





GGATAAAGTTGCAGGACCACTTCTGCG





CTCGGCCCTTCCGGCTGGCTGGTTTAT





TGCTGATAAATCTGGAGCCGGTGAGCG





TGGAAGCCGCGGTATCATTGCAGCACT





GGGGCCAGATGGTAAGCCCTCCCGTAT





CGTAGTTATCTACACGACGGGGAGTCA





GGCAACTATGGATGAACGAAATAGACA





GATCGCTGAGATAGGTGCCTCACTGAT





TAAGCATTGGTAACTGTCAGACCAAGT





TTACTCATATATACTTTAG






62
LVDP-CArG-RE-GPR
atggatagcactgagaacgtcatcaag
dsRED-Express2




cccttcatgcgcttcaaggtgcacatg
fragment source to




gagggctccgtgaacggccacgagttc
clone in GEMS




gagatcgagggcgagggcgagggcaag
Addgene #89762,




ccctacgagggcacccagaccgccaag
the sequence of




ctgcaggtgaccaagggcggccccctg
which is accessible




cccttcgcctgggacatcctgtccccc
at addgene.org/




cagttccagtacggctccaaggtgtac
89762/ and




gtgaagcaccccgccgacatccccgac
incorporated




tacaagaagctgtccttccccgagggc
by reference




ttcaagtgggagcgcgtgatgaacttc





gaggacggcggcgtggtgaccgtgacc





caggactcctccctgcaggacggcacc





ttcatctaccacgtgaagttcatcggc





gtgaacttcccctccgacggccccgta





atgcagaagaagactctgggctgggag





ccctccaccgagcgcctgtacccccgc





gacggcgtgctgaagggcgagatccac





aaggcgctgaagctgaagggcggcggc





cactacctggtggagttcaagtcaatc





tacatggccaagaagcccgtgaagctg





cccggctactactacgtggactccaag





ctggacatcacctcccacaacgaggac





tacaccgtggtggagcagtacgagcgc





gccgaggcccgccaccacctgttccag





tag






63
EGFP sequence
ATGGTGAGCAAGGGCGAGGAGCTGTTC





ACCGGGGTGGTGCCCATCCTGGTCGAG





CTGGATGGCGATGTAAATGGCCACAAG





TTCAGCGTGTCCGGCGAGGGCGAGGGC





GATGCCACCTACGGCAAGCTCACCCTG





AAGTTCATCTGCACCACCGGCAAGCTG





CCCGTGCCCTGGCCCACCCTCGTCACC





ACCCTCACCTACGGCGTGCAGTGCTTC





AGCCGCTACCCCGATCACATGAAGCAG





CACGATTTCTTCAAGTCCGCCATGCCC





GAAGGCTACGTCCAGGAGCGCACCATC





TTCTTCAAGGATGATGGCAATTACCGT





ACCCGCGCCGAGGTGAAGTTCGAGGGC





GATACCCTGGTGAATCGCATCGAGCTG





AAGGGCATCGATTTCAAGGAGGATGGC





AATATCCTGGGGCACAAGCTGGAGTAC





AATTACAATAGCCACAATGTCTATATC





ATGGCCGATAAGCAGAAGAATGGCATC





AAGGTGAATTTCAAGATCCGCCACAAT





ATCGAGGATGGCAGCGTGCAGCTCGCC





GATCACTACCAGCAGAATACCCCCATC





GGCGATGGCCCCGTGCTGCTGCCCGAT





AATCACTACCTGAGCACCCAGTCCGCC





CTGAGCAAAGATCCCAATGAGAAGCGC





GATCACATGGTCCTGCTGGAGTTCGTC





ACCGCCGCCGGGATCACTCTCGGCATG





GATGAGCTGTACAAG






64
m6A sensor linker
GCGGACTTACGACAGTTGCGTTACACC




sequence
CTTTCTCGACAAAACCTAACTTGCGCA





GAAAACATGCCAATCTCATCTTGGCTT






65
PEST domain
TTGCTTAGCCATGGCTTCCCGCCGGAG




sequence
GTGGAGGAGCAGGATGATGGCACGCTG





CCCATGTCTTGTGCCCAGGAGAGCGGG





ATGGACCGTCACCCTGCAGCCTGTGCT





TCTGCTAGGATCAATGTGTTAGATGCG






66
m6A sensor sequence
GACTTACGACAG






67
amino acid sequence
PHPVLEKLRSINNYNPKDFDWNLKHGR




of YTHDF2-YTH
VFIIKSYSEDDIHRSIKYNIWCSTEHG





NKRLDAAYRSMNGKGPVYLLFSVNGSG





HFCGVAEMKSAVDYNTCAGVWSQDKWK





GRFDVRWIFVKDVPNSQLRHIRLENNE





NKPVTNSRDTQEVPLEKAKQVLKIIAS





YKHTTSIFDDFSHYEKRQEEEESVKKE





RQGRGK






68
amino acid sequence
PHPVLEKLRSINNYNPKDFDWNLKHGR




of YTHDF2-
VFIIKSYSEDDIHRSIKYNIACSTEHG




YTH_W432A_W486A
NKRLDAAYRSMNGKGPVYLLFSVNGSG





HFCGVAEMKSAVDYNTCAGVASQDKWK





GRFDVRWIFVKDVPNSQLRHIRLENNE





NKPVTNSRDTQEVPLEKAKQVLKIIAS





YKHTTSIFDDFSHYEKRQEEEESVKKE





RQGRGK






69
amino acid sequence
GRVFIIKSYSEDDIHRSIKYNIWCSTE




of YTHDF2-YTHmut
HGNKRLDAAYRSMNGKGPVYLLFSVNG





SGHFCGVAEMKSAVDYNTCAGVWSQDK





WKGRFDVRWIFVKDVPNSQLRHIRLEN





NENKPVTNSRDTQEVPLEKAKQVLKII





ASYKHTTSIFDDFSHYEKRQEEEESVK





KERQGRGK






70
amino acid sequence
GRVFIIKSYSEDDIHRSIKYNIACSTE




of YTHDF2-YTHmut2
HGNKRLDAAYRSMNGKGPVYLLFSVNG





SGHFCGVAEMKSAVDYNTCAGVASQDK





WKGRFDVRWIFVKDVPNSQLRHIRLEN





NENKPVTNSRDTQEVPLEKAKQVLKII





ASYKHTTSIFDDFSHYEKRQEEEESVK





KERQGRGK






71
amino acid sequence
PHPVLEKLRSINNYNPKDFDWNLKHGR




of YTHDF2-YTH
VFIIKSYSENDIHRSIKYNIWCSTEHG




D422N
NKRLDAAYRSMNGKGPVYLLFSVNGSG





HFCGVAEMKSAVDYNTCAGVWSQDKWK





GRFDVRWIFVKDVPNSQLRHIRLENNE





NKPVTNSRDTQEVPLEKAKQVLKIIAS





YKHTTSIFDDFSHYEKRQEEEESVKKE





RQGRGK






72
amino acid sequence
HPVLEKLKAAHSYNPKEFEWNLKSGRV




of YTHDF1
FIIKSYSEDDIHRSIKYSIWCSTEHGN





KRLDSAFRCMSSKGPVYLLFSVNGSGH





FCGVAEMKSPVDYGTSAGVWSQDKWKG





KFDVQWIFVKDVPNNQLRHIRLENNDN





KPVTNSRDTQEVPLEKAKQVLKIISSY





KHTTSIFDDFAHYEKRQEEEEVVRKER





QSRNKQ









73
amino acid sequence
GRVFIIKSYSEDDIHRSIKYSIWCSTE




of YTHDF1mut
HGNKRLDSAFRCMSSKGPVYLLFSVNG





SGHFCGVAEMKSPVDYGTSAGVWSQDK





WKGKFDVQWIFVKDVPNNQLRHIRLEN





NDNKPVTNSRDTQEVPLEKAKQVLKII





SSYKHTTSIFDDFAHYEKRQEEEEVVR





KERQSRNKQ






74
amino acid sequence
HPVLEKLKAAHSYNPKEFEWNLKSGRV




of YTHDF1 D401N
FIIKSYSEDNIHRSIKYSIWCSTEHGN





KRLDSAFRCMSSKGPVYLLFSVNGSGH





FCGVAEMKSPVDYGTSAGVWSQDKWKG





KFDVQWIFVKDVPNNQLRHIRLENNDN





KPVTNSRDTQEVPLEKAKQVLKIISSY





KHTTSIFDDFAHYEKRQEEEEVVRKER





QSRNKQ






75
amino acid sequence
VHPVLEKLKAINNYNPKDFDWNLKNGR




of YTHDF3
VFIIKSYSEDDIHRSIKYSIWCSTEHG





NKRLDAAYRSLNGKGPLYLLFSVNGSG





HFCGVAEMKSVVDYNAYAGVWSQDKWK





GKFEVKWIFVKDVPNNQLRHIRLENND





NKPVTNSRDTQEVPLEKAKQVLKIIAT





FKHTTSIFDDFAHYEKRQEEEEAMRRE





RNRNKQ






76
amino acid sequence
SKLKYVLQDARFFLIKSNNHENVSLAK




of YTHDC1
AKGVWSTLPVNEKKLNLAFRSARSVIL





IFSVRESGKFQGFARLSSESHHGGSPI





HWVLPAGMSAKMLGGVFKIDWICRREL





PFTKSAHLTNPWNEHKPVKIGRDGQEI





ELECGTQLCLLFPPDESIDLYQVIHKM





RHK






77
amino acid sequence
PVRYFIMKSSNLRNLEISQQKGIWSTT




of YTHDC2
PSNERKLNRAFWESSIVYLVFSVQGSG





HFQGFSRMSSEIGREKSQDWGSAGLGG





VFKVEWIRKESLPFQFAHHLLNPWNDN





KKVQISRDGQELEPLVGEQLLQLWERL





PLGEKNTTD






78
amino acid sequence
MSSETGPVAVDPTLRRRIEPHEFEVFF




of rAPOBEC1
DPRELRKETCLLYEINWGGRHSIWRHT





SQNTNKHVEVNFIEKFTTERYFCPNTR





CSITWFLSWSPCGECSRAITEFLSRYP





HVTLFIYIARLYHHADPRNRQGLRDLI





SSGVTIQIMTEQESGYCWRNFVNYSPS





NEAHWPRYPHLWVRLYVLELYCIILGL





PPCLNILRRKQPQLTFFTIALQSCHYQ





RLPPHILWATGLK






79
amino acid sequence
MDSLLMNRRKFLYQFKNVRWAKGRRET




of hAICDA
YLCYVVKRRDSATSFSLDFGYLRNKNG





CHVELLFLRYISDWDLDPGRCYRVTWF





TSWSPCYDCARHVADFLRGNPNLSLRI





FTARLYFCEDRKAEPEGLRRLHRAGVQ





IAIMTFKDYFYCWNTFVENHERTFKAW





EGLHENSVRLSRQLRRILLPLYEVDDL





RDAFRTLGL






80
amino acid sequence
MEASPASGPRHLMDPHIFTSNFNNGIG




of hAPOBEC3A
RHKTYLCYEVERLDNGTSVKMDQHRGF





LHNQAKNLLCGFYGRHAELRFLDLVPS





LQLDPAQIYRVTWFISWSPCFSWGCAG





EVRAFLQENTHVRLRIFAARIYDYDPL





YKEALQMLRDAGAQVSIMTYDEFKHCW





DTFVDHQGCPFQPWDGLDEHSQALSGR





LRAILQNQGN






81
amino acid sequence
QLHLPQVLADAVSRLVLGKFGDLTDNF




of catalytic domain
SSPHARRKVLAGVVMTTGTDVKDAKVI




of ADAR2
SVSTGTKCINGEYMSDRGLALNDCHAE





IISRRSLLRFLYTQLELYLNNKDDQKR





SIFQKSERGGFRLKENVQFHLYISTSP





CGDARIFSPHEPILEEPADRHPNRKAR





GQLRTKIESGQGTIPVRSNASIQTWDG





VLQGERLLTMSCSDKIARWNVVGIQGS





LLSIFVEPIYFSSIILGSLYHGDHLSR





AMYQRISNIEDLPPLYTLNKPLLSGIS





NAEARQPGKAPNFSVNWTVGDSAIEVI





NATTGKDELGRASRLCKHALYCRWMRV





HGKVPSHLLRSKITKPNVYHESKLAAK





EYQAAKARLFTAFIKAGLGAWVEKPTE





QDQFSLT






82

SGSETPGTSESATPE






83

SGSETPGTSESATPES






84

(GGGGS)3






85

((GGGGS)10)






86

(A(EAAAK)3A)






87

E. coli codon

ATGAGCAGCGAAACCGGTCCGGTGGCG




optimized APOBEC1-
GTTGACCCGACCCTGCGTCGTCGTATT




YTH for protein
GAGCCGCACGAGTTCGAAGTGTTCTTT




purification
GATCCGCGTGAGCTGCGTAAGGAAACC





TGCCTGCTGTACGAAATTAACTGGGGT





GGCCGTCACAGCATCTGGCGTCACACC





AGCCAGAACACCAACAAGCACGTTGAG





GTGAACTTCATCGAAAAATTTACCACC





GAGCGTTACTTCTGCCCGAACACCCGT





TGCAGCATTACCTGGTTTCTGAGCTGG





AGCCCGTGCGGTGAATGCAGCCGTGCG





ATCACCGAGTTCCTGAGCCGTTATCCG





CACGTTACCCTGTTTATCTACATTGCG





CGTCTGTATCACCACGCGGACCCGCGT





AACCGTCAAGGTCTGCGTGATCTGATC





AGCAGCGGCGTGACCATCCAGATTATG





ACCGAGCAAGAAAGCGGTTACTGCTGG





CGTAACTTCGTTAACTATAGCCCGAGC





AACGAAGCGCATTGGCCGCGTTACCCG





CACCTGTGGGTGCGTCTGTACGTTCTG





GAGCTGTATTGCATCATTCTGGGCCTG





CCGCCGTGCCTGAACATTCTGCGTCGT





AAGCAGCCGCAACTGACCTTCTTTACC





ATCGCGCTGCAGAGCTGCCACTACCAA





CGTCTGCCGCCGCACATTCTGTGGGCG





ACCGGTCTGAAGAGCGGCAGCGAAACC





CCGGGTACCAGCGAAAGCGCGACCCCG





GAGCCGCACCCGGTGCTGGAGAAACTG





CGTAGCATCAACAACTATAACCCGAAG





GACTTCGATTGGAACCTGAAACACGGT





CGTGTTTTTATCATTAAGAGCTACAGC





GAAGACGATATCCACCGTAGCATTAAA





TATAACATCTGGTGCAGCACCGAGCAC





GGCAACAAGCGTCTGGACGCGGCGTAC





CGTAGCATGAACGGTAAAGGCCCGGTG





TATCTGCTGTTCAGCGTTAACGGTAGC





GGCCACTTTTGCGGTGTGGCGGAAATG





AAAAGCGCGGTTGATTACAACACCTGC





GCGGGTGTGTGGAGCCAGGACAAGTGG





AAAGGCCGTTTCGATGTTCGTTGGATT





TTTGTGAAGGACGTTCCGAACAGCCAA





CTGCGTCACATCCGTCTGGAGAACAAC





GAAAACAAACCGGTGACCAACAGCCGT





GATACCCAGGAAGTGCCGCTGGAAAAG





GCGAAACAAGTTCTGAAGATCATTGCG





AGCTACAAACACACCACCAGCATCTTC





GACGATTTTAGCCACTATGAGAAGCGT





CAGGAAGAGGAAGAGAGCGTGAAGAAG





GAGCGTCAAGGTCGTGGCAAACTGGAG





TACCCGTATGACGTTCCGGATTATGCG





TAAATTGGAAGTGGATAA






88

E. coli codon

ATGAGCAGCGAAACCGGTCCGGTGGCG




optimized APOBEC1-
GTTGACCCGACCCTGCGTCGTCGTATT




YTHmut for protein
GAGCCGCACGAGTTCGAAGTGTTCTTT




purification
GATCCGCGTGAGCTGCGTAAGGAAACC





TGCCTGCTGTACGAAATTAACTGGGGT





GGCCGTCACAGCATCTGGCGTCACACC





AGCCAGAACACCAACAAGCACGTTGAG





GTGAACTTCATCGAAAAATTTACCACC





GAGCGTTACTTCTGCCCGAACACCCGT





TGCAGCATTACCTGGTTTCTGAGCTGG





AGCCCGTGCGGTGAATGCAGCCGTGCG





ATCACCGAGTTCCTGAGCCGTTATCCG





CACGTTACCCTGTTTATCTACATTGCG





CGTCTGTATCACCACGCGGACCCGCGT





AACCGTCAAGGTCTGCGTGATCTGATC





AGCAGCGGCGTGACCATCCAGATTATG





ACCGAGCAAGAAAGCGGTTACTGCTGG





CGTAACTTCGTTAACTATAGCCCGAGC





AACGAAGCGCATTGGCCGCGTTACCCG





CACCTGTGGGTGCGTCTGTACGTTCTG





GAGCTGTATTGCATCATTCTGGGCCTG





CCGCCGTGCCTGAACATTCTGCGTCGT





AAGCAGCCGCAACTGACCTTCTTTACC





ATCGCGCTGCAGAGCTGCCACTACCAA





CGTCTGCCGCCGCACATTCTGTGGGCG





ACCGGTCTGAAGAGCGGCAGCGAAACC





CCGGGTACCAGCGAAAGCGCGACCCCG





GAGGGTCGTGTTTTTATCATTAAGAGC





TACAGCGAAGACGATATCCACCGTAGC





ATTAAATATAACATCTGGTGCAGCACC





GAGCACGGCAACAAGCGTCTGGACGCG





GCGTACCGTAGCATGAACGGTAAAGGC





CCGGTGTATCTGCTGTTCAGCGTTAAC





GGTAGCGGCCACTTTTGCGGTGTGGCG





GAAATGAAAAGCGCGGTTGATTACAAC





ACCTGCGCGGGTGTGTGGAGCCAGGAC





AAGTGGAAAGGCCGTTTCGATGTTCGT





TGGATTTTTGTGAAGGACGTTCCGAAC





AGCCAACTGCGTCACATCCGTCTGGAG





AACAACGAAAACAAACCGGTGACCAAC





AGCCGTGATACCCAGGAAGTGCCGCTG





GAAAAGGCGAAACAAGTTCTGAAGATC





ATTGCGAGCTACAAACACACCACCAGC





ATCTTCGACGATTTTAGCCACTATGAG





AAGCGTCAGGAAGAGGAAGAGAGCGTG





AAGAAGGAGCGTCAAGGTCGTGGCAAA





CTGGAGTACCCGTATGACGTTCCGGAT





TATGCGTAAATTGGAAGTGGATAA






89

PKKKRKV






90

LPPLERLTL






91

MDPVVVLGLCLSCLLLLSLWKQSYGGG






92

METDTLLLWVLLLWVPGSTGD






93

EQKLISEEDL






94

GKPIPNPLLGLDST






95

IPNPLLGLD






96

DYKDDDDK






97

DYKDHDGDYKDHDIDYKDDDDK






98
DHFR domain
ISLIAALAVDHVIGMETVMPWNLPADL





AWFKRNTLNKPVIMGRHTWESIGRPLP





GRKNIILSSQPSTDDRVTWVKSVDEAI





AACGDVPEIMVIGGGRVYEQFLPKAQK





LYLTHIDAEVEGDTHFPDYEPDDWESV





FSEFHDADAQNSHSYCFEILERR






99

GACUUAUGACAG






100

GACUUACGACAG






101

GGACTTACGACAGTT






102

GGCUUACGACAG






103

GACUUACGAGAG






104

HHHHHH






105
Construct comprising
gtcgacggatcgggagatctcccgatc




a nucleic acid
ccctatggtgcactctcagtacaatct




encoding GFP, a m6A
gctctgatgccgcatagttaagccagt




reporter sequence and
atctgctccctgcttgtgtgttggagg




DHFR; and a nucleic
tcgctgagtagtgcgcgagcaaaattt




acid encoding
aagctacaacaaggcaaggcttgaccg




APOBEC1-YTH (5′-3′)
acaattgcatgaagaatctgcttaggg





ttaggcgttttgcgctgcttcgcgatg





tacgggccagatatacgcgttgacatt





gattattgactagttattaatagtaat





caattacggggtcattagttcatagcc





catatatggagttccgcgttacataac





ttacggtaaatggcccgcctggctgac





cgcccaacgacccccgcccattgacgt





caataatgacgtatgttcccatagtaa





cgccaatagggactttccattgacgtc





aatgggtggagtatttacggtaaactg





cccacttggcagtacatcaagtgtatc





atatgccaagtacgccccctattgacg





tcaatgacggtaaatggcccgcctggc





attatgcccagtacatgaccttatggg





actttcctacttggcagtacatctacg





tattagtcatcgctattaccatggtga





tgcggttttggcagtacatcaatgggc





gtggatagcggtttgactcacggggat





ttccaagtctccaccccattgacgtca





atgggagtttgttttggcaccaaaatc





aacgggactttccaaaatgtogtaaca





actccgccccattgacgcaaatgggcg





gtaggcgtgtacggtgggaggtctata





taagcagagctggtttagtgaaccgtc





agatccgctagagatccgcggccgcgc





tagcgtttaaacgggccctctagagcc





gccatggtgagcaagggcgaggagctg





ttcaccggggtggtgcccatcctggtc





gagctggatggcgatgtaaatggccac





aagttcagcgtgtccggcgagggcgag





ggcgatgccacctacggcaagctcacc





ctgaagttcatctgcaccaccggcaag





ctgcccgtgccctggcccaccctcgtc





accaccctcacctacggcgtgcagtgc





ttcagccgctaccccgatcacatgaag





cagcacgatttcttcaagtccgccatg





cccgaaggctacgtccaggagcgcacc





atcttcttcaaggatgatggcaattac





cgtacccgcgccgaggtgaagttcgag





ggcgataccctggtgaatcgcatcgag





ctgaagggcatcgatttcaaggaggat





ggcaatatcctggggcacaagctggag





tacaattacaatagccacaatgtctat





atcatggccgataagcagaagaatggc





atcaaggtgaatttcaagatccgccac





aatatcgaggatggcagcgtgcagctc





gccgatcactaccagcagaataccccc





atcggcgatggccccgtgctgctgccc





gataatcactacctgagcacccagtcc





gccctgagcaaagatcccaatgagaag





cgcgatcacatggtcctgctggagttc





gtcaccgccgccgggatcactctcggc





atggatgagctgtacaaggcggactta





cgacagttgcgttacaccctttctcga





caaaacctaacttgcgcagaaaacatg





ccaatctcatcttggcttatcagtctg





attgcggcgttagcggtagatcacgtt





atcggcatggaaaccgtcatgccgtgg





aacctgcctgccgatctcgcctggttt





aaacgcaacaccttaaataaacccgtg





attatgggccgccatacctgggaatca





atcggtcgtccgttgccaggacgcaaa





aatattatcctcagcagtcaaccgagt





acggacgatcgcgtaacgtgggtgaag





tcggtggatgaagccatcgcggcgtgt





ggtgacgtaccagaaatcatggttatt





ggcggcggtcgcgtttatgaacagttc





ttgccaaaagcgcaaaaactgtatctg





acgcatatcgacgcagaagtggaaggc





gacacccatttcccggattacgagccg





gatgactgggaatcggtattcagcgaa





ttccacgatgctgatgcgcagaactct





cacagctattgctttgagattctggag





cggcgataagcctcattgtgcattctc





tcgagtacccctacgacgtgcccgact





acgcctgagggacccgacaggcccgaa





ggaatagaagaagaaggtggagagaga





gacagagacagatccattcgattagtg





aacggatcggcactgcgtgcgccaatt





ctgcagacaaatggcagtattcatcca





caattttaaaagaaaaggggggattgg





ggggtacagtgcaggggaaagaatagt





agacataatagcaacagacatacaaac





taaagaattacaaaaacaaattacaaa





aattcaaaattttcgggtttattacag





ggacagcagagatccagtttggttacc





agtgtgatggatatctgcagaattcgc





ccttggatccgaattcctgcagccccg





actttcacttttctctatcactgatag





ggagtggtaaactcgactttcactttt





ctctatcactgatagggagtggtaaac





tcgactttcacttttctctatcactga





tagggagtggtaaactcgactttcact





tttctctatcactgatagggagtggta





aactcgactttcacttttctctatcac





tgatagggagtggtaaactcgactttc





acttttctctatcactgatagggagtg





gtaaactcgactttcacttttctctat





cactgatagggagtggtaaactcgagg





gggatccactagcatgaagggcgaatt





ccagcacactggtaacccgtgtcggct





ccagatctggcctccgcgccgggtttt





ggcgcctcccgcgggcgcccccctcct





cacggcgagccgcgttgacattgatta





ttgactaggcttttgcaaaaagctttg





caaagatggataaagttttaaacagag





aggaatctttgcagctaatggaccttc





taggtcttgaaaggagtgggaattggc





tccggtgcccgtcagtgggcagagcgc





acatcgcccacagtccccgagaagttg





gggggaggggtcggcaattgaaccggt





gcctagagaaggtggcgcggggtaaac





tgggaaagtgatgtcgtgtactggctc





cgcctttttcccgagggtgggggagaa





ccgtatataagtgcagtagtcgccgtg





aacgttctttttcgcaacgggtttgcc





gccagaacacaggtaagtgccgtgtgt





ggttcccgcgggcctggcctctttacg





ggttatggcccttgcgtgccttgaatt





acttccacctggctgcagtacgtgatt





cttgatcccgagcttcgggttggaagt





gggtgggagagttcgaggccttgcgct





taaggagccccttcgcctcgtgcttga





gttgaggcctggcctgggcgctggggc





cgccgcgtgcgaatctggtggcacctt





cgcgcctgtctcgctgctttcgataag





tctctagccatttaaaatttttgatga





cctgctgcgacgctttttttctggcaa





gatagtcttgtaaatgcgggccaagat





ctgcacactggtatttcggtttttggg





gccgcgggcggcgacggggcccgtgcg





tcccagcgcacatgttcggcgaggcgg





ggcctgcgagcgcggccaccgagaatc





ggacgggggtagtctcaagctggccgg





cctgctctggtgcctggcctcgcgccg





ccgtgtatcgccccgccctgggcggca





aggctggcccggtcggcaccagttgcg





tgagcggaaagatggccgcttcccggc





cctgctgcagggagctcaaaatggagg





acgcggcgctcgggagagcgggcgggt





gagtcacccacacaaaggaaaagggcc





tttccgtcctcagccgtcgcttcatgt





gactccacggagtaccgggcgccgtcc





aggcacctcgattagttctcgagcttt





tggagtacgtcgtctttaggttggggg





gaggggttttatgcgatggagtttccc





cacactgagtgggtggagactgaagtt





aggccagcttggcacttgatgtaattc





tccttggaatttgccctttttgagttt





ggatcttggttcattctcaagcctcag





acagtggttcaaagtttttttcttcca





tttcaggtgtcgtgaggaattagcttg





gtactaatacgactcactatagggaga





cccaagctggctaggtaagcttggtac





cgagctcggatccactagtccagtgtg





gtggaattctgcagatatccagcacag





tggggtttagtgaaccgtcagatccgc





tagagatccgcggccgctaatacgact





cactatagggagagccgccaccatgag





ctcagagactggcccagtggctgtgga





ccccacattgagacggcggatcgagcc





ccatgagtttgaggtattcttcgatcc





gagagagctccgcaaggagacctgcct





gctttacgaaattaattgggggggccg





gcactccatttggcgacatacatcaca





gaacactaacaagcacgtcgaagtcaa





cttcatcgagaagttcacgacagaaag





atatttctgtccgaacacaaggtgcag





cattacctggtttctcagctggagccc





atgcggcgaatgtagtagggccatcac





tgaattcctgtcaaggtatccccacgt





cactctgtttatttacatcgcaaggct





gtaccaccacgctgacccccgcaatcg





acaaggcctgcgggatttgatctcttc





aggtgtgactatccaaattatgactga





gcaggagtcaggatactgctggagaaa





ctttgtgaattatagcccgagtaatga





agcccactggcctaggtatccccatct





gtgggtacgactgtacgttcttgaact





gtactgcatcatactgggcctgcctcc





ttgtctcaacattctgagaaggaagca





gccacagctgacattctttaccatcgc





tcttcagtcttgtcattaccagcgact





gcccccacacattctctgggccaccgg





gttgaaaagcggcagcgagactcccgg





gacctcagagtccgccacaccagaacc





ccacccagtgttggagaagcttcggtc





cattaataactataaccccaaagattt





tgactggaatctgaaacatggccgggt





tttcatcattaagagctactctgagga





cgatattcaccgttccattaagtataa





tatttggtgcagcacagagcatggtaa





caagagactggatgctgcttatcgttc





catgaacgggaaaggccccgtttactt





acttttcagtgtcaacggcagtggaca





cttctgtggcgtggcagaaatgaaatc





tgctgtggactacaacacatgtgcagg





tgtgtggtcccaggacaaatggaaggg





tcgttttgatgtcaggtggatttttgt





gaaggacgttcccaatagccaactgcg





acacattcgcctagagaacaacgagaa





taaaccagtgaccaactctagggacac





tcaggaagtgcctctggaaaaggctaa





gcaggtgttgaaaattatagccagcta





caagcacaccacttccatttttgatga





cttctcacactatgagaaacgccaaga





ggaagaagaaagtgttaaaaaggaacg





tcaaggtcgtgggaaactcgagtaccc





ctacgacgtgcccgactacgcctgagt





ttaaaatcgatggtacactcgaggtta





acgaattctaccgggtaggggaggcgc





ttttcccaaggcagtctggagcatgcg





ctttagcagccccgctgggcacttggc





gctacacaagtggcctctggcctcgca





cacattccacatccaccggtaggcgcc





aaccggctccgttctttggtggcccct





tcgcgccaccttctactcctcccctag





tcaggaagttcccccccgccccgcagc





tcgcgtcgtgcaggacgtgacaaatgg





aagtagcacgtctcactagtctcgtgc





agatggacagcaccgctgagcaatgga





agcgggtaggcctttggggcagcggcc





aatagcagctttgctccttcgctttct





gggctcagaggctgggaagggggggtc





cgggggcgggctcaggggcgggctcag





gggcggggcgggcgcccgaaggtcctc





cggaggcccggcattctgcacgcttca





aaagcgcacgtctgccgcgctgttctc





ctcttcctcatctccgggcctttcgac





ctgcatcccgccaccatgaccgagtac





aagcccacggtgcgcctcgccacccgc





gacgacgtccccagggccgtacgcacc





ctcgccgccgegttcgccgactacccc





gccacgegccacaccgtcgatccggac





cgccacatcgaggggtcaccgagctgc





aagaactcttcctcacgcgcgtcgggc





tcgacatcggcaaggtgtgggtcgcgg





acgacggcgccgggtggcggtctggac





cacgccggagagcgtcgaagcgggggc





ggtgttcgccgagatcggcccgcgcat





ggccgagttgagcggttcccggctggc





cgcgcagcaacagatggaaggcctcct





ggcgccgcaccggcccaaggagcccgc





gtggttcctggccaccgtcggagtctc





gcccgaccaccagggcaagggtctggg





cagcgccgtcgtgctccccggagtgga





ggcggccgagcgcgccggggtgcccgc





cttcctggagacctccgcgccccgcaa





cctccccttctacgagcggctcggctt





caccgtcaccgccgacgtcgaggtgcc





cgaaggaccgcgcacctggtgcatgac





ccgcaagcccggtgccggttccggcgc





aacaaacttctctctgctgaaacaagc





cggagatgtcgaagagaatcctggacc





gatggctagattagataaaagtaaagt





gattaacagcgcattagagctgcttaa





tgaggtcggaatcgaaggtttaacaac





ccgtaaactegcccagaagctaggtgt





agagcagcctacattgtattggcatgt





aaaaaataagcgggctttgctcgacgc





cttagccattgagatgttagataggca





ccatactcacttttgccctttagaagg





ggaaagctggcaagattttttacgtaa





taacgctaaaagttttagatgtgcttt





actaagtcatcgcgatggagcaaaagt





acatttaggtacacggcctacagaaaa





acagtatgaaactctcgaaaatcaatt





agcctttttatgccaacaaggtttttc





actagagaatgcattatatgcactcag





cgctgtggggcattttactttaggttg





cgtattggaagatcaagagcatcaagt





cgctaaagaagaaagggaaacacctac





tactgatagtatgccgccattattacg





acaagctatcgaattatttgatcacca





aggtgcagagccagccttcttattcgg





ccttgaattgatcatatgcggattaga





aaaacaacttaaatgtgaaagtgggtc





gccaaaaaagaagagaaaggtcgacgg





cggtggtgctttgtctcctcagcactc





tgctgtcactcaaggaagtatcatcaa





gaacaaggagggcatggatgctaagtc





actaactgcctggtcccggacactggt





gaccttcaaggatgtatttgtggactt





caccagggaggagtggaagctgctgga





cactgctcagcagatcgtgtacagaaa





tgtgatgctggagaactataagaacct





ggtttccttgggttatcagcttactaa





gccagatgtgatcctccggttggagaa





gggagaagagccctggctggtgtaaag





tagatgccgaccgaacaagagctgatt





tcgagaacgcctcagccagcaactcgc





gcgagcctagcaaggcaaatgcgagag





aacggccttacgcttggtggcacagtt





ctcgtccacagttcgctaagctcgctc





ggctgggtcgcgggagggccggtcgca





gtgattcaggcccttctggattgtgtt





ggtccccagggcacgattgtcatgccc





acgcactcgggtgatctgactgatccc





gcagattggagatcgccgcccgtgcct





gccgattgggtgcagatccgtcgagtt





aacaaaagaaaaggggggactggaagg





gctaattcactcccaacgaagacaaga





tatcataacttcgtatagcatacatta





tacgaagttatcggctagctggtccgg





actgtactgggtctctctggttagacc





agatctgagcctgggagctctctggct





aactagggaacccactgcttaagcctc





aataaagcttgccttgagtgcttcaag





tagtgtgtgcccgtctgttgtgtgact





ctggtaactagagatccctcagaccct





tttagtcagtgtggaaaatctctagca





gggcccgtttaaacccgctgatcagcc





tcgactgtgccttctagttgccagcca





tctgttgtttgcccctcccccgtgcct





tccttgaccctggaaggtgccactccc





actgtcctttcctaataaaatgaggaa





attgcatcgcattgtctgagtaggtgt





cattctattctggggggtggggtgggg





caggacagcaagggggaggattgggaa





gacaatagcaggcatgctggggatgcg





gtgggctctatggcttctgaggcggaa





agaaccagctggggctctagggggtat





ccccacgcgccctgtagcggcgcatta





agcgcggcgggtgtggtggttacgcgc





agcgtgaccgctacacttgccagcgcc





ctagcgcccgctcctttcgctttcttc





ccttcctttctcgccacgttcgccggc





tttccccgtcaagctctaaatcggggg





ctccctttagggttccgatttagtgct





ttacggcacctcgaccccaaaaaactt





gattagggtgatggttcacgtagtggg





ccatcgccctgatagacggtttttcgc





cctttgacgttggagtccacgttcttt





aatagtggactcttgttccaaactgga





acaacactcaaccctatctcggtctat





tcttttgatttataagggattttgccg





atttcggcctattggttaaaaaatgag





ctgatttaacaaaaatttaacgcgaat





taattctgtggaatgtgtgtcagttag





ggtgtggaaagtccccaggctccccag





caggcagaagtatgcaaagcatgcatc





tcaattagtcagcaaccaggtgtggaa





agtccccaggctccccagcaggcagaa





gtatgcaaagcatgcatctcaattagt





cagcaaccatagtcccgcccctaactc





cgcccatcccgcccctaactccgccca





gttccgcccattetccgccccatggct





gactaattttttttatttatgcagagg





ccgaggccgcctctgcctctgagctat





tccagaagtagtgaggaggcttttttg





gaggcctaggcttttgcaaaaagctcc





cgggagcttgtatatccattttcggat





ctgatcagcacgtgttgacaattaatc





atcggcatagtatatcggcatagtata





atacgacaaggtgaggaactaaaccat





ggccaagttgaccagtgccgttccggt





gctcaccgcgcgcgacgtcgccggagc





ggtcgagttctggaccgaccggctcgg





gttctcccgggacttcgtggaggacga





cttcgccggtgtggtccgggacgacgt





gaccctgttcatcagcgcggtccagga





ccaggtggtgccggacaacaccctggc





ctgggtgtgggtgcgcggcctggacga





gctgtacgccgagtggtcggaggtcgt





gtccacgaacttccgggacgcctccgg





gccggccatgaccgagatcggcgagca





gccgtggggggggagttcgccctgcgc





gacccggccggcaactgcgtgcacttc





gtggccgaggagcaggactgacacgtg





ctacgagatttcgattccaccgccgcc





ttctatgaaaggttgggcttcggaatc





gttttccgggacgccggctggatgatc





ctccagcgcggggatctcatgctggag





ttcttcgcccaccccaacttgtttatt





gcagcttataatggttacaaataaagc





aatagcatcacaaatttcacaaataaa





gcatttttttcactgcattctagttgt





ggtttgtccaaactcatcaatgtatot





tatcatgtctgtataccgtcgacctct





agctagagcttggcgtaatcatggtca





tagctgtttcctgtgtgaaattgttat





ccgctcacaattccacacaacatacga





gccggaagcataaagtgtaaagcctgg





ggtgcctaatgagtgagctaactcaca





ttaattgcgttgcgctcactgcccgct





ttccagtcgggaaacctgtcgtgccag





ctgcattaatgaatcggccaacgcgcg





gggagaggcggtttgcgtattgggcgc





tcttccgcttcctcgctcactgactcg





ctgcgctcggtcgttcggctgcggcga





gcggtatcagctcactcaaaggcggta





atacggttatccacagaatcaggggat





aacgcaggaaagaacatgtgagcaaaa





ggccagcaaaaggccaggaaccgtaaa





aaggccgcgttgctggcgtttttccat





aggctccgcccccctgacgagcatcac





aaaaatcgacgctcaagtcagaggtgg





cgaaacccgacaggactataaagatac





caggcgtttccccctggaagctccctc





gtgcgctctcctgttccgaccctgccg





cttaccggatacctgtccgcctttctc





ccttcgggaagcgtggcgctttctcat





agctcacgctgtaggtatctcagttcg





gtgtaggtcgttcgctccaagctgggc





tgtgtgcacgaaccccccgttcagccc





gaccgctgcgccttatccggtaactat





cgtcttgagtccaacccggtaagacac





gacttatcgccactggcagcagccact





ggtaacaggattagcagagcgaggtat





gtaggcggtgctacagagttcttgaag





tggtggcctaactacggctacactaga





agaacagtatttggtatctgcgctctg





ctgaagccagttaccttcggaaaaaga





gttggtagctcttgatccggcaaacaa





accaccgctggtagcggtggttttttt





gtttgcaagcagcagattacgcgcaga





aaaaaaggatctcaagaagatcctttg





atcttttctacggggtctgacgctcag





tggaacgaaaactcacgttaagggatt





ttggtcatgagattatcaaaaaggatc





ttcacctagatccttttaaattaaaaa





tgaagttttaaatcaatctaaagtata





tatgagtaaacttggtctgacagttac





caatgcttaatcagtgaggcacctatc





tcagcgatctgtctatttcgttcatcc





atagttgcctgactccccgtcgtgtag





ataactacgatacgggagggcttacca





tctggccccagtgctgcaatgataccg





cgagacccacgctcaccggctccagat





ttatcagcaataaaccagccagccgga





agggccgagcgcagaagtggtcctgca





actttatccgcctccatccagtctatt





aattgttgccgggaagctagagtaagt





agttcgccagttaatagtttgcgcaac





gttgttgccattgctacaggcatcgtg





gtgtcacgctcgtcgtttggtatggct





tcattcagctccggttcccaacgatca





aggcgagttacatgatcccccatgttg





tgcaaaaaagcggttagctccttcggt





cctccgatcgttgtcagaagtaagttg





gccgcagtgttatcactcatggttatg





gcagcactgcataattctcttactgtc





atgccatccgtaagatgcttttctgtg





actggtgagtactcaaccaagtcattc





tgagaatagtgtatgcggcgaccgagt





tgctcttgcccggcgtcaatacgggat





aataccgcgccacatagcagaacttta





aaagtgctcatcattggaaaacgttct





tcggggcgaaaactctcaaggatctta





ccgctgttgagatccagttcgatgtaa





cccactcgtgcacccaactgatcttca





gcatcttttactttcaccagcgtttct





gggtgagcaaaaacaggaaggcaaaat





gccgcaaaaaagggaataagggcgaca





cggaaatgttgaatactcatactcttc





ctttttcaatattattgaagcatttat





cagggttattgtctcatgagcggatac





atatttgaatgtatttagaaaaataaa





caaataggggttccgcgcacatttccc





cgaaaagtgccacctgac






106
Construct comprising
agggagtggtaaactcgactttcactt




a nucleic acid
ttctctatcactgatagggagtggtaa




sequence encoding
actcgactttcacttttctctatcact




GFP, a m6A reporter
gatagggagtggtaaactcgactttca




sequence, and DHFR;
cttttctctatcactgatagggagtgg




a nucleic acid
taaactcgactttcacttttctctatc




sequence encoding
actgatagggagtggtaaactcgaggg




APOBEC1-YTH (5′-3′);
ggatccactagcatgaagggcgaattc




and a nucleic acid
cagcacactggtaacccgtgtcggctc




sequence encoding
cagatctggcctccgcgccgggttttg




dsRed
gcgcctcccgcgggcgcccccctcctc





acggcgagccgcgttgacattgattat





tgactaggcttttgcaaaaagctttgc





aaagatggataaagttttaaacagaga





ggaatctttgcagctaatggaccttct





aggtcttgaaaggagtgggaattggct





ccggtgcccgtcagtgggcagagcgca





catcgcccacagtccccgagaagttgg





ggggaggggtcggcaattgaaccggtg





cctagagaaggtggcgcggggtaaact





gggaaagtgatgtcgtgtactggctcc





gcctttttcccgagggtgggggagaac





cgtatataagtgcagtagtcgccgtga





acgttctttttcgcaacgggtttgccg





ccagaacacaggtaagtgccgtgtgtg





gttcccgcgggcctggcctctttacgg





gttatggcccttgcgtgccttgaatta





cttccacctggctgcagtacgtgattc





ttgatcccgagcttcgggttggaagtg





gggggagagttcgaggccttgcgctta





aggagccccttcgcctcgtgcttgagt





tgaggcctggcctgggcgctggggccg





ccgcgtgcgaatctggtggcaccttcg





cgcctgtctcgctgctttcgataagtc





tctagccatttaaaatttttgatgacc





tgctgcgacgctttttttctggcaaga





tagtcttgtaaatgcgggccaagatct





gcacactggtatttcggtttttggggc





cgcgggcggcgacggggcccgtgcgtc





ccagcgcacatgttcggcgaggcgggg





cctgcgagcgcggccaccgagaatcgg





acgggggtagtctcaagctggccggcc





tgctctggtgcctggcctcgcgccgcc





gtgtatcgccccgccctgggcggcaag





gctggcccggtcggcaccagttgcgtg





agcggaaagatggccgcttcccggccc





tgctgcagggagctcaaaatggaggac





gcggcgctcgggagagcggggggtgag





tcacccacacaaaggaaaagggccttt





ccgtcctcagccgtcgcttcatgtgac





tccacggagtaccgggcgccgtccagg





cacctcgattagttctcgagcttttgg





agtacgtcgtctttaggttggggggag





gggttttatgcgatggagtttccccac





actgagtgggtggagactgaagttagg





ccagcttggcacttgatgtaattctcc





ttggaatttgccctttttgagtttgga





tcttggttcattctcaagcctcagaca





gtggttcaaagtttttttcttccattt





caggtgtcgtgaggaattagcttggta





ctaatacgactcactatagggagaccc





aagctggctaggtaagcttggtaccga





gctcggatccactagtccagtgtggtg





gaattctgcagatatccagcacagtgg





ggtttagtgaaccgtcagatccgctag





agatccgcggccgctaatacgactcac





tatagggagagccgccaccatgagctc





agagactggcccagtggctgtggaccc





cacattgagacggcggatcgagcccca





tgagtttgaggtattcttcgatccgag





agagctccgcaaggagacctgcctgct





ttacgaaattaattgggggggccggca





ctccatttggcgacatacatcacagaa





cactaacaagcacgtcgaagtcaactt





catcgagaagttcacgacagaaagata





tttctgtccgaacacaaggtgcagcat





tacctggtttctcagctggagcccatg





cggcgaatgtagtagggccatcactga





attcctgtcaaggtatccccacgtcac





tctgtttatttacatcgcaaggctgta





ccaccacgctgacccccgcaatcgaca





aggcctgcgggatttgatctcttcagg





tgtgactatccaaattatgactgagca





ggagtcaggatactgctggagaaactt





tgtgaattatagcccgagtaatgaagc





ccactggcctaggtatccccatctgtg





ggtacgactgtacgttcttgaactgta





ctgcatcatactgggcctgcctccttg





tctcaacattctgagaaggaagcagcc





acagctgacattctttaccatcgctct





tcagtcttgtcattaccagegactgcc





cccacacattctctgggccaccgggtt





gaaaagcggcagcgagactcccgggac





ctcagagtccgccacaccagaacccca





cccagtgttggagaagcttcggtccat





taataactataaccccaaagattttga





ctggaatctgaaacatggccgggtttt





catcattaagagctactctgaggacga





tattcaccgttccattaagtataatat





ttggtgcagcacagagcatggtaacaa





gagactggatgctgcttatcgttccat





gaacgggaaaggccccgtttacttact





tttcagtgtcaacggcagtggacactt





ctgtggcgtggcagaaatgaaatctgc





tgtggactacaacacatgtgcaggtgt





gtggtcccaggacaaatggaagggtcg





ttttgatgtcaggtggatttttgtgaa





ggacgttoccaatagccaactgcgaca





cattcgcctagagaacaacgagaataa





accagtgaccaactctagggacactca





ggaagtgcctctggaaaaggctaagca





ggtgttgaaaattatagccagctacaa





gcacaccacttccatttttgatgactt





ctcacactatgagaaacgccaagagga





agaagaaagtgttaaaaaggaacgtca





aggtcgtgggaaactcgagtaccccta





cgacgtgcccgactacgcctgagttta





aaatcgatggtacactcgaggttaacg





aattctaccttacccagagtgcaggtg





tgtggagatccctcctgccttgacatt





gagcagccttagagggtgggggaggct





caggggtcaggtctctgttcctgctta





ttggggagttcctggcctggcccttct





atgtctccccaggtaccccagtttttc





tgggttcacccagagtgcagatgcttg





aggaggtgggaagggactatttggggg





tgtctggctcaggtgccatgcctcact





ggggctggttggcacctgcatttcctg





ggagtggggctgtctcagggtagctgg





gcacggtgttcccttgagtgggggtgt





agtgagtgttcctagctgccacgcctt





tgccttcacctatgggatcgtggctgt





cagttaattaaccttccgcgggagctc





acggggagagccccccgccaaagcccc





cagggatgtaattgcatccctcttccg





ctagggggcagcagcgagccgcccggg





gctccgctccggtccggcgctcccccc





gcatccccgagccggagccggcagcgt





gcggggacagcccggcacggggaaggt





ggcacgcgatcgctttcctctgaacgc





ttctcgctgctctttgagcctgcagac





acctggggggatacggggaaaaagctt





taggctgaaagagagatttagaatgac





agaatcatagaatggcctgggttgcaa





aggagcacagtgctcacccagctccaa





ccccctgctatgtgcagggtcgccaac





cagcagcccaggctgcccagagccaca





tccagcctggccttgaatgcctgcagg





gatggggcatccacagcctccttgggc





aacctgttcagtgcgtcacggatccaa





ttccacggggttggggttgcgcctttt





ccaaggcagccctgggtttgcgcaggg





acgcggctgctctgggcgtggttccgg





gaaacgcagcggcgccgaccctgggtc





tcgcacattcttcacgtccgttcgcag





cgtcacccggatcttcgccgctaccct





tgtgggccccccggcgacgcttcctgc





tccgcccctaagtcgggaaggttcctt





gcggttcgcggcgtgccggacgtgaca





aacggaagccgcacgtctcactagtac





cctcgcagacggacagcgccagggagc





aatggcagcgcgccgaccgcgatgggc





tgtggccaatagcggctgctcagcagg





gcgcgccgagagcagcggccgggaagg





ggcggtgcgggaggcggggtgtggggc





ggtagtgtgggccctgttcctgcccgc





gcggtgttccgcattctgcaagcctcc





ggagcgcacgtcggcagtcggctccct





cgttgaccgaatcaccgacctctctcc





ccagctgtagctagcacaaccatggat





agcactgagaacgtcatcaagcccttc





atgcgcttcaaggtgcacatggagggc





tccgtgaacggccacgagttcgagatc





gagggcgagggcgagggcaagccctac





gagggcacccagaccgccaagctgcag





gtgaccaagggcggccccctgcccttc





gcctgggacatcctgtccccccagttc





cagtacggctccaaggtgtacgtgaag





caccccgccgacatccccgactacaag





aagctgtccttccccgagggcttcaag





tgggagcgcgtgatgaacttcgaggac





ggcggcgtggtgaccgtgacccaggac





tcctccctgcaggacggcaccttcatc





taccacgtgaagttcatcggcgtgaac





ttcccctccgacggccccgtaatgcag





aagaagactctgggctgggagccctcc





accgagcgcctgtacccccgcgacggc





gtgctgaagggcgagatccacaaggcg





ctgaagctgaagggcggcggccactac





ctggtggagttcaagtcaatctacatg





gccaagaagcccgtgaagctgcccggc





tactactacgtggactccaagctggac





atcacctcccacaacgaggactacacc





gtggtggagcagtacgagcgcgccgag





gcccgccaccacctgttccagtagggc





tagctggtccggactgtactgggtctc





tctggttagaccagatctgagcctggg





agctctctggctaactagggaacccac





tgcttaagcctcaataaagcttgcctt





gagtgcttcaagtagtgtgtgcccgtc





tgttgtgtgactctggtaactagagat





ccctcagacccttttagtcagtgtgga





aaatctctagcagggcccgtttaaacc





cgctgatcagcctcgactgtgccttct





agttgccagccatctgttgtttgcccc





tcccccgtgccttccttgaccctggaa





ggtgccactcccactgtcctttcctaa





taaaatgaggaaattgcatcgcattgt





ctgagtaggtgtcattctattctgggg





ggtggggtggggcaggacagcaagggg





gaggattgggaagacaatagcaggcat





gctggggatgcggtgggctctatggct





tctgaggcggaaagaaccagctggggc





tctagggggtatccccacgcgccctgt





agcggcgcattaagcgcggcgggtgtg





gtggttacgcgcagcgtgaccgctaca





cttgccagcgccctagegcccgctcct





ttcgctttettcccttcctttctcgcc





acgttcgccggctttccccgtcaagct





ctaaatcgggggctccctttagggttc





cgatttagtgctttacggcacctcgac





cccaaaaaacttgattagggtgatggt





tcacgtagtgggccatcgccctgatag





acggtttttcgccctttgacgttggag





tccacgttctttaatagtggactcttg





ttccaaactggaacaacactcaaccct





atctcggtctattcttttgatttataa





gggattttgccgatttcggcctattgg





ttaaaaaatgagctgatttaacaaaaa





tttaacgcgaattaattctgtggaatg





tgtgtcagttagggtgtggaaagtccc





caggctccccagcaggcagaagtatgc





aaagcatgcatctcaattagtcagcaa





ccaggtgtggaaagtccccaggctccc





cagcaggcagaagtatgcaaagcatgc





atctcaattagtcagcaaccatagtcc





cgcccctaactcegcccatccegcccc





taactccgcccagttccgcccattctc





cgccccatggctgactaatttttttta





tttatgcagaggccgaggccgcctctg





cctctgagctattccagaagtagtgag





gaggcttttttggaggcctaggctttt





gcaaaaagctcccgggagcttgtatat





ccattttcggatctgatcagcacgtgt





tgacaattaatcatcggcatagtatat





cggcatagtataatacgacaaggtgag





gaactaaaccatggccaagttgaccag





tgccgttccggtgctcaccgcgcgcga





cgtcgccggagcggtcgagttctggac





cgaccggctcgggttctcccgggactt





cgtggaggacgacttcgccggtgtggt





ccgggacgacgtgaccctgttcatcag





cgcggtccaggaccaggtggtgccgga





caacaccctggcctgggtgtgggtgcg





cggcctggacgagctgtacgccgagtg





gtcggaggtcgtgtccacgaacttccg





ggacgcctccgggccggccatgaccga





gatcggcgagcagccgtggggggggag





ttcgccctgcgcgacccggccggcaac





tgcgtgcacttcgtggccgaggagcag





gactgacacgtgctacgagatttcgat





tccaccgccgccttctatgaaaggttg





ggcttcggaatcgttttccgggacgcc





ggctggatgatcctccagcgcggggat





ctcatgctggagttcttcgcccacccc





aacttgtttattgcagcttataatggt





tacaaataaagcaatagcatcacaaat





ttcacaaataaagcatttttttcactg





cattctagttgtggtttgtccaaactc





atcaatgtatcttatcatgtctgtata





ccgtcgacctctagctagagcttggcg





taatcatggtcatagctgtttcctgtg





tgaaattgttatccgctcacaattcca





cacaacatacgagccggaagcataaag





tgtaaagcctggggtgcctaatgagtg





agctaactcacattaattgcgttgcgc





tcactgcccgctttccagtcgggaaac





ctgtcgtgccagctgcattaatgaatc





ggccaacgcgcggggagaggcggtttg





cgtattgggcgctcttccgcttcctcg





ctcactgactcgctgcgctcggtcgtt





cggctgcggcgagcggtatcagctcac





tcaaaggcggtaatacggttatccaca





gaatcaggggataacgcaggaaagaac





atgtgagcaaaaggccagcaaaaggcc





aggaaccgtaaaaaggccgcgttgctg





gcgtttttccataggctccgcccccct





gacgagcatcacaaaaatcgacgctca





agtcagaggtggcgaaacccgacagga





ctataaagataccaggcgtttccccct





ggaagctccctcgtgegctctcctgtt





ccgaccctgccgcttaccggatacctg





tccgcctttctcccttcgggaagcgtg





gcgctttctcatagctcacgctgtagg





tatctcagttcggtgtaggtcgttcgc





tccaagctgggctgtgtgcacgaaccc





cccgttcagcccgaccgctgcgcctta





tccggtaactatcgtcttgagtccaac





ccggtaagacacgacttatcgccactg





gcagcagccactggtaacaggattagc





agagcgaggtatgtaggcggtgctaca





gagttcttgaagtggtggcctaactac





ggctacactagaagaacagtatttggt





atctgcgctctgctgaagccagttacc





ttcggaaaaagagttggtagctcttga





tccggcaaacaaaccaccgctggtagc





ggtggtttttttgtttgcaagcagcag





attacgcgcagaaaaaaaggatctcaa





gaagatcctttgatcttttctacgggg





tctgacgctcagtggaacgaaaactca





cgttaagggattttggtcatgagatta





tcaaaaaggatcttcacctagatcctt





ttaaattaaaaatgaagttttaaatca





atctaaagtatatatgagtaaacttgg





tctgacagttaccaatgcttaatcagt





gaggcacctatctcagcgatctgtcta





tttcgttcatccatagttgcctgactc





cccgtcgtgtagataactacgatacgg





gagggcttaccatctggccccagtgct





gcaatgataccgcgagacccacgctca





ccggctccagatttatcagcaataaac





cagccagccggaagggccgagcgcaga





agtggtcctgcaactttatccgcctcc





atccagtctattaattgttgccgggaa





gctagagtaagtagttcgccagttaat





agtttgcgcaacgttgttgccattgct





acaggcatcgtggtgtcacgctcgtcg





tttggtatggcttcattcagctccggt





tcccaacgatcaaggcgagttacatga





tcccccatgttgtgcaaaaaagcggtt





agctccttcggtcctccgatcgttgtc





agaagtaagttggccgcagtgttatca





ctcatggttatggcagcactgcataat





tctcttactgtcatgccatccgtaaga





tgcttttctgtgactggtgagtactca





accaagtcattctgagaatagtgtatg





cggcgaccgagttgctcttgcccggcg





tcaatacgggataataccgcgccacat





agcagaactttaaaagtgctcatcatt





ggaaaacgttcttcggggcgaaaactc





tcaaggatcttaccgctgttgagatcc





agttcgatgtaacccactcgtgcaccc





aactgatcttcagcatcttttactttc





accagcgtttctgggtgagcaaaaaca





ggaaggcaaaatgccgcaaaaaaggga





ataagggcgacacggaaatgttgaata





ctcatactcttcctttttcaatattat





tgaagcatttatcagggttattgtctc





atgagcggatacatatttgaatgtatt





tagaaaaataaacaaataggggttccg





cgcacatttccccgaaaagtgccacct





gac






107
Construct comprising
agggagtggtaaactcgactttcactt




a nucleic acid
ttctctatcactgatagggagtggtaa




sequence encoding
actcgactttcacttttctctatcact




GFP-PEST, a m6A
gatagggagtggtaaactcgactttca




reporter sequence,
cttttctctatcactgatagggagtgg




and DHFR; a nucleic
taaactcgactttcacttttctctatc




acid sequenc
actgatagggagtggtaaactcgaggg




encoding APOBEC1-YTH
ggatccactagcatgaagggcgaattc




(5′-3′)
cagcacactggtaacccgtgtcggctc





cagatctggcctccgcgccgggttttg





gcgcctcccgcgggcgcccccctectc





acggcgagccgcgttgacattgattat





tgactaggcttttgcaaaaagctttgc





aaagatggataaagttttaaacagaga





ggaatctttgcagctaatggaccttct





aggtcttgaaaggagtgggaattggct





ccggtgcccgtcagtgggcagagcgca





catcgcccacagtccccgagaagttgg





ggggaggggtcggcaattgaaccggtg





cctagagaaggtggcgcggggtaaact





gggaaagtgatgtcgtgtactggctcc





gcctttttcccgaggggggggagaacc





gtatataagtgcagtagtcgccgtgaa





cgttctttttcgcaacgggtttgccgc





cagaacacaggtaagtgccgtgtgtgg





ttcccgcgggcctggcctctttacggg





ttatggcccttgcgtgccttgaattac





ttccacctggctgcagtacgtgattct





tgatcccgagcttcgggttggaagtgg





ggggagagttcgaggccttgcgcttaa





ggagccccttcgcctcgtgcttgagtt





gaggcctggcctgggcgctggggccgc





cgcgtgcgaatctggtggcaccttcgc





gcctgtctcgctgctttcgataagtct





ctagccatttaaaatttttgatgacct





gctgcgacgctttttttctggcaagat





agtcttgtaaatgcgggccaagatctg





cacactggtatttcggtttttggggcc





gcgggcggcgacggggcccgtgcgtcc





cagcgcacatgttcggcgaggcggggc





ctgcgagcgcggccaccgagaatcgga





cgggggtagtctcaagctggccggcct





gctctggtgcctggcctcgcgccgccg





tgtatcgccccgccctgggcggcaagg





ctggcccggtcggcaccagttgcgtga





gcggaaagatggccgcttcccggccct





gctgcagggagctcaaaatggaggacg





cggcgctcgggagagcggggggtgagt





cacccacacaaaggaaaagggcctttc





cgtcctcagccgtcgcttcatgtgact





ccacggagtaccgggcgccgtccaggc





acctogattagttctcgagcttttgga





gtacgtcgtctttaggttggggggagg





ggttttatgcgatggagtttccccaca





ctgagtgggtggagactgaagttaggc





cagcttggcacttgatgtaattctcct





tggaatttgccctttttgagtttggat





cttggttcattctcaagcctcagacag





tggttcaaagtttttttcttccatttc





aggtgtcgtgaggaattagcttggtac





taatacgactcactatagggagaccca





agctggctaggtaagcttggtaccgag





ctcggatccactagtccagtgtggtgg





aattctgcagatatccagcacagtggg





gtttagtgaaccgtcagatccgctaga





gatccgcggccgctaatacgactcact





atagggagagccgccaccatgagctca





gagactggcccagtggctgtggacccc





acattgagacggcggatcgagccccat





gagtttgaggtattcttcgatccgaga





gagctccgcaaggagacctgcctgctt





tacgaaattaattgggggggccggcac





tccatttggcgacatacatcacagaac





actaacaagcacgtcgaagtcaacttc





atcgagaagttcacgacagaaagatat





ttctgtccgaacacaaggtgcagcatt





acctggtttctcagctggagcccatgc





ggcgaatgtagtagggccatcactgaa





ttcctgtcaaggtatccccacgtcact





ctgtttatttacatcgcaaggctgtac





caccacgctgacccccgcaatcgacaa





ggcctgcgggatttgatctcttcaggt





gtgactatccaaattatgactgagcag





gagtcaggatactgctggagaaacttt





gtgaattatagcccgagtaatgaagcc





cactggcctaggtatccccatctgtgg





gtacgactgtacgttcttgaactgtac





tgcatcatactgggcctgcctccttgt





ctcaacattctgagaaggaagcagcca





cagctgacattctttaccatcgctctt





cagtcttgtcattaccagcgactgccc





ccacacattctctgggccaccgggttg





aaaagcggcagcgagactcccgggacc





tcagagtccgccacaccagaaccccac





ccagtgttggagaagcttcggtccatt





aataactataaccccaaagattttgac





tggaatctgaaacatggccgggttttc





atcattaagagctactctgaggacgat





attcaccgttccattaagtataatatt





tggtgcagcacagagcatggtaacaag





agactggatgctgcttatcgttccatg





aacgggaaaggccccgtttacttactt





ttcagtgtcaacggcagtggacacttc





tgtggcgtggcagaaatgaaatctgct





gtggactacaacacatgtgcaggtgtg





tggtcccaggacaaatggaagggtcgt





tttgatgtcaggtggatttttgtgaag





gacgttcccaatagccaactgcgacac





attcgcctagagaacaacgagaataaa





ccagtgaccaactctagggacactcag





gaagtgcctctggaaaaggctaagcag





gtgttgaaaattatagccagctacaag





cacaccacttccatttttgatgacttc





tcacactatgagaaacgccaagaggaa





gaagaaagtgttaaaaaggaacgtcaa





ggtcgtgggaaactcgagtacccctac





gacgtgcccgactacgcctgagtttaa





aatcgatggtacactcgaggttaacga





attctaccttacccagagtgcaggtgt





gtggagatccctcctgccttgacattg





agcagccttagaggggggggaggctca





ggggtcaggtctctgttcctgcttatt





ggggagttcctggcctggcccttctat





gtctccccaggtaccccagtttttctg





ggttcacccagagtgcagatgcttgag





gaggtgggaagggactatttgggggtg





tctggctcaggtgccatgcctcactgg





ggctggttggcacctgcatttcctggg





agtggggctgtctcagggtagctgggc





acggtgttcccttgagtgggggtgtag





tgagtgttcctagctgccacgcctttg





ccttcacctatgggatcgtggctgtca





gttaattaaccttccgcgggagctcac





ggggagagccccccgccaaagccccca





gggatgtaattgcatccctcttccgct





agggggcagcagcgagccgcccggggc





tccgctccggtccggcgctccccccgc





atccccgagccggagccggcagcgtgc





ggggacagcccggcacggggaaggtgg





cacgcgatcgctttcctctgaacgctt





ctcgctgctctttgagcctgcagacac





ctggggggatacggggaaaaagcttta





ggctgaaagagagatttagaatgacag





aatcatagaatggcctgggttgcaaag





gagcacagtgctcacccagctccaacc





ccctgctatgtgcagggtcgccaacca





gcagcccaggctgcccagagccacatc





cagcctggccttgaatgcctgcaggga





tggggcatccacagcctccttgggcaa





cctgttcagtgcgtcacggatccaatt





ccacggggttggggttgcgccttttcc





aaggcagccctgggtttgcgcagggac





gcggctgctctgggcgtggttccggga





aacgcagcggcgccgaccctgggtctc





gcacattcttcacgtccgttcgcagcg





tcacccggatcttcgccgctacccttg





tgggccccccggcgacgcttcctgctc





cgcccctaagtcgggaaggttccttgc





ggttcgcggcgtgccggacgtgacaaa





cggaagccgcacgtctcactagtaccc





tcgcagacggacagcgccagggagcaa





tggcagcgcgccgaccgcgatgggctg





tggccaatagcggctgctcagcagggc





gcgccgagagcagcggccgggaagggg





cggtgcgggaggcggggtgtggggcgg





tagtgtgggccctgttcctgcccgcgc





ggtgttccgcattctgcaagcctccgg





agcgcacgtcggcagtcggctccctcg





ttgaccgaatcaccgacctctctcccc





agctgtagctagcacaaccatggatag





cactgagaacgtcatcaagcccttcat





gcgcttcaaggtgcacatggagggctc





cgtgaacggccacgagttcgagatcga





gggcgagggcgagggcaagccctacga





gggcacccagaccgccaagctgcaggt





gaccaagggggccccctgcccttcgcc





tgggacatcctgtccccccagttccag





tacggctccaaggtgtacgtgaagcac





cccgccgacatccccgactacaagaag





ctgtccttccccgagggcttcaagtgg





gagcgcgtgatgaacttcgaggacggc





ggcgtggtgaccgtgacccaggactcc





tccctgcaggacggcaccttcatctac





cacgtgaagttcatcggcgtgaacttc





ccctccgacggccccgtaatgcagaag





aagactctgggctgggagccctccacc





gagcgcctgtacccccgcgacggcgtg





ctgaagggcgagatccacaaggcgctg





aagctgaagggcggcggccactacctg





gtggagttcaagtcaatctacatggcc





aagaagcccgtgaagctgcccggctac





tactacgtggactccaagctggacatc





acctcccacaacgaggactacaccgtg





gtggagcagtacgagcgcgccgaggcc





cgccaccacctgttccagtagggctag





ctggtccggactgtactgggtctctct





ggttagaccagatctgagcctgggagc





tctctggctaactagggaacccactgc





ttaagcctcaataaagcttgccttgag





tgcttcaagtagtgtgtgcccgtctgt





tgtgtgactctggtaactagagatccc





tcagacccttttagtcagtgtggaaaa





tctctagcagggcccgtttaaacccgc





tgatcagcctcgactgtgccttctagt





tgccagccatctgttgtttgcccctcc





cccgtgccttccttgaccctggaaggt





gccactcccactgtcctttcctaataa





aatgaggaaattgcatcgcattgtctg





agtaggtgtcattctattctggggggt





ggggggggcaggacagcaagggggagg





attgggaagacaatagcaggcatgctg





gggatgcggtgggctctatggcttctg





aggcggaaagaaccagctggggctcta





gggggtatccccacgcgccctgtagcg





gcgcattaagcgcggcgggtgtggtgg





ttacgcgcagcgtgaccgctacacttg





ccagcgccctagcgcccgctcctttcg





ctttcttcccttcctttctcgccacgt





tcgccggctttccccgtcaagctctaa





atcgggggctccctttagggttccgat





ttagtgctttacggcacctcgacccca





aaaaacttgattagggtgatggttcac





gtagtgggccatcgccctgatagacgg





tttttcgccctttgacgttggagtcca





cgttctttaatagtggactcttgttcc





aaactggaacaacactcaaccctatct





cggtctattcttttgatttataaggga





ttttgccgatttcggcctattggttaa





aaaatgagctgatttaacaaaaattta





acgcgaattaattctgtggaatgtgtg





tcagttagggtgtggaaagtccccagg





ctccccagcaggcagaagtatgcaaag





catgcatctcaattagtcagcaaccag





gtgtggaaagtccccaggctccccagc





aggcagaagtatgcaaagcatgcatct





caattagtcagcaaccatagtcccgcc





cctaactcegcccatccegcccctaac





tccgcccagttccgcccattctccgcc





ccatggctgactaattttttttattta





tgcagaggccgaggccgcctctgcctc





tgagctattccagaagtagtgaggagg





cttttttggaggcctaggcttttgcaa





aaagctcccgggagcttgtatatccat





tttcggatctgatcagcacgtgttgac





aattaatcatcggcatagtatatcggc





atagtataatacgacaaggtgaggaac





taaaccatggccaagttgaccagtgcc





gttccggtgctcaccgcgcgcgacgtc





gccggagcggtcgagttctggaccgac





cggctcgggttctcccgggacttcgtg





gaggacgacttcgccggtgtggtccgg





gacgacgtgaccctgttcatcagcgcg





gtccaggaccaggtggtgccggacaac





accctggcctgggtgtgggtgcgcggc





ctggacgagctgtacgccgagtggtcg





gaggtcgtgtccacgaacttccgggac





gcctccgggccggccatgaccgagatc





ggcgagcagccgtggggggggagttcg





ccctgcgcgacccggccggcaactgcg





tgcacttcgtggccgaggagcaggact





gacacgtgctacgagatttcgattcca





ccgccgccttctatgaaaggttgggct





tcggaatcgttttccgggacgccggct





ggatgatcctccagcgcggggatctca





tgctggagttcttcgcccaccccaact





tgtttattgcagcttataatggttaca





aataaagcaatagcatcacaaatttca





caaataaagcatttttttcactgcatt





ctagttgtggtttgtccaaactcatca





atgtatcttatcatgtctgtataccgt





cgacctctagctagagcttggcgtaat





catggtcatagctgtttcctgtgtgaa





attgttatccgctcacaattccacaca





acatacgagccggaagcataaagtgta





aagcctggggtgcctaatgagtgagct





aactcacattaattgcgttgcgctcac





tgcccgctttccagtcgggaaacctgt





cgtgccagctgcattaatgaatcggcc





aacgcgcggggagaggcggtttgcgta





ttgggcgctcttccgcttcctcgctca





ctgactcgctgcgctcggtcgttcggc





tgcggcgagcggtatcagctcactcaa





aggcggtaatacggttatccacagaat





caggggataacgcaggaaagaacatgt





gagcaaaaggccagcaaaaggccagga





accgtaaaaaggccgcgttgctggcgt





ttttccataggctccgcccccctgacg





agcatcacaaaaatcgacgctcaagtc





agaggtggcgaaacccgacaggactat





aaagataccaggcgtttccccctggaa





gctccctcgtgcgctctcctgttccga





ccctgccgcttaccggatacctgtccg





cctttctcccttcgggaagcgtggcgc





tttctcatagctcacgctgtaggtatc





tcagttcggtgtaggtcgttcgctcca





agctgggctgtgtgcacgaaccccccg





ttcagcccgaccgctgcgccttatccg





gtaactatcgtcttgagtccaacccgg





taagacacgacttatcgccactggcag





cagccactggtaacaggattagcagag





cgaggtatgtaggcggtgctacagagt





tcttgaagtggtggcctaactacggct





acactagaagaacagtatttggtatct





gcgctctgctgaagccagttaccttcg





gaaaaagagttggtagctcttgatccg





gcaaacaaaccaccgctggtagcggtg





gtttttttgtttgcaagcagcagatta





cgcgcagaaaaaaaggatctcaagaag





atcctttgatcttttctacggggtctg





acgctcagtggaacgaaaactcacgtt





aagggattttggtcatgagattatcaa





aaaggatcttcacctagatccttttaa





attaaaaatgaagttttaaatcaatct





aaagtatatatgagtaaacttggtctg





acagttaccaatgcttaatcagtgagg





cacctatctcagcgatctgtctatttc





gttcatccatagttgcctgactccccg





tcgtgtagataactacgatacgggagg





gcttaccatctggccccagtgctgcaa





tgataccgcgagacccacgctcaccgg





ctccagatttatcagcaataaaccagc





cagccggaagggccgagcgcagaagtg





gtcctgcaactttatccgcctccatcc





agtctattaattgttgccgggaagcta





gagtaagtagttcgccagttaatagtt





tgcgcaacgttgttgccattgctacag





gcatcgtggtgtcacgctcgtcgtttg





gtatggcttcattcagctccggttccc





aacgatcaaggcgagttacatgatccc





ccatgttgtgcaaaaaagcggttagct





ccttcggtcctccgatcgttgtcagaa





gtaagttggccgcagtgttatcactca





tggttatggcagcactgcataattctc





ttactgtcatgccatccgtaagatgct





tttctgtgactggtgagtactcaacca





agtcattctgagaatagtgtatgcggc





gaccgagttgctcttgcccggcgtcaa





tacgggataataccgcgccacatagca





gaactttaaaagtgctcatcattggaa





aacgttcttcggggcgaaaactctcaa





ggatcttaccgctgttgagatccagtt





cgatgtaacccactcgtgcacccaact





gatcttcagcatcttttactttcacca





gcgtttctgggtgagcaaaaacaggaa





ggcaaaatgccgcaaaaaagggaataa





gggcgacacggaaatgttgaatactca





tactcttcctttttcaatattattgaa





gcatttatcagggttattgtctcatga





gcggatacatatttgaatgtatttaga





aaaataaacaaataggggttccgcgca





catttccccgaaaagtgccacctgac






108
Exemplary m6A
GCGGACTTACGACAGTTGCGTTACACC




sensor sequence-5′-3′
CTTTCTCGACAAAACCTAACTTGCGCA





GAAAACATGCCAATCTCATCTTGGCTT






109
Exemplary m6A
GCGGCGTTACGACAGTTGCGTTACACC




sensor sequence-5′-3′
CTTTCTCGACAAAACCTAACTTGCGCA





GAAAACATGCCAATCTCATCTTGGCTT






110
Exemplary m6A
GCGGACTTACGTCAGTTGCGTTACACC




sensor sequence-5′-3′
CTTTCTCGACAAAACCTAACTTGCGCA





GAAAACATGCCAATCTCATCTTGGCTT






111
Exemplary m6A
GCGGAGTTACGACAGTTGCGTTACACC




sensor sequence-5′-3′
CTTTCTCGTCAAAACCTAACTTGCGCA





GAAAACATGCCAATCTCATCTTGGCTT






112
Exemplary m6A
GCGGAGTTACGACAGTTGCGTTACACC




sensor sequence-5′-3′
CTTTCTCGACAAAGCCTAACTTGCGCA





GAAAACATGCCAATCTCATCTTGGCTT






113
Exemplary m6A
GCGGAGTTACGACAGTTGCGTTACACC




sensor sequence-5′-3′
CTTTCTCGACAAAACCTAGCTTGCGCA





GAAAACATGCCAATCTCATCTTGGCTT






114
Exemplary m6A
GCGGAGTTACGACAGTTGCGTTACACC




sensor sequence-5′-3′
CTTTCTCGACAAAACCTAACTTGCGCA





GAAAGCATGCCAATCTCATCTTGGCTT






115
Exemplary m6A
GCGGACTTACGACAGTTGCGTCCAATC




sensor sequence-5′-3′
TCATCTTGGCTT






116
Exemplary m6A
GCGGCCTTACGTCAGTTGCGTTACACC




sensor sequence-5′-3′
CTTTCTCGGCAAAGCCTAGCTTGCGCA





GAAAGCATGCCAATCTCATCTTGGCTT






117

(GGGGS)20






118
deaminase domain of
TSNFNNGIGRHKTYLCYEVERLDNGTS




hAPOBEC3A
VKMDQHRGFLHNQAKNLLCGFYGRHAE





LRFLDLVPSLQLDPAQIYRVTWFISWS





PCFSWGCAGEVRAFLQENTHVRLRIFA





ARIYDYDPLYKEALQMLRDAGAQVSIM





TYDEFKHCWDTFVDHQGCPFQPWDGLD





EHSQALSGRLR






119
catalytic domain of
MDSLLMNRREFLYQFKNVRWAKGRRET




ADAR2
YLCYVVKRRDSATSFSLDFGYLRNKNG





CHVELLFLRYISDWDLDPGRCYRVTWF





ISWSPCYDCARHVADFLRGNPNLSLRI





FTARLYFCEAGRREPEGLRRLHRAGVQ





IAIMTFKDYFYCWNTFVENHGRTFKAW





EGLHENSVRLSRQLRRILL






120
deaminase domain of
RRRIEPHEFEVFFDPRELRKETCLLYE




rAPOBEC1
INWGGRHSIWRHTSQNTNKHVEVNFIE





KFTTERYFCPNTRCSITWFLSWSPCGE





CSRAITEFLSRYPHVTLFIYIARLYHH





ADPRNRQGLRDLISSGVTIQIMTEQES





GYCWRNFVNYSPSNEAHWPRYPHLWVR





LYVLELYCIILGLPPCLNILRRKQPQL





TFFTIALQSCHYQRLPPHILWATGLK






121
hADAR1
MNPRQGYSLSGYYTHPFQGYEHRQLRY





QQPGPGSSPSSFLLKQIEFLKGQLPEA





PVIGKQTPSLPPSLPGLRPRFPVLLAS





STRGRQVDIRGVPRGVHLRSQGLQRGF





QHPSPRGRSLPQRGVDCLSSHFQELSI





YQDQEQRILKFLEELGEGKATTAHDLS





GKLGTPKKEINRVLYSLAKKGKLQKEA





GTPPLWKIAVSTQAWNQHSGVVRPDGH





SQGAPNSDPSLEPEDRNSTSVSEDLLE





PFIAVSAQAWNQHSGVVRPDSHSQGSP





NSDPGLEPEDSNSTSALEDPLEFLDMA





EIKEKICDYLFNVSDSSALNLAKNIGL





TKARDINAVLIDMERQGDVYRQGTTPP





IWHLTDKKRERMQIKRNTNSVPETAPA





AIPETKRNAEFLTCNIPTSNASNNMVT





TEKVENGQEPVIKLENRQEARPEPARL





KPPVHYNGPSKAGYVDFENGQWATDDI





PDDLNSIRAAPGEFRAIMEMPSFYSHG





LPRCSPYKKLTECQLKNPISGLLEYAQ





FASQTCEFNMIEQSGPPHEPRFKFQVV





INGREFPPAEAGSKKVAKQDAAMKAMT





ILLEEAKAKDSGKSEESSHYSTEKESE





KTAESQTPTPSATSFFSGKSPVTTLLE





CMHKLGNSCEFRLLSKEGPAHEPKFQY





CVAVGAQTFPSVSAPSKKVAKQMAAEE





AMKALHGEATNSMASDNQPEGMISESL





DNLESMMPNKVRKIGELVRYLNTNPVG





GLLEYARSHGFAAEFKLVDQSGPPHEP





KFVYQAKVGGRWFPAVCAHSKKQGKQE





AADAALRVLIGENEKAERMGFTEVTPV





TGASLRRTMLLLSRSPEAQPKTLPLTG





STFHDQIAMLSHRCFNTLTNSFQPSLL





GRKILAAIIMKKDSEDMGVVVSLGTGN





RCVKGDSLSLKGETVNDCHAEIISRRG





FIRFLYSELMKYNSQTAKDSIFEPAKG





GEKLQIKKTVSFHLYISTAPCGDGALF





DKSCSDRAMESTESRHYPVFENPKQGK





LRTKVENGEGTIPVESSDIVPTWDGIR





LGERLRTMSCSDKILRWNVLGLQGALL





THFLQPIYLKSVTLGYLFSQGHLTRAI





CCRVTRDGSAFEDGLRHPFIVNHPKVG





RVSIYDSKRQSGKTKETSVNWCLADGY





DLEILDGTRGTVDGPRNELSRVSKKNI





FLLFKKLCSFRYRRDLLRLSYGEAKKA





ARDYETAKNYFKKGLKDMGYGNWISKP





QEEKNFYLCPV






122
hADAR1 catalytic
LPLTGSTFHDQIAMLSHRCFNTLTNSF




domain
QPSLLGRKILAAIIMKKDSEDMGVVVS





LGTGNRCVKGDSLSLKGETVNDCHAEI





ISRRGFIRFLYSELMKYNSQTAKDSIF





EPAKGGEKLQIKKTVSFHLYISTAPCG





DGALFDKSCSDRAMESTESRHYPVFEN





PKQGKLRTKVENGEGTIPVESSDIVPT





WDGIRLGERLRTMSCSDKILRWNVLGL





QGALLTHFLQPIYLKSVTLGYLFSQGH





LTRAICCRVTRDGSAFEDGLRHPFIVN





HPKVGRVSIYDSKRQSGKTKETSVNWC





LADGYDLEILDGTRGTVDGPRNELSRV





SKKNIFLLFKKLCSFRYRRDLLRLSYG





EAKKAARDYETAKNYFKKGLKDMGYGN





WISKPQEEKNFYLCPV






123

A(EAAAK)10A






124

A(EAAAK)20A






125
human TP53 amino
MEEPQSDPSVEPPLSQETFSDLWKLLP
canonical human



acid sequence
ENNVLSPLPSQAMDDLMLSPDDIEQWF
p53 sequence




TEDPGPDEAPRMPEAAPPVAPAPAAPT
(accessed at




PAAPAPAPSWPLSSSVPSQKTYQGSYG
uniprot.org/uniprot




FRLGFLHSGTAKSVTCTYSPALNKMFC
kb/P04637/entry#s




QLAKTCPVQLWVDSTPPPGTRVRAMAI
equences)




YKQSQHMTEVVRRCPHHERCSDSDGLA





PPQHLIRVEGNLRVEYLDDRNTFRHSV





VVPYEPPEVGSDCTTIHYNYMCNSSCM





GGMNRRPILTIITLEDSSGNLLGRNSF





EVRVCACPGRDRRTEEENLRKKGEPHH





ELPPGSTKRALPNNTSSSPQPKKKPLD





GEYFTLQIRGRERFEMFRELNEALELK





DAQAGKEPGGSRAHSSHLKSKKGQSTS





RHKKLMFKTEGPDSD






126
human SOCS2 amino
MTLRCLEPSGNGGEGTRSQWGTAGSAE
canonical human



acid sequence
EPSPQAARLAKALRELGQTGWYWGSMT
p53 sequence




VNEAKEKLKEAPEGTFLIRDSSHSDYL
(accessed at




LTISVKTSAGPTNLRIEYQDGKFRLDS
uniprot.org/uniprot




IICVKSKLKQFDSVVHLIDYYVQMCKD
kb/O14508/entry#s




KRTGPEAPRNGTVHLYLTKPLYTSAPS
equences)




LQHLCRLTINKCTGAIWGLPLPTRLKD





YLEEYKFQV









Disclosed are materials, compositions, and components that can be used for, can be used in conjunction with, can be used in preparation for, or are products of the disclosed embodiments. These and other materials are disclosed herein, and it is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each various individual and collective combinations and permutations of these compositions may not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a method is disclosed and discussed and a number of modifications that can be made to a number of molecules included in the method are discussed, each and every combination and permutation of the method, and the modifications that are possible are specifically contemplated unless specifically indicated to the contrary. Likewise, any subset or combination of these is also specifically contemplated and disclosed. This concept applies to all aspects of this disclosure including, but not limited to, steps in methods using the disclosed compositions. Thus, if there are a variety of additional steps that can be performed, it is understood that each of these additional steps can be performed with any specific method steps or combination of method steps of the disclosed methods, and that each such combination or subset of combinations is specifically contemplated and should be considered disclosed.


One skilled in the art will readily appreciate that the present disclosure is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The present disclosure described herein are presently representative of preferred embodiments, are exemplary, and are not intended as limitations on the scope of the present disclosure. Changes therein and other uses will occur to those skilled in the art which are encompassed within the spirit of the present disclosure as defined by the scope of the claims.


No admission is made that any reference, including any non-patent or patent document cited in this specification, constitutes prior art. In particular, it will be understood that, unless otherwise stated, reference to any document herein does not constitute an admission that any of these documents forms part of the common general knowledge in the art in the United States or in any other country. Any discussion of the references states what their authors assert, and the applicant reserves the right to challenge the accuracy and pertinence of any of the documents cited herein. All references cited herein are fully incorporated by reference, unless explicitly indicated otherwise. The present disclosure shall control in the event there are any disparities between any definitions and/or description found in the cited references.

Claims
  • 1. An expression system comprising: (a) a first DNA construct comprising a polynucleotide encoding a fusion protein, wherein the fusion protein comprises an N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase or a catalytic domain of an adenosine deaminase; and(b) a second DNA construct comprising a polynucleotide encoding a heterologous polypeptide, the polynucleotide encoding a heterologous polypeptide comprising: i. a polynucleotide encoding an effector protein;ii. a polynucleotide encoding a m6A sensor sequence; andiii. a polynucleotide encoding a dihydrofolate reductase (DHFR).
  • 2. The expression system of claim 1, wherein the m6A binding domain comprises a sequence having at least 90% or greater sequence identity to SEQ ID NOs: 66 or 108-116.
  • 3. The expression system of claim 1, wherein the m6A binding domain is fused to the catalytic domain via a peptide linker.
  • 4. The expression system of claim 1, wherein the catalytic domain comprises a polypeptide having at least 95% identity to SEQ ID NO 78 or a catalytic fragment thereof, SEQ ID NO: 79 or a catalytic fragment thereof; SEQ ID NO: 80 or a catalytic fragment thereof; or SEQ ID NO: 81.
  • 5. The expression system of claim 1, wherein a vector comprises the first DNA construct.
  • 6. The expression system of claim 1, wherein a vector comprises the second DNA construct.
  • 7. The expression system of claim 1, wherein a vector comprises the first DNA construct and the second DNA construct.
  • 8. The expression system of claim 1, wherein the nucleic acid sequence encoding a fusion protein, the nucleic acid sequence encoding a heterologous polypeptide and a polypeptide encoding dihydrofolate reductase (DHFR), or both, are operably linked to a first promoter.
  • 9. The expression system of claim 9, wherein the system further comprises a nucleic acid sequence encoding a selectable marker operably linked to a second promoter.
  • 10. The expression system of claim 9, wherein the first promoter is a constitutive or an inducible promoter.
  • 11. The expression system of claim 1, wherein the cytidine deaminase is APOBEC-1.
  • 12. The expression system of claim 1, wherein the effector protein is a tumor suppressor protein or a catalytically dead RNA-guided endonuclease.
  • 13. The expression system of claim 13, wherein the tumor suppressor protein is suppressor of cytokine signaling 2 (SOC2) or p53 or one of the proteins listed in Table 1.
  • 14. The expression system of claim 13, wherein the catalytically dead RNA-guided endonuclease is a dCas9 or a dCas13.
  • 15. A polynucleotide comprising a nucleic acid sequence encoding an effector protein polypeptide, a m6A sensor sequence, and a polypeptide encoding dihydrofolate reductase (DHFR).
  • 16. A vector comprising the expression system of claim 1.
  • 17. A host cell comprising the expression system of claim 1 or the vector of claim 17.
  • 18. A non-human transgenic animal comprising the host cell of claim 18.
  • 19. A kit comprising the expression system of claim 1.
  • 20. A method of increasing expression of a tumor suppressor protein in one or more cells, comprising introducing the expression system of claim 1 into the one or more cells.
  • 21. The method of claim 21, wherein the one or more cells are hepatocellular carcinoma (HCC) cells.
  • 22. (canceled)
  • 23. A method of reducing M6A effector regulator expression, comprising: introducing an expression system into a subject having or suspected of having a cancer, wherein the expression system comprises:(a) a first DNA construct comprising a polynucleotide encoding a fusion protein, wherein the fusion protein comprises an N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase or a catalytic domain of an adenosine deaminase; and(b) a second DNA construct comprising a polynucleotide encoding a heterologous polypeptide, the polynucleotide encoding a heterologous polypeptide comprising: iv. a polynucleotide encoding a catalytically-dead RNA-guided endonuclease;v. a polynucleotide encoding a m6A sensor sequence; andvi. a polynucleotide encoding a dihydrofolate reductase (DHFR);(c) an sgRNA configured to bind to an m6a regulator.
  • 24. (canceled)
  • 25. (canceled)
  • 26. (canceled)
  • 27. (canceled)
  • 28. A method of reducing M6A hypermethylation, comprising: introducing an expression system into a subject having or suspected of having a cancer, wherein the expression system comprises:(a) a first DNA construct comprising a polynucleotide encoding a fusion protein, wherein the fusion protein comprises an N6-methyladenosine (m6A) binding domain of a YT521-B homology (YTH) domain-containing protein fused to a catalytic domain of a cytidine deaminase or a catalytic domain of an adenosine deaminase; and(b) a second DNA construct comprising a polynucleotide encoding a heterologous polypeptide, the polynucleotide encoding a heterologous polypeptide comprising: vii. a polynucleotide encoding a catalytically-dead RNA-guided endonuclease;viii. a polynucleotide encoding a m6A sensor sequence; andix. a polynucleotide encoding a dihydrofolate reductase (DHFR);(c) an sgRNA configured to bind to an m6a regulator.
  • 28. (canceled)
  • 29. (canceled)
  • 30. (canceled)
  • 31. (canceled)
  • 32. A method of inhibiting a cancer cell, the method comprising: introducing the expression system of claim 1 into the cancer cell, wherein the cancer cell comprises m6A RNA hypermethylation, and wherein the second DNA construct comprising a polynucleotide encoding an effector protein, the effector protein comprising a tumor suppressor protein.
  • 33. (canceled)
  • 34. (canceled)
  • 35. (canceled)
  • 36. (canceled)
  • 37. (canceled)
  • 38. (canceled)
  • 39. (canceled)
  • 40. (canceled)
  • 41. (canceled)
  • 42. (canceled)
  • 43. A method of treating a subject having a cancer characterized by m6A RNA hypermethylation, the method comprising inhibiting a cancer cell according to the method of claim 32.
  • 44. The method of claim 43, wherein the cancer comprises at least one of acute myeloid leukemia (AML), glioblastoma (GBM), lung cancer, endometrial cancer, cervical cancer, ovarian cancer, breast cancer, colorectal cancer (CRC), a hepatocellular carcinoma (HCC), pancreatic cancer, gastric cancer, prostate cancer, or renal cell carcinoma.
  • 45. (canceled)
  • 46. (canceled)
  • 47. (canceled)
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims priority to, and the benefit of, U.S. Provisional Application No. 63/415,395, filed on Oct. 12, 2022, and U.S. Provisional Application No. 63/531,948, filed on Aug. 10, 2023, the entire contents of both of which are incorporated by reference as if fully set forth herein.

STATEMENT REGARDING FEDERALLY FUNDED RESEARCH

This invention was made with government support under Grant Nos. DP1DA046584 and R0IMH118366 awarded by the National Institutes of Health/National Institute on Drug Abuse and National Institutes of Health/National Institute of Mental Health, respectively. The government has certain rights in the invention.

Provisional Applications (2)
Number Date Country
63531948 Aug 2023 US
63415395 Oct 2022 US