WO2023039528A1 - Nanobody-mediated control of gene expression and epigenetic memory - Google Patents

Nanobody-mediated control of gene expression and epigenetic memory Download PDF

Info

Publication number
WO2023039528A1
WO2023039528A1 PCT/US2022/076208 US2022076208W WO2023039528A1 WO 2023039528 A1 WO2023039528 A1 WO 2023039528A1 US 2022076208 W US2022076208 W US 2022076208W WO 2023039528 A1 WO2023039528 A1 WO 2023039528A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
nanobody
amino acid
cells
acid sequence
Prior art date
Application number
PCT/US2022/076208
Other languages
French (fr)
Inventor
Mike Vi VAN
Jun Wan
Lacramioara Bintu
Original Assignee
The Board Of Trustees Of The Leland Stanford Junior University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Board Of Trustees Of The Leland Stanford Junior University filed Critical The Board Of Trustees Of The Leland Stanford Junior University
Publication of WO2023039528A1 publication Critical patent/WO2023039528A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B30/00Methods of screening libraries
    • C40B30/06Methods of screening libraries by measuring effects on living organisms, tissues or cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present invention relates to methods and systems of controlling gene expression and epigenetic memory utilizing synthetic fusion proteins comprising a DNA binding protein or domain or fragment thereof and a nanobody configured to bind a chromatin regulator.
  • CROSS-REFERENCE TO RELATED APPLICATIONS [0002] This application claims the benefit of U.S. Provisional Application No. 63/242,898, filed September 10, 2021, the content of which is herein incorporated by reference in its entirety.
  • chromatin regulators are large, consisting of multiple domains that are necessary for their function, either to stimulate catalytic function or to mediate interactions with other members of the complex.
  • adeno-associated virus AAV
  • lentivirus payload limit ⁇ 9.7 kb
  • AAV adeno-associated virus
  • Cas9 lentivirus
  • Methods which overcome the size and viral packaging challenges often result in poor editing efficiency due to the inability to effectively deliver all the components for gene editing to any one given cell.
  • the methods comprise identifying and selecting nanobodies for a target chromatin regulator comprising at least one or all of: a) preparing a yeast nanobody library comprising yeast cells each displaying a single nanobody on its surface; b) mixing the yeast nanobody library with a target chromatin regulator wherein the target chromatin regulator is configured to bind to a solid surface; c) separating yeast cells bound to target chromatin regulator from unbound yeast cells; d) amplifying nanobody DNA sequences from the yeast cells bound to target chromatin regulator; e) cloning the nanobody sequences into a human lentiviral vector to prepare a nanobody lentiviral library, wherein each nanobody DNA sequence is expressed as a fusion protein with a DNA binding domain; f) transforming the nanobody lentiviral library into reporter cells, wherein a reporter cell comprises a two-part reporter gene comprising a surface marker and a fluorescent protein, wherein the two-part reporter gene is capable of being silenced or induced by the
  • the methods further comprise repeating steps a-d one or more times.
  • the chromatin regulator comprises a methylase, a demethylase, an acetylase, or ATP-dependent chromatin remodeling complex.
  • the DNA binding domain is an inducible DNA binding domain and the method further comprising treating the reporter cells with an agent configured to induce the inducible DNA binding domain for a length of time.
  • the two-part reporter gene is under the control of a strong promoter and capable of being silenced by a functional chromatin repressor.
  • the two-part reporter gene is under the control of a weak promoter and capable of being induced by a functional chromatin activator.
  • fusion proteins comprising a DNA binding protein, DNA binding domain, or a functional fragment thereof covalently linked to at least one nanobody configured to bind a chromatin regulator.
  • one or more of at least one nanobody is identified by a method disclosed herein.
  • the chromatin regulator comprises DNA (cytosine-5)-methyltransferase 3A (DNMT3A), or complex thereof.
  • the chromatin regulator comprises a ten-eleven translocation methylcytosine 1/2/3 (TET1/2/3) complex.
  • the at least one nanobody comprises at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFXXXXM (SEQ ID NO: 13), EZVAXIXXGXXTNY (SEQ ID NO: 14), and AAXXXXXYYXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid.
  • the at least one nanobody comprises a first CDR comprising an amino acid sequence of GTIFXXXM (SEQ ID NO: 13), a second CDR comprising an amino acid sequence of EZVAXIXXGXXTNY (SEQ ID NO: 14), and a third CDR comprising an amino acid sequence of AAXXXXXYYXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid.
  • the at least one nanobody is configured to bind DNA (cytosine-5)-methyltransferase 3A (DNMT3A) complexes.
  • the nanobody comprises at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), GNIFDGASM (SEQ ID NO: 4), EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), AAGRYYYPGHGY (SEQ ID NO: 8), AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 12).
  • GTIFAHSRM SEQ ID NO: 1
  • GTISSDGYM SEQ ID NO: 2
  • GTIFYFFGM SEQ ID NO: 3
  • the at least one nanobody comprises: a first CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), and GNIFDGASM (SEQ ID NO: 4); a second CDR comprising an amino acid sequence selected from the group consisting of EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), and AAGRYYYPGHGY (SEQ ID NO: 8); a third CDR comprising an amino acid sequence selected from the group consisting of AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO:
  • the nanobody comprises an amino acid sequence at least 70% (e.g., 80%, 85%, 90%, 95%, 98%, 99%) similar to any of SEQ ID NOs: 16-20.
  • the at least one nanobody comprises at least one CDR having an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933-5959.
  • the at least one nanobody comprises: a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933-5941; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5942- 5950; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5951-5959; or a combination thereof.
  • the nanobody comprises an amino acid sequence at least 70% similar to any of SEQ ID NOs: 5960-5968.
  • the nanobody comprises an amino acid sequence of SEQ ID NOs: 5960-5968.
  • the fusion protein further comprises a transcription factor or regulator.
  • the transcription factor or regulator comprises Krüppel- associated box (KRAB).
  • KRAB Krüppel- associated box
  • DNA (cytosine-5)-methyltransferase 3A (DNMT3A) binding proteins e.g., antibodies and fragments thereof
  • fusion proteins thereof comprising at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFXXXXM (SEQ ID NO: 13), EZVAXIXXGXXTNY (SEQ ID NO: 14), and AAXXXXXYYXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid.
  • the at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), GNIFDGASM (SEQ ID NO: 4), EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), AAGRYYYPGHGY (SEQ ID NO: 8), AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), AND AAKPSRVYWRDYRFFY (SEQ ID NO: 12).
  • GTIFAHSRM SEQ ID NO: 1
  • GTISSDGYM SEQ ID NO: 2
  • GTIFYFFGM SEQ ID NO: 3
  • GNIFDGASM S
  • the DNMT3A binding protein comprises an amino acid sequence at least 70% similar to SEQ ID NOs: 16-20.
  • TET1/2/3 complex binding proteins e.g., antibodies and fragments thereof
  • fusion proteins thereof e.g., antibodies and fragments thereof
  • the TET1/2/3 complex binding proteins comprises at least one CDR having an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933- 5959.
  • the TET1/2/3 complex binding proteins comprises: a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933- 5941; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5942-5950; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5951-5959; or a combination thereof.
  • the TET1/2/3 complex binding proteins comprises an amino acid sequence at least 70% similar to any of SEQ ID NOs: 5960-5968.
  • the TET1/2/3 complex binding proteins comprises an amino acids sequence of any of SEQ ID NOs: 5960-5968.
  • the DNMT3A binding protein or TET1/2/3 complex binding protein is covalently attached to a DNA binding protein, DNA binding domain, or a functional fragment thereof.
  • Nucleic acids and vectors encoding the fusion proteins, nanobodies, DNMT3A binding proteins, and TET1/2/3 complex binding proteins, and compositions and cells comprising thereof are provided.
  • methods for modifying gene expression, epigenetic memory, or a combination thereof of at least one target nucleic acid may comprise contacting a target nucleic acid with a fusion protein as described herein.
  • the methods comprise contacting the target nucleic acid with at least one fusion protein comprising a DNA binding protein, DNA binding domain, or a functional fragment thereof covalently linked to a nanobody configured to bind a chromatin regulator.
  • the gene expression and/or epigenetic memory of at least two genes are modulated.
  • the at least one target nucleic acid is in a cell and the chromatin regulator is endogenous to the cell.
  • the contacting comprises introducing into the cell the fusion protein, or a nucleic acid encoding thereof.
  • the introducing comprises administering to a subject.
  • the methods further comprise contacting the target nucleic acid with an exogenous chromatin regulator.
  • the endogenous or exogenous chromatin regulator comprises a methylase, a demethylase, an acetylase, or ATP-dependent chromatin remodeling complex.
  • the nanobody is identified or selected by a method as disclosed herein.
  • the chromatin regulator comprises DNA (cytosine-5)- methyltransferase 3A (DNMT3A).
  • the chromatin regulator comprises a TET1/2/3 complex.
  • the methods further comprise contacting the target nucleic acid with a transcription factor or regulator.
  • the fusion protein further comprises the transcription factor or regulator.
  • the transcription factor or regulator comprises a transcriptional repressor.
  • the transcription factor comprises Krüppel-associated box (KRAB).
  • KRAB Krüppel-associated box
  • 1A is a schematic of an exemplary construct for constitutive coexpression (under the pGK promoter) of H2B-mIFP and antiGFP nanobody fused with the rTetR DNA-binding protein (separated by the self-cleaving peptide T2A) randomly integrated into HEK293T cells by PiggyBac (blue box, top). These cells also contain a TagRFP reporter gene integrated at the AAVS1 “safe harbor” locus and driven by the pEF promoter (bottom). Five TetO-binding sites allow binding of rTetR upstream of the reporter gene upon dox addition.
  • the nuclear localization signal (NLS) and H2B domains localize fluorescent protein signals to the nucleus, improving quantification during time-lapse imaging.
  • Plasmids expressing GFP- tagged CRs (HP1 ⁇ , HP1 ⁇ , HP1 ⁇ , and HDAC5) were transiently transfected into cells (green box, right).
  • FIG. 1B is time-lapse imaging of cells upon recruitment of GFP-tagged HP1 ⁇ (top) and HDAC5 (bottom).
  • Cells stably expressing the reporter (TagRFP, red) and the rTetR-antiGFP fusion (mIFP, purple) were transiently transfected with GFP-HP1 ⁇ or GFP-HDAC5 (GFP, green).
  • FIG. 1C shows cells that still have GFP-CR expression (yellow circles) by day 5 of recruitment remain silenced. Images and analysis in FIG. 1B and 1C are from one biological replicate. Scale bars: 20 ⁇ m.
  • FIG.1D, left is a graph of fluorescence distributions measured by flow cytometry showing reporter silencing after recruitment of GFPCRs (+dox) for 5 days. Cells were gated for the presence of both GFP-CR (GFP positive) and rTetR-antiGFP (mIFP positive).
  • FIG.1D right is a graph of mean percentage of cells silenced upon presence or absence of dox for 5 days.
  • GFP-HP1 ⁇ : **p 0.0031; GFP-HP1 ⁇ vs.
  • FIGS. 2A-2E show that nanobody-mediated recruitment of endogenous chromatin regulators can silence gene expression and confer memory.
  • FIG. 2A is a schematic of an exemplary construct for constitutive coexpression of H2B-mIFP and nanobody against CR (antiHP1 or antiDNMT1) fused with the rTetR DNA-binding protein (blue box, top) expressed in HEK293T cells containing a TagRFP reporter (bottom).
  • FIG. 2B left is a graph of fluorescence distributions of the TagRFP reporter after 5 days of recruitment (dox treatment) in cells stably expressing the nanobody constructs or rTetR-KRAB were analyzed by flow cytometry to determine the percentage of cells with the reporter silenced (left of the red dotted line).
  • Statistical analysis by two-tailed Tukey’s test antiHP1 vs.
  • FIG.2C is targeted bisulfite sequencing of the reporter after 5 days of recruitment with antiDNMT1, DNMT1, and DNMT3B (+dox), compared to untreated cells expressing the same effectors ( ⁇ dox). Dox-treated cells were sorted based on the silencing of the TagRFP reporter labeled as +dox ON and +dox OFF (see FIG. 12A for representative gating).
  • FIG. 2D shows the experimental design for investigating epigenetic memory: rTetR-effectors were recruited to the reporter for 5 days (+dox) and then released ( ⁇ dox). Memory was monitored after dox removal via flow cytometry throughout 30 days.
  • FIG. 2E is a graph of silencing and memory dynamics data (right) for the experiment described in FIG. 2D with representative flow cytometry histogram for antiDNMT1 at day 0, 15, 30 after dox removal (left). The percentage of cells silenced was normalized to the no dox control to adjust for any background silencing. Means are from three replicates.
  • FIGS. 3A-3F show nanobody-mediated enhancement of KRAB and DNMT3A repression.
  • FIG. 3A, left is a graph of fluorescence distributions of the reporter gene after transient expression of rTetR-effector fusions and recruitment by dox treatment for 5 days, measured using flow cytometry.
  • FIG. 3C is fluorescence distributions after transient expression and targeting of dCas9-effector fusions to the TetO sites upstream of the reporter gene (+) or to a safe-targeting control site ( ⁇ ) for 5 days. Means are from two biological replicates. After targeting the dCas9-effector fusions for 5 days, silenced cells were sorted, and memory dynamics was measured by flow cytometry throughout 40 days (FIG. 3D).
  • FIGS. 4A-4G show nanobodies as signal detection and recording tools.
  • FIG. 4A is a schematic of an exemplary device for measuring and recording signal duration.
  • the input signal is coupled to the recruitment of rTetR-antiDNMT1 near the pEF promoter to silence an output gene.
  • FIG. 4B is a graph of the percentages of cells with TagRFP reporter silenced as measured by flow cytometry at the end of the indicated dox signal durations in a cell line stably expressing rTetR-antiDNMT1. Means and standard deviations (SDs) of experimental data from three replicates (red dots) and linear fit (black).
  • FIG. 4C shows the percentage of cells with TagRFP silenced after different signal (dox treatment) durations: 14 days (top) and 5 days (bottom). The gray shaded regions (negative numbers) indicate the period with dox.
  • FIG. 4D is a schematic of the three-state model of silencing by antiDNMT1 during recruitment (+dox, top) and during release ( ⁇ dox, bottom).
  • FIG. 4E is a graph of the percentage of cells irreversibly silent after different durations of recruitment (dox treatment) predicted by the model in FIG. 4D plotted as a black line. Experimental data recorded at 7 days during the release period shown as black dots if used for model fitting (14 days), or blue diamonds if not used in the fit.
  • FIG. 4D is a schematic of the three-state model of silencing by antiDNMT1 during recruitment (+dox, top) and during release ( ⁇ dox, bottom).
  • FIG. 4E is a graph of the percentage of cells irreversibly silent after different durations of recruitment (dox treatment) predicted by the model in FIG. 4D plotted as a black line. Experimental data recorded at 7 days during the release period shown as black dots if used for model fitting (14 days), or blue diamonds if not used in the fit.
  • FIG. 4D is a schematic
  • FIG. 4F shows the percentages of cells silenced relative to no dox controls for pulsed recruitment (top: 3 days +dox, 2 days ⁇ dox, 2 days +dox) compared to continuous recruitment for the same duration (bottom: 5 days +dox, 2 days ⁇ dox).
  • FIG. 4G shows the percentages of cells silenced relative to no dox controls for pulsed recruitment vs. continuous recruitment, recorded at the same time after dox removal, plotted as in FIG. 4F.
  • FIGS. 5A and 5B show schematics for a platform for selecting nanobodies capable of gene regulation in human cells.
  • FIG. 5A left shows yeast cells (black circles) carrying a diverse library of genetically encoded nanobodies (McMahon et al. Nature Structural & Molecular Biology Volume 25, pages 289–296 (2016), incorporated herein by reference in its entirety). Each yeast cell expresses and displays a single nanobody on its surface (colored lollipops). Yeast cells are mixed with FLAG-tagged chromatin regulator (CR) complexes (brown) purified from human cells.
  • CR chromatin regulator
  • FIG. 5A right shows the extraction of genomic DNA from the enriched yeast library, amplification of the DNA sequences encoding for the selected nanobodies, and recloning as a pool into a human expression lentiviral vector (Lentiviral Nanobody Library).
  • An exemplary lentiviral cloning vector is shown on the bottom, where each nanobody in the library is cloned downstream of the DNA binding domain rTetR.
  • FIG. 5B shows human cells infected with the pooled nanobody lentiviral library (hexagons), at a low infection rate, such that each cell expresses a single nanobody.
  • Each nanobody is expressed as a fusion protein with the DNA binding domain rTetR, and in the presence of doxycycline (+dox) can bind to the TetO sites upstream of a pEF promoter driving the expression of a reporter gene (Tycko J. et al., Cell. 2020 Dec 23;183(7):2020-2035, incorporated herein by reference in its entirety).
  • the reporter gene consists of a citrine fluorophore and a surface marker used for magnetic separation.
  • FIGS. 6A-6D show that yeast display against DNMT3A results in nanobody library enrichment.
  • FIG. 6A is a schematic showing experimental workflow: the naive nanobody library and the enriched libraries after each round of selection against DNMT3A are extracted from yeast using a genomic DNA extraction kit and sequenced using NGS Illumina sequencing (NGS seq).
  • FIG. 6B is graphs of the results of NGS sequencing showing the number of nanobodies in each library and their respective frequencies.
  • FIG. 6C is a schematic showing yeast display setup for measuring yeast library enrichment against the DNMT3A complex by flow cytometry. Yeast cells from Round 3 of yeast display selection against DNMT3A are incubated with cell lysate from human HEK-293T cells expressing FLAG- tagged DNMT3A (as well as other human contaminant proteins). Addition of antiFLAG antibody conjugated with the fluorescent dye FITC stains the yeast cells that bind the DNMT3A.
  • FIGS. 7A-7E show the characterization of individual antiDNMT3A nanobodies for gene silencing in human cells. Twenty-four nanobodies (see Table 2 for sequences) were chosen at random from the Lentiviral Nanobody Library cloned after Round 3 enrichment against DNMT3A.
  • FIG. 7A is a western blot showing co-immunoprecipitation of each rTetR-3xFlag- Nanobody and 3xHA-DNMT3A in HEK-293T cells.
  • FIGS. 7B shows the percentage of cells with the citrine reporter silenced (off) in (top) K562 and (bottom) HEK-293T cells after 5 days of dox-mediated recruitment of fusions between rTetR and each of the 24 nanobodies, as measured by flow cytometry.
  • rTetR without a nanobody is used as a negative control (NC).
  • the rTetR- KRAB is used as a positive control for repression.
  • Nanobody #8 has the strongest repressive effect.
  • FIGS. 7C, 7D, and 7E left are graphs showing the percentage of cells with the citrine reporter off after recruitment of rTetR, rTetR-KRAB, rTetR-nanobody2 (NB#2), and rTetR- nanobody8 (NB#8) for 3 days (blue) or 5 days (red) in K562 (FIG. 7C), HEK-293 (FIG. 7D), and Hela cells (FIG. 7E).
  • FIG. 8A and 8B show NB #8 can silence gene expression when fused to dCas9.
  • FIG. 8A is a graph of GFP fluorescent distributions of an pSV40-GFP reporter as measured by flow cytometry after recruitment of NB#8-dCas9 with a sgRNA that targets the pSV40 promoter (red).
  • dCas9 alone black
  • KRAB-dCas9+sgRNA is a positive control (CRISPRi).
  • FIG. 8B is a graph of the percentages of cells with pSV40-GFP silenced calculated from flow cytometry distributions upon targeting of the pSV40 promoter with the indicated fusions. [0038]
  • FIGS. 9A-9C show that NB#8 recruitment can lead to epigenetic memory in certain cell types.
  • Percentages of HEK-293 (FIG. 9A), K562 (FIG. 9B), and Hela (FIG. 9C) cells with the citrine reporter silenced are calculated from flow cytometry distributions during 5 days of dox-mediated recruitment (Dox+) and after release (Dox-) of rTetR fused NB#8 or NB#2.
  • rTetR without a nanobody is used as a negative control (NC).
  • the rTetR-KRAB is used as a positive control.
  • 10A-10D show that the disclosed high-throughput recruitment assay can identify antiDNMT3A nanobodies that repress gene expression in human cells.
  • the entire Lentiviral Nanobody Library obtained by pooled Golden Gate cloning of the Round 3 nanobodies selected against DNMT3A was used to produce lentivirus and infect HEK-293 cells at a low MOI (FIG. 10A).
  • Cells were selected with puromycin for integration of the nanobody constructs and dox was added for 5 days to induce recruitment of each nanobody upstream of a fluorescent-magnetic reporter gene, as described in FIG. 5B. Cells with the gene OFF were separated from those that remained ON using magnetic separation.
  • Nanobody NB#8 pink star
  • NB#25, NB#26, NB#27, and NB#28 also shown as stars.
  • FIG. 10B shows the validation of individual anti-DNMT3A nanobodies recovered from the high-throughput screen in HEK-293 cells. Gene blocks encoding for each nanobody sequence were individually cloned as fusions with rTetR and were delivered to HEK-293 cells by lentivirus to perform silencing experiments. All 5 nanobodies efficiently silence gene expression when recruited to the reporter by dox addition for 2 (green) or 5 (red) days compared to the no dox controls (black). Correlation between log2(OFF:ON) screen scores and the fraction of cells with citrine off as measured individually by flow cytometry is shown in FIG.
  • FIGS. 7B and 10B show all nanobodies listed in FIGS. 7B and 10B. Nanobodies were collapsed on sequence identity when applicable, and nanobodies with too few sequencing reads to compute reliable screen scores were excluded from this analysis.
  • FIG. 10D is multiple sequence alignment with the five validated nanobodies that exhibit strong repressive activity. CDR locations are indicated. Nanobody NB#8, NB#25, NB#26, NB#27, and NB#28 are SEQ ID NOs: 16-20, respectively. Concensus sequence is SEQ ID NO: 5932. [0040] FIGS 11A-11F show localization dynamics of GFP-tagged chromatin regulators and recruitment GFP-tagged chromatin regulators.
  • FIG. 11A Time-lapse images of cells transiently expressed with GFP-tagged HP1 ⁇ (FIG. 11A) and HDAC5 (FIG. 11B).
  • Cells undergoing cell division are represented at time 0 hours. Yellow boxes highlight the re-entry of GFP-tagged chromatin regulators into the nucleus.
  • Time-lapse experiment from 1 biological replicate White scale bars represent 10 ⁇ m.
  • FIG. 11C is a representative example of mIFP and GFP gating for data in FIG. 1D.
  • FIG. 11D shows the data from FIG. 1D gated based on different GFP expression levels and analyzed for the percentage of cells with the TagRFP reporter silenced at day 4 of dox.
  • FIG. 11C is a representative example of mIFP and GFP gating for data in FIG. 1D.
  • FIG. 11D shows the data from FIG. 1D gated based on different GFP expression levels and analyzed for the percentage of cells with the TagRFP reporter silenced at day 4 of dox.
  • FIG. 11E is a graph of the percentage of cells with reporter silenced after recruitment of an 8x repeat array of antiGFP nanobodies (+dox for 5 days).
  • FIG. 11F is a graph of the percentage of cells with TagRFP silenced after four days of recruitment with antiGFP (grey) or antiHP1 nanobody (orange).
  • FIGS. 12A-12E show targeted bisulfite sequencing controls and treatment of cells with DNA or histone methylation inhibitors.
  • FIG. 12A shows representative sorting for targeted bisulfite sequencing after 5 days of recruitment with antiDNMT1, DNMT1, and DNMT3B (left) and in the absence of dox (right).
  • FIG. 12A shows representative sorting for targeted bisulfite sequencing after 5 days of recruitment with antiDNMT1, DNMT1, and DNMT3B (left) and in the absence of dox (right).
  • 12B is the targeted bisulfite sequencing of two control genes, IGF2 (silent gene with imprinted DNA methylation) and ACTB (active gene, no DNA methylation expected) in the same cell populations as in FIG. 2C. Also included are non- methylated DNA controls from the human HCT116 DKO cell line that contains knockouts of DNA methyltransferases DNMT1 and DNMT3B. Methylated DNA was obtained from the non- methylated HCT116 DKO genomic DNA by in vitro treatment with the M. SssI methyltransferase.
  • FIG. 12E is a schematic for an exemplary expression vector for H2B-mIFP and the rTetR-antiDNMT1- antiHP1 fusion under a pGK constitutive promoter with sizes of the DNA encoding for the nanobodies shown in base pairs (bp).
  • FIGS. 13A and 13B show transient expression and recruitment of nanobodies can also silence gene expression and confer memory.
  • FIG. 13A is graphs of reporter fluorescent distributions(left) and percent cells silent (right) after transient expression of rTetR-effector fusions and 5 days of dox treatment.
  • FIG. 14A-14E show separate co-recruitment of regulators to the reporter gene and CXCR4 endogenous gene silencing.
  • FIG. 14A is a schematic of an expression vector for rTetR- KRAB-antiDNMT1 compared to the previously published KRAB-DNMT3A-3L fusion.
  • FIG. 14B is a graph of the percent cells with reporter silenced (relative to no dox controls) after co- recruitment of separate fusion of rTetR-effectors at the TagRFP reporter gene. Experimental setup is the same as in FIGS. 2A and 2D. Included for reference are the percentages of cells permanently silenced after individual recruitment with KRAB, antiDNMT1, or antiHP1 (dashed lines) taken from FIG. 2E.
  • FIG. 1 is a schematic of an expression vector for rTetR- KRAB-antiDNMT1 compared to the previously published KRAB-DNMT3A-3L fusion.
  • FIG. 14B is a graph
  • FIG. 14C is a schematic of an expression vector for dCas9-KRAB- antiDNMT1 under a CMV constitutive promoter.
  • the sgRNA targeting either the TetO site or a safe genomic site with no annotated function
  • FIG. 14D is a diagram of sgRNA binding sites for the targeting of dCas9-KRAB-antiDNMT1 to the endogenous CXCR4 gene.
  • CXCR4 is a cell surface transmembrane protein, which enables us to use conjugated fluorescent antibodies with flow cytometry to quantify gene expression.
  • sgRNAs Five sgRNAs were cloned spanning the upstream region of the transcriptional start site of this gene, targeting either the template or non- template strand.
  • the dCas9 and sgRNA constructs were modified to express mCitrine and mCherry, respectively, to allow for cell sorting. After transient expression and targeting at the endogenous CXCR4 gene for 4 days, cells were sorted for the presence of both dCas9 (mCitrine positive) and sgRNA (mCherry positive). Cells were then immunostained for CXCR4 expression and analyzed by flow cytometry (FIG. 14E, left). Means of percent cells with silent CXCR4 from 2 replicates are shown throughout 17 days after sorting (FIG.
  • FIG. 16A is a graph of predictions of 3-state silencing model for pulsed recruitment with different values of the second lag time between dox addition and start of silencing (Tlag2).
  • FIGS. 17A-17D shows generation of cells with an epigenetically silenced citrine reporter gene.
  • FIGS. 17B-17D are flow cytometry measurements of citrine levels histograms (top) and cell forward scatter against citrine levels (bottom) for wild type HEK293A cells without a reporter(FIG.
  • FIGS. 18A-18C show the yeast display screen against TET1/2/3 complexes.
  • FIG. 18A is a schematic of the yeast display was performed against chromatin regulator complexes containing full length, FLAG-tagged human TET1, mouse TET2, and human TET3 that were overexpressed in HEK293T cells and immunoprecipitated using anti-FLAG coated magnetic beads.
  • the same protocol was used as for yeast display against, except only two rounds of selection were performed. Briefly, yeast cells containing a na ⁇ ve nanobody library were mixed with the FLAG-TET complexes, and two rounds of selection were performed using anti-FLAG magnetic beads.
  • FIG. 18B shows measurement of nanobody enrichment. Yeast cells were mixed with the FLAG-TET1/2/3 complexes and stained with fluorescently-labeled anti-FLAG antibody (488nm wavelength, y axis).
  • the x axis measures nanobody expression using an anti-HA antibody (647nm wavelength). An enrichment of anti-FLAG signal (in quadrant Q2) is observed after round 1 and round 2 of selection, indicating successful enrichment for nanobodies that can bind to the TET1/2/3 complexes.
  • FIG. 18C genomic DNA from the na ⁇ ve and round 2 nanobody libraries was sequenced and the frequency of each detected nanobody was quantified. An increased frequency of a subset of nanobodies was observed, indicating the size of the library decreased due to the yeast display selection. The round 2 nanobodies against TET1/2/3 complexes were used for further studies in human cells. [0048] FIGS.
  • FIG. 19A-19D show that a high-throughput recruitment assay can identify anti- Tet1/2/3 complex nanobodies that activate gene expression in human cells.
  • the library of nanobodies against TET1/2/3 complexes from round 2 of yeast display were cloned fused to rTetR, and delivered via lentivirus to HEK293A cells containing the epigenetically silenced reporter (FIG. 19A). Viral delivery was performed at low MOI, to ensure delivery of a single nanobody per cell. After dox addition, each nanobody is recruited to the silenced reporter, and can activate the gene. Cells are then separated into re-activated (citrine ON) and still-silenced (OFF). Nanobodies that reactivate the gene are enriched in the ON population.
  • FIG. 19B is a graph of Log2(ON:OFF) scores per replicate of the anit-TET1/2/3 complex nanobody recruitment screen in HEK293 cells. Higher scores indicate enrichment of a particular nanobody sequence in the ON population. Nanobodies chosen for follow-up are denoted as stars.
  • 19D is a timecourse of dox-mediated activation starting from day 3 of dox addition (nanobody recruitment at the silenced reporter), plotted as fraction of cells that have the reporter re-activated (citrine ON). Gray curve at the bottom indicates stable epigenetic silencing of the citrine reporter with the rTetR alone control (no nanobody).
  • nanobodies also referred to as single-domain antibodies
  • CRs chromatin regulators
  • the target gene or nucleic acid are in a cell and the methods comprise introducing into a cell (e.g., administering to a subject) the disclosed fusion proteins comprising the nanobody and the DNA binding proteins, or domains or functional fragments thereof, or a nucleic acid thereof to allow recruitment endogenous CRs from the existing cellular chromatin network, thus obviating the need for introducing a large exogenous chromatin remodeling complex.
  • the methods and system described herein enhance the functionality of commonly used transcriptional effectors, e.g., KRAB.
  • combining nanobodies together or with other regulators, such as DNMT3A or KRAB may enhance silencing speed and epigenetic memory.
  • This technology leverages a yeast nanobody library and a high-throughput screen to characterize tens of thousands of potential nanobodies. Multiple rounds of negative and positive selection can be used to reduce the yeast nanobody library size to a few thousand members for use in the high- throughput screening platform, as necessary. Since only a couple of nanobodies against chromatin regulators exist, the disclosed methods allow identification of human chromatin regulator complexes that act as efficient modulators of gene expression in human cells for the development of efficient genetic engineering tools.
  • Exemplary applications for the methods disclosed herein include, but are not limited to: targeted chromatin remodeling with persistent memory, gene and cell therapy (e.g., to silence a pathogenic transcript in a patient) or in research, and use in genetic circuits, e.g., inducible gene expression or more complex circuits. These circuits find use in gene therapy (e.g., AAV delivery of antibodies) and cell therapy (e.g., ex vivo engineering of CAR-T cells) to achieve therapeutic gene expression outputs in response to environmental and small molecule inputs.
  • gene therapy e.g., AAV delivery of antibodies
  • cell therapy e.g., ex vivo engineering of CAR-T cells
  • antibody refers to a protein that is endogenously used by the immune system to identify and neutralize foreign objects, such as bacteria and viruses.
  • an antibody is a protein that comprises at least one complementarity determining region (CDR).
  • the CDRs form the “hypervariable region” of an antibody, which is responsible for antigen binding (discussed further below).
  • a whole antibody typically consists of four polypeptides: two identical copies of a heavy (H) chain polypeptide and two identical copies of a light (L) chain polypeptide.
  • Each of the heavy chains contains one N-terminal variable (V H ) region and three C-terminal constant (C H1 , C H2 , and C H3 ) regions
  • each light chain contains one N-terminal variable (V L ) region and one C-terminal constant (C L ) region.
  • the light chains of antibodies can be assigned to one of two distinct types, either kappa ( ⁇ ) or lambda ( ⁇ ), based upon the amino acid sequences of their constant domains.
  • each light chain is linked to a heavy chain by disulfide bonds, and the two heavy chains are linked to each other by disulfide bonds.
  • the light chain variable region is aligned with the variable region of the heavy chain, and the light chain constant region is aligned with the first constant region of the heavy chain.
  • the remaining constant regions of the heavy chains are aligned with each other.
  • the variable regions of each pair of light and heavy chains form the antigen binding site of an antibody.
  • the V H and V L regions have the same general structure, with each region comprising four framework (FW or FR) regions.
  • framework region refers to the relatively conserved amino acid sequences within the variable region which are located between the CDRs.
  • the framework regions form the ⁇ sheets that provide the structural framework of the variable region (see, e.g., C. A. Janeway et al. (eds.), Immunobiology, 5th Ed., Garland Publishing, New York, N.Y. (2001)).
  • the framework regions are connected by three CDRs. As discussed above, the three CDRs, known as CDR1, CDR2, and CDR3, form the “hypervariable region” of an antibody, which is responsible for antigen binding.
  • the CDRs form loops connecting, and in some cases comprising part of, the beta-sheet structure formed by the framework regions. While the constant regions of the light and heavy chains are not directly involved in binding of the antibody to an antigen, the constant regions can influence the orientation of the variable regions. The constant regions also exhibit various effector functions, such as participation in antibody-dependent complement-mediated lysis or antibody-dependent cellular toxicity via interactions with effector molecules and cells.
  • fragment of an antibody “antibody fragment,” and “antigen-binding fragment” of an antibody are used interchangeably herein to refer to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (see, generally, Holliger et al., Nat.
  • the antibody fragment desirably comprises, for example, one or more CDRs, the variable region (or portions thereof), the constant region (or portions thereof), or combinations thereof.
  • antibody fragments include, but are not limited to, (i) a Fab fragment, which is a monovalent fragment consisting of the V L , V H , C L , and C H1 domains, (ii) a F(ab’) 2 fragment, which is a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region, (iii) a Fv fragment consisting of the V L and V H domains of a single arm of an antibody, (iv) a Fab’ fragment, which results from breaking the disulfide bridge of an F(ab’) 2 fragment using mild reducing conditions, (v) a disulfide-stabilized Fv fragment (dsFv), and (vi) a domain antibody (dAb), which is an antibody single variable region domain (V H or V L ) polypeptide that specifically binds antigen.
  • a Fab fragment which is a monovalent fragment consisting of the V L , V H , C L , and C H1
  • a “nanobody,” as used herein, refers to polypeptides comprising the variable region of a heavy chain of an antibody.
  • a nanobody is functionally similar to a single domain antibody with only one heavy chain variable region.
  • the antigen-binding properties of a nanobody can be described by three variable regions (CDRs) divided by four framework regions (FRs) with the general structure as shown below: FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4 in which FR1 to FR4 refer to framework regions 1 to 4, respectively, and in which CDR1 to CDR3 refer to the complementarity determining regions 1 to 3.
  • CDRs three variable regions
  • FRs framework regions
  • the CDRs normally form a loop structure in which the ⁇ -sheets formed by the FRs therebetween are spatially close to each other, constituting the antigen-binding site of the nanobody.
  • the amino acid sequences of the same type of nanobodies can be compared to determine which amino acids constitute the FR or CDR regions.
  • the present invention includes not only intact nanobodies but also fragment(s) of immunologically active nanobody or fusion protein(s) formed from nanobodies with other sequences. Therefore, the present invention also includes fragments, derivatives, and analogs of the nanobodies.
  • nucleic acid or a “nucleic acid sequence” refers to a polymer or oligomer of pyrimidine and/or purine bases, preferably cytosine, thymine, and uracil, and adenine and guanine, respectively (See Albert L. Lehninger, Principles of Biochemistry, at 793- 800 (Worth Pub. 1982)).
  • the present technology contemplates any deoxyribonucleotide, ribonucleotide, or peptide nucleic acid component, and any chemical variants thereof, such as methylated, hydroxymethylated, or glycosylated forms of these bases, and the like.
  • the polymers or oligomers may be heterogenous or homogenous in composition and may be isolated from naturally occurring sources or may be artificially or synthetically produced.
  • the nucleic acids may be DNA or RNA, or a mixture thereof, and may exist permanently or transitionally in single-stranded or double-stranded form, including homoduplex, heteroduplex, and hybrid states.
  • a nucleic acid or nucleic acid sequence comprises other kinds of nucleic acid structures such as, for instance, a DNA/RNA helix, peptide nucleic acid (PNA), morpholino nucleic acid (see, e.g., Braasch and Corey, Biochemistry, 41(14): 4503-4510 (2002)) and U.S. Pat.
  • LNA locked nucleic acid
  • cyclohexenyl nucleic acids see Wang, J. Am. Chem. Soc., 122: 8595-8602 (2000), and/or a ribozyme.
  • nucleic acid or “nucleic acid sequence” may also encompass a chain comprising non-natural nucleotides, modified nucleotides, and/or non- nucleotide building blocks that can exhibit the same function as natural nucleotides (e.g., “nucleotide analogs”); further, the term “nucleic acid sequence” as used herein refers to an oligonucleotide, nucleotide or polynucleotide, and fragments or portions thereof, and to DNA or RNA of genomic or synthetic origin, which may be single or double- stranded, and represent the sense or antisense strand.
  • nucleic acid refers to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof.
  • a “peptide” or “polypeptide” is a linked sequence of two or more amino acids linked by peptide bonds.
  • the peptide or polypeptide can be natural, synthetic, or a modification or combination of natural and synthetic.
  • Polypeptides include proteins such as binding proteins, receptors, and antibodies. The proteins may be modified by the addition of sugars, lipids or other moieties not included in the amino acid chain.
  • polypeptide and “protein,” are used interchangeably herein.
  • percent sequence identity refers to the percentage of nucleotides or nucleotide analogs in a nucleic acid sequence, or amino acids in an amino acid sequence, that is identical with the corresponding nucleotides or amino acids in a reference sequence after aligning the two sequences and introducing gaps, if necessary, to achieve the maximum percent identity.
  • additional nucleotides in the nucleic acid, that do not align with the reference sequence are not taken into account for determining sequence identity.
  • a number of mathematical algorithms for obtaining the optimal alignment and calculating identity between two or more sequences are known and incorporated into a number of available software programs. Examples of such programs include CLUSTAL-W, T-Coffee, and ALIGN (for alignment of nucleic acid and amino acid sequences), BLAST programs (e.g., BLAST 2.1, BL2SEQ, and later versions thereof) and FASTA programs (e.g., FASTA3x, FASTM, and SSEARCH) (for sequence alignment and sequence similarity searches). Sequence alignment algorithms also are disclosed in, for example, Altschul et al., J. Molecular Biol., 215(3): 403-410 (1990), Beigert et al., Proc. Natl. Acad.
  • a “vector” or “expression vector” is a replicon, such as plasmid, phage, virus, or cosmid, to which another DNA segment may be attached or incorporated so as to bring about the replication of the attached segment in a cell.
  • Method for identifying and selecting nanobodies Disclosed herein are methods for identifying and selecting nanobodies. The methods can be utilized to identify nanobodies against different endogenous protein targets including but not limited to: transcription or translation factors, chromatin regulators, or any protein that modulates cellular processes for which reporters are easily available, including but not limited to: DNA repair, mRNA or protein stability, and immune responses.
  • the methods may comprise any or all of: preparing a yeast nanobody library comprising yeast cells each displaying a single nanobody on its surface; mixing the yeast nanobody library with a protein target wherein the protein target is configured to bind to a solid surface; separating yeast cells bound to the protein target from unbound yeast cells; amplifying nanobody DNA sequences from the yeast cells bound to protein target; cloning the nanobody sequences into a human lentiviral vector to prepare a nanobody lentiviral library; and identifying the nanobody as a binding protein for the protein target based on a functional output.
  • the methods are directed to identifying and selecting nanobodies for a target chromatin regulator.
  • the chromatin regulator comprises a methylase, a demethylase, an acetylase, or ATP-dependent chromatin remodeling complex.
  • the methods comprise: preparing a yeast nanobody library comprising yeast cells each displaying a single nanobody on its surface; mixing the yeast nanobody library with a target chromatin regulator wherein the target chromatin regulator is configured to bind to a surface (e.g., microparticle, plate, membrane); separating yeast cells bound to target chromatin regulator from unbound yeast cells; amplifying nanobody DNA sequences from the yeast cells bound to target chromatin regulator; cloning the nanobody sequences into a human lentiviral vector to prepare a nanobody lentiviral library, wherein each nanobody DNA sequence is expressed as a fusion protein with a DNA binding domain; transforming the nanobody lentiviral library into reporter cells, wherein a reporter cell comprises a two-part reporter gene comprising a surface marker and a fluorescent protein
  • the methods further comprise repeating the preparing a yeast nanobody library, mixing the yeast nanobody library with a target chromatin regulator, separating yeast cells bound to target chromatin regulator from unbound yeast cells, and amplifying nanobody DNA sequences from the yeast cells bound to target chromatin regulator one or more times.
  • the DNA binding domain is an inducible DNA binding domain.
  • the inducible DNA binding domain may use any system for induction of DNA binding, including, but not limited to, tetracycline Tet,/DOX inducible systems, light inducible systems, Abscisic acid (ABA) inducible systems, cumate systems, 40HT/estrogen inducible systems, ecdysone-based inducible systems, and FKBP12/FRAP (FKBP12-rapamycin complex) inducible systems.
  • the methods may further comprise treating the reporter cells with an agent configured to induce the inducible DNA binding domain for a length of time.
  • the reporter cells are treated with the agent at least 24 hours.
  • the reporter cells may be treated with the agent for at least 24 hours, at least 36 hours, at least 48 hours (2 days), at least 3 days, at least 4 days, at least 5 days, at least 6 days, at least 7 days, at least 8 days, at least 9 days, at least 10 days, at least 14 days, or more.
  • the reporter cells at treated with the agent for 3-12 days, 3-10 days, 3-7 days, or 3- 5 days.
  • the inducible DNA binding domain comprises a tag.
  • the tag may include any tag known in the art, including tags removable by chemical or enzymatic means.
  • Suitable tags for use in the present method include chitin binding protein (CBP), maltose binding protein (MBP), Strep-tag, glutathione-S-transferase (GST), a polyhistidine (PolyHis) tag, an ALFA-tag, a V5-tag, a Myc-tag, a hemagglutinin(HA)-tag, a Spot-tag, a T7-tag, an NE-tag, a Calmodulin-tag, a polyglutamate tag, a polyarginine tag, a FLAG tag, and the like.
  • CBP chitin binding protein
  • MBP maltose binding protein
  • GST glutathione-S-transferase
  • PolyHis polyhistidine
  • ALFA-tag ALFA-tag
  • V5-tag V5-tag
  • Myc-tag a hemagglutinin(HA)-tag
  • Spot-tag a T7-tag
  • the methods comprise transforming reporter cells with the domain library, wherein the reporter cell comprises a two-part reporter gene comprising a surface marker and a fluorescent protein under the control of a promoter.
  • the two-part reporter gene is capable of being modulated by the target chromatin regulator following treatment with an agent configured to induce the inducible DNA binding domain and allowing recruitment of the chromatin regulator by the putative nanobody.
  • the promoter may confer a high rate of transcription (a strong promoter) or confer a low rate of transcription (weak promoter). Many promoter libraries have been established experimentally and choice of promoter and promoter strength is dependent on cell type. In some embodiments, when identifying nanobodies configured to bind chromatin activators, a weak promoter may be used.
  • a strong promoter when identifying nanobodies configured to bind chromatin repressors, a strong promoter may be used.
  • Cell surface markers include proteins and carbohydrates which are attached to the cellular membrane. Cell surface markers are generally known in the art for a variety of cell types and can be expressed in a reporter cell of choice based on known molecular biology methods.
  • the surface marker may be a synthetic surface marker comprising marker polypeptide attached to a transmembrane domain.
  • the marker polypeptide may include an antibody or a fragment thereof (e.g., Fc region) attached to a transmembrane domain.
  • the marker polypeptide is human IgG1 Fc region and the synthetic surface marker comprises human IgG1 Fc region attached to a transmembrane domain.
  • Fluorescent proteins are well known in the art and include proteins adapted to fluoresce in various cellular compartments and as a result of varying wavelengths of incoming light. Examples of fluorescent proteins include: phycobiliproteins, cyan fluorescent protein (CFP), green fluorescent protein (GFP), yellow fluorescent protein (YFP or citrine), enhanced orange fluorescent protein (OFP), enhanced green fluorescent protein (eGFP), modified green fluorescent protein (emGFP), enhanced yellow fluorescent protein (eYFP) and/or monomeric red fluorescent protein (mRFP) and derivatives and variants thereof.
  • CFP cyan fluorescent protein
  • GFP green fluorescent protein
  • YFP or citrine yellow fluorescent protein
  • OFP enhanced orange fluorescent protein
  • eGFP enhanced green fluorescent protein
  • emGFP modified green fluorescent protein
  • eYFP enhanced yellow fluorescent protein
  • mRFP monomeric red fluorescent protein
  • the methods comprise separating reporter cells based on presence or absence of the surface marker, the fluorescent protein, or a combination thereof.
  • a number of cell separation techniques are known in the art are suitable for use with the methods disclosed herein, including, for example, immunomagnetic cell separation, fluorescent-activated cell sorting (FACS), and microfluidic cell sorting.
  • cell separation comprises immunomagnetic cell separation.
  • the chromatin regulator is identified as a functional chromatin repressor (e.g., able to bind to the nanobody and modulate transcription and/or epigenetic memory) when recruited by the nanobody due to enrichment of cells with an absence of the surface marker, the fluorescent protein, or a combination thereof.
  • the chromatin regulator is identified as a functional chromatin activator when recruited by the nanobody due to enrichment of cells with a presence of the surface marker, the fluorescent protein, or a combination thereof.
  • the present disclosure also provides fusion proteins comprising a DNA binding protein, DNA binding domain, or a functional fragment thereof covalently linked to a nanobody configured to bind a chromatin regulator.
  • the fusion protein comprises two nanobodies configured to bind the same or different chromatin regulator. The two nanobodies may be linked by a covalent linker.
  • the term “chromatin regulator” refers to a protein or polypeptide that interacts with, directly or indirectly, specific DNA sequences to modify histones, DNA, or histone-DNA complexes (e.g., through methylation, acetylation, phosphorylation, adenosine diphosphate–ribosylation, glycosylation, sumoylation, or ubiquitylation or remodel DNA-histone structure with energy from ATP hydrolysis).
  • the chromatin regulator comprises a methylase, a demethylase, an acetylase, or ATP-dependent chromatin remodeling complex.
  • one or more of the at least one nanobody is identified and/or selected by the methods described herein.
  • the nanobody is configured to bind DNA (cytosine-5)- methyltransferase 3A (DNMT3A) complexes.
  • the nanobody configured to bind DNMT3A complexes may comprise, consist essentially of, or consist of any of the amino acid sequences shown in FIG. 10D, or fragments thereof.
  • the DNMT3A complex nanobody comprises at least one CDR comprising an amino acid sequence as shown in the consensus sequences in FIG. 10D.
  • the at least one CDR may be selected from the group consisting of GTIFXXXXM (SEQ ID NO: 13), EZVAXIXXGXXTNY (SEQ ID NO: 14), and AAXXXXXYYXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid.
  • the DNMT3A complex nanobody comprises a first CDR comprising an amino acid sequence of GTIFXXXM (SEQ ID NO: 13), a second CDR comprising an amino acid sequence of EZVAXIXXGXXTNY (SEQ ID NO: 14), and a third CDR comprising an amino acid sequence of AAXXXXYYXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid.
  • the DNMT3A complex nanobody comprises at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), GNIFDGASM (SEQ ID NO: 4), EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), AAGRYYYPGHGY (SEQ ID NO: 8), AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 12).
  • GTIFAHSRM SEQ ID NO: 1
  • GTISSDGYM SEQ ID NO: 2
  • GTIFYFFGM SEQ ID NO:
  • the DNMT3A complex nanobody comprises a first CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), and GNIFDGASM (SEQ ID NO: 4); a second CDR comprising an amino acid sequence selected from the group consisting of EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), and AAGRYYYPGHGY (SEQ ID NO: 8); a third CDR comprising an amino acid sequence selected from the group consisting of AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY
  • GTIFAHSRM S
  • the DNMT3A complex nanobody comprises, consists essentially of, or consists of an amino acid sequence at least 70% (e.g., at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98% or at least 99%) similar to any of SEQ ID NOs: 16-20.
  • the DNMT3A complex nanobody comprises, consists essentially of, or consists of an amino acid sequence at least 70% similar to any of SEQ ID NO: 5932.
  • the nanobody is configured to bind ten-eleven translocation methylcytosine 1/2/3 (TET1/2/3) complexes.
  • the nanobody configured to bind TET1/2/3 complexes may comprise, consist essentially of, or consist of any of the amino acid sequences of SEQ ID NOs: 5960-5968.
  • the nanobody configured to bind TET1/2/3 complexes comprises at least one CDR may be selected from the group consisting of GXIZ 1 XXXXM, EZ 2 VAXIXXGXXTZ 3 Y (SEQ ID NO: 5978), and AZ 4 XXXXYXXXXY, wherein Z 1 is S or F, Z 2 is F or L, Z 3 is N or Y, Z 4 is A or V, and each X is any amino acid.
  • the nanobody configured to bind TET1/2/3 complexes comprises a first CDR comprising an amino acid sequence of GXIZ 1 XXXXM, a second CDR comprising an amino acid sequence of EZ 2 VAXIXXGXXTZ 3 Y (SEQ ID NO: 5978), and a third CDR comprising an amino acid sequence of AZ 4 XXXXYXXXXY, wherein Z 1 is S or F, Z 2 is F or L, Z 3 is N or Y, Z 4 is A or V, and each X is any amino acid.
  • the TET1/2/3 complex nanobody comprises at least one CDR having an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933-5959.
  • the TET1/2/3 complex nanobody comprises a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933-5941; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5942-5950; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5951-5959; or a combination thereof.
  • the TET1/2/3 complex nanobody comprises a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5935 or 5936; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5944 or 5945; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5953- 5954; or a combination thereof.
  • the TET1/2/3 complex nanobody comprises, consists essentially of, or consists of an amino acid sequence at least 70% (e.g., at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98% or at least 99%) similar to any of SEQ ID NOs: 5960-5968.
  • the fusion protein further comprises a transcription factor or regulator (e.g., a transcriptional repressor, a transcriptional activator).
  • the transcription factor comprises Krüppel-associated box (KRAB).
  • KRAB Krüppel-associated box
  • DNMT3A binding protein encompasses proteins which bind DNMT3A and/or complexes comprising DNMT3A.
  • DNMT3A forms a catalytically active dimer in a DNMT3A:DNMT3L complex.
  • DNMT3A is capable of further homo- and hetero- oligomerization with DNMT3B.
  • DNMT3A forms complexes with proteins outside of the DNMT3 family. For example, a direct interaction between EZH2 of the Polycomb-repressive complex 2 (PRC2) and DNMT3A has been demonstrated.
  • PRC2 Polycomb-repressive complex 2
  • fusion proteins of the DNMT3A binding protein covalently linked to a DNA binding protein, DNA binding domain, or a functional fragment thereof are provided.
  • the DNMT3A binding protein is an antibody or a fragment thereof.
  • the DNMT3A binding protein is a nanobody.
  • the DNMT3A binding protein may comprise, consist essentially of, or consist of any of the amino acid sequences shown in FIG. 10D, or fragments thereof.
  • the DNMT3A binding protein comprises at least one CDR comprising an amino acid sequence as shown in the consensus sequences in FIG. 10D.
  • the at least one CDR may be selected from the group consisting of GTIFXXXXM (SEQ ID NO: 13), EZVAXIXXGXXTNY (SEQ ID NO: 14), and AAXXXXXYYXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid.
  • the DNMT3A binding protein comprises the at least one nanobody comprises a first CDR comprising an amino acid sequence of GTIFXXXXM (SEQ ID NO: 13), a second CDR comprising an amino acid sequence of EZVAXIXXGXXTNY (SEQ ID NO: 14), and a third CDR comprising an amino acid sequence of AAXXXXYYXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid.
  • the DNMT3A binding protein comprises at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), GNIFDGASM (SEQ ID NO: 4), EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), AAGRYYYPGHGY (SEQ ID NO: 8), AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 12).
  • GTIFAHSRM SEQ ID NO: 1
  • GTISSDGYM SEQ ID NO: 2
  • GTIFYFFGM SEQ ID NO: 3
  • the DNMT3A binding protein comprises a first CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), and GNIFDGASM (SEQ ID NO: 4); a second CDR comprising an amino acid sequence selected from the group consisting of EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), and AAGRYYYPGHGY (SEQ ID NO: 8); a third CDR comprising an amino acid sequence selected from the group consisting of AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 1), GT
  • the DNMT3A binding protein comprises, consists essentially of, or consists of an amino acid sequence at least 70% (e.g., at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98% or at least 99%) similar to any of SEQ ID NOs: 16-20.
  • the DNMT3A binding protein comprises, consists essentially of, or consists of an amino acid sequence at least 70% similar to any of SEQ ID NO: 5932.
  • a ten-eleven translocation methylcytosine 1/2/3 (TET1/2/3) complex binding protein is also provided.
  • fusion proteins of the TET1/2/3 complex binding protein covalently linked to a DNA binding protein, DNA binding domain, or a functional fragment thereof are provided.
  • the TET1/2/3 complex binding protein is an antibody or a fragment thereof.
  • the TET1/2/3 complex binding protein is a nanobody.
  • the nanobody is configured to bind ten-eleven translocation methylcytosine 1/2/3 (TET1/2/3) complexes.
  • the TET1/2/3 complex binding protein may comprise, consist essentially of, or consist of any of the amino acid sequences of SEQ ID NOs: 5960-5968.
  • the TET1/2/3 complex binding protein comprises at least one CDR may be selected from the group consisting of GXIZ 1 XXXXM, EZ 2 VAXIXXGXXTZ 3 Y (SEQ ID NO: 5978), and AZ 4 XXXXYXXXXY, wherein Z 1 is S or F, Z 2 is F or L, Z 3 is N or Y, Z 4 is A or V, and each X is any amino acid.
  • the TET1/2/3 complex binding protein comprises a first CDR comprising an amino acid sequence of GXIZ 1 XXXXM, a second CDR comprising an amino acid sequence of EZ 2 VAXIXXGXXTZ 3 Y (SEQ ID NO: 5978), and a third CDR comprising an amino acid sequence of AZ 4 XXXXYXXXXY, wherein Z 1 is S or F, Z 2 is F or L, Z 3 is N or Y, Z 4 is A or V, and each X is any amino acid. [00101] In some embodiments, the TET1/2/3 complex binding protein comprises at least one CDR having an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933- 5959.
  • the TET1/2/3 complex binding protein comprises a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933- 5941; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5942-5950; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5951-5959; or a combination thereof.
  • the TET1/2/3 complex binding protein comprises a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5935 or 5936; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5944 or 5945; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5953-5954; or a combination thereof.
  • the TET1/2/3 complex binding protein comprises, consists essentially of, or consists of an amino acid sequence at least 70% (e.g., at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98% or at least 99%) similar to any of SEQ ID NOs: 5960-5968.
  • the DNA binding protein, domain, or functional fragment thereof is any polypeptide which is capable of binding double- or single-stranded DNA, generally or with sequence specificity.
  • DNA binding proteins and domains include those polypeptides having helix-turn- helix motifs, zinc fingers, leucine zippers, HMG-box (high mobility group box) domains, winged helix region, winged helix-turn-helix region, helix-loop-helix region, immunoglobulin fold, B3 domain, Wor3 domain, TAL effector DNA-binding domain and the like.
  • the DNA binding proteins or domains may be a natural binding domain.
  • the DNA binding domain comprises a programmable DNA binding proteins or domains, e.g., a DNA binding protein or domain engineered, for example by altering one or more amino acid of a natural DNA binding protein or domain to bind to a predetermined nucleotide sequence.
  • the DNA binding protein, domain, or functional fragment thereof is capable of binding directly to the target DNA sequences.
  • the DNA binding protein, domain, or functional fragment thereof may be derived from domains found in naturally occurring Transcription activator-like effectors (TALEs), such as AvrBs3, Hax2, Hax3 or Hax4 (Bonas et al.1989. Mol Gen Genet 218(1): 127-36; Kay et al. 2005 Mol Plant Microbe Interact 18(8): 838-48).
  • TALEs have a modular DNA-binding domain consisting of repetitive sequences of residues; each repeat region consists of 34 amino acids.
  • the TALE DNA binding domains may be engineered using known methods to provide a DNA binding domain with chosen specificity for any target sequence.
  • the DNA binding domain may comprise multiple (e.g., 2, 3, 4, 5, 6, 10, 20, or more) Tal effector DNA-binding motifs.
  • any number of nucleotide-specific Tal effector motifs can be combined to form a sequence-specific DNA-binding domain to be employed in the present transcription factor.
  • the DNA binding protein, domain, or functional fragment thereof associates with the target DNA in concert with an exogenous factor.
  • the DNA binding protein, domain, or functional fragment thereof is an inducible DNA binding protein, domain, or functional fragment thereof, as described and exemplified elsewhere herein.
  • the DNA binding protein, domain, or functional fragment thereof is derived from a Clustered Regularly Interspaced Short Palindromic Repeats associated (Cas) protein (e.g., catalytically dead Cas9) and associates with the target DNA through a guide RNA.
  • Cas Clustered Regularly Interspaced Short Palindromic Repeats associated
  • the gRNA itself comprises a sequence complementary to one strand of the DNA target sequence and a scaffold sequence which binds and recruits Cas9 to the target DNA sequence.
  • the transcription factors described herein may be useful for CRISPR interference (CRISPRi) or CRISPR activation (CRISPRa).
  • the guide RNA may be a crRNA, crRNA/tracrRNA (or single guide RNA, sgRNA).
  • the gRNA may be a non-naturally occurring gRNA.
  • the terms “gRNA,” “guide RNA” and “guide sequence” may be used interchangeably throughout and refer to a nucleic acid comprising a sequence that determines the binding specificity of the Cas protein.
  • a gRNA hybridizes to (complementary to, partially or completely) the DNA target sequence.
  • the gRNA or portion thereof that hybridizes to the target nucleic acid (a target site) may be any length necessary for selective hybridization.
  • gRNAs or sgRNA(s) can be between about 5 and about 100 nucleotides long, or longer (e.g., 5, 6, 7, 8, 9, 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, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 5960, 61, 62, 63, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86
  • sgRNA(s) there are many publicly available software tools that can be used to facilitate the design of sgRNA(s); including but not limited to, Genscript Interactive CRISPR gRNA Design Tool, WU-CRISPR, and Broad Institute GPP sgRNA Designer.
  • Genscript Interactive CRISPR gRNA Design Tool WU-CRISPR
  • WU-CRISPR WU-CRISPR
  • Broad Institute GPP sgRNA Designer There are also publicly available pre-designed gRNA sequences to target many genes and locations within the genomes of many species (human, mouse, rat, zebrafish, C. elegans), including but not limited to, IDT DNA Predesigned Alt-R CRISPR-Cas9 guide RNAs, Addgene Validated gRNA Target Sequences, and GenScript Genome-wide gRNA databases.
  • the present disclosure also provides nucleic acids encoding a nanobody fusion protein, DNMT3A binding protein, DNMT3A binding protein fusion protein, TET1/2/3 complex binding protein, and TET1/2/3 complex binding protein fusion protein as disclosed herein.
  • the at least one CDR of the nanobody or DNMT3A binding protein may be encoded by a nucleic acid sequence of any of those disclosed in Table 2 (SEQ ID NOs: 21- 92), Table 3, or SEQ ID NOs: 93-5928.
  • the nanobody, nanobody fusion protein, or DNMT3A binding protein or fusion thereof comprises a nucleic acid sequence at least 70% similar to any of SEQ ID NOs: 21-5928.
  • the nanobody or DNMT3A binding protein may comprise a combination of two or three CDRs, each individually encoded by a nucleic acid sequence having at least 70% similar to any of SEQ ID NOs: 21-5928.
  • a single nanobody or DNMT3A binding protein may be encoded by a nucleic acid sequence comprising one, two, or three, individual sequences having at least 70% similar to any of SEQ ID NOs: 21-5928. See for example, those nanobodies listed in Table 3 here or in Table 3 of U.S. Provisional Application No. 63/242,898, incorporated by reference in its entirety.
  • the nanobody, nanobody fusion protein, or TET1/2/3 complex binding protein or fusion thereof comprises a nucleic acid sequence at least 70% similar to any of SEQ ID NOs: 5969-5977, or fragments thereof.
  • Nucleic acids of the present disclosure can comprise any of a number of promoters known to the art, wherein the promoter is constitutive, regulatable or inducible, cell type specific, tissue-specific, or species specific.
  • a promoter sequence of the invention can also include sequences of other regulatory elements that are involved in modulating transcription (e.g., enhancers, Kozak sequences and introns).
  • promoter/regulatory sequences useful for driving constitutive expression of a gene include, but are not limited to, for example, CMV (cytomegalovirus promoter), EF1a (human elongation factor 1 alpha promoter), SV40 (simian vacuolating virus 40 promoter), PGK (mammalian phosphoglycerate kinase promoter), Ubc (human ubiquitin C promoter), human beta-actin promoter, rodent beta-actin promoter, CBh (chicken beta-actin promoter), CAG (hybrid promoter contains CMV enhancer, chicken beta actin promoter, and rabbit beta- globin splice acceptor), TRE (Tetracycline response element promoter), H1 (human polymerase III RNA promoter), U6 (human U6 small nuclear promoter), and the like.
  • CMV cytomegalovirus promoter
  • EF1a human elongation factor 1 alpha promoter
  • SV40 simi
  • Additional promoters that can be used for expression of the components of the present system, include, without limitation, cytomegalovirus (CMV) intermediate early promoter, a viral LTR such as the Rous sarcoma virus LTR, HIV-LTR, HTLV-1 LTR, Maloney murine leukemia virus (MMLV) LTR, myeoloproliferative sarcoma virus (MPSV) LTR, spleen focus-forming virus (SFFV) LTR, the simian virus 40 (SV40) early promoter, herpes simplex tk virus promoter, elongation factor 1- alpha (EF1- ⁇ ) promoter with or without the EF1- ⁇ intron.
  • CMV cytomegalovirus
  • a viral LTR such as the Rous sarcoma virus LTR, HIV-LTR, HTLV-1 LTR, Maloney murine leukemia virus (MMLV) LTR, myeoloproliferative sarcoma virus (MPSV)
  • any regulatable promoter may be used, such that its expression can be modulated within a cell.
  • inducible expression can be accomplished by placing the nucleic acid encoding such a molecule under the control of an inducible promoter/regulatory sequence. Promoters that are well known in the art can be induced in response to inducing agents such as metals, glucocorticoids, tetracycline, hormones, and the like, are also contemplated for use with the invention. Thus, it will be appreciated that the present disclosure includes the use of any promoter/regulatory sequence known in the art that is capable of driving expression of the desired protein operably linked thereto.
  • the present disclosure also provides for vectors containing the nucleic acids and cells containing the nucleic acids or vectors, thereof.
  • the vectors may be used to propagate the nucleic acid in an appropriate cell and/or to allow expression from the nucleic acid (e.g., an expression vector).
  • an expression vector e.g., an expression vector.
  • the person of ordinary skill in the art would be aware of the various vectors available for propagation and expression of a nucleic acid sequence.
  • expression vectors for stable or transient expression may be constructed via conventional methods and introduced into cells.
  • nucleic acids encoding the nanobodies or fusion proteins thereof, or other nucleic acids or proteins may be cloned into a suitable expression vector, such as a plasmid or a viral vector in operable linkage to a suitable promoter.
  • a suitable expression vector such as a plasmid or a viral vector in operable linkage to a suitable promoter.
  • the selection of expression vectors/plasmids/viral vectors should be suitable for integration and replication in eukaryotic cells.
  • vectors of the present disclosure can drive the expression of one or more sequences in mammalian cells using a mammalian expression vector.
  • mammalian expression vectors examples include pCDM8 (Seed, Nature (1987) 329:840, incorporated herein by reference) and pMT2PC (Kaufman, et al., EMBO J. (1987) 6:187, incorporated herein by reference).
  • the expression vector's control functions are typically provided by one or more regulatory elements.
  • commonly used promoters are derived from polyoma, adenovirus 2, cytomegalovirus, simian virus 40, and others disclosed herein and known in the art.
  • the vectors of the present disclosure may direct the expression of the nucleic acid in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid).
  • tissue-specific regulatory elements include promoters that may be tissue specific or cell specific.
  • tissue specific refers to a promoter that is capable of directing selective expression of a nucleotide sequence of interest to a specific type of tissue (e.g., seeds) in the relative absence of expression of the same nucleotide sequence of interest in a different type of tissue.
  • tissue type specific refers to a promoter that is capable of directing selective expression of a nucleotide sequence of interest in a specific type of cell in the relative absence of expression of the same nucleotide sequence of interest in a different type of cell within the same tissue.
  • cell type specific when applied to a promoter also means a promoter capable of promoting selective expression of a nucleotide sequence of interest in a region within a single tissue.
  • Cell type specificity of a promoter may be assessed using methods well known in the art, e.g., immunohistochemical staining.
  • the vector may contain, for example, some or all of the following: a selectable marker gene for selection of stable or transient transfectants in host cells; transcription termination and RNA processing signals; 5’-and 3’-untranslated regions; internal ribosome binding sites (IRESes), versatile multiple cloning sites; and reporter gene for assessing expression of the chimeric receptor.
  • Suitable vectors and methods for producing vectors containing transgenes are well known and available in the art.
  • Selectable markers include chloramphenicol resistance, tetracycline resistance, spectinomycin resistance, neomycin resistance, puromycin resistance, streptomycin resistance, erythromycin resistance, rifampicin resistance, bleomycin resistance, thermally adapted kanamycin resistance, gentamycin resistance, hygromycin resistance, trimethoprim resistance, dihydrofolate reductase (DHFR), GPT; the URA3, HIS4, LEU2, and TRP1 genes of S. cerevisiae.
  • the vectors When introduced into a cell, the vectors may be maintained as an autonomously replicating sequence or extrachromosomal element or may be integrated into host DNA.
  • the disclosure further provides for cells comprising a nanobody or a fusion protein thereof, a nucleic acid, or a vector, as disclosed herein.
  • Conventional viral and non-viral based gene transfer methods can be used to introduce the nucleic acids into cells, tissues, or a subject. Such methods can be used to administer the nucleic acids to cells in culture, or in a host organism.
  • Non-viral vector delivery systems include DNA plasmids, cosmids, RNA (e.g., a transcript of a vector described herein), a nucleic acid, and a nucleic acid complexed with a delivery vehicle.
  • Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after delivery to the cell. A variety of viral constructs may be used to deliver the present nucleic acids to the cells, tissues and/or a subject.
  • Viral vectors include, for example, retroviral, lentiviral, adenoviral, adeno-associated and herpes simplex viral vectors.
  • Nonlimiting examples of such recombinant viruses include recombinant adeno- associated virus (AAV), recombinant adenoviruses, recombinant lentiviruses, recombinant retroviruses, recombinant herpes simplex viruses, recombinant poxviruses, phages, etc.
  • AAV adeno- associated virus
  • the present disclosure provides vectors capable of integration in the host genome, such as retrovirus or lentivirus. See, e.g., Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York, 1989; Kay, M. A., et al., 2001 Nat. Medic. 7(1):33-40; and Walther W.
  • nucleic acids, nanobodies or fusion proteins thereof may be delivered by any suitable means.
  • the nucleic acids or proteins thereof are delivered in vivo.
  • the nucleic acids or proteins thereof are delivered to isolated/cultured cells in vitro or ex vivo to provide modified cells useful for in vivo delivery to patients afflicted with a disease or condition.
  • Vectors according to the present disclosure can be transformed, transfected, or otherwise introduced into a wide variety of host cells. Transfection refers to the taking up of a vector by a cell whether or not any coding sequences are in fact expressed.
  • Transduction refers to entry of a virus into the cell and expression (e.g., transcription and/or translation) of sequences delivered by the viral vector genome.
  • transduction generally refers to entry of the recombinant viral vector into the cell and expression of a nucleic acid of interest delivered by the vector genome.
  • Methods of delivering vectors to cells are well known in the art and may include DNA or RNA electroporation, transfection reagents such as liposomes or nanoparticles to delivery DNA or RNA; delivery of DNA, RNA, or protein by mechanical deformation (see, e.g., Sharei et al. Proc. Natl. Acad. Sci. USA (2013) 110(6): 2082-2087, incorporated herein by reference); or viral transduction.
  • the vectors are delivered to host cells by viral transduction.
  • Nucleic acids can be delivered as part of a larger construct, such as a plasmid or viral vector, or directly, e.g., by electroporation, lipid vesicles, viral transporters, microinjection, and biolistics (high-speed particle bombardment).
  • delivery vehicles such as nanoparticle- and lipid-based delivery systems can be used. Further examples of delivery vehicles include lentiviral vectors, ribonucleoprotein (RNP) complexes, lipid-based delivery system, gene gun, hydrodynamic, electroporation or nucleofection microinjection, and biolistics.
  • RNP ribonucleoprotein
  • the disclosure provides an isolated cell comprising the vector(s) or nucleic acid(s) disclosed herein.
  • Preferred cells are those that can be easily and reliably grown, have reasonably fast growth rates, have well characterized expression systems, and can be transformed or transfected easily and efficiently.
  • suitable prokaryotic cells include, but are not limited to, cells from the genera Bacillus (such as Bacillus subtilis and Bacillus brevis), Escherichia (such as E. coli), Pseudomonas, Streptomyces, Salmonella, and Envinia.
  • Suitable eukaryotic cells include, for example, yeast cells, insect cells, and mammalian cells.
  • yeast cells include those from the genera Kluyveromyces, Pichia, Rhino-sporidium, Saccharomyces, and Schizosaccharomyces.
  • Exemplary insect cells include Sf-9 and HIS (Invitrogen, Carlsbad, Calif.) and are described in, for example, Kitts et al., Biotechniques, 14: 810-817 (1993); Lucklow, Curr. Opin. Biotechnol., 4: 564-572 (1993); and Lucklow et al., J. Virol., 67: 4566-4579 (1993), incorporated herein by reference.
  • the cell is a mammalian cell, and in some embodiments, the cell is a human cell.
  • suitable mammalian and human host cells are known in the art, and many are available from the American Type Culture Collection (ATCC, Manassas, Va.).
  • suitable mammalian cells include, but are not limited to, Chinese hamster ovary cells (CHO) (ATCC No. CCL61), CHO DHFR-cells (Urlaub et al., Proc. Natl. Acad. Sci. USA, 97: 4216-4220 (1980)), human embryonic kidney (HEK) 293 or 293T cells (ATCC No. CRL1573), and 3T3 cells (ATCC No. CCL92).
  • suitable mammalian cell lines are the monkey COS-1 (ATCC No. CRL1650) and COS-7 cell lines (ATCC No. CRL1651), as well as the CV-1 cell line (ATCC No. CCL70).
  • Further exemplary mammalian host cells include primate, rodent, and human cell lines, including transformed cell lines. Normal diploid cells, cell strains derived from in vitro culture of primary tissue, as well as primary explants, are also suitable.
  • Other suitable mammalian cell lines include, but are not limited to, mouse neuroblastoma N2A cells, HeLa, HEK, A549, HepG2, mouse L- 929 cells, and BHK or HaK hamster cell lines.
  • compositions or systems comprising a nanobody or a fusion protein thereof, a nucleic acid, a vector, or a cell, as described herein.
  • the compositions or system comprises two or more fusion proteins, nucleic acids, vectors, or cells, as described herein.
  • the composition or system further comprises a transcription factor or regulator (e.g., a transcriptional repressor, a transcriptional activator).
  • the transcription factor comprises Krüppel-associated box (KRAB).
  • the composition or system further comprises a chromatin regulator.
  • the composition or system further comprises a gRNA.
  • the gRNA may be encoded on the same nucleic acid as the nanobody or fusion protein thereof or a different nucleic acid.
  • the vector encoding a nanobody or fusion protein thereof may further encode a gRNA, under the same or different promoter.
  • the gRNA is encoded on its own vector, separated from that of nanobody or fusion protein thereof. 4.
  • the methods comprise contacting the target nucleic acid with at least one fusion protein comprising a DNA binding protein, DNA binding domain, or a functional fragment thereof covalently linked to a nanobody configured to bind a chromatin regulator.
  • the gene expression and/or epigenetic memory of at least two genes is modulated. Descriptions provided elsewhere herein with regards to the fusion protein and components thereof are applicable to the present methods.
  • Modulation of expression comprises increasing or decreasing gene expression compared to normal gene expression for the target gene. When the gene expression of at least two genes is modulation, both genes may have increased gene expression, both gene may have decreased gene expression, or one gene may have increased gene expression and the other may have decreased gene expression.
  • the epigenetic memory of a cell is defined by the set of modifications to the cell’s DNA that does not necessarily alter the coding sequence but rather alters gene expression by the chemical modification (e.g., methylation, acetylation, demethylation, deacetylation) of the DNA and related histones.
  • Modulation of epigenetic memory comprises the chemical modification of the DNA and related histones such that the gene expression is regulated.
  • the epigenetic memory is modulated over long time scales and even over generations and is considered persistent or stable.
  • the epigenetic memory is modulated in the short-term and is still dynamic.
  • the methods described herein may result in changes to epigenetic memory which result in increased persistence (as measured by the number or percentage of cells maintaining the modification and/or gene expression changes) for at least 30 days when compared to modifications of other transcriptional regulators or without any induced modulation.
  • the disclose methods may result in greater than 30% (e.g., greater than 35%, greater than 40%, greater than 45%, greater than 50%, greater than 55%, greater than 60%, greater than 65%, greater than 70%, greater than 75%, greater than 80%, or more) of the cells maintain the modulation of epigenetic memory for greater than 30 days.
  • the at least one target nucleic acid is in a cell and the chromatin regulator is endogenous to the cell.
  • the cell may be a prokaryotic or eukaryotic cell. In some embodiments, the cell is a eukaryotic cell. In some embodiments, the cell is in vitro. In some embodiments, the cell is ex vivo. [00141] In some embodiments, the cell is in an organism or host, such that introducing the disclosed systems, compositions, vectors into the cell comprises administration to a subject. The method may comprise providing or administering to the subject, in vivo, or by transplantation of ex vivo treated cells, at least one synthetic transcription factor, nucleic acid, vector, or composition or system as described herein.
  • a “subject” may be human or non-human and may include, for example, animal strains or species used as “model systems” for research purposes, such a mouse model as described herein. Likewise, subject may include either adults or juveniles (e.g., children). Moreover, subject may mean any living organism, preferably a mammal (e.g., human or non- human) that may benefit from the administration of compositions contemplated herein.
  • mammals include, but are not limited to, any member of the Mammalian class: humans, non- human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice, and guinea pigs, and the like.
  • non- mammals include, but are not limited to, birds, fish, and the like.
  • the mammal is a human.
  • the terms “providing”, “administering,” and “introducing,” are used interchangeably herein and refer to the placement of the systems of the disclosure into a subject by a method or route which results in at least partial localization of the system to a desired site.
  • the systems can be administered by any appropriate route which results in delivery to a desired location in the subject.
  • the method further comprises contacting the target nucleic acid with an exogenous chromatin regulator.
  • the methods further comprise contacting the target nucleic acid with a transcription factor or regulator.
  • the transcription factor comprises Krüppel-associated box (KRAB). 5.
  • kits including at least one or all of a nanobody, a fusion protein thereof, or nucleic acid or vector encoding thereof, a composition or system as described herein, a cell comprising a nanobody, a fusion protein thereof, or nucleic acid or vector encoding thereof, a reporter cell as described herein, a yeast cell as described herein, and a two-part reporter gene as described herein or a nucleic acid encoding thereof.
  • the kits can also comprise instructions for using the components of the kit. The instructions are relevant materials or methodologies pertaining to the kit.
  • the materials may include any combination of the following: background information, list of components, brief or detailed protocols for using the compositions, trouble-shooting, references, technical support, and any other related documents.
  • Instructions can be supplied with the kit or as a separate member component, either as a paper form or an electronic form which may be supplied on computer readable memory device or downloaded from an internet website, or as recorded presentation.
  • the kit may include instructions for use in any of the methods described herein.
  • the instructions can comprise a description of use of the components for the methods of identifying and selecting nanobodies or methods of modulating gene expression.
  • the kits provided herein are in suitable packaging.
  • Kits optionally may provide additional components such as buffers and interpretive information. Normally, the kit comprises a container and a label or package insert(s) on or associated with the container. In some embodiment, the disclosure provides articles of manufacture comprising contents of the kits described above.
  • the kit may further comprise a device for holding or administering the present proteins, nucleic acids, or composition.
  • the device may include an infusion device, an intravenous solution bag, a hypodermic needle, a vial, and/or a syringe.
  • the present disclosure also provides for kits for performing the methods or producing the components in vitro.
  • the kit may include the components of the present system.
  • Optional components of the kit include one or more of the following: (1) buffer constituents, (2) control plasmid, (3) sequencing primers. 6. Examples Materials and Methods Plasmid construction [00153]
  • the TagRFP reporter (5xTetO-pEF-TagRFP-3xNLS) construct was assembled using a AAV zinc finger donor vector backbone (Addgene #22212) containing a promoter-less splice- acceptor upstream of a puromycin resistance gene and homology arms against the AAVS1 locus.
  • Three elements of the reporter were amplified from the following sources: five TetO-binding sites upstream of a pEF promoter from PhiC31-Neo-ins-5xTetO-pEF-H2B-Citrine-ins (Addgene #78099), TagRFP-T from pEN_ERK.KTR-tagRFP-T, and 3xNLS from pEN_mCherry-NLS (both gifts from Joydeb Sinha & Mary Teruel, Stanford). These components were cloned into the AAV donor vector backbone using Gibson Assembly.
  • the plasmids containing the rTetR-effector fusions were cloned into the PBCMV- MCS-EF1 ⁇ -Puro PiggyBac vector backbone (System Biosciences #PB510B-1), which was further modified via Gibson Assembly with the following components: PGK promoter from pSLQ2818, mIFP from pSLQ2837-1 (both gifts from Tony Gao & Stanley Qi, Stanford)67, and H2B-rTetR-Zeo from pEx1-pEF-H2B-mCherry- T2A-rTetR-KRAB-Zeo (Addgene #78352).
  • Plasmids containing the dCas9-effector fusions were derived from the dCas9-KRAB vector backbone (Addgene #110820) and modified by Gibson Assembly with their respective effectors from sources listed above.
  • the dCas9-effector fusions containing KRAB or KRAB- antiDNMT1 were further modified with mCitrine-NLS upstream of the dCas9 to allow for cell sorting and analysis of endogenous gene silencing.
  • the sgRNA cloning vector was modified to express mIFP or mCherry.
  • sgRNA sequence was cloned into the plasmid using the BlpI and BstXI cloning sites, as previously described.
  • the AAVS1 TagRFP reporter donor vector (Addgene #163083) and rTetRantiGFP recruitment vector (Addgene #163084) have been deposited to Addgene. Plasmids containing antiDNMT1 and/or antiHP1 are available on request upon signing an MTA with ChromoTek and Institut Curie, respectively.
  • Cell culture [00157] Cells were cultured at 37 °C under a humidified atmosphere with 5% CO 2 .
  • HEK293T cells (Takara Bio #632180) were maintained in Dulbecco’s modified Eagle medium (DMEM; Gibco #10569010) supplemented with 25mM D-glucose (Gibco), 1mM sodium pyruvate (Gibco), 1 ⁇ GlutaMAXTM (Gibco), and 10% Tet Approved FBS (Clontech Laboratories). When cells reached 80% confluence, they were gently washed with 1 ⁇ DPBS (Life Technologies) and passaged using 0.25% Trypsin (Life Technologies).
  • DMEM Dulbecco’s modified Eagle medium
  • the reporter cell line was created by integrating the TagRFP fluorescent reporter at the first intron of the constitutively expressed gene PPP1R12C at the AAVS1 locus in HEK293T cells.
  • the integration of the reporter was performed by co-transfecting 1000 ng TagRFP reporter (5 ⁇ TetO-pEF-TagRFP-3 ⁇ NLS) donor plasmid and 500 ng of each TALEN arm (AAVS1- TALEN-L (Addgene #35431) targeting 5 ⁇ -TGTCCCCTCCACCCCACA-3 ⁇ (SEQ ID NO: 5929) and AAVS1-TALEN-R (Addgene #35432) targeting 5 ⁇ -TTTCTGTCACCAATCCTG-3 ⁇ (SEQ ID NO: 5930)). Cells were selected with 500 ng/mL puromycin (InvivoGen) starting 48 h post transfection for ⁇ 5 days or until all of the negative control cells died.
  • Cells positive for TagRFP had two peaks representing the monoallelic and bi-allelic integration of the reporter at the AAVS1 locus.
  • Cells with the lower fluorescence peak (monoallelic) were sorted by fluorescence-activated cell sorting using a Sony SH800 Cell Sorter with a 100 ⁇ m disposable chip.
  • Each of the individual rTetR-effector plasmids was randomly integrated into this reporter line by co-transfecting 250 ng Super PiggyBac Transposase expression vector (System Biosciences #PB200PA-1) and 750 ng of rTetR-effector donor vector. These cells were selected with 60 ⁇ g/mL zeocin (InvivoGen) starting 48 h post transfection.
  • transfections were performed in 24-well plates using Lipofectamine 2000 (Invitrogen).
  • Transient transfections Approximately 70,000 cells were seeded per well in a 24-well plate and the next day cells were transfected using Lipofectamine 2000 (Invitrogen) according to manufacturer instructions.
  • 1000 ng of each GFP-CR was delivered.
  • 1000 ng of rTetR-effector expression vector was delivered to each well. 600 ng of dCas9-effector and 400 ng of sgRNA were co-delivered for the silencing and reactivation experiments involving dCas9 fusions.
  • Imaging was done using a Leica DMi8 fluorescence microscope with Adaptive Focus Control, a ⁇ 20 or ⁇ 40 dry objective, and a Leica DFC9000 GT sCMOS camera. Fluorophores were excited using a Lumencor SOLA SE II light source. Images were automatically acquired every 15min, using LAS X software (Leica Microsystems).
  • the imaging media was changed daily for ⁇ 5 days (until the cells became too confluent to continue movies).
  • Time-lapse movies were analyzed using ImageJ by visually tracking individual cell lineages and manually circling the area corresponding to the cell’s nuclei 1 h after each cell division. Average fluorescence intensities of mIFP, TagRFP, and GFP within these contours of the cell nuclei were calculated and plotted based on their cell lineage using MATLAB (MathWorks).
  • Gene expression analysis via flow cytometry [00161] Cells expressing stably integrated or transiently transfected rTetR-effectors were assayed by flow cytometry during and after 5 days of 1 ⁇ g/mL dox (Tocris) treatment.
  • cells were also treated with 1 ⁇ M 5-Aza-2’ (Sigma) or 100 nM chaetocin (Cayman Chemical). Media containing small molecules were replaced daily.
  • dCas9-effectors cells were analyzed 5 days post transfection, and after being sorted for silencing (TagRFP-negative cells). On the day of flow cytometry analysis, cells were collected using 0.25% Trypsin (Life Technologies). A fraction of the cells (varying between one half to one twentieth, depending on cell density) were replated for the next time point.
  • the remaining cells were resuspended in flow buffer (1 ⁇ Hank’s balanced salt solution (Life Technologies) and 2.5 mg/mL bovine serum albumin (BSA) (Sigma)) and filtered through a 40 ⁇ m strainer (Corning) to remove cell clumps.
  • Cellular fluorescence distributions were measured with the CytoFLEX S Flow Cytometer (Beckman Coulter) and the CytExpert Software (Beckman Coulter).
  • the resulting data were analyzed with a custom MATLAB program called EasyFlow (antebilab(dot)github(dot)io/easyflow/).
  • BV421-labeled IgG2a (clone MOPC-173 (1 : 20); Biolegend) served as an isotype control. Afterwards, cells were washed three times with 1% BSA/DPBS and then analyzed by flow cytometry for cells that were double positive for dCas9 (mCitrine) and sgRNA (mCherry). Targeted bisulfite sequencing [00163] Reporter cells stably expressing rTetRantiDNMT1, rTetR-DNMT1, or rTetR- DNMT3B were treated with 1 ⁇ g/mL dox for 5 days.
  • each treated cell line was sorted in TagRFP-negative (+dox OFF) and TagRFP-positive (+dox ON) cells using a SONY SH800 Cell Sorter.
  • Total genomic DNA was extracted from these cells with the DNeasy Blood & Tissue Kit (Qiagen) according to the manufacturer’s instructions and quantified using a NanoDrop 8000 spectrophotometer (Thermo).
  • DNA was also extracted from cells not treated with dox ( ⁇ dox) grown at the same time as the +dox cells for each cell line and sorted to include the entire population (which is >97% ON for all cell lines).
  • Assays were designed targeting CpG sites in 100–300 nucleotide regions at the reporter gene and two control genes, IGF2 (positive) and ACTB (negative), using primers created with Rosefinch, Zymo Research’s proprietary sodium bisulfite converted DNA-specific primer design tool.
  • primers were designed to avoid annealing to CpG sites in the region of interest.
  • C or T pyrimidine
  • a or G purine
  • becomes ⁇ ⁇ ag, for recruitment times larger than ⁇ ag, and no changes in the fractions of silent/active cells are allowed at shorter times.
  • HEK-293 cells were plated in 4 x 10cm plates in DMEM media + 10% FBS supplemented with L-glutamine and Pen/Strep. At the time of transfection, cells were about 70-80% confluent. The following day (20-24 hours later), cells were transfected with pRetro-CMV2-TO-3xFLAG-DNMT3A plasmid. Before transfection: in each 10cm plate, medium was changed to 20 ml plain DMEM (no FBS, no Pen/Strep).
  • the first set contained 450 ⁇ l 2X HBS (50 mM HEPES, pH 7.05, 10 mM KCl, 12 mM dextrose, 280 mM NaCl, 1.5mM Na 2 PO 4 ).
  • the other set contained 25 ⁇ g Plasmid DNA (pRetro-CMV2-TO-3xFLAG-DNMT3A) + 65 ⁇ l 2M CaCl 2 into 0.1XTE (450 ⁇ l total).
  • the DNA and CaCl 2 were mixed by pipetting up and down with a 200 ⁇ l pipet and added dropwise to the 2X HBS. This mixture was incubated at room temp for 1 min.
  • the DNA-Calcium phosphate co-precipitate was added dropwise to the surface of the media containing the cells.
  • the plate was swirled gently to mix.
  • 48-72hrs post transfection transfected cells were harvested for lysis and DNMT3A protein immunoprecipitation on magnetic beads.
  • anti-Flag Magnetic Beads (No. B26101, Bimake) suspension was transferred to a new tube with 0.5 mL TBS buffer (50 mM Tris HCl, 150 mM NaCl, pH 7.4). The mixture was gently pipetted 5 times. The tube was placed on a magnet for 10 seconds to separate the beads from the solution, and the supernatant was discarded. This step was repeated 2 times.
  • TBS buffer 50 mM Tris HCl, 150 mM NaCl, pH 7.4
  • the tube was placed on a magnet for 10 seconds to separate the beads from the solution, and the supernatant was discarded. This step was repeated 2 times.
  • To bind the DNMT3A complex to the magnetic beads ⁇ 500 ⁇ L of cell lysate was added to the washed magnetic beads. The tubes were gently rotated for 2 h at room temperature or overnight at 4°C.
  • the tubes were placed on the magnet to separate the beads from the solution for 2 minutes and the supernatant was transferred into a new tube for detecting whether Flag-tagged protein is left unbound.
  • 500 ⁇ L PBST was added to the tube (NaCl 136.89 mM; KCl 2.67 mM; Na 2 HPO 4 8.1 mM; KH 2 PO 4 1.76 mM; 0.5% Tween20), and the magnetic beads were resuspended by pipetting gently. Then the tube was rotated for 5 min, and, after that, placed on the magnet to separate the beads from solution for 2 minutes to remove the supernatant. This wash step was repeated 2 times.
  • yeast growth and induction The yeast nanobody library was maintained in Yglc4.5 - Trp medium (1 liter: 3.8 g of -Trp drop-out media supplement (US Biological), 6.7 g Yeast Nitrogen Base, 10.4 g Sodium Citrate 7.4 g Citric Acid Monohydrate, 10 mL Pen-Strep (10,000 units/mL stock), and 20 g glucose, pH4.5).
  • Nanobody expression was under the control of the GAL1 promoter such that nanobodies were produced on the cell surface when yeast was grown in a galactose-containing medium.
  • Expression of the nanobody library was induced by dilution of a yeast aliquot into -Trp +galactose medium (1 liter: 3.8 g -Trp drop-out media supplement (US Biological), 6.7 g Yeast Nitrogen Base, 10 mL Pen-Strep (10,000 units/mL stock), 20 g glucose or galactose (glucose for normal growth and galactose for induction of nanobodies), pH 6) followed by shaking for 48 hours, at 25 °C, 220 rpm.
  • BBE Bead-Based Enrichment of nanobodies using yeast surface display DNMT3A- coated beads were prepared by immunoprecipitation as described above. 150 ⁇ l washed beads were removed from the magnet and the beads were resuspended in 1000 ⁇ l ice-cold selection buffer (20 ⁇ mM HEPES, pH 7.5, 150 ⁇ mM sodium chloride, 2% (w/v) BSA, 1 ⁇ mM EDTA), and placed on ice until needed.
  • Yeast negative selection Each round of BBE selection began with a negative selection step which involved incubating the yeast with non-antigen-coated beads to remove yeast-expressing nanobodies that bound nonspecifically to the magnetic beads. Specifically, 150 ⁇ L resuspended Flag-conjugated beads were added to the yeast cells induced with galactose. Cells were incubated on the rotary wheel at 4°C for 2 h.
  • the tube was placed on the magnet, taking care to transfer any liquid lodged in the cap of the tube to the bottom portion of the tube. After 2 minutes, the supernatant was carefully removed from the tube and transferred into a fresh 10ml tube. The supernatant served as the input for the next selection step.
  • the beads were resuspended in 1ml ice-cold selection buffer with a pipette and placed on the magnet for 2min. The supernatant was removed from the washed beads and discarded. The beads were resuspended in 1ml ice-cold selection buffer and set aside as negative#1 to enable estimation of the number of cells captured by the negative selection.
  • a second negative selection was performed using the supernatant from the previous step as input, before proceeding to the following step with the resulting depleted supernatant.
  • Yeast positive selection After the negative selection, DNMT3A-binding nanobodies were enriched over 3 rounds of BBE selection by staining the yeast with DNMT3A complex- coated beads. Specifically, the yeast cells after negative selection were mixed with the DNMT3A-coated magnetic beads and incubated on the rotary wheel at 4°C for 2h. Upon completion of the incubation, the tube was placed on a magnet, taking care to transfer any liquid lodged in the cap of the tube to the bottom portion of the tube. The cells and the beads were incubated on the magnet for 2min.
  • the sample (5ul) was diluted into 995 ⁇ L - Trp4.5_media (200x dilution) and set aside (tube labeled as positive#1) for a later analysis step.
  • the cells on the beads were grown at 30°C with shaking for 48 hours.
  • the saved beads from the negative sorts (negative#1) were vortexed and 100 ⁇ L beads were transferred into 400 ⁇ L fresh -Trp4.5_media. Take these diluted samples, vortex, and transfer 5 ⁇ L of each sample into 995 ⁇ L -Trp4.5_media (200x dilution) (negative #2).
  • Thee 200x dilutions of the negative sort(negative#2) and the positive sorts (positive#1) were vortex and 10 ⁇ L from each population was transferred into 190 ⁇ L - Trp4.5_media (4000x dilution).
  • a -Trp4.5_media plate was divided into four regions using a permanent marker. Each dilution was vortex and 20 ⁇ L was plated. The plate was incubated at 30°C for 3 days and the colonies were counted. One colony in the 200x and 4000x dilutions represents 5x10 4 and 1x10 6 cells recovered, respectively.
  • the cells After overnight growth, measure the OD600 of the cells. If the OD600 was still low, the cells were allowed to grow another day. Once the culture approaches saturation, the cells were pelleted (at 900xg for 5 minutes) and the supernatant was aspirated. The pellet was resuspended in 1ml Trp4.5_media and the cells were transferred to a 2ml tube. The supernatant was recovered following magnetic precipitation and the cells were diluted into two cultures for further expansion - 2.5x10 8 cells into 25ml -Trp4.5_media for growth and induction and remaining cells into 25ml -Trp4.5_media for overnight growth and temporary storage at 4 degree in case the first selected population needs to be induced and selected again.
  • nanobody-expressing yeast were incubated with the DNMT3A complex, washed, and then stained with Anti-DYKDDDDK Tag (DYKDDDDK (SEQ ID NO: 5931) tag) Mouse Monoclonal antibody (FITC (Fluorescein)) (GenScript, A01632, 1:50 dilution), and HA-Tag (6E2) Mouse mAb (Alexa Fluor® 647 Conjugate) (Cell Signaling Technology, 3444S, 1:50 dilution). DNMT3A binding was confirmed and analyzed by flow cytometry (ZE5) to verify the enrichment for nanobody binders compared to the naive yeast library.
  • FITC Fluorescein
  • HA-Tag 6E2
  • Mouse mAb Alexa Fluor® 647 Conjugate
  • the library of nanobody plasmids was extracted from the enriched yeast library by Zymoprep Yeast Plasmid Miniprep II (Cat# D2004). High-throughput screening of nanobodies capable of silencing in human cells [00205] Pooled library cloning of selected nanobodies into a lentiviral construct The library of nanobody plasmids was extracted after three rounds of yeast display enrichment, and then PCR amplified. 8x 50 ⁇ L reactions were set up in a clean PCR hood to avoid amplifying contaminating DNA.
  • the resulting dsDNA libraries were gel extracted by loading ⁇ 4 lanes of a 2% TAE gel, excising the band at the expected length (around 400 bp), and using a QIAgen gel extraction kit.
  • the libraries were cloned into a lentiviral recruitment vector pWJ036 with 4x10 ⁇ l GoldenGate reactions (75 ng of pre-digested and gel-extracted backbone plasmid, 5 ng of library (2:1 molar ratio of insert:backbone), 0.13 ml of T4 DNA ligase (NEB, 20000 U/ml), 0.75 ⁇ l of Esp3I-HF (NEB), and 1 ⁇ l of 10x T4 DNA ligase buffer) with 30 cycles of digestion at 37°C and ligation at 16°C for 5 minutes each, followed by a final 5 minute digestion at 37°C and then 20 minutes of heat inactivation at 70°C.
  • the domains were amplified from the plasmid pool and from the original oligo pool by PCR with primers with extensions that include Illumina adapters and sequenced.
  • the PCR and sequencing protocol were the same as described below for sequencing from genomic DNA, except these PCRs use 10 ng of input DNA and 17 cycles. These sequencing datasets were analyzed as described below to determine the uniformity of coverage and synthesis quality of the libraries.
  • 20-30 colonies from the transformations were Sanger sequenced (Quintara) to estimate the cloning efficiency and the proportion of empty backbone plasmids in the pools.
  • HEK293T cells were plated on four 10-cm tissue culture plates.
  • HEK293T cells were plated in 10 mL of DMEM, grown overnight, and then transfected with a mixture of the three third-generation packaging plasmids (6.5 ⁇ g pMDLG/pRRE, 5 ⁇ g Rev, 3.5 ⁇ g VSVG) and 10 ⁇ g of rTetR- Nanobody library vectors using the calcium phosphate method.
  • a mixture of the three third-generation packaging plasmids 6.5 ⁇ g pMDLG/pRRE, 5 ⁇ g Rev, 3.5 ⁇ g VSVG
  • 10 ⁇ g of rTetR- Nanobody library vectors using the calcium phosphate method.
  • ⁇ Lentivirus was harvested at 48 hours and 72 hours.
  • the pooled lentivirus was filtered through a 0.45-mm PVDF filter (Millipore) to remove any cellular debris.
  • Nanobody library targeting the DNMT3A complex screen 8x10cm plates with HEK-293 reporter cells at the density of 250 cells/mm 2 were infected with the lentiviral library for two separate biological replicates of the infection. Infected cells grew for 3 days and then the cells were selected with 2 ⁇ g/mL puromycin. Infection and selection efficiency were monitored every other day using flow cytometry to measure mScarlet (and thus nanobody) positive cells (ZE5). After 3 days of selection with puromycin, cells in each 10cm plate were transferred to 15cm plate to make the maintenance coverage > 25,000 x cells per library element (a very high coverage level that compensates for losses due to incomplete puromycin selection, library preparation, and library synthesis errors).
  • nanobody recruitment at the reporter was induced by treating the cells with 1 ⁇ g/ml doxycycline (Fisher Scientific) for 5 days. Cells were split every other day and measured for maintenance coverage on ZE5.
  • Magnetic separation of reporter cells The reporter included a synthetic surface marker, consisting of the human IgG1 Fc region linked to an Igk leader and PDGFRb transmembrane domain, to enable magnetic separation of OFF from ON cells.
  • HEK-293 cells were trypsinized and spun down at 300 x g for 5 minutes.
  • Beads (60 ⁇ L) were prepared for every 1 x 10 7 cells, by adding 1 mL of buffer per 200 ⁇ L of beads, vortexing for 5 s, placing on a magnetic tube rack, waiting one minute, removing supernatant, and finally removing the beads from the magnet and resuspending in 100-600 ⁇ l of blocking buffer per initial 60 ⁇ L of beads. After incubation, the bead and cell mixture were placed on the magnetic rack for > 2 minutes. The unbound supernatant was transferred to a new tube, placed on the magnet again for > 2 minutes to remove any remaining beads, and then the supernatant was transferred and saved as the unbound fraction.
  • the beads were resuspended in the same volume of blocking buffer, magnetically separated again, the supernatant was discarded, and the tube with the beads was kept as the bound fraction.
  • the bound fraction was resuspended in a blocking buffer or PBS to dilute the cells (the unbound fraction is already dilute).
  • Flow cytometry (ZE5) was performed using a small portion of each fraction to estimate the number of cells in each fraction (to ensure library coverage was maintained) and to confirm separation based on citrine reporter levels (the bound fraction should be > 90% citrine positive, while the unbound fraction is more variable depending on the initial distribution of reporter levels).
  • the samples were spun down and the pellets were frozen at -80 °C until genomic DNA extraction.
  • Genomic library preparation and next generation sequencing Genomic DNA was extracted with the QIAgen Blood Maxi Kit following the manufacturer’s instructions with up to 1.25 x 10 8 cells per column. DNA was eluted in EB and not AE to avoid subsequence PCR inhibition. The domain sequences were amplified by PCR with primers containing Illumina adapters as extensions. A test PCR was performed using 400 ng of genomic DNA in a 50 ⁇ L (half size) reaction to verify if the PCR conditions would result in a visible band at the expected size for each sample. Then, 25x 50 ⁇ L reactions were set up on ice (in a clean PCR hood to avoid amplifying contaminating DNA), with the number of reactions depending on the amount of genomic DNA available in each experiment.
  • thermocycling protocol 400 ng of genomic DNA, 23 ⁇ L H 2 O, 1 of each 10 uM primer, and 25 ⁇ L of Q5 Hot Start High-Fidelity 2X Master Mix (NEB) was used in each reaction.
  • the thermocycling protocol was to preheat the thermocycler to 98 °C, then add samples for 3 minutes at 98 °C, then 32x cycles of 98 °C for 10 s, 55 °C for 30 s, 72 °C for 50 s, and then a final step of 72 °C for 10 minutes.
  • PCR reactions were pooled and ⁇ 140 ⁇ L were run in at least three lanes of a 2% TAE gel alongside a 100-bp ladder for at least one hour, the library band around 400 bp was cut out, and DNA was purified using the QIAquick Gel Extraction kit (QIAgen) with a 30 ⁇ L elution into non-stick tubes (Ambion). A confirmatory gel was run to verify that small products were removed.
  • QIAquick Gel Extraction kit QIAgen
  • a confirmatory gel was run to verify that small products were removed.
  • These libraries were then quantified with a Qubit HS kit (Thermo Fisher) and sequenced on an Illumina NextSeq with a High output kit using a paired end (forward read 200 and reversed read 100) and 8 cycle index reads.
  • Reads with one or more undetected CDR and/or with mean quality scores less than 30 were filtered out. Reads with identical CDR combinations at the DNA-sequence level were grouped and counted. This process was repeated for each sample sequenced. The enrichments for each nanobody (CDR combination) between OFF and ON samples were computed using the script ‘makeRhos.py’. In this script, nanobodies with fewer than 5 reads in both samples for a given replicate were filtered out, whereas nanobodies with fewer than 5 reads in one sample would have those reads adjusted to 5 to avoid inflating enrichment values due to low sequencing depth.
  • Counts were normalized to the sum of counts in that sample to account for differences in sequencing depth (in effect, frequencies were computed) prior to computing log2(OFF:ON) enrichment scores.
  • Individual validations of nanobody function in human cells [00210] Interaction assay between nanobodies and DNMT3A Individual nanobodies were synthesized (gBlock, IDT) and cloned as fusions with rTetR(SE-G72P) with a 3xFLAG, upstream of a P2A-mScarlet and puromycin selection marker using Gibson assembly cloning into the lentivirus backbones pWJ036.
  • DNMT3A with 3xHA was cloned into the pRetro-CMV2-TO-puromycin vector using Gibson assembly. 5 ⁇ 10 6 HEK-293T cells were seeded in 10 ⁇ cm cell culture dishes and grown for 24 ⁇ h.
  • the plasmid expressing 3xFLAG- rTetR(SE-G72P)-Nanobody (12.5ug) and the plasmid expressing 3XHA-DNMT3A(12.5ug) were co-transfected into HEK-293T cells using Calcium phosphate method.
  • lysis buffer 50 ⁇ mM Tris–HCl, pH 8.0, 1 ⁇ mM EDTA, 150 ⁇ mM NaCl, 1% NP-40, 1xNEM and 1 ⁇ PMSF
  • cell lysates were cleared by centrifugation at 10,000 ⁇ g for 10 ⁇ min.
  • Immunoprecipitations were performed with Anti-Flag Magnetic Beads (No. B26101, Bimake) for 5 ⁇ h at 4 ⁇ °C. Samples were washed 5 ⁇ 30 ⁇ min in lysis buffer.
  • Proteins from the immunoprecipitates and from whole-cell lysates were separated by 12% SDS-PAGE, transferred to nitrocellulose membranes, blocked with 5% milk in TBST and analyzed by immunoblotting with mouse anti-FLAG M2 (Sigma, 1:1000) and mouse anti-HA (#901501, Biolegend, 1:1000). Staining of tubulin from whole-cell lysates with mouse anti-tubulin (12G10, Developmental Studies Hybridoma Bank, 1:5000) was used as the loading control.
  • doxycycline (1ug/ml) to recruit rTetR- Nanobody to the reporter or left untreated.
  • doxycycline was removed by spinning down the cells, replacing media with DPBS (GIBCO) to dilute any remaining doxycycline, and then spinning down the cells again and transferring them to fresh media.
  • Time points were measured every 2-3 days by flow cytometry analysis of > 30,000 cells on a ZE5 flow cytometer (BioRad). Data was analyzed using FlowJo. Events were gated for viability and for mScarlet as a delivery marker.
  • Example 1 Nanobodies against GFP-CRs are used to control gene expression [00212] To test whether nanobodies can be used to recruit CRs for efficient gene expression control, a nanobody against green fluorescent protein (GFP) was fused to a reverse tetracycline repressor (rTetR) DNA-binding domain and used to recruit various GFP-tagged CRs (GFP-CR) to a TagRFP fluorescent reporter gene located at the AAVS1 locus in HEK293T cells (FIG. 1A). The reporter contained five Tet operator (TetO)-binding sites upstream of a constitutive pEF promoter driving the expression of the fluorescent gene.
  • GFP green fluorescent protein
  • rTetR reverse tetracycline repressor
  • HP1 ⁇ , HP1 ⁇ , and HDAC5 led to a higher fraction of cells silenced compared to HP1 ⁇ , consistent with their reported roles in silencing and association with heterochromatin. Nevertheless, HP1 ⁇ , which can associate with either heterochromatin or actively transcribed regions, still led to silencing in a majority of cells (FIG. 1D).
  • lower levels of GFP- CRs led to a higher fraction of cells silenced after 4 days of recruitment for all four CRs (FIG. 11D).
  • increasing the number of anti-GFP nanobodies fused to a single rTetR to 8 did not increase the fraction of cells silenced (FIG. 11E), suggestive that a single nanobody is sufficient for silencing in the reporter system.
  • Example 2 Nanobodies against DNMT1 and HP1 can silence a Reporter Gene and Confer Epigenetic Memory
  • Two existing nanobodies against endogenous CRs, antiHP1 and antiDNMT1 were tested for their capacity to silence and induce epigenetic memory.
  • the antiHP1 nanobody was shown to bind to all three isoforms of HP1 in cell lysate by western blotting and in cells by immunofluorescence.
  • the antiDNMT1 nanobody has been used to immunoprecipitate endogenous DNMT1 from whole cell lysate and visualize endogenous DNMT1 in live cells.
  • the silenced (TagRFP-negative) cells were sorted at the end of 5 days of dox treatment and measure their persistence of silencing for 30 days. Similar to stable expression, the antiDNMT1-antiHP1 fusion had improved epigenetic memory over KRAB, with 35.4% cells still silent at 30 days post sorting vs. 15% cells, respectively (FIG. 13B). Transient expression of nanobodies can also lead to reporter silencing and impart epigenetic memory, but less efficiently than stable expression.
  • Example 3 Recruitment of antiDNMT1 improves silencing speed and epigenetic memory of other CRs at the reporter. [00220] The antiDNMT1 nanobody was tested in combination with KRAB (FIG. 14A).
  • the KRAB-antiDNMT1 fusion was tested with the dCas9 system at the reporter gene (FIG. 14C).
  • Single-guide RNAs sgRNAs
  • sgRNAs Single-guide RNAs
  • dCas9-KRAB- antiDNMT1 demonstrated improved memory over dCas9-KRAB and over a combined repressor, dCas9-KRABMeCP2, (FIG. 3D; 27.2% vs. 17.5%).
  • KRABMeCP2 had the same memory as KRAB alone (FIG. 3D; 18.1% vs. 17.5%) and the addition of antiDNMT1 to this fusion resulted in a similar improvement in memory as when added to KRAB (FIG. 3D; 33% for KRAB- MeCP2-antiDNMT1 vs. 27.2% for KRAB-antiDNMT1).
  • the antiDNMT1-DNMT3A-3L triple fusion was by far the strongest (FIG. 3E; dark blue) resulting in about 87% of the cells being silenced at 5 days of dox.
  • the antiDNMT1 nanobody improved the speed of silencing in all combinations with DNMT3A (FIG. 3F).
  • All fusions containing rTetR-DNMT3A, including the ones containing antiDNMT1 led to permanent epigenetic memory at the reporter gene (FIG. 15A).
  • a similar increase in the speed of silencing of the reporter gene was seen when antiDNMT1 was fused to the HDAC enzyme HDAC4 (FIG. 15B).
  • Example 4 Nanobody-mediated recruitment of CRs for synthetic circuit control.
  • These nanobody-based tools for controlling gene expression and epigenetic memory may be suitable to serve as devices in synthetic circuits for detecting and recording signals.
  • Cellular stopwatches and recording devices are important components of synthetic biology circuits.
  • the response of the antiDNMT1 nanobody presents a unique opportunity of implementing a very compact stopwatch that can both measure and record the duration of a signal.
  • the desired signal can be coupled to the expression of rTetR-antiDNMT1, which in turn can be recruited upstream of an output gene encoding for fluorescence, signaling molecules, or proteins involved in cell death or survival (FIG. 4A).
  • the addition of dox starts the time recording session, while removal of dox ends it.
  • active cells silence at a slow rate (k S ) during recruitment by antiDNMT1. They first reach a reversible silent state (R) and can transition from this to an irreversibly silent state (I) with a rate k I . After release of the nanobody, the reversibly silent population reactivates at a rate k A , while the irreversibly silent cells remain silent.
  • T lag T lag
  • the three-state phenomenological model can be used to predict the fraction of cells silent over time for different types of signals without changing the three transition rates and initial lag time.
  • the model predicted that the fraction of cells silenced at the end of a 5-day pulsed signal (3 days of dox, 2 days of no dox, and then 2 days of dox) was approximately the same as at the end of a continuous 5-day signal (5 days of dox and then 2 days of no dox), matching experimental data (FIG. 4F).
  • This model also predicted that continuous signals result in a small but systematically higher level of cells permanently silenced compared to interrupted signals of the same total duration, which was also consistent with experimental data (FIG. 4G).
  • a yeast display against a chromatin regulator (CR) that is known to act as a repressor was used to reduce library size to about ⁇ 10 4 sequences.
  • This library was recloned into a lentiviral vector, and lentiviral infection was used to deliver the pooled enriched library to human cells.
  • CR chromatin regulator
  • a nanobody sequence was identified (antiDNMT3A NB#8).
  • antiDNMT3A NB#8 acted as a strong gene silencer when fused to rTetR in multiple cell types (FIG. 7), silenced a reporter gene when fused to dCas9 (FIG. 8), and imparted epigenetic memory (FIG.
  • the silencing ability of nanobodies can be measured in a high-throughput manner in human cells (FIG. 10), and this method was used to select for nanobodies against the DNMT3A complex that can silence gene expression. From the high-throughput measurement in human cells, of the approximately 3000 nanobodies obtained at the end of three rounds of yeast display against DNMT3A and lentiviral delivery in human cells, only about ⁇ 60 (2%) had a strong effect on gene expression. Besides NB#8, several other individual nanobodies (NB#25, #26, #27, #28) that were identified in the high-throughput screening were validated as strong gene silencers (FIG. 10).
  • This method allowed selection of nanobodies that can perform the desired silencing function in human cells. While many nanobodies can bind an endogenous repressive complex, few silence in human cells. For example, in a small-scale test, only 1/24 nanobodies that bound DNMT3A silenced a reporter gene in the majority of cells (Table 2, FIG. 7). In contrast, 5/5 nanobodies chosen from the top hits from the functional silencing screen in human cells (Table 3, FIG. 10A) silenced >90% of the cells (FIG. 10C).
  • Table 2 antiDNMT3A nanobodies
  • Table 3 Exemplary individual anti-DNMT3A nanobodies recovered from the disclosed high-throughput screen, as shown by SEQ ID NO encoding each of 3 CDRs Each CDR is separated by ‘ ’

Abstract

Provided herein are compositions, systems, and methods for controlling gene expression and epigenetic memory utilizing synthetic fusion proteins comprising a DNA binding protein or domain or fragment thereof and a nanobody configured to bind a transcriptional regulator (e.g., a chromatin regulator).

Description

NANOBODY-MEDIATED CONTROL OF GENE EXPRESSION AND EPIGENETIC MEMORY FIELD [0001] The present invention relates to methods and systems of controlling gene expression and epigenetic memory utilizing synthetic fusion proteins comprising a DNA binding protein or domain or fragment thereof and a nanobody configured to bind a chromatin regulator. CROSS-REFERENCE TO RELATED APPLICATIONS [0002] This application claims the benefit of U.S. Provisional Application No. 63/242,898, filed September 10, 2021, the content of which is herein incorporated by reference in its entirety. STATEMENT REGARDING FEDERALLY-SPONSORED RESEARCH [0003] This invention was made with Government support under contract GM128947 awarded by the National Institutes of Health. The Government has certain rights in the invention. SEQUENCE LISTING STATEMENT [0004] The contents of the electronic sequence listing titled STDU2-38758-601.xml (Size: 5,344,904 bytes; and Date of Creation: September 9, 2022) is herein incorporated by reference in its entirety. BACKGROUND [0005] Controlling gene expression and epigenetic memory is important for many biological processes including development and cancer, as well as for developing synthetic biology applications that rely on genetic engineering of cells (e.g., human cells). Thus, development of tools that can target a specific gene and alter its expression in a controlled manner is desirable for studying these biological process and genetic engineering. Existing tools include one or more effector domains from transcription factors or chromatin regulators fused with sequence specific DNA binding elements to target select genes in a genome. In mammalian cells, the most efficient way to control gene expression and impart epigenetic memory is with chromatin regulators. However, chromatin regulators are large, consisting of multiple domains that are necessary for their function, either to stimulate catalytic function or to mediate interactions with other members of the complex. Due to this large size, it is difficult to build compact gene regulatory tools for delivery to cells via viral delivery vectors such as adeno-associated virus (AAV) (payload limit ^ 4.7 kb) or lentivirus (payload limit ^ 9.7 kb). This challenge is exacerbated due to the size of the sequence specific DNA binding elements, e.g., Cas9 is over 4.5 kb. Methods which overcome the size and viral packaging challenges often result in poor editing efficiency due to the inability to effectively deliver all the components for gene editing to any one given cell. SUMMARY [0006] In some embodiments, provided herein are methods for identifying and selecting nanobodies. In some embodiments, the methods comprise identifying and selecting nanobodies for a target chromatin regulator comprising at least one or all of: a) preparing a yeast nanobody library comprising yeast cells each displaying a single nanobody on its surface; b) mixing the yeast nanobody library with a target chromatin regulator wherein the target chromatin regulator is configured to bind to a solid surface; c) separating yeast cells bound to target chromatin regulator from unbound yeast cells; d) amplifying nanobody DNA sequences from the yeast cells bound to target chromatin regulator; e) cloning the nanobody sequences into a human lentiviral vector to prepare a nanobody lentiviral library, wherein each nanobody DNA sequence is expressed as a fusion protein with a DNA binding domain; f) transforming the nanobody lentiviral library into reporter cells, wherein a reporter cell comprises a two-part reporter gene comprising a surface marker and a fluorescent protein, wherein the two-part reporter gene is capable of being silenced or induced by the target chromatin regulator; g) separating the reporter cells based on presence or absence of the surface marker, the fluorescent protein, or a combination thereof; and h) identifying the nanobody as a binding protein for the target chromatin regulator and capable of regulating gene expression. In some embodiments, the methods further comprise repeating steps a-d one or more times. [0007] In some embodiments, the chromatin regulator comprises a methylase, a demethylase, an acetylase, or ATP-dependent chromatin remodeling complex. [0008] In some embodiments, the DNA binding domain is an inducible DNA binding domain and the method further comprising treating the reporter cells with an agent configured to induce the inducible DNA binding domain for a length of time. [0009] In some embodiments, the two-part reporter gene is under the control of a strong promoter and capable of being silenced by a functional chromatin repressor. In some embodiments, the two-part reporter gene is under the control of a weak promoter and capable of being induced by a functional chromatin activator. [0010] Also provided herein are fusion proteins comprising a DNA binding protein, DNA binding domain, or a functional fragment thereof covalently linked to at least one nanobody configured to bind a chromatin regulator. In some embodiments, one or more of at least one nanobody is identified by a method disclosed herein. In some embodiments, the chromatin regulator comprises DNA (cytosine-5)-methyltransferase 3A (DNMT3A), or complex thereof. In some embodiments, the chromatin regulator comprises a ten-eleven translocation methylcytosine 1/2/3 (TET1/2/3) complex. [0011] In some embodiments, the at least one nanobody comprises at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFXXXXM (SEQ ID NO: 13), EZVAXIXXGXXTNY (SEQ ID NO: 14), and AAXXXXXYYXXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid. In certain embodiments, the at least one nanobody comprises a first CDR comprising an amino acid sequence of GTIFXXXXM (SEQ ID NO: 13), a second CDR comprising an amino acid sequence of EZVAXIXXGXXTNY (SEQ ID NO: 14), and a third CDR comprising an amino acid sequence of AAXXXXXYYXXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid. [0012] In some embodiments, the at least one nanobody is configured to bind DNA (cytosine-5)-methyltransferase 3A (DNMT3A) complexes. In some embodiments, the nanobody comprises at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), GNIFDGASM (SEQ ID NO: 4), EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), AAGRYYYPGHGY (SEQ ID NO: 8), AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 12). In certain embodiments,^the at least one nanobody comprises: a first CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), and GNIFDGASM (SEQ ID NO: 4); a second CDR comprising an amino acid sequence selected from the group consisting of EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), and AAGRYYYPGHGY (SEQ ID NO: 8); a third CDR comprising an amino acid sequence selected from the group consisting of AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 12); or a combination thereof. [0013] In some embodiments, the nanobody comprises an amino acid sequence at least 70% (e.g., 80%, 85%, 90%, 95%, 98%, 99%) similar to any of SEQ ID NOs: 16-20. [0014] In some embodiments, the at least one nanobody comprises at least one CDR having an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933-5959. In certain embodiments, the at least one nanobody comprises: a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933-5941; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5942- 5950; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5951-5959; or a combination thereof. [0015] In some embodiments, the nanobody comprises an amino acid sequence at least 70% similar to any of SEQ ID NOs: 5960-5968. In some embodiments, the nanobody comprises an amino acid sequence of SEQ ID NOs: 5960-5968. [0016] In some embodiments, the fusion protein further comprises a transcription factor or regulator. In select embodiments, the transcription factor or regulator comprises Krüppel- associated box (KRAB). [0017] Further provided are DNA (cytosine-5)-methyltransferase 3A (DNMT3A) binding proteins (e.g., antibodies and fragments thereof), and fusion proteins thereof, comprising at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFXXXXM (SEQ ID NO: 13), EZVAXIXXGXXTNY (SEQ ID NO: 14), and AAXXXXXYYXXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid. In some embodiments, the at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), GNIFDGASM (SEQ ID NO: 4), EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), AAGRYYYPGHGY (SEQ ID NO: 8), AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), AND AAKPSRVYWRDYRFFY (SEQ ID NO: 12). In select embodiments, the DNMT3A binding protein comprises an amino acid sequence at least 70% similar to SEQ ID NOs: 16-20. [0018] Further provided are TET1/2/3 complex binding proteins (e.g., antibodies and fragments thereof), and fusion proteins thereof. [0019] In some embodiments, the TET1/2/3 complex binding proteins comprises at least one CDR having an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933- 5959. In certain embodiments, the TET1/2/3 complex binding proteins comprises: a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933- 5941; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5942-5950; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5951-5959; or a combination thereof. [0020] In some embodiments, the TET1/2/3 complex binding proteins comprises an amino acid sequence at least 70% similar to any of SEQ ID NOs: 5960-5968. In some embodiments, the TET1/2/3 complex binding proteins comprises an amino acids sequence of any of SEQ ID NOs: 5960-5968. [0021] In some embodiments, the DNMT3A binding protein or TET1/2/3 complex binding protein is covalently attached to a DNA binding protein, DNA binding domain, or a functional fragment thereof. [0022] Nucleic acids and vectors encoding the fusion proteins, nanobodies, DNMT3A binding proteins, and TET1/2/3 complex binding proteins, and compositions and cells comprising thereof are provided. [0023] Additionally provided are methods for modifying gene expression, epigenetic memory, or a combination thereof of at least one target nucleic acid. The methods may comprise contacting a target nucleic acid with a fusion protein as described herein. In some embodiments, the methods comprise contacting the target nucleic acid with at least one fusion protein comprising a DNA binding protein, DNA binding domain, or a functional fragment thereof covalently linked to a nanobody configured to bind a chromatin regulator. In some embodiments, the gene expression and/or epigenetic memory of at least two genes are modulated. [0024] In some embodiments, the at least one target nucleic acid is in a cell and the chromatin regulator is endogenous to the cell. Thus, in some embodiments, the contacting comprises introducing into the cell the fusion protein, or a nucleic acid encoding thereof. In some embodiments, the introducing comprises administering to a subject. [0025] In some embodiments, the methods further comprise contacting the target nucleic acid with an exogenous chromatin regulator. [0026] In some embodiments, the endogenous or exogenous chromatin regulator comprises a methylase, a demethylase, an acetylase, or ATP-dependent chromatin remodeling complex. [0027] In some embodiments, the nanobody is identified or selected by a method as disclosed herein. In select embodiments, the chromatin regulator comprises DNA (cytosine-5)- methyltransferase 3A (DNMT3A). In select embodiments, the chromatin regulator comprises a TET1/2/3 complex. [0028] In some embodiments, the methods further comprise contacting the target nucleic acid with a transcription factor or regulator. In select embodiments, the fusion protein further comprises the transcription factor or regulator. In some embodiments, the transcription factor or regulator comprises a transcriptional repressor. In some embodiments, the transcription factor comprises Krüppel-associated box (KRAB). [0029] Other aspects and embodiments of the disclosure will be apparent in light of the following detailed description. BRIEF DESCRIPTION OF THE DRAWINGS [0030] FIGS. 1A-1D show that nanobodies against GFP-tagged chromatin regulators allow for gene expression control. FIG. 1A is a schematic of an exemplary construct for constitutive coexpression (under the pGK promoter) of H2B-mIFP and antiGFP nanobody fused with the rTetR DNA-binding protein (separated by the self-cleaving peptide T2A) randomly integrated into HEK293T cells by PiggyBac (blue box, top). These cells also contain a TagRFP reporter gene integrated at the AAVS1 “safe harbor” locus and driven by the pEF promoter (bottom). Five TetO-binding sites allow binding of rTetR upstream of the reporter gene upon dox addition. The nuclear localization signal (NLS) and H2B domains localize fluorescent protein signals to the nucleus, improving quantification during time-lapse imaging. Plasmids expressing GFP- tagged CRs (HP1Į, HP1ȕ, HP1Ȗ, and HDAC5) were transiently transfected into cells (green box, right). FIG. 1B is time-lapse imaging of cells upon recruitment of GFP-tagged HP1Į (top) and HDAC5 (bottom). Cells stably expressing the reporter (TagRFP, red) and the rTetR-antiGFP fusion (mIFP, purple) were transiently transfected with GFP-HP1Į or GFP-HDAC5 (GFP, green). Cells were treated with dox throughout the movie starting at time 0 hours. Example images across the time-lapse movie (left) and single-cell traces for individual cell lineages as they undergo up to eight cell divisions (right). FIG. 1C shows cells that still have GFP-CR expression (yellow circles) by day 5 of recruitment remain silenced. Images and analysis in FIG. 1B and 1C are from one biological replicate. Scale bars: 20 ^m. FIG.1D, left is a graph of fluorescence distributions measured by flow cytometry showing reporter silencing after recruitment of GFPCRs (+dox) for 5 days. Cells were gated for the presence of both GFP-CR (GFP positive) and rTetR-antiGFP (mIFP positive). The red dotted line was used to determine the percentage of cells silenced shown on the right. FIG.1D, right is a graph of mean percentage of cells silenced upon presence or absence of dox for 5 days. Each dot is an independently transfected biological replicate (GFP-HP1Į: n = 7; GFP-HP1ȕ: n = 3; GFP-HP1Ȗ: n = 3; GFP- HDAC5: n = 7; GFP: n = 3). Statistical analysis by two-tailed Tukey’s test (GFP-HP1Į vs. GFP- HP1Ȗ: ****p = 4.8eí^^^^*)3-HP1ȕ vs. GFP-HP1Ȗ: **p =0.0031; GFP-HP1Ȗ vs. GFP-HDAC5: ****p = 9.0e − 6). [0031] FIGS. 2A-2E show that nanobody-mediated recruitment of endogenous chromatin regulators can silence gene expression and confer memory. FIG. 2A is a schematic of an exemplary construct for constitutive coexpression of H2B-mIFP and nanobody against CR (antiHP1 or antiDNMT1) fused with the rTetR DNA-binding protein (blue box, top) expressed in HEK293T cells containing a TagRFP reporter (bottom). FIG. 2B, left is a graph of fluorescence distributions of the TagRFP reporter after 5 days of recruitment (dox treatment) in cells stably expressing the nanobody constructs or rTetR-KRAB were analyzed by flow cytometry to determine the percentage of cells with the reporter silenced (left of the red dotted line). FIG. 2B, right is a graph of the percentages of cells silenced as calculated from 3 replicates. Statistical analysis by two-tailed Tukey’s test (antiHP1 vs. antiDNMT1: ****p=5.6eí^^^DQWL+3^^YV^^ antiDNMT1-antiHP1: ****p=2.5eí^^^^DQWL'107^^YV^^DQWL'107^-antiHP1: ****p=1.8eí^^^^ antiDNMT1-antiHP1 vs. KRAB: ****p=1.7e−7). FIG.2C is targeted bisulfite sequencing of the reporter after 5 days of recruitment with antiDNMT1, DNMT1, and DNMT3B (+dox), compared to untreated cells expressing the same effectors (−dox). Dox-treated cells were sorted based on the silencing of the TagRFP reporter labeled as +dox ON and +dox OFF (see FIG. 12A for representative gating). CpG positions are calculated relative to the start of the most upstream TetO site. Positive and negative controls for DNA methylation are shown in FIG. 12B. FIG. 2D shows the experimental design for investigating epigenetic memory: rTetR-effectors were recruited to the reporter for 5 days (+dox) and then released (−dox). Memory was monitored after dox removal via flow cytometry throughout 30 days. FIG. 2E is a graph of silencing and memory dynamics data (right) for the experiment described in FIG. 2D with representative flow cytometry histogram for antiDNMT1 at day 0, 15, 30 after dox removal (left). The percentage of cells silenced was normalized to the no dox control to adjust for any background silencing. Means are from three replicates. SDs are plotted but are too small to show for most data points. Statistical analysis by two-tailed Tukey’s test at day 30 after dox removal (antiHP1 vs. antiDNMT1: **p=0.0076; antiDNMT1 vs. KRAB: ***p=0.00026; KRAB vs. antiDNMT1- antiHP1: *p=0.043). [0032] FIGS. 3A-3F show nanobody-mediated enhancement of KRAB and DNMT3A repression. FIG. 3A, left is a graph of fluorescence distributions of the reporter gene after transient expression of rTetR-effector fusions and recruitment by dox treatment for 5 days, measured using flow cytometry. FIG. 3A, right is the means of percent cells silenced (to the left of the red dotted line in the left-side plots) are shown as bars. Each dot represents an independently transfected biological replicate (KRAB: n = 5; KRAB-antiDNMT1: n = 5; KRAB-antiDNMT1-antiHP1: n= 3; statistical analysis by one-way ANOVA). After recruitment with rTetR-effector fusions for 5 days, silenced cells were sorted and memory dynamics were measured by flow cytometry throughout 30 days (FIG. 3B). Data represent three biological replicates and are fitted with an exponential decay curve. Two-tailed Tukey’s test analysis at day 30 after sorting (KRAB vs. KRABantiDNMT1: ***p= 0.00022; KRAB vs. KRAB-antiDNMT1- antiHP1: **p = 0.0016). FIG. 3C is fluorescence distributions after transient expression and targeting of dCas9-effector fusions to the TetO sites upstream of the reporter gene (+) or to a safe-targeting control site (−) for 5 days. Means are from two biological replicates. After targeting the dCas9-effector fusions for 5 days, silenced cells were sorted, and memory dynamics was measured by flow cytometry throughout 40 days (FIG. 3D). Each dot is a biological replicate (KRAB: n = 2; KRAB-antiDNMT1: n =3; KRAB-MeCP2: n= 3; KRAB-MeCP2- antiDNMT1: n = 2) and are fitted with an exponential decay curve (“Methods”). Two-tailed Tukey’s test at day 40 after sorting (KRAB-MeCP2 vs. KRAB-antiDNMT1: *p = 0.040). FIG. 3E is a graph of the transient expression and recruitment of rTetR-based DNA methyltransferase combinations to the reporter gene for 5 days (DNMT3A: n = 2; antiDNMT1-DNMT3A: n = 4; DNMT3A-3L: n = 4; antiDNMT1-DNMT3A-3L: n = 6). Statistical analysis by two-tailed Tukey’s test (antiDNMT1-DNMT3A vs. antiDNMT1-DNMT3A-3L: ***p =0.00044; DNMT3A-3L vs. antiDNMT1-DNMT3A-3L: ***p =0.00015). FIG. 3F is graphs of corresponding silencing dynamics of rTetR-based combinations in FIG. 3E throughout 5 days of recruitment (n = 3 replicates). Statistical analysis by two-tailed Tukey’s test (DNMT3A vs. antiDNMT1-DNMT3A: ***p2 = 0.00023, ***p3 = 0.00051, ****p4 = 4.3e − 8, ****p5 = 3.5e− 7 | DNMT3A-3L vs. antiDNMT1-DNMT3A-3L: ****p2 =5.3e − 8, **p3 =0.0033, ****p4 = 5.1e − 5, **p5 = 0.0019). [0033] FIGS. 4A-4G show nanobodies as signal detection and recording tools. FIG. 4A is a schematic of an exemplary device for measuring and recording signal duration. The input signal is coupled to the recruitment of rTetR-antiDNMT1 near the pEF promoter to silence an output gene. FIG. 4B is a graph of the percentages of cells with TagRFP reporter silenced as measured by flow cytometry at the end of the indicated dox signal durations in a cell line stably expressing rTetR-antiDNMT1. Means and standard deviations (SDs) of experimental data from three replicates (red dots) and linear fit (black). FIG. 4C shows the percentage of cells with TagRFP silenced after different signal (dox treatment) durations: 14 days (top) and 5 days (bottom). The gray shaded regions (negative numbers) indicate the period with dox. Dashed lines indicate dox removal. Continuous lines represent fits to the model in FIG. 4D for the 14 days data and predictions of the model for the 5-day data. FIG. 4D is a schematic of the three-state model of silencing by antiDNMT1 during recruitment (+dox, top) and during release (−dox, bottom). FIG. 4E is a graph of the percentage of cells irreversibly silent after different durations of recruitment (dox treatment) predicted by the model in FIG. 4D plotted as a black line. Experimental data recorded at 7 days during the release period shown as black dots if used for model fitting (14 days), or blue diamonds if not used in the fit. FIG. 4F shows the percentages of cells silenced relative to no dox controls for pulsed recruitment (top: 3 days +dox, 2 days −dox, 2 days +dox) compared to continuous recruitment for the same duration (bottom: 5 days +dox, 2 days −dox). Experimental data from three replicates shown as black dots, means as gray bars, and model predictions with 95% CI in red (“Methods”). FIG. 4G shows the percentages of cells silenced relative to no dox controls for pulsed recruitment vs. continuous recruitment, recorded at the same time after dox removal, plotted as in FIG. 4F. [0034] FIGS. 5A and 5B show schematics for a platform for selecting nanobodies capable of gene regulation in human cells. FIG. 5A, left shows yeast cells (black circles) carrying a diverse library of genetically encoded nanobodies (McMahon et al. Nature Structural & Molecular Biology Volume 25, pages 289–296 (2018), incorporated herein by reference in its entirety). Each yeast cell expresses and displays a single nanobody on its surface (colored lollipops). Yeast cells are mixed with FLAG-tagged chromatin regulator (CR) complexes (brown) purified from human cells. FIG. 5A, middle shows the FLAG tag immobilization of the CR complexes to the surface of magnetic beads (green) coated with anti-FLAG antibodies (Yeast Display). Yeast cells containing nanobodies that bind to the CR complex are enriched after magnetic separation (Enriched Yeast Nanobody Library). FIG. 5A, right shows the extraction of genomic DNA from the enriched yeast library, amplification of the DNA sequences encoding for the selected nanobodies, and recloning as a pool into a human expression lentiviral vector (Lentiviral Nanobody Library). An exemplary lentiviral cloning vector is shown on the bottom, where each nanobody in the library is cloned downstream of the DNA binding domain rTetR. FIG. 5B shows human cells infected with the pooled nanobody lentiviral library (hexagons), at a low infection rate, such that each cell expresses a single nanobody. Each nanobody is expressed as a fusion protein with the DNA binding domain rTetR, and in the presence of doxycycline (+dox) can bind to the TetO sites upstream of a pEF promoter driving the expression of a reporter gene (Tycko J. et al., Cell. 2020 Dec 23;183(7):2020-2035, incorporated herein by reference in its entirety). The reporter gene consists of a citrine fluorophore and a surface marker used for magnetic separation. In cells expressing a nanobody that can recruit endogenous repressive CRs (brown) without inhibiting their function, the citrine reporter gene will turn off. DNA encoding for functional nanobodies will be enriched in the OFF vs. the ON population. [0035] FIGS. 6A-6D show that yeast display against DNMT3A results in nanobody library enrichment. FIG. 6A is a schematic showing experimental workflow: the naive nanobody library and the enriched libraries after each round of selection against DNMT3A are extracted from yeast using a genomic DNA extraction kit and sequenced using NGS Illumina sequencing (NGS seq). FIG. 6B is graphs of the results of NGS sequencing showing the number of nanobodies in each library and their respective frequencies. Nanobody sequences to the right of the brown line are enriched in Round 2 and 3 compared to the Round 1 and Naive libraries. FIG. 6C is a schematic showing yeast display setup for measuring yeast library enrichment against the DNMT3A complex by flow cytometry. Yeast cells from Round 3 of yeast display selection against DNMT3A are incubated with cell lysate from human HEK-293T cells expressing FLAG- tagged DNMT3A (as well as other human contaminant proteins). Addition of antiFLAG antibody conjugated with the fluorescent dye FITC stains the yeast cells that bind the DNMT3A. FIG. 6D is a probability distribution showing the number of yeast cells with a particular FITC fluorescence as measured by flow cytometry after Round 3 of selection (green) vs. the Naive Library. [0036] FIGS. 7A-7E show the characterization of individual antiDNMT3A nanobodies for gene silencing in human cells. Twenty-four nanobodies (see Table 2 for sequences) were chosen at random from the Lentiviral Nanobody Library cloned after Round 3 enrichment against DNMT3A. FIG. 7A is a western blot showing co-immunoprecipitation of each rTetR-3xFlag- Nanobody and 3xHA-DNMT3A in HEK-293T cells. FIG. 7B shows the percentage of cells with the citrine reporter silenced (off) in (top) K562 and (bottom) HEK-293T cells after 5 days of dox-mediated recruitment of fusions between rTetR and each of the 24 nanobodies, as measured by flow cytometry. rTetR without a nanobody is used as a negative control (NC). The rTetR- KRAB is used as a positive control for repression. Nanobody #8 has the strongest repressive effect. FIGS. 7C, 7D, and 7E, left are graphs showing the percentage of cells with the citrine reporter off after recruitment of rTetR, rTetR-KRAB, rTetR-nanobody2 (NB#2), and rTetR- nanobody8 (NB#8) for 3 days (blue) or 5 days (red) in K562 (FIG. 7C), HEK-293 (FIG. 7D), and Hela cells (FIG. 7E). FIGS. 7C, 7D, and 7E, right are citrine fluorescent distributions measured using flow cytometry and used to calculate the percentages on the left. [0037] FIGS. 8A and 8B show NB #8 can silence gene expression when fused to dCas9. FIG. 8A is a graph of GFP fluorescent distributions of an pSV40-GFP reporter as measured by flow cytometry after recruitment of NB#8-dCas9 with a sgRNA that targets the pSV40 promoter (red). dCas9 alone (black) serves as a negative control and KRAB-dCas9+sgRNA is a positive control (CRISPRi). FIG. 8B is a graph of the percentages of cells with pSV40-GFP silenced calculated from flow cytometry distributions upon targeting of the pSV40 promoter with the indicated fusions. [0038] FIGS. 9A-9C show that NB#8 recruitment can lead to epigenetic memory in certain cell types. Percentages of HEK-293 (FIG. 9A), K562 (FIG. 9B), and Hela (FIG. 9C) cells with the citrine reporter silenced are calculated from flow cytometry distributions during 5 days of dox-mediated recruitment (Dox+) and after release (Dox-) of rTetR fused NB#8 or NB#2. rTetR without a nanobody is used as a negative control (NC). The rTetR-KRAB is used as a positive control. [0039] FIGS. 10A-10D show that the disclosed high-throughput recruitment assay can identify antiDNMT3A nanobodies that repress gene expression in human cells. The entire Lentiviral Nanobody Library obtained by pooled Golden Gate cloning of the Round 3 nanobodies selected against DNMT3A was used to produce lentivirus and infect HEK-293 cells at a low MOI (FIG. 10A). Cells were selected with puromycin for integration of the nanobody constructs and dox was added for 5 days to induce recruitment of each nanobody upstream of a fluorescent-magnetic reporter gene, as described in FIG. 5B. Cells with the gene OFF were separated from those that remained ON using magnetic separation. Genomic DNA was extracted, PCR amplified for the region encoding for the nanobodies, and sequenced using NGS Illumina sequencing. The frequency of reads for each nanobody sequence was measured in the bound (ON) and unbound (OFF) populations and used to calculate an OFF:ON score. Log2(OFF:ON) scores from 2 biological replicates are plotted against each other. Nanobody NB#8 (pink star), which was known to efficiently repress gene expression from the miniscreen in FIG. 7, appears among the hits with high OFF:ON ratios. Four other nanobodies among the hits were selected for further validation: NB#25, NB#26, NB#27, and NB#28, also shown as stars. The other nanobodies that were individually tested in FIG. 7B did not efficiently silence gene expression are shown as dark gray dots, and also exhibit low OFF:ON scores in the screen. FIG. 10B shows the validation of individual anti-DNMT3A nanobodies recovered from the high-throughput screen in HEK-293 cells. Gene blocks encoding for each nanobody sequence were individually cloned as fusions with rTetR and were delivered to HEK-293 cells by lentivirus to perform silencing experiments. All 5 nanobodies efficiently silence gene expression when recruited to the reporter by dox addition for 2 (green) or 5 (red) days compared to the no dox controls (black). Correlation between log2(OFF:ON) screen scores and the fraction of cells with citrine off as measured individually by flow cytometry is shown in FIG. 10C for all nanobodies listed in FIGS. 7B and 10B. Nanobodies were collapsed on sequence identity when applicable, and nanobodies with too few sequencing reads to compute reliable screen scores were excluded from this analysis. FIG. 10D is multiple sequence alignment with the five validated nanobodies that exhibit strong repressive activity. CDR locations are indicated. Nanobody NB#8, NB#25, NB#26, NB#27, and NB#28 are SEQ ID NOs: 16-20, respectively. Concensus sequence is SEQ ID NO: 5932. [0040] FIGS 11A-11F show localization dynamics of GFP-tagged chromatin regulators and recruitment GFP-tagged chromatin regulators. Time-lapse images of cells transiently expressed with GFP-tagged HP1Į (FIG. 11A) and HDAC5 (FIG. 11B). Cells undergoing cell division are represented at time 0 hours. Yellow boxes highlight the re-entry of GFP-tagged chromatin regulators into the nucleus. Time-lapse experiment from 1 biological replicate. White scale bars represent 10 ^m. FIG. 11C is a representative example of mIFP and GFP gating for data in FIG. 1D. FIG. 11D shows the data from FIG. 1D gated based on different GFP expression levels and analyzed for the percentage of cells with the TagRFP reporter silenced at day 4 of dox. FIG. 11E is a graph of the percentage of cells with reporter silenced after recruitment of an 8x repeat array of antiGFP nanobodies (+dox for 5 days). The experimental set up is the same as in FIGS. 1A and 1D, with the reporter and nanobody array constructs stably integrated, and GFP-CRs transiently transfected. Each dot is an independently transfected biological replicate (GFP-HP1Į: n = 6; GFP-HP1ȕ: n = 3; GFP-HP1Ȗ: n = 3; GFP-HDAC5: n = 6). FIG. 11F is a graph of the percentage of cells with TagRFP silenced after four days of recruitment with antiGFP (grey) or antiHP1 nanobody (orange). Cells were also co-expressed with GFP or GFP-HP1Į (diagonal lines). Means are from 3 replicates; statistical analysis by two-tailed unpaired t-test (antiGFP + GFP-HP1Į vs. antiHP1 + GFP: ****p = 8.8e-7; antiGFP + GFP-HP1Į vs. antiHP1 + GFP-
Figure imgf000014_0001
[0041] FIGS. 12A-12E show targeted bisulfite sequencing controls and treatment of cells with DNA or histone methylation inhibitors. FIG. 12A shows representative sorting for targeted bisulfite sequencing after 5 days of recruitment with antiDNMT1, DNMT1, and DNMT3B (left) and in the absence of dox (right). FIG. 12B is the targeted bisulfite sequencing of two control genes, IGF2 (silent gene with imprinted DNA methylation) and ACTB (active gene, no DNA methylation expected) in the same cell populations as in FIG. 2C. Also included are non- methylated DNA controls from the human HCT116 DKO cell line that contains knockouts of DNA methyltransferases DNMT1 and DNMT3B. Methylated DNA was obtained from the non- methylated HCT116 DKO genomic DNA by in vitro treatment with the M. SssI methyltransferase. Graphs comparing the percentage of cells with TagRFP silenced after four days of recruitment with doxycycline and 5-aza-2’-deoxycytidine (5-Aza-2’), a DNA methyltransferase inhibitor (FIG. 12C), or chaetocin, a broad-spectrum inhibitor of lysine histone methyltransferases (FIG. 12D). Means are from 3 replicates; statistical analysis by two-tailed unpaired t-test (5-Aza-2’: antiDNMT1 ****p = 8.0e-14; antiHP1 **p = 0.0046 | Chaetocin: antiDNMT1 ****p = 2.8e-5; KRAB ****p = 3.5e-5, antiHP1 ***p = 0.00019). FIG. 12E is a schematic for an exemplary expression vector for H2B-mIFP and the rTetR-antiDNMT1- antiHP1 fusion under a pGK constitutive promoter with sizes of the DNA encoding for the nanobodies shown in base pairs (bp). [0042] FIGS. 13A and 13B show transient expression and recruitment of nanobodies can also silence gene expression and confer memory. FIG. 13A is graphs of reporter fluorescent distributions(left) and percent cells silent (right) after transient expression of rTetR-effector fusions and 5 days of dox treatment. Each dot is an independently transfected biological replicate (antiDNMT1: n = 1; antiHP1: n = 1; antiDNMT1-antiHP1: n = 5; KRAB: n = 5). FIG. 13B is a graph of the cells silenced by KRAB and antiDNMT1-antiHP1 sorted after 5 days of dox treatment and analyzed by flow cytometry for memory. Each time point contains 3 biological replicates (individual dots). Data were fitted with an exponential decay curve (lines, Methods). Statistical analysis by two-tailed Tukey-test at day 30 after sorting (KRAB vs. antiDNMT1- antiHP1: ***p = 0.00060). [0043] FIGS. 14A-14E show separate co-recruitment of regulators to the reporter gene and CXCR4 endogenous gene silencing. FIG. 14A is a schematic of an expression vector for rTetR- KRAB-antiDNMT1 compared to the previously published KRAB-DNMT3A-3L fusion. FIG. 14B is a graph of the percent cells with reporter silenced (relative to no dox controls) after co- recruitment of separate fusion of rTetR-effectors at the TagRFP reporter gene. Experimental setup is the same as in FIGS. 2A and 2D. Included for reference are the percentages of cells permanently silenced after individual recruitment with KRAB, antiDNMT1, or antiHP1 (dashed lines) taken from FIG. 2E. FIG. 14C is a schematic of an expression vector for dCas9-KRAB- antiDNMT1 under a CMV constitutive promoter. The sgRNA (targeting either the TetO site or a safe genomic site with no annotated function) was expressed on a different vector driven by the mouse U6 promoter and contained a mIFP marker. FIG. 14D is a diagram of sgRNA binding sites for the targeting of dCas9-KRAB-antiDNMT1 to the endogenous CXCR4 gene. CXCR4 is a cell surface transmembrane protein, which enables us to use conjugated fluorescent antibodies with flow cytometry to quantify gene expression. Five sgRNAs were cloned spanning the upstream region of the transcriptional start site of this gene, targeting either the template or non- template strand. The dCas9 and sgRNA constructs were modified to express mCitrine and mCherry, respectively, to allow for cell sorting. After transient expression and targeting at the endogenous CXCR4 gene for 4 days, cells were sorted for the presence of both dCas9 (mCitrine positive) and sgRNA (mCherry positive). Cells were then immunostained for CXCR4 expression and analyzed by flow cytometry (FIG. 14E, left). Means of percent cells with silent CXCR4 from 2 replicates are shown throughout 17 days after sorting (FIG. 14E, right). [0044] FIGS. 15A and 15B show a fusion of antiDNMT1 nanobody to DNMT3A-3L and HDAC4. After the rTetR-based DNA methyltransferases combinations were transiently transfected and treated with dox for 5 days, silenced cells were sorted, and reactivation was measured by flow cytometry throughout 30 days (FIG. 15A). Each dot is a biological replicate (DNMT3A-3L: n = 1; antiDNMT1-DNMT3A: n = 1; antiDNMT1-DNMT3L: n = 1; antiDNMT1-DNMT3A-3L: n = 3) and the data are fitted with an exponential decay curve. FIG. 15B is a graph of the transient expression and recruitment of the rTetR-HDAC4-antiDNMT1 fusion to the reporter gene throughout 5 days (n = 4 replicates). Statistical analysis by two-tailed unpaired t-test (HDAC4 vs. HDAC4-antiDNMT1: *p3 = 0.043, ***p4 = 0.00035, ***p5 = 0.00024). [0045] FIGS. 16A and 16B show comparisons of different models of antiDNMT1-mediated silencing for pulsed recruitment FIG. 16A is a graph of predictions of 3-state silencing model for pulsed recruitment with different values of the second lag time between dox addition and start of silencing (Tlag2). The predictions of the model shown in red (Tlag2 = 0) are plotted next to experimental data in FIGS 4F-4G. FIG. 16B are graphs of the comparison of model predictions and experimental data for the 5 days (left) or 7 day (right) pulsed data for the model where the lag time between dox addition and silencing on the second recruitment pulse is equal to the one on the first pulse. Experimental data from 3 replicates shown as black dots, means as gray bars, and model predictions with 95% CI in blue. [0046] FIGS. 17A-17D shows generation of cells with an epigenetically silenced citrine reporter gene. FIG. 17A is a schematic of human DNMT3A fused to rTetR and transfected transiently into HEK293A cells expressing the 9xTetO-pEF-surface marker-citrine reporter. Dox was added during the transient transfection, leading to recruitment of DNMT3A at the reporter and silencing of the citrine gene. Dox was then removed, cells were passaged for >20 days, and cells that maintained the reporter silenced in the absence of dox (indicating epigenetic silencing) were sorted and expanded. FIGS. 17B-17D are flow cytometry measurements of citrine levels histograms (top) and cell forward scatter against citrine levels (bottom) for wild type HEK293A cells without a reporter(FIG. 17B), HEK293A cells with the citrine reporter integrated and rTetR transiently transfected (FIG. 17C), and HEK293A cells with the citrine reporter integrated and rTetR-DNMT3A transiently transfected (FIG. 17D). Data shown was collected 20 days after transient transfection and dox removal. Cells with the citrine reporter silenced after transient DNMT3a transfection were sorted (citrine OFF, quadrant Q4), and a stable population was maintained and used for screening and individual validations of nanobodies that can activate an epigenetically silenced gene. [0047] FIGS. 18A-18C show the yeast display screen against TET1/2/3 complexes. FIG. 18A is a schematic of the yeast display was performed against chromatin regulator complexes containing full length, FLAG-tagged human TET1, mouse TET2, and human TET3 that were overexpressed in HEK293T cells and immunoprecipitated using anti-FLAG coated magnetic beads. The same protocol was used as for yeast display against, except only two rounds of selection were performed. Briefly, yeast cells containing a naïve nanobody library were mixed with the FLAG-TET complexes, and two rounds of selection were performed using anti-FLAG magnetic beads. FIG. 18B shows measurement of nanobody enrichment. Yeast cells were mixed with the FLAG-TET1/2/3 complexes and stained with fluorescently-labeled anti-FLAG antibody (488nm wavelength, y axis). The x axis measures nanobody expression using an anti-HA antibody (647nm wavelength). An enrichment of anti-FLAG signal (in quadrant Q2) is observed after round 1 and round 2 of selection, indicating successful enrichment for nanobodies that can bind to the TET1/2/3 complexes. In FIG. 18C, genomic DNA from the naïve and round 2 nanobody libraries was sequenced and the frequency of each detected nanobody was quantified. An increased frequency of a subset of nanobodies was observed, indicating the size of the library decreased due to the yeast display selection. The round 2 nanobodies against TET1/2/3 complexes were used for further studies in human cells. [0048] FIGS. 19A-19D show that a high-throughput recruitment assay can identify anti- Tet1/2/3 complex nanobodies that activate gene expression in human cells. The library of nanobodies against TET1/2/3 complexes from round 2 of yeast display were cloned fused to rTetR, and delivered via lentivirus to HEK293A cells containing the epigenetically silenced reporter (FIG. 19A). Viral delivery was performed at low MOI, to ensure delivery of a single nanobody per cell. After dox addition, each nanobody is recruited to the silenced reporter, and can activate the gene. Cells are then separated into re-activated (citrine ON) and still-silenced (OFF). Nanobodies that reactivate the gene are enriched in the ON population. FIG. 19B is a graph of Log2(ON:OFF) scores per replicate of the anit-TET1/2/3 complex nanobody recruitment screen in HEK293 cells. Higher scores indicate enrichment of a particular nanobody sequence in the ON population. Nanobodies chosen for follow-up are denoted as stars. FIG. 19C shows individual validations for the 9 chosen nanobodies shown in (FIG. 19B). Flow cytometry measurements of citrine fluorescence distributions on day 10 of dox recruitment. VP64 was chosen as an activating positive control, rTetR alone (with no nanobody fused) was used as a negative control. Grey curves = no dox added, purple curves = dox added. FIG. 19D is a timecourse of dox-mediated activation starting from day 3 of dox addition (nanobody recruitment at the silenced reporter), plotted as fraction of cells that have the reporter re-activated (citrine ON). Gray curve at the bottom indicates stable epigenetic silencing of the citrine reporter with the rTetR alone control (no nanobody). DETAILED DESCRIPTION [0049] Systems and methods that use nanobodies (also referred to as single-domain antibodies) to recruit chromatin regulators (CRs) to a target gene or nucleic acid for silencing gene expression and imparting epigenetic memory are provided. Further, in some embodiments, the nanobodies are fused onto DNA binding proteins, or domains or functional fragments thereof. In some embodiments, the target gene or nucleic acid are in a cell and the methods comprise introducing into a cell (e.g., administering to a subject) the disclosed fusion proteins comprising the nanobody and the DNA binding proteins, or domains or functional fragments thereof, or a nucleic acid thereof to allow recruitment endogenous CRs from the existing cellular chromatin network, thus obviating the need for introducing a large exogenous chromatin remodeling complex. In some embodiments, the methods and system described herein enhance the functionality of commonly used transcriptional effectors, e.g., KRAB. For example, combining nanobodies together or with other regulators, such as DNMT3A or KRAB, may enhance silencing speed and epigenetic memory. [0050] Also provided are methods for identifying and selecting nanobodies which are capable of binding CRs and facilitating chromatin modifications, e.g., in a human cell. This technology leverages a yeast nanobody library and a high-throughput screen to characterize tens of thousands of potential nanobodies. Multiple rounds of negative and positive selection can be used to reduce the yeast nanobody library size to a few thousand members for use in the high- throughput screening platform, as necessary. Since only a couple of nanobodies against chromatin regulators exist, the disclosed methods allow identification of human chromatin regulator complexes that act as efficient modulators of gene expression in human cells for the development of efficient genetic engineering tools. The methods can be very easily extended to identify nanobodies against different endogenous protein targets that modulate other cellular processes for which reporters are easily available, including but not limited to: DNA repair, mRNA or protein stability, and immune responses. [0051] Exemplary applications for the methods disclosed herein include, but are not limited to: targeted chromatin remodeling with persistent memory, gene and cell therapy (e.g., to silence a pathogenic transcript in a patient) or in research, and use in genetic circuits, e.g., inducible gene expression or more complex circuits. These circuits find use in gene therapy (e.g., AAV delivery of antibodies) and cell therapy (e.g., ex vivo engineering of CAR-T cells) to achieve therapeutic gene expression outputs in response to environmental and small molecule inputs. 1. Definitions [0052] The terms “comprise(s),” “include(s),” “having,” “has,” “can,” “contain(s),” and variants thereof, as used herein, are intended to be open-ended transitional phrases, terms, or words that do not preclude the possibility of additional acts or structures. As used herein, comprising a certain sequence or a certain SEQ ID NO usually implies that at least one copy of said sequence is present in recited peptide or polynucleotide. However, two or more copies are also contemplated. The singular forms “a,” “and” and “the” include plural references unless the context clearly dictates otherwise. The present disclosure also contemplates other embodiments “comprising,” “consisting of” and “consisting essentially of,” the embodiments or elements presented herein, whether explicitly set forth or not. [0053] For the recitation of numeric ranges herein, each intervening number there between with the same degree of precision is explicitly contemplated. For example, for the range of 6-9, the numbers 7 and 8 are contemplated in addition to 6 and 9, and for the range 6.0-7.0, the number 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, and 7.0 are explicitly contemplated. [0054] Unless otherwise defined herein, scientific, and technical terms used in connection with the present disclosure shall have the meanings that are commonly understood by those of ordinary skill in the art. The meaning and scope of the terms should be clear; in the event, however of any latent ambiguity, definitions provided herein take precedent over any dictionary or extrinsic definition. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. [0055] The term “antibody,” as used herein, refers to a protein that is endogenously used by the immune system to identify and neutralize foreign objects, such as bacteria and viruses. Typically, an antibody is a protein that comprises at least one complementarity determining region (CDR). The CDRs form the “hypervariable region” of an antibody, which is responsible for antigen binding (discussed further below). A whole antibody typically consists of four polypeptides: two identical copies of a heavy (H) chain polypeptide and two identical copies of a light (L) chain polypeptide. Each of the heavy chains contains one N-terminal variable (VH) region and three C-terminal constant (CH1, CH2, and CH3) regions, and each light chain contains one N-terminal variable (VL) region and one C-terminal constant (CL) region. The light chains of antibodies can be assigned to one of two distinct types, either kappa (^) or lambda (^), based upon the amino acid sequences of their constant domains. In a typical antibody, each light chain is linked to a heavy chain by disulfide bonds, and the two heavy chains are linked to each other by disulfide bonds. The light chain variable region is aligned with the variable region of the heavy chain, and the light chain constant region is aligned with the first constant region of the heavy chain. The remaining constant regions of the heavy chains are aligned with each other. The variable regions of each pair of light and heavy chains form the antigen binding site of an antibody. The VH and VL regions have the same general structure, with each region comprising four framework (FW or FR) regions. The term “framework region,” as used herein, refers to the relatively conserved amino acid sequences within the variable region which are located between the CDRs. There are four framework regions in each variable domain, which are designated FR1, FR2, FR3, and FR4. The framework regions form the ȕ sheets that provide the structural framework of the variable region (see, e.g., C. A. Janeway et al. (eds.), Immunobiology, 5th Ed., Garland Publishing, New York, N.Y. (2001)). The framework regions are connected by three CDRs. As discussed above, the three CDRs, known as CDR1, CDR2, and CDR3, form the “hypervariable region” of an antibody, which is responsible for antigen binding. The CDRs form loops connecting, and in some cases comprising part of, the beta-sheet structure formed by the framework regions. While the constant regions of the light and heavy chains are not directly involved in binding of the antibody to an antigen, the constant regions can influence the orientation of the variable regions. The constant regions also exhibit various effector functions, such as participation in antibody-dependent complement-mediated lysis or antibody-dependent cellular toxicity via interactions with effector molecules and cells. [0056] The terms “fragment of an antibody,” “antibody fragment,” and “antigen-binding fragment” of an antibody are used interchangeably herein to refer to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (see, generally, Holliger et al., Nat. Biotech., 23(9): 1126-1129 (2005)). Any antigen-binding fragment of the antibody described herein is within the scope of the invention. The antibody fragment desirably comprises, for example, one or more CDRs, the variable region (or portions thereof), the constant region (or portions thereof), or combinations thereof. Examples of antibody fragments include, but are not limited to, (i) a Fab fragment, which is a monovalent fragment consisting of the VL, VH, CL, and CH1 domains, (ii) a F(ab’)2 fragment, which is a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region, (iii) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (iv) a Fab’ fragment, which results from breaking the disulfide bridge of an F(ab’)2 fragment using mild reducing conditions, (v) a disulfide-stabilized Fv fragment (dsFv), and (vi) a domain antibody (dAb), which is an antibody single variable region domain (VH or VL) polypeptide that specifically binds antigen. [0057] A “nanobody,” as used herein, refers to polypeptides comprising the variable region of a heavy chain of an antibody. A nanobody is functionally similar to a single domain antibody with only one heavy chain variable region. In general, the antigen-binding properties of a nanobody can be described by three variable regions (CDRs) divided by four framework regions (FRs) with the general structure as shown below: FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4 in which FR1 to FR4 refer to framework regions 1 to 4, respectively, and in which CDR1 to CDR3 refer to the complementarity determining regions 1 to 3. The amino acid sequences of four FRs are relatively conservative and do not directly participate in binding reactions. The CDRs normally form a loop structure in which the ȕ-sheets formed by the FRs therebetween are spatially close to each other, constituting the antigen-binding site of the nanobody. The amino acid sequences of the same type of nanobodies can be compared to determine which amino acids constitute the FR or CDR regions. The present invention includes not only intact nanobodies but also fragment(s) of immunologically active nanobody or fusion protein(s) formed from nanobodies with other sequences. Therefore, the present invention also includes fragments, derivatives, and analogs of the nanobodies. [0058] As used herein, a “nucleic acid” or a “nucleic acid sequence” refers to a polymer or oligomer of pyrimidine and/or purine bases, preferably cytosine, thymine, and uracil, and adenine and guanine, respectively (See Albert L. Lehninger, Principles of Biochemistry, at 793- 800 (Worth Pub. 1982)). The present technology contemplates any deoxyribonucleotide, ribonucleotide, or peptide nucleic acid component, and any chemical variants thereof, such as methylated, hydroxymethylated, or glycosylated forms of these bases, and the like. The polymers or oligomers may be heterogenous or homogenous in composition and may be isolated from naturally occurring sources or may be artificially or synthetically produced. In addition, the nucleic acids may be DNA or RNA, or a mixture thereof, and may exist permanently or transitionally in single-stranded or double-stranded form, including homoduplex, heteroduplex, and hybrid states. In some embodiments, a nucleic acid or nucleic acid sequence comprises other kinds of nucleic acid structures such as, for instance, a DNA/RNA helix, peptide nucleic acid (PNA), morpholino nucleic acid (see, e.g., Braasch and Corey, Biochemistry, 41(14): 4503-4510 (2002)) and U.S. Pat. No.5,034,506), locked nucleic acid (LNA; see Wahlestedt et al., Proc. Natl. Acad. Sci. U.S.A., 97: 5633-5638 (2000)), cyclohexenyl nucleic acids (see Wang, J. Am. Chem. Soc., 122: 8595-8602 (2000)), and/or a ribozyme. Hence, the term “nucleic acid” or “nucleic acid sequence” may also encompass a chain comprising non-natural nucleotides, modified nucleotides, and/or non- nucleotide building blocks that can exhibit the same function as natural nucleotides (e.g., “nucleotide analogs”); further, the term “nucleic acid sequence” as used herein refers to an oligonucleotide, nucleotide or polynucleotide, and fragments or portions thereof, and to DNA or RNA of genomic or synthetic origin, which may be single or double- stranded, and represent the sense or antisense strand. The terms “nucleic acid,” “polynucleotide,” “nucleotide sequence,” and “oligonucleotide” are used interchangeably. They refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof. [0059] A “peptide” or “polypeptide” is a linked sequence of two or more amino acids linked by peptide bonds. The peptide or polypeptide can be natural, synthetic, or a modification or combination of natural and synthetic. Polypeptides include proteins such as binding proteins, receptors, and antibodies. The proteins may be modified by the addition of sugars, lipids or other moieties not included in the amino acid chain. The terms “polypeptide” and “protein,” are used interchangeably herein. [0060] As used herein, the term “percent sequence identity” refers to the percentage of nucleotides or nucleotide analogs in a nucleic acid sequence, or amino acids in an amino acid sequence, that is identical with the corresponding nucleotides or amino acids in a reference sequence after aligning the two sequences and introducing gaps, if necessary, to achieve the maximum percent identity. Hence, in case a nucleic acid according to the technology is longer than a reference sequence, additional nucleotides in the nucleic acid, that do not align with the reference sequence, are not taken into account for determining sequence identity. A number of mathematical algorithms for obtaining the optimal alignment and calculating identity between two or more sequences are known and incorporated into a number of available software programs. Examples of such programs include CLUSTAL-W, T-Coffee, and ALIGN (for alignment of nucleic acid and amino acid sequences), BLAST programs (e.g., BLAST 2.1, BL2SEQ, and later versions thereof) and FASTA programs (e.g., FASTA3x, FAS™, and SSEARCH) (for sequence alignment and sequence similarity searches). Sequence alignment algorithms also are disclosed in, for example, Altschul et al., J. Molecular Biol., 215(3): 403-410 (1990), Beigert et al., Proc. Natl. Acad. Sci. USA, 106(10): 3770-3775 (2009), Durbin et al., eds., Biological Sequence Analysis: Probabilistic Models of Proteins and Nucleic Acids, Cambridge University Press, Cambridge, UK (2009), Soding, Bioinformatics, 21(7): 951-960 (2005), Altschul et al., Nucleic Acids Res., 25(17): 3389-3402 (1997), and Gusfield, Algorithms on Strings, Trees and Sequences, Cambridge University Press, Cambridge UK (1997)). [0061] A “vector” or “expression vector” is a replicon, such as plasmid, phage, virus, or cosmid, to which another DNA segment may be attached or incorporated so as to bring about the replication of the attached segment in a cell. 2. Method for identifying and selecting nanobodies [0062] Disclosed herein are methods for identifying and selecting nanobodies. The methods can be utilized to identify nanobodies against different endogenous protein targets including but not limited to: transcription or translation factors, chromatin regulators, or any protein that modulates cellular processes for which reporters are easily available, including but not limited to: DNA repair, mRNA or protein stability, and immune responses. [0063] The methods may comprise any or all of: preparing a yeast nanobody library comprising yeast cells each displaying a single nanobody on its surface; mixing the yeast nanobody library with a protein target wherein the protein target is configured to bind to a solid surface; separating yeast cells bound to the protein target from unbound yeast cells; amplifying nanobody DNA sequences from the yeast cells bound to protein target; cloning the nanobody sequences into a human lentiviral vector to prepare a nanobody lentiviral library; and identifying the nanobody as a binding protein for the protein target based on a functional output. [0064] In some embodiments, the methods are directed to identifying and selecting nanobodies for a target chromatin regulator. In some embodiments, the chromatin regulator comprises a methylase, a demethylase, an acetylase, or ATP-dependent chromatin remodeling complex. [0065] In some embodiments, the methods comprise: preparing a yeast nanobody library comprising yeast cells each displaying a single nanobody on its surface; mixing the yeast nanobody library with a target chromatin regulator wherein the target chromatin regulator is configured to bind to a surface (e.g., microparticle, plate, membrane); separating yeast cells bound to target chromatin regulator from unbound yeast cells; amplifying nanobody DNA sequences from the yeast cells bound to target chromatin regulator; cloning the nanobody sequences into a human lentiviral vector to prepare a nanobody lentiviral library, wherein each nanobody DNA sequence is expressed as a fusion protein with a DNA binding domain; transforming the nanobody lentiviral library into reporter cells, wherein a reporter cell comprises a two-part reporter gene comprising a surface marker and a fluorescent protein, wherein the two- part reporter gene is capable of being silenced or induced by the target chromatin regulator; separating the reporter cells based on presence or absence of the surface marker, the fluorescent protein, or a combination thereof; and identifying the nanobody as a binding protein for the target chromatin regulator and capable of regulating gene expression. [0066] In some embodiments, the methods further comprise repeating the preparing a yeast nanobody library, mixing the yeast nanobody library with a target chromatin regulator, separating yeast cells bound to target chromatin regulator from unbound yeast cells, and amplifying nanobody DNA sequences from the yeast cells bound to target chromatin regulator one or more times. [0067] In some embodiments, the DNA binding domain is an inducible DNA binding domain. The inducible DNA binding domain may use any system for induction of DNA binding, including, but not limited to, tetracycline Tet,/DOX inducible systems, light inducible systems, Abscisic acid (ABA) inducible systems, cumate systems, 40HT/estrogen inducible systems, ecdysone-based inducible systems, and FKBP12/FRAP (FKBP12-rapamycin complex) inducible systems. [0068] In such instances in which an inducible DNA binding domain is used, the methods may further comprise treating the reporter cells with an agent configured to induce the inducible DNA binding domain for a length of time. [0069] In some embodiments, the reporter cells are treated with the agent at least 24 hours. For, example the reporter cells may be treated with the agent for at least 24 hours, at least 36 hours, at least 48 hours (2 days), at least 3 days, at least 4 days, at least 5 days, at least 6 days, at least 7 days, at least 8 days, at least 9 days, at least 10 days, at least 14 days, or more. In some embodiments, the reporter cells at treated with the agent for 3-12 days, 3-10 days, 3-7 days, or 3- 5 days. [0070] In some embodiments, the inducible DNA binding domain comprises a tag. The tag may include any tag known in the art, including tags removable by chemical or enzymatic means. Suitable tags for use in the present method include chitin binding protein (CBP), maltose binding protein (MBP), Strep-tag, glutathione-S-transferase (GST), a polyhistidine (PolyHis) tag, an ALFA-tag, a V5-tag, a Myc-tag, a hemagglutinin(HA)-tag, a Spot-tag, a T7-tag, an NE-tag, a Calmodulin-tag, a polyglutamate tag, a polyarginine tag, a FLAG tag, and the like. [0071] The methods comprise transforming reporter cells with the domain library, wherein the reporter cell comprises a two-part reporter gene comprising a surface marker and a fluorescent protein under the control of a promoter. The two-part reporter gene is capable of being modulated by the target chromatin regulator following treatment with an agent configured to induce the inducible DNA binding domain and allowing recruitment of the chromatin regulator by the putative nanobody. [0072] The promoter may confer a high rate of transcription (a strong promoter) or confer a low rate of transcription (weak promoter). Many promoter libraries have been established experimentally and choice of promoter and promoter strength is dependent on cell type. In some embodiments, when identifying nanobodies configured to bind chromatin activators, a weak promoter may be used. In some embodiments, when identifying nanobodies configured to bind chromatin repressors, a strong promoter may be used. [0073] Cell surface markers include proteins and carbohydrates which are attached to the cellular membrane. Cell surface markers are generally known in the art for a variety of cell types and can be expressed in a reporter cell of choice based on known molecular biology methods. The surface marker may be a synthetic surface marker comprising marker polypeptide attached to a transmembrane domain. For example, the marker polypeptide may include an antibody or a fragment thereof (e.g., Fc region) attached to a transmembrane domain. In some embodiments, the marker polypeptide is human IgG1 Fc region and the synthetic surface marker comprises human IgG1 Fc region attached to a transmembrane domain. [0074] Fluorescent proteins are well known in the art and include proteins adapted to fluoresce in various cellular compartments and as a result of varying wavelengths of incoming light. Examples of fluorescent proteins include: phycobiliproteins, cyan fluorescent protein (CFP), green fluorescent protein (GFP), yellow fluorescent protein (YFP or citrine), enhanced orange fluorescent protein (OFP), enhanced green fluorescent protein (eGFP), modified green fluorescent protein (emGFP), enhanced yellow fluorescent protein (eYFP) and/or monomeric red fluorescent protein (mRFP) and derivatives and variants thereof. [0075] The methods comprise separating reporter cells based on presence or absence of the surface marker, the fluorescent protein, or a combination thereof. A number of cell separation techniques are known in the art are suitable for use with the methods disclosed herein, including, for example, immunomagnetic cell separation, fluorescent-activated cell sorting (FACS), and microfluidic cell sorting. In some embodiments, cell separation comprises immunomagnetic cell separation. [0076] The chromatin regulator is identified as a functional chromatin repressor (e.g., able to bind to the nanobody and modulate transcription and/or epigenetic memory) when recruited by the nanobody due to enrichment of cells with an absence of the surface marker, the fluorescent protein, or a combination thereof. [0077] The chromatin regulator is identified as a functional chromatin activator when recruited by the nanobody due to enrichment of cells with a presence of the surface marker, the fluorescent protein, or a combination thereof. 3. Fusion Proteins [0078] The present disclosure also provides fusion proteins comprising a DNA binding protein, DNA binding domain, or a functional fragment thereof covalently linked to a nanobody configured to bind a chromatin regulator. [0079] In some embodiments, the fusion protein comprises two nanobodies configured to bind the same or different chromatin regulator. The two nanobodies may be linked by a covalent linker. [0080] As used herein, the term “chromatin regulator” refers to a protein or polypeptide that interacts with, directly or indirectly, specific DNA sequences to modify histones, DNA, or histone-DNA complexes (e.g., through methylation, acetylation, phosphorylation, adenosine diphosphate–ribosylation, glycosylation, sumoylation, or ubiquitylation or remodel DNA-histone structure with energy from ATP hydrolysis). In some embodiments, the chromatin regulator comprises a methylase, a demethylase, an acetylase, or ATP-dependent chromatin remodeling complex. [0081] In some embodiments, one or more of the at least one nanobody is identified and/or selected by the methods described herein. [0082] In some embodiments, the nanobody is configured to bind DNA (cytosine-5)- methyltransferase 3A (DNMT3A) complexes. The nanobody configured to bind DNMT3A complexes may comprise, consist essentially of, or consist of any of the amino acid sequences shown in FIG. 10D, or fragments thereof. [0083] In some embodiments, the DNMT3A complex nanobody comprises at least one CDR comprising an amino acid sequence as shown in the consensus sequences in FIG. 10D. For example, the at least one CDR may be selected from the group consisting of GTIFXXXXM (SEQ ID NO: 13), EZVAXIXXGXXTNY (SEQ ID NO: 14), and AAXXXXXYYXXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid. In some embodiments, the DNMT3A complex nanobody comprises a first CDR comprising an amino acid sequence of GTIFXXXXM (SEQ ID NO: 13), a second CDR comprising an amino acid sequence of EZVAXIXXGXXTNY (SEQ ID NO: 14), and a third CDR comprising an amino acid sequence of AAXXXXXYYXXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid. [0084] In some embodiments, the DNMT3A complex nanobody comprises at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), GNIFDGASM (SEQ ID NO: 4), EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), AAGRYYYPGHGY (SEQ ID NO: 8), AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 12). [0085] In some embodiments, the DNMT3A complex nanobody comprises a first CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), and GNIFDGASM (SEQ ID NO: 4); a second CDR comprising an amino acid sequence selected from the group consisting of EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), and AAGRYYYPGHGY (SEQ ID NO: 8); a third CDR comprising an amino acid sequence selected from the group consisting of AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 12); or a combination thereof. [0086] In some embodiments, the DNMT3A complex nanobody comprises, consists essentially of, or consists of an amino acid sequence at least 70% (e.g., at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98% or at least 99%) similar to any of SEQ ID NOs: 16-20. In some embodiments, the DNMT3A complex nanobody comprises, consists essentially of, or consists of an amino acid sequence at least 70% similar to any of SEQ ID NO: 5932. [0087] In some embodiments, the nanobody is configured to bind ten-eleven translocation methylcytosine 1/2/3 (TET1/2/3) complexes. The nanobody configured to bind TET1/2/3 complexes may comprise, consist essentially of, or consist of any of the amino acid sequences of SEQ ID NOs: 5960-5968. [0088] In some embodiments, the nanobody configured to bind TET1/2/3 complexes comprises at least one CDR may be selected from the group consisting of GXIZ1XXXXM, EZ2VAXIXXGXXTZ3Y (SEQ ID NO: 5978), and AZ4XXXXXYXXXXXY, wherein Z1 is S or F, Z2 is F or L, Z3 is N or Y, Z4 is A or V, and each X is any amino acid. In some embodiments, the nanobody configured to bind TET1/2/3 complexes comprises a first CDR comprising an amino acid sequence of GXIZ1XXXXM, a second CDR comprising an amino acid sequence of EZ2VAXIXXGXXTZ3Y (SEQ ID NO: 5978), and a third CDR comprising an amino acid sequence of AZ4XXXXXYXXXXXY, wherein Z1 is S or F, Z2 is F or L, Z3 is N or Y, Z4 is A or V, and each X is any amino acid. [0089] In some embodiments, the TET1/2/3 complex nanobody comprises at least one CDR having an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933-5959. In some embodiments, the TET1/2/3 complex nanobody comprises a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933-5941; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5942-5950; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5951-5959; or a combination thereof. In some embodiments, the TET1/2/3 complex nanobody comprises a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5935 or 5936; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5944 or 5945; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5953- 5954; or a combination thereof. [0090] In some embodiments, the TET1/2/3 complex nanobody comprises, consists essentially of, or consists of an amino acid sequence at least 70% (e.g., at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98% or at least 99%) similar to any of SEQ ID NOs: 5960-5968. [0091] In some embodiments, the fusion protein further comprises a transcription factor or regulator (e.g., a transcriptional repressor, a transcriptional activator). In some embodiments, the transcription factor comprises Krüppel-associated box (KRAB). The nanobody and the transcription factor or regulator may be linked by a covalent linker. [0092] Also provided is a DNA (cytosine-5)-methyltransferase 3A (DNMT3A) binding protein. The term “DNMT3A binding protein” encompasses proteins which bind DNMT3A and/or complexes comprising DNMT3A. DNMT3A forms a catalytically active dimer in a DNMT3A:DNMT3L complex. In addition, DNMT3A is capable of further homo- and hetero- oligomerization with DNMT3B. DNMT3A forms complexes with proteins outside of the DNMT3 family. For example, a direct interaction between EZH2 of the Polycomb-repressive complex 2 (PRC2) and DNMT3A has been demonstrated. [0093] Further provided are fusion proteins of the DNMT3A binding protein covalently linked to a DNA binding protein, DNA binding domain, or a functional fragment thereof. In some embodiments, the DNMT3A binding protein is an antibody or a fragment thereof. In select embodiments, the DNMT3A binding protein is a nanobody. [0094] The DNMT3A binding protein may comprise, consist essentially of, or consist of any of the amino acid sequences shown in FIG. 10D, or fragments thereof. In some embodiments, the DNMT3A binding protein comprises at least one CDR comprising an amino acid sequence as shown in the consensus sequences in FIG. 10D. For example, the at least one CDR may be selected from the group consisting of GTIFXXXXM (SEQ ID NO: 13), EZVAXIXXGXXTNY (SEQ ID NO: 14), and AAXXXXXYYXXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid. In some embodiments, the DNMT3A binding protein comprises the at least one nanobody comprises a first CDR comprising an amino acid sequence of GTIFXXXXM (SEQ ID NO: 13), a second CDR comprising an amino acid sequence of EZVAXIXXGXXTNY (SEQ ID NO: 14), and a third CDR comprising an amino acid sequence of AAXXXXXYYXXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid. [0095] In some embodiments, the DNMT3A binding protein comprises at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), GNIFDGASM (SEQ ID NO: 4), EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), AAGRYYYPGHGY (SEQ ID NO: 8), AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 12). [0096] In some embodiments, the DNMT3A binding protein comprises a first CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), and GNIFDGASM (SEQ ID NO: 4); a second CDR comprising an amino acid sequence selected from the group consisting of EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), and AAGRYYYPGHGY (SEQ ID NO: 8); a third CDR comprising an amino acid sequence selected from the group consisting of AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 12); or a combination thereof. [0097] In some embodiments, the DNMT3A binding protein comprises, consists essentially of, or consists of an amino acid sequence at least 70% (e.g., at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98% or at least 99%) similar to any of SEQ ID NOs: 16-20. In some embodiments, the DNMT3A binding protein comprises, consists essentially of, or consists of an amino acid sequence at least 70% similar to any of SEQ ID NO: 5932. [0098] Also provided is a ten-eleven translocation methylcytosine 1/2/3 (TET1/2/3) complex binding protein. Further provided are fusion proteins of the TET1/2/3 complex binding protein covalently linked to a DNA binding protein, DNA binding domain, or a functional fragment thereof. In some embodiments, the TET1/2/3 complex binding protein is an antibody or a fragment thereof. In select embodiments, the TET1/2/3 complex binding protein is a nanobody. [0099] In some embodiments, the nanobody is configured to bind ten-eleven translocation methylcytosine 1/2/3 (TET1/2/3) complexes. The TET1/2/3 complex binding protein may comprise, consist essentially of, or consist of any of the amino acid sequences of SEQ ID NOs: 5960-5968. [00100] In some embodiments, the TET1/2/3 complex binding protein comprises at least one CDR may be selected from the group consisting of GXIZ1XXXXM, EZ2VAXIXXGXXTZ3Y (SEQ ID NO: 5978), and AZ4XXXXXYXXXXXY, wherein Z1 is S or F, Z2 is F or L, Z3 is N or Y, Z4 is A or V, and each X is any amino acid. In some embodiments, the TET1/2/3 complex binding protein comprises a first CDR comprising an amino acid sequence of GXIZ1XXXXM, a second CDR comprising an amino acid sequence of EZ2VAXIXXGXXTZ3Y (SEQ ID NO: 5978), and a third CDR comprising an amino acid sequence of AZ4XXXXXYXXXXXY, wherein Z1 is S or F, Z2 is F or L, Z3 is N or Y, Z4 is A or V, and each X is any amino acid. [00101] In some embodiments, the TET1/2/3 complex binding protein comprises at least one CDR having an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933- 5959. In some embodiments, the TET1/2/3 complex binding protein comprises a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933- 5941; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5942-5950; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5951-5959; or a combination thereof. In select embodiments, the TET1/2/3 complex binding protein comprises a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5935 or 5936; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5944 or 5945; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5953-5954; or a combination thereof. [00102] In some embodiments, the TET1/2/3 complex binding protein comprises, consists essentially of, or consists of an amino acid sequence at least 70% (e.g., at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 98% or at least 99%) similar to any of SEQ ID NOs: 5960-5968. [00103] The DNA binding protein, domain, or functional fragment thereof, is any polypeptide which is capable of binding double- or single-stranded DNA, generally or with sequence specificity. DNA binding proteins and domains include those polypeptides having helix-turn- helix motifs, zinc fingers, leucine zippers, HMG-box (high mobility group box) domains, winged helix region, winged helix-turn-helix region, helix-loop-helix region, immunoglobulin fold, B3 domain, Wor3 domain, TAL effector DNA-binding domain and the like. The DNA binding proteins or domains may be a natural binding domain. In some embodiments, the DNA binding domain comprises a programmable DNA binding proteins or domains, e.g., a DNA binding protein or domain engineered, for example by altering one or more amino acid of a natural DNA binding protein or domain to bind to a predetermined nucleotide sequence. [00104] In some embodiments, the DNA binding protein, domain, or functional fragment thereof, is capable of binding directly to the target DNA sequences. [00105] The DNA binding protein, domain, or functional fragment thereof, may be derived from domains found in naturally occurring Transcription activator-like effectors (TALEs), such as AvrBs3, Hax2, Hax3 or Hax4 (Bonas et al.1989. Mol Gen Genet 218(1): 127-36; Kay et al. 2005 Mol Plant Microbe Interact 18(8): 838-48). TALEs have a modular DNA-binding domain consisting of repetitive sequences of residues; each repeat region consists of 34 amino acids. A pair of residues at the 12th and 13th position of each repeat region determines the nucleotide specificity and combining of the regions allows synthesis of sequence-specific TALE DNA- binding domains. In some embodiments, the TALE DNA binding domains may be engineered using known methods to provide a DNA binding domain with chosen specificity for any target sequence. The DNA binding domain may comprise multiple (e.g., 2, 3, 4, 5, 6, 10, 20, or more) Tal effector DNA-binding motifs. In particular, any number of nucleotide-specific Tal effector motifs can be combined to form a sequence-specific DNA-binding domain to be employed in the present transcription factor. [00106] In some embodiments, the DNA binding protein, domain, or functional fragment thereof, associates with the target DNA in concert with an exogenous factor. [00107] In some embodiments, the DNA binding protein, domain, or functional fragment thereof, is an inducible DNA binding protein, domain, or functional fragment thereof, as described and exemplified elsewhere herein. [00108] In some embodiments, the DNA binding protein, domain, or functional fragment thereof, is derived from a Clustered Regularly Interspaced Short Palindromic Repeats associated (Cas) protein (e.g., catalytically dead Cas9) and associates with the target DNA through a guide RNA. The gRNA itself comprises a sequence complementary to one strand of the DNA target sequence and a scaffold sequence which binds and recruits Cas9 to the target DNA sequence. The transcription factors described herein may be useful for CRISPR interference (CRISPRi) or CRISPR activation (CRISPRa). [00109] The guide RNA (gRNA) may be a crRNA, crRNA/tracrRNA (or single guide RNA, sgRNA). The gRNA may be a non-naturally occurring gRNA. The terms “gRNA,” “guide RNA” and “guide sequence” may be used interchangeably throughout and refer to a nucleic acid comprising a sequence that determines the binding specificity of the Cas protein. A gRNA hybridizes to (complementary to, partially or completely) the DNA target sequence. [00110] The gRNA or portion thereof that hybridizes to the target nucleic acid (a target site) may be any length necessary for selective hybridization. gRNAs or sgRNA(s) can be between about 5 and about 100 nucleotides long, or longer (e.g., 5, 6, 7, 8, 9, 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, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 5960, 61, 62, 63, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 9192, 93, 94, 95, 96, 97, 98, 99, or 100 nucleotides in length, or longer). [00111] To facilitate gRNA design, many computational tools have been developed (See Prykhozhij et al. (PLoS ONE, 10(3): (2015)); Zhu et al. (PLoS ONE, 9(9) (2014)); Xiao et al. (Bioinformatics. Jan 21 (2014)); Heigwer et al. (Nat Methods, 11(2): 122–123 (2014)). Methods and tools for guide RNA design are discussed by Zhu (Frontiers in Biology, 10 (4) pp 289-296 (2015)), which is incorporated by reference herein. Additionally, there are many publicly available software tools that can be used to facilitate the design of sgRNA(s); including but not limited to, Genscript Interactive CRISPR gRNA Design Tool, WU-CRISPR, and Broad Institute GPP sgRNA Designer. There are also publicly available pre-designed gRNA sequences to target many genes and locations within the genomes of many species (human, mouse, rat, zebrafish, C. elegans), including but not limited to, IDT DNA Predesigned Alt-R CRISPR-Cas9 guide RNAs, Addgene Validated gRNA Target Sequences, and GenScript Genome-wide gRNA databases. [00112] The present disclosure also provides nucleic acids encoding a nanobody fusion protein, DNMT3A binding protein, DNMT3A binding protein fusion protein, TET1/2/3 complex binding protein, and TET1/2/3 complex binding protein fusion protein as disclosed herein. [00113] For example, the at least one CDR of the nanobody or DNMT3A binding protein may be encoded by a nucleic acid sequence of any of those disclosed in Table 2 (SEQ ID NOs: 21- 92), Table 3, or SEQ ID NOs: 93-5928. In some embodiments, the nanobody, nanobody fusion protein, or DNMT3A binding protein or fusion thereof comprises a nucleic acid sequence at least 70% similar to any of SEQ ID NOs: 21-5928. [00114] In some embodiments, the nanobody or DNMT3A binding protein may comprise a combination of two or three CDRs, each individually encoded by a nucleic acid sequence having at least 70% similar to any of SEQ ID NOs: 21-5928. Thus, a single nanobody or DNMT3A binding protein may be encoded by a nucleic acid sequence comprising one, two, or three, individual sequences having at least 70% similar to any of SEQ ID NOs: 21-5928. See for example, those nanobodies listed in Table 3 here or in Table 3 of U.S. Provisional Application No. 63/242,898, incorporated by reference in its entirety. [00115] In some embodiments, the nanobody, nanobody fusion protein, or TET1/2/3 complex binding protein or fusion thereof comprises a nucleic acid sequence at least 70% similar to any of SEQ ID NOs: 5969-5977, or fragments thereof. [00116] Nucleic acids of the present disclosure can comprise any of a number of promoters known to the art, wherein the promoter is constitutive, regulatable or inducible, cell type specific, tissue-specific, or species specific. In addition to the sequence sufficient to direct transcription, a promoter sequence of the invention can also include sequences of other regulatory elements that are involved in modulating transcription (e.g., enhancers, Kozak sequences and introns). Many promoter/regulatory sequences useful for driving constitutive expression of a gene are available in the art and include, but are not limited to, for example, CMV (cytomegalovirus promoter), EF1a (human elongation factor 1 alpha promoter), SV40 (simian vacuolating virus 40 promoter), PGK (mammalian phosphoglycerate kinase promoter), Ubc (human ubiquitin C promoter), human beta-actin promoter, rodent beta-actin promoter, CBh (chicken beta-actin promoter), CAG (hybrid promoter contains CMV enhancer, chicken beta actin promoter, and rabbit beta- globin splice acceptor), TRE (Tetracycline response element promoter), H1 (human polymerase III RNA promoter), U6 (human U6 small nuclear promoter), and the like. Additional promoters that can be used for expression of the components of the present system, include, without limitation, cytomegalovirus (CMV) intermediate early promoter, a viral LTR such as the Rous sarcoma virus LTR, HIV-LTR, HTLV-1 LTR, Maloney murine leukemia virus (MMLV) LTR, myeoloproliferative sarcoma virus (MPSV) LTR, spleen focus-forming virus (SFFV) LTR, the simian virus 40 (SV40) early promoter, herpes simplex tk virus promoter, elongation factor 1- alpha (EF1-Į) promoter with or without the EF1-Į intron. Additional promoters include any constitutively active promoter. Alternatively, any regulatable promoter may be used, such that its expression can be modulated within a cell. [00117] Moreover, inducible expression can be accomplished by placing the nucleic acid encoding such a molecule under the control of an inducible promoter/regulatory sequence. Promoters that are well known in the art can be induced in response to inducing agents such as metals, glucocorticoids, tetracycline, hormones, and the like, are also contemplated for use with the invention. Thus, it will be appreciated that the present disclosure includes the use of any promoter/regulatory sequence known in the art that is capable of driving expression of the desired protein operably linked thereto. [00118] The present disclosure also provides for vectors containing the nucleic acids and cells containing the nucleic acids or vectors, thereof. The vectors may be used to propagate the nucleic acid in an appropriate cell and/or to allow expression from the nucleic acid (e.g., an expression vector). The person of ordinary skill in the art would be aware of the various vectors available for propagation and expression of a nucleic acid sequence. [00119] To construct cells that express the present nanobodies or fusion proteins thereof, expression vectors for stable or transient expression may be constructed via conventional methods and introduced into cells. For example, nucleic acids encoding the nanobodies or fusion proteins thereof, or other nucleic acids or proteins, may be cloned into a suitable expression vector, such as a plasmid or a viral vector in operable linkage to a suitable promoter. The selection of expression vectors/plasmids/viral vectors should be suitable for integration and replication in eukaryotic cells. [00120] In certain embodiments, vectors of the present disclosure can drive the expression of one or more sequences in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed, Nature (1987) 329:840, incorporated herein by reference) and pMT2PC (Kaufman, et al., EMBO J. (1987) 6:187, incorporated herein by reference). When used in mammalian cells, the expression vector's control functions are typically provided by one or more regulatory elements. For example, commonly used promoters are derived from polyoma, adenovirus 2, cytomegalovirus, simian virus 40, and others disclosed herein and known in the art. For other suitable expression systems for both prokaryotic and eukaryotic cells see, e.g., Chapters 16 and 17 of Sambrook, et al., MOLECULAR CLONING: A LABORATORY MANUAL. 2nd eds., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989, incorporated herein by reference. [00121] The vectors of the present disclosure may direct the expression of the nucleic acid in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid). Such regulatory elements include promoters that may be tissue specific or cell specific. The term “tissue specific” as it applies to a promoter refers to a promoter that is capable of directing selective expression of a nucleotide sequence of interest to a specific type of tissue (e.g., seeds) in the relative absence of expression of the same nucleotide sequence of interest in a different type of tissue. The term “cell type specific” as applied to a promoter refers to a promoter that is capable of directing selective expression of a nucleotide sequence of interest in a specific type of cell in the relative absence of expression of the same nucleotide sequence of interest in a different type of cell within the same tissue. The term “cell type specific” when applied to a promoter also means a promoter capable of promoting selective expression of a nucleotide sequence of interest in a region within a single tissue. Cell type specificity of a promoter may be assessed using methods well known in the art, e.g., immunohistochemical staining. [00122] Additionally, the vector may contain, for example, some or all of the following: a selectable marker gene for selection of stable or transient transfectants in host cells; transcription termination and RNA processing signals; 5’-and 3’-untranslated regions; internal ribosome binding sites (IRESes), versatile multiple cloning sites; and reporter gene for assessing expression of the chimeric receptor. Suitable vectors and methods for producing vectors containing transgenes are well known and available in the art. Selectable markers include chloramphenicol resistance, tetracycline resistance, spectinomycin resistance, neomycin resistance, puromycin resistance, streptomycin resistance, erythromycin resistance, rifampicin resistance, bleomycin resistance, thermally adapted kanamycin resistance, gentamycin resistance, hygromycin resistance, trimethoprim resistance, dihydrofolate reductase (DHFR), GPT; the URA3, HIS4, LEU2, and TRP1 genes of S. cerevisiae. [00123] When introduced into a cell, the vectors may be maintained as an autonomously replicating sequence or extrachromosomal element or may be integrated into host DNA. [00124] Thus, the disclosure further provides for cells comprising a nanobody or a fusion protein thereof, a nucleic acid, or a vector, as disclosed herein. [00125] Conventional viral and non-viral based gene transfer methods can be used to introduce the nucleic acids into cells, tissues, or a subject. Such methods can be used to administer the nucleic acids to cells in culture, or in a host organism. Non-viral vector delivery systems include DNA plasmids, cosmids, RNA (e.g., a transcript of a vector described herein), a nucleic acid, and a nucleic acid complexed with a delivery vehicle. [00126] Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after delivery to the cell. A variety of viral constructs may be used to deliver the present nucleic acids to the cells, tissues and/or a subject. Viral vectors include, for example, retroviral, lentiviral, adenoviral, adeno-associated and herpes simplex viral vectors. Nonlimiting examples of such recombinant viruses include recombinant adeno- associated virus (AAV), recombinant adenoviruses, recombinant lentiviruses, recombinant retroviruses, recombinant herpes simplex viruses, recombinant poxviruses, phages, etc. The present disclosure provides vectors capable of integration in the host genome, such as retrovirus or lentivirus. See, e.g., Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York, 1989; Kay, M. A., et al., 2001 Nat. Medic. 7(1):33-40; and Walther W. and Stein U., 2000 Drugs, 60(2): 249-71, incorporated herein by reference. [00127] The nucleic acids, nanobodies or fusion proteins thereof may be delivered by any suitable means. In certain embodiments, the nucleic acids or proteins thereof are delivered in vivo. In other embodiments, the nucleic acids or proteins thereof are delivered to isolated/cultured cells in vitro or ex vivo to provide modified cells useful for in vivo delivery to patients afflicted with a disease or condition. [00128] Vectors according to the present disclosure can be transformed, transfected, or otherwise introduced into a wide variety of host cells. Transfection refers to the taking up of a vector by a cell whether or not any coding sequences are in fact expressed. Numerous methods of transfection are known to the ordinarily skilled artisan, for example, lipofectamine, calcium phosphate co-precipitation, electroporation, DEAE-dextran treatment, microinjection, viral infection, and other methods known in the art. Transduction refers to entry of a virus into the cell and expression (e.g., transcription and/or translation) of sequences delivered by the viral vector genome. In the case of a recombinant vector, “transduction” generally refers to entry of the recombinant viral vector into the cell and expression of a nucleic acid of interest delivered by the vector genome. [00129] Methods of delivering vectors to cells are well known in the art and may include DNA or RNA electroporation, transfection reagents such as liposomes or nanoparticles to delivery DNA or RNA; delivery of DNA, RNA, or protein by mechanical deformation (see, e.g., Sharei et al. Proc. Natl. Acad. Sci. USA (2013) 110(6): 2082-2087, incorporated herein by reference); or viral transduction. In some embodiments, the vectors are delivered to host cells by viral transduction. Nucleic acids can be delivered as part of a larger construct, such as a plasmid or viral vector, or directly, e.g., by electroporation, lipid vesicles, viral transporters, microinjection, and biolistics (high-speed particle bombardment). [00130] Additionally, delivery vehicles such as nanoparticle- and lipid-based delivery systems can be used. Further examples of delivery vehicles include lentiviral vectors, ribonucleoprotein (RNP) complexes, lipid-based delivery system, gene gun, hydrodynamic, electroporation or nucleofection microinjection, and biolistics. Various gene delivery methods are discussed in detail by Nayerossadat et al. (Adv Biomed Res. 2012; 1: 27) and Ibraheem et al. (Int J Pharm. 2014 Jan 1;459(1-2):70-83), incorporated herein by reference. [00131] As such, the disclosure provides an isolated cell comprising the vector(s) or nucleic acid(s) disclosed herein. Preferred cells are those that can be easily and reliably grown, have reasonably fast growth rates, have well characterized expression systems, and can be transformed or transfected easily and efficiently. Examples of suitable prokaryotic cells include, but are not limited to, cells from the genera Bacillus (such as Bacillus subtilis and Bacillus brevis), Escherichia (such as E. coli), Pseudomonas, Streptomyces, Salmonella, and Envinia. Suitable eukaryotic cells are known in the art and include, for example, yeast cells, insect cells, and mammalian cells. Examples of suitable yeast cells include those from the genera Kluyveromyces, Pichia, Rhino-sporidium, Saccharomyces, and Schizosaccharomyces. Exemplary insect cells include Sf-9 and HIS (Invitrogen, Carlsbad, Calif.) and are described in, for example, Kitts et al., Biotechniques, 14: 810-817 (1993); Lucklow, Curr. Opin. Biotechnol., 4: 564-572 (1993); and Lucklow et al., J. Virol., 67: 4566-4579 (1993), incorporated herein by reference. Desirably, the cell is a mammalian cell, and in some embodiments, the cell is a human cell. A number of suitable mammalian and human host cells are known in the art, and many are available from the American Type Culture Collection (ATCC, Manassas, Va.). Examples of suitable mammalian cells include, but are not limited to, Chinese hamster ovary cells (CHO) (ATCC No. CCL61), CHO DHFR-cells (Urlaub et al., Proc. Natl. Acad. Sci. USA, 97: 4216-4220 (1980)), human embryonic kidney (HEK) 293 or 293T cells (ATCC No. CRL1573), and 3T3 cells (ATCC No. CCL92). Other suitable mammalian cell lines are the monkey COS-1 (ATCC No. CRL1650) and COS-7 cell lines (ATCC No. CRL1651), as well as the CV-1 cell line (ATCC No. CCL70). Further exemplary mammalian host cells include primate, rodent, and human cell lines, including transformed cell lines. Normal diploid cells, cell strains derived from in vitro culture of primary tissue, as well as primary explants, are also suitable. Other suitable mammalian cell lines include, but are not limited to, mouse neuroblastoma N2A cells, HeLa, HEK, A549, HepG2, mouse L- 929 cells, and BHK or HaK hamster cell lines. [00132] Methods for selecting suitable mammalian cells and methods for transformation, culture, amplification, screening, and purification of cells are known in the art. [00133] The present invention is also directed to compositions or systems comprising a nanobody or a fusion protein thereof, a nucleic acid, a vector, or a cell, as described herein. In some embodiments, the compositions or system comprises two or more fusion proteins, nucleic acids, vectors, or cells, as described herein. [00134] In some embodiments, the composition or system further comprises a transcription factor or regulator (e.g., a transcriptional repressor, a transcriptional activator). In some embodiments, the transcription factor comprises Krüppel-associated box (KRAB). [00135] In some embodiments, the composition or system further comprises a chromatin regulator. [00136] In some embodiments, the composition or system further comprises a gRNA. The gRNA may be encoded on the same nucleic acid as the nanobody or fusion protein thereof or a different nucleic acid. In some embodiments, the vector encoding a nanobody or fusion protein thereof may further encode a gRNA, under the same or different promoter. In some embodiments, the gRNA is encoded on its own vector, separated from that of nanobody or fusion protein thereof. 4. Methods of Modifying Gene Expression and/or Epigenetic Memory [00137] The present disclosure also provides methods of modulating gene expression and/or epigenetic memory of at least one target gene. The methods comprise contacting the target nucleic acid with at least one fusion protein comprising a DNA binding protein, DNA binding domain, or a functional fragment thereof covalently linked to a nanobody configured to bind a chromatin regulator. In some embodiments, the gene expression and/or epigenetic memory of at least two genes is modulated. Descriptions provided elsewhere herein with regards to the fusion protein and components thereof are applicable to the present methods. [00138] Modulation of expression comprises increasing or decreasing gene expression compared to normal gene expression for the target gene. When the gene expression of at least two genes is modulation, both genes may have increased gene expression, both gene may have decreased gene expression, or one gene may have increased gene expression and the other may have decreased gene expression. [00139] The epigenetic memory of a cell is defined by the set of modifications to the cell’s DNA that does not necessarily alter the coding sequence but rather alters gene expression by the chemical modification (e.g., methylation, acetylation, demethylation, deacetylation) of the DNA and related histones. Modulation of epigenetic memory comprises the chemical modification of the DNA and related histones such that the gene expression is regulated. In some embodiments, the epigenetic memory is modulated over long time scales and even over generations and is considered persistent or stable. In some embodiments, the epigenetic memory is modulated in the short-term and is still dynamic. For example, the methods described herein may result in changes to epigenetic memory which result in increased persistence (as measured by the number or percentage of cells maintaining the modification and/or gene expression changes) for at least 30 days when compared to modifications of other transcriptional regulators or without any induced modulation. For example, the disclose methods may result in greater than 30% (e.g., greater than 35%, greater than 40%, greater than 45%, greater than 50%, greater than 55%, greater than 60%, greater than 65%, greater than 70%, greater than 75%, greater than 80%, or more) of the cells maintain the modulation of epigenetic memory for greater than 30 days. [00140] In some embodiments, the at least one target nucleic acid is in a cell and the chromatin regulator is endogenous to the cell. The cell may be a prokaryotic or eukaryotic cell. In some embodiments, the cell is a eukaryotic cell. In some embodiments, the cell is in vitro. In some embodiments, the cell is ex vivo. [00141] In some embodiments, the cell is in an organism or host, such that introducing the disclosed systems, compositions, vectors into the cell comprises administration to a subject. The method may comprise providing or administering to the subject, in vivo, or by transplantation of ex vivo treated cells, at least one synthetic transcription factor, nucleic acid, vector, or composition or system as described herein. [00142] A “subject” may be human or non-human and may include, for example, animal strains or species used as “model systems” for research purposes, such a mouse model as described herein. Likewise, subject may include either adults or juveniles (e.g., children). Moreover, subject may mean any living organism, preferably a mammal (e.g., human or non- human) that may benefit from the administration of compositions contemplated herein. Examples of mammals include, but are not limited to, any member of the Mammalian class: humans, non- human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice, and guinea pigs, and the like. Examples of non- mammals include, but are not limited to, birds, fish, and the like. In one embodiment of the methods and compositions provided herein, the mammal is a human. [00143] As used herein, the terms “providing”, “administering,” and “introducing,” are used interchangeably herein and refer to the placement of the systems of the disclosure into a subject by a method or route which results in at least partial localization of the system to a desired site. The systems can be administered by any appropriate route which results in delivery to a desired location in the subject. [00144] In some embodiments, the method further comprises contacting the target nucleic acid with an exogenous chromatin regulator. [00145] In some embodiments, the methods further comprise contacting the target nucleic acid with a transcription factor or regulator. In some embodiments, the transcription factor comprises Krüppel-associated box (KRAB). 5. Kits [00146] Also within the scope of the present disclosure are kits including at least one or all of a nanobody, a fusion protein thereof, or nucleic acid or vector encoding thereof, a composition or system as described herein, a cell comprising a nanobody, a fusion protein thereof, or nucleic acid or vector encoding thereof, a reporter cell as described herein, a yeast cell as described herein, and a two-part reporter gene as described herein or a nucleic acid encoding thereof. [00147] The kits can also comprise instructions for using the components of the kit. The instructions are relevant materials or methodologies pertaining to the kit. The materials may include any combination of the following: background information, list of components, brief or detailed protocols for using the compositions, trouble-shooting, references, technical support, and any other related documents. Instructions can be supplied with the kit or as a separate member component, either as a paper form or an electronic form which may be supplied on computer readable memory device or downloaded from an internet website, or as recorded presentation. [00148] It is understood that the disclosed kits can be employed in connection with the disclosed methods. The kit may include instructions for use in any of the methods described herein. The instructions can comprise a description of use of the components for the methods of identifying and selecting nanobodies or methods of modulating gene expression. [00149] The kits provided herein are in suitable packaging. Suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging, and the like. [00150] Kits optionally may provide additional components such as buffers and interpretive information. Normally, the kit comprises a container and a label or package insert(s) on or associated with the container. In some embodiment, the disclosure provides articles of manufacture comprising contents of the kits described above. [00151] The kit may further comprise a device for holding or administering the present proteins, nucleic acids, or composition. The device may include an infusion device, an intravenous solution bag, a hypodermic needle, a vial, and/or a syringe. [00152] The present disclosure also provides for kits for performing the methods or producing the components in vitro. The kit may include the components of the present system. Optional components of the kit include one or more of the following: (1) buffer constituents, (2) control plasmid, (3) sequencing primers. 6. Examples Materials and Methods Plasmid construction [00153] The TagRFP reporter (5xTetO-pEF-TagRFP-3xNLS) construct was assembled using a AAV zinc finger donor vector backbone (Addgene #22212) containing a promoter-less splice- acceptor upstream of a puromycin resistance gene and homology arms against the AAVS1 locus. Three elements of the reporter were amplified from the following sources: five TetO-binding sites upstream of a pEF promoter from PhiC31-Neo-ins-5xTetO-pEF-H2B-Citrine-ins (Addgene #78099), TagRFP-T from pEN_ERK.KTR-tagRFP-T, and 3xNLS from pEN_mCherry-NLS (both gifts from Joydeb Sinha & Mary Teruel, Stanford). These components were cloned into the AAV donor vector backbone using Gibson Assembly. [00154] The plasmids containing the rTetR-effector fusions were cloned into the PBCMV- MCS-EF1Į-Puro PiggyBac vector backbone (System Biosciences #PB510B-1), which was further modified via Gibson Assembly with the following components: PGK promoter from pSLQ2818, mIFP from pSLQ2837-1 (both gifts from Tony Gao & Stanley Qi, Stanford)67, and H2B-rTetR-Zeo from pEx1-pEF-H2B-mCherry- T2A-rTetR-KRAB-Zeo (Addgene #78352). Each effector was PCR amplified from the following plasmid: antiGFP nanobody68; antiDNMT1 nanobody—purchased from ChromoTek; antiHP1 nanobody35; KRAB—Addgene #84241; MeCP2—Addgene #110821; and DNMT3A-3L—Addgene #71827. A list of all CRs used in this study can be found in Table 1.
Figure imgf000043_0001
Figure imgf000044_0001
[00155] Plasmids containing the dCas9-effector fusions were derived from the dCas9-KRAB vector backbone (Addgene #110820) and modified by Gibson Assembly with their respective effectors from sources listed above. The dCas9-effector fusions containing KRAB or KRAB- antiDNMT1 were further modified with mCitrine-NLS upstream of the dCas9 to allow for cell sorting and analysis of endogenous gene silencing. The sgRNA cloning vector was modified to express mIFP or mCherry. Each sgRNA sequence was cloned into the plasmid using the BlpI and BstXI cloning sites, as previously described. [00156] The AAVS1 TagRFP reporter donor vector (Addgene #163083) and rTetRantiGFP recruitment vector (Addgene #163084) have been deposited to Addgene. Plasmids containing antiDNMT1 and/or antiHP1 are available on request upon signing an MTA with ChromoTek and Institut Curie, respectively. Cell culture [00157] Cells were cultured at 37 °C under a humidified atmosphere with 5% CO2. HEK293T cells (Takara Bio #632180) were maintained in Dulbecco’s modified Eagle medium (DMEM; Gibco #10569010) supplemented with 25mM D-glucose (Gibco), 1mM sodium pyruvate (Gibco), 1× GlutaMAXTM (Gibco), and 10% Tet Approved FBS (Clontech Laboratories). When cells reached 80% confluence, they were gently washed with 1× DPBS (Life Technologies) and passaged using 0.25% Trypsin (Life Technologies). For long-term storage, cells were resuspended in freezing media (10% dimethyl sulfoxide (Sigma) and cell media) in a cryovial and frozen at −80 °C. Stable cell lines construction [00158] The reporter cell line was created by integrating the TagRFP fluorescent reporter at the first intron of the constitutively expressed gene PPP1R12C at the AAVS1 locus in HEK293T cells. The integration of the reporter was performed by co-transfecting 1000 ng TagRFP reporter (5×TetO-pEF-TagRFP-3×NLS) donor plasmid and 500 ng of each TALEN arm (AAVS1- TALEN-L (Addgene #35431) targeting 5ƍ-TGTCCCCTCCACCCCACA-3ƍ (SEQ ID NO: 5929) and AAVS1-TALEN-R (Addgene #35432) targeting 5ƍ-TTTCTGTCACCAATCCTG-3ƍ(SEQ ID NO: 5930)). Cells were selected with 500 ng/mL puromycin (InvivoGen) starting 48 h post transfection for ~5 days or until all of the negative control cells died. Cells positive for TagRFP had two peaks representing the monoallelic and bi-allelic integration of the reporter at the AAVS1 locus. Cells with the lower fluorescence peak (monoallelic) were sorted by fluorescence-activated cell sorting using a Sony SH800 Cell Sorter with a 100 ^m disposable chip. Each of the individual rTetR-effector plasmids was randomly integrated into this reporter line by co-transfecting 250 ng Super PiggyBac Transposase expression vector (System Biosciences #PB200PA-1) and 750 ng of rTetR-effector donor vector. These cells were selected with 60 ^g/mL zeocin (InvivoGen) starting 48 h post transfection. All transfections were performed in 24-well plates using Lipofectamine 2000 (Invitrogen). Transient transfections [00159] Approximately 70,000 cells were seeded per well in a 24-well plate and the next day cells were transfected using Lipofectamine 2000 (Invitrogen) according to manufacturer instructions. For experiments in the antiGFP nanobody cell line, 1000 ng of each GFP-CR was delivered. For the transient silencing and reactivation experiments, 1000 ng of rTetR-effector expression vector was delivered to each well. 600 ng of dCas9-effector and 400 ng of sgRNA were co-delivered for the silencing and reactivation experiments involving dCas9 fusions. A maximum of 1000 ng of DNA vector was used per transfection. Acquisition of time-lapse movies [00160] Approximately 200,000 reporter cells stably expressing rTetR-antiGFP were seeded per well in a 24-well plate, and the next day were transfected with 1000 ng GFP-CR plasmids using Lipofectamine LTX (Invitrogen), according to the manufacturer’s instructions. Transfection with Lipofectamine LTX helped reduce cell death during time-lapse imaging. Six hours after transfection, cells were re-seeded at a density of ~20,000 cells in a 24-well imaging plate (Ibidi #82406) coated with 2% Matrigel (Corning), left overnight in the incubator to adhere, and imaging was started early the next day. Imaging was done using a Leica DMi8 fluorescence microscope with Adaptive Focus Control, a ×20 or ×40 dry objective, and a Leica DFC9000 GT sCMOS camera. Fluorophores were excited using a Lumencor SOLA SE II light source. Images were automatically acquired every 15min, using LAS X software (Leica Microsystems). Cells were grown in low-fluorescence imaging media, which consisted of Fluoro- BriteTM DMEM (Gibco) supplemented with 25mM D-glucose (Gibco), 1mM sodium pyruvate (Gibco), 1× GlutaMAXTM (Gibco), 10% Tet Approved FBS (Clontech Laboratories), and 1× Penicillin/Streptomycin (Gibco). The microscope was enclosed in an environmental control chamber (OkoLab) kept at 37 °C and 5% CO2. Dox (Tocris) was added to the imaging media to a final concentration of 1 ^g/mL. The imaging media was changed daily for ~5 days (until the cells became too confluent to continue movies). Time-lapse movies were analyzed using ImageJ by visually tracking individual cell lineages and manually circling the area corresponding to the cell’s nuclei 1 h after each cell division. Average fluorescence intensities of mIFP, TagRFP, and GFP within these contours of the cell nuclei were calculated and plotted based on their cell lineage using MATLAB (MathWorks). Gene expression analysis via flow cytometry [00161] Cells expressing stably integrated or transiently transfected rTetR-effectors were assayed by flow cytometry during and after 5 days of 1 ^g/mL dox (Tocris) treatment. When indicated, cells were also treated with 1 ^M 5-Aza-2’ (Sigma) or 100 nM chaetocin (Cayman Chemical). Media containing small molecules were replaced daily. For experiments involving dCas9-effectors, cells were analyzed 5 days post transfection, and after being sorted for silencing (TagRFP-negative cells). On the day of flow cytometry analysis, cells were collected using 0.25% Trypsin (Life Technologies). A fraction of the cells (varying between one half to one twentieth, depending on cell density) were replated for the next time point. The remaining cells were resuspended in flow buffer (1× Hank’s balanced salt solution (Life Technologies) and 2.5 mg/mL bovine serum albumin (BSA) (Sigma)) and filtered through a 40 ^m strainer (Corning) to remove cell clumps. Cellular fluorescence distributions were measured with the CytoFLEX S Flow Cytometer (Beckman Coulter) and the CytExpert Software (Beckman Coulter). The resulting data were analyzed with a custom MATLAB program called EasyFlow (antebilab(dot)github(dot)io/easyflow/). After cells were gated based on forward and side scatter, a manual gate was imposed on the TagRFP fluorescence to determine the percentage of silent cells for each sample. The gate was selected to contain 1–5% of the positive TagRFP signal in untreated cells. A total minimum of 20,000 events were recorded for each sample. [00162] For experiments analyzing the expression of the endogenous CXCR4 gene, cells were first trypsinized and then washed with 1% BSA (Sigma) in 1× DPBS (Life Technologies). Cells were then incubated on ice for 1 h with monoclonal Brilliant Violet 421 (BV421)-labeled anti- CXCR4 antibody (clone 12G5 (1 : 20); BioLegend). BV421-labeled IgG2a (clone MOPC-173 (1 : 20); Biolegend) served as an isotype control. Afterwards, cells were washed three times with 1% BSA/DPBS and then analyzed by flow cytometry for cells that were double positive for dCas9 (mCitrine) and sgRNA (mCherry). Targeted bisulfite sequencing [00163] Reporter cells stably expressing rTetRantiDNMT1, rTetR-DNMT1, or rTetR- DNMT3B were treated with 1 ^g/mL dox for 5 days. Afterwards, each treated cell line was sorted in TagRFP-negative (+dox OFF) and TagRFP-positive (+dox ON) cells using a SONY SH800 Cell Sorter. Total genomic DNA was extracted from these cells with the DNeasy Blood & Tissue Kit (Qiagen) according to the manufacturer’s instructions and quantified using a NanoDrop 8000 spectrophotometer (Thermo). As a control, DNA was also extracted from cells not treated with dox (−dox) grown at the same time as the +dox cells for each cell line and sorted to include the entire population (which is >97% ON for all cell lines). A minimum of 2000 ng of DNA for each sample was submitted to Zymo Research Corporation (Irvine, CA) for processing and analysis using the MethylCheckTM Service: Targeted Bisulfite Sequencing. Purified, non- methylated DNA from human HCT116 DKO cell line that contains knockouts of DNA methyltransferases, DNMT1 and DNMT3B, and in vitro-methylated DNA was obtained from Zymo Research (#D5014). [00164] Assays were designed targeting CpG sites in 100–300 nucleotide regions at the reporter gene and two control genes, IGF2 (positive) and ACTB (negative), using primers created with Rosefinch, Zymo Research’s proprietary sodium bisulfite converted DNA-specific primer design tool. In addition, primers were designed to avoid annealing to CpG sites in the region of interest. In the event that CpG sites were absolutely necessary for target amplification, primers were synthesized with a pyrimidine (C or T) at the CpG cytosine in the forward primer, or a purine (A or G) in the reverse primer to minimize amplification bias to either the methylated or unmethylated allele. Following primer validation, samples were bisulfite converted using the EZ DNA Methylation-Lightning™ Kit (Zymo Research, #D5030). Amplification of all samples using region-specific primer pairs was performed; the resulting amplicons were pooled and subsequently barcoded using PCR. After barcoding, samples were purified (ZR-96 DNA Clean & Concentrator, #D4023) and then prepared for massively parallel sequencing using a MiSeq V2 300 bp Reagent Kit (Illumina) and paired-end sequencing protocol according to the manufacturer’s guidelines. [00165] Sequence reads were identified using standard Illumina base-calling software and then analyzed using a Zymo Research proprietary analysis pipeline, which is written in Python. Low quality nucleotides and adapter sequences were trimmed off during analysis QC. Sequence reads were aligned back to the reference genome using Bismark (www(dot)bioinformatics(dot)babraham(dot)ac(dot)uk/projects/bismark/), an aligner optimized for bisulfite sequence data and methylation calling. Paired-end alignment was used as default thus requiring both read 1 and read 2 be aligned within a certain distance, otherwise both read 1 and read 2 were discarded. Index files were constructed using the bismark_genome_preparation command and the entire reference genome. The non_directional parameter was applied while running Bismark. All other parameters were set to default. Nucleotides in primers were trimmed off from amplicons during methylation calling. The methylation level of each sampled cytosine was estimated as the number of reads reporting a C, divided by the total number of reads reporting a C or T. Statistical analysis [00166] Data are displayed as individual points or as mean ± SD, with sample size indicated in the figure legend. Statistical significance was evaluated using GraphPad Prism 8. A one-way analysis of variance with post hoc Tukey’s honestly significant difference test or Student’s two- sample unpaired t-test were used for statistical analysis. Statistical significance of the data is indicated as follows: *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001; ns = not significant. Gene control modeling and data fitting [00167] The percentage of cells in each of the 3 states in the gene control model (FIG. 4D) was derived by solving the differential equations associated with the kinetic model (See below), and all fits were done based on these equations, as follows: [00168] In FIGS. 2E, 3B, 3D, 13B, and 15A, the percentages of cells silent during the release period IJ are fit to an exponential decay:
Figure imgf000049_0001
where I1 is the percentage of cells irreversibly silenced at the end of the recruitment period, and kA is the rate of reactivation. [00169] For FIG. 4C, the fraction of cells active (A = (1− fraction cells silenced)/100) after a recruitment signal (+dox) of duration t and reactivation period (−dox) of duration IJ, were fit to the following equation (derived below):
Figure imgf000049_0002
for all recruitment times ^^ ^^ag, and ^(^,^)=1 for ^< ^^ag. [00170] For silencing time points, since IJ = 0, the equation above simplifies to:
Figure imgf000049_0003
[00171] All experimental data points from three independent replicates across 14 days of silencing (red points in FIG. 4C, top) and 30 days of reactivation (black points in FIG. 4C, top) were recorded in MATLAB as three vectors: A (percent of cells silent), t (time with dox), and IJ (time after dox removal). For the silencing data points (+dox) t = 0, 1, . . . , 14 and IJ = 0, 0, . . . , 0. For reactivation data points (−dox) t = 14, 14, . . . , 14 and IJ = 0, 1, 2, . . . , 30. These data were fit to the A(t,IJ) eq. (2) above using MATLAB’s fit function with the NonlinearLeastSquares method, with kS, kI, kA, and Tlag as free parameters. [00172] For pulsing experiments predictions in FIGS. 4F and 4G the full solutions
Figure imgf000049_0004
^^^^^^^^ǡ^^ൌ^^(see below) were used to calculate the fractions of cells in each state for each period. For the first +dox period, the starting conditions were^^0ൌ^ǡ^^^ൌ^ǡ^^^ൌ^ǡ^and cells started silencing after a lag period Tlag, as such eq. (4) below was used. For the next periods, general eq. (3) (below) was applied using as starting conditions for each period the end points from the previous one. The Tlag period was only applied in the first dox induction period. [00173] The linear fit in FIG. 4B was performed using MATLAB’s polyfit function with coefficient 1 and the goodness of fit was calculated using
Figure imgf000049_0005
where y is the mean percent cells silenced at each time point from three replicates and N is the number of time points. Modeling of Gene Control [00174] Gene silencing and memory upon recruitment of antiDNMT1 at a gene can be described by a kinetic model consisting of 3 gene states (FIG. 4D). The time evolution for the fraction of cells in each of the three states - active (A), reversibly silent (R), and irreversibly silent (I) - can be calculated from the set of differential equations associated with the kinetic rates in FIG. 4D, during recruitment and release, respectively. The cells in each state with a “+” subscript (e.g., ^+), to indicate when the equations describe the “+dox” silencing period, and with a “-” subscript for the “-dox” period (e.g., ^−). At the end, they were combined into a single function that describes the behavior across the two periods (no subscript, e.g., A). [00175] Derivation of the fractions of cells in each gene state during antiDNMT1 recruitment (+dox) During recruitment, the time of dox induction was denoted as t, and can describe the fraction of cells in each state according to the kinetic model in FIG. 4D, top:
Figure imgf000050_0001
[00176] The fraction of cells active during recruitment can be solved directly from the first equation above:
Figure imgf000050_0002
where ^0 is the fraction of cells active at the beginning of recruitment. Since the total fractions of silent and active cells must add up to 1, the total fraction of silent cells (S) can be described as: ^ This equation is sufficient to describe and fit the silencing phase. Note
Figure imgf000050_0003
that in order to express percentages of cell silent as a function of time, each equation was multiplied by 100. [00177] To understand how many cells are committed to long term memory (and thus the behavior after CR release), how silent cells partition into reversibly versus irreversibly silent over time was solved: ^
Figure imgf000050_0004
is replaced into
Figure imgf000050_0005
Figure imgf000050_0006
[00178] This differential equation was solved to get:
Figure imgf000050_0007
[00179] The fraction of irreversible silenced cells over time is: ^+(^)=1−^+(^)−^+(^). [00180] In summary, the function describing the fraction of cells in each of the three states for a given silencing dox signal of duration ^^(and no reactivation period):
Figure imgf000051_0001
[00181] Derivation of the fractions of cells in each gene state during antiDNMT1 release (- dox) [00182] During release (-dox), the time since dox was removed was denoted as ^, and can describe the fraction of cells in each state according to the kinetic model in FIG. 4D, bottom:
Figure imgf000051_0002
[00183] The fraction of cells reversibly silent during release is solved from the ^^ିȀ^^ equation above:
Figure imgf000051_0003
where ^1 is the fraction of reversibly silenced cells at the beginning of release (end of recruitment). [00184] During release, the irreversible fraction stays constant, at the value reached at the end of recruitment, ^1: ି^ (^)=^1 [00185] The total fraction of cells silent over time in the release phase, ^ି^ɒ^ ൌ
Figure imgf000051_0006
^ି^ ^ ɒ ^ , is described by an exponential decay to the irreversible fraction: ^ି ^ ɒ ^
Figure imgf000051_0007
[00186] The fraction of cells active is:
Figure imgf000051_0005
[00187] In summary:
Figure imgf000051_0004
[00188] General solution and additional considerations for fitting the data [00189] The results of the two previous sections can be combined to obtain general solutions that describe the cells in each state across both recruitment and release times. The starting fractions of cells ^1, ^1, at the beginning of the –dox release, depend on the duration of +dox period ^: ^1=^+(^), and ^1=^+(^). Therefore, based on the solutions derived in the release section above (equations (2)):
Figure imgf000052_0002
[00190] By replacing ^+(^), ^
Figure imgf000052_0001
, and ^+(^) with the set of equations (1) derived in the recruitment section, the general solution after a recruitment signal (+dox) of duration t, and reactivation period (-dox) of duration IJ becomes:
Figure imgf000052_0003
[00191] During recruitment, a time lag was observed between dox addition and the onset of silencing (^^ag). Therefore, for fitting purposes, in the equations above ^ becomes ^− ^^ag, for recruitment times larger than ^^ag, and no changes in the fractions of silent/active cells are allowed at shorter times. The final equations used to calculate the fraction of cells silent over time during a period of +dox induction are:
Figure imgf000052_0004
[00192] For fitting a continuous dox signal and the reactivation following it, all cells are started as silenced: ^0=1, ^0=0, ^0=0. Therefore, the equations for silencing simplify to:
Figure imgf000053_0001
[00193] The general solution in the case when all cells are started as active (^0=1, ^0=0, ^0=0) becomes:
Figure imgf000053_0002
for all recruitment times ^^ ^^ag, and ^(^,^)=1, ^(^,^)=0, ^(^,^)=0 for ^< ^^ag. Yeast Display [00194] Expression of FLAG-tagged DNMT3A in HEK-293 cells Full length DNMT3A with 3xFLAG was cloned into the pRetro-CMV2-TO-puromycin vector using Gibson assembly. Twenty-four hours prior to transfection, HEK-293 cells were plated in 4 x 10cm plates in DMEM media + 10% FBS supplemented with L-glutamine and Pen/Strep. At the time of transfection, cells were about 70-80% confluent. The following day (20-24 hours later), cells were transfected with pRetro-CMV2-TO-3xFLAG-DNMT3A plasmid. Before transfection: in each 10cm plate, medium was changed to 20 ml plain DMEM (no FBS, no Pen/Strep). For transfection, two sets of 1.5 ml tubes were prepared: The first set contained 450 μl 2X HBS (50 mM HEPES, pH 7.05, 10 mM KCl, 12 mM dextrose, 280 mM NaCl, 1.5mM Na2PO4).The other set contained 25μg Plasmid DNA (pRetro-CMV2-TO-3xFLAG-DNMT3A) + 65μl 2M CaCl2 into 0.1XTE (450 μl total). The DNA and CaCl2 were mixed by pipetting up and down with a 200 μl pipet and added dropwise to the 2X HBS. This mixture was incubated at room temp for 1 min. The DNA-Calcium phosphate co-precipitate was added dropwise to the surface of the media containing the cells. The plate was swirled gently to mix. Ten to twelve hours after transfection, the medium was gently aspired, and 10 ml of pre-warmed DMEM medium (+10%FBS, no Pen/Strep) was added to the plates, trying not to disturb the fine DNA-Calcium phosphate precipitates on the bottom of the plate. 48-72hrs post transfection, transfected cells were harvested for lysis and DNMT3A protein immunoprecipitation on magnetic beads. [00195] Preparation of DNMT3A-coated magnetic beads for yeast display To prepare the HEK-293 cell lysis, cells were scraped in lysis buffer (50ௗmM Tris–HCl, pH 8.0, 1ௗmM EDTA, 150ௗmM NaCl, 1% NP40, 1× PMSF, and 1 x NEM) and cell lysates kept on ice for 30ௗmin. Cell lysates were cleared by centrifugation at 10,000ௗ×ௗg for 1ௗmin. The supernatant was kept on ice before the immunoprecipitation. To prepare magnetic beads for immunoprecipitation, the anti- Flag magnetic beads were resuspended in the vial (pipet gently 10 times, don’t vortex). 150 μL (the amount may be scaled up or down as required) anti-Flag Magnetic Beads (No. B26101, Bimake) suspension was transferred to a new tube with 0.5 mL TBS buffer (50 mM Tris HCl, 150 mM NaCl, pH 7.4). The mixture was gently pipetted 5 times. The tube was placed on a magnet for 10 seconds to separate the beads from the solution, and the supernatant was discarded. This step was repeated 2 times. To bind the DNMT3A complex to the magnetic beads, ~500 μL of cell lysate was added to the washed magnetic beads. The tubes were gently rotated for 2 h at room temperature or overnight at 4°C. Then the tubes were placed on the magnet to separate the beads from the solution for 2 minutes and the supernatant was transferred into a new tube for detecting whether Flag-tagged protein is left unbound. To wash any non-specifically bound proteins from the beads, 500 μL PBST was added to the tube (NaCl 136.89 mM; KCl 2.67 mM; Na2HPO48.1 mM; KH2PO41.76 mM; 0.5% Tween20), and the magnetic beads were resuspended by pipetting gently. Then the tube was rotated for 5 min, and, after that, placed on the magnet to separate the beads from solution for 2 minutes to remove the supernatant. This wash step was repeated 2 times. If any non-specific proteins still bound to the beads (as measured by Western blot), please extend the wash time, increase the number of washes, or the detergent content in the wash solution. [00196] Yeast growth and induction The yeast nanobody library was maintained in Yglc4.5 - Trp medium (1 liter: 3.8 g of -Trp drop-out media supplement (US Biological), 6.7 g Yeast Nitrogen Base, 10.4 g Sodium Citrate 7.4 g Citric Acid Monohydrate, 10 mL Pen-Strep (10,000 units/mL stock), and 20 g glucose, pH4.5). Nanobody expression was under the control of the GAL1 promoter such that nanobodies were produced on the cell surface when yeast was grown in a galactose-containing medium. Expression of the nanobody library was induced by dilution of a yeast aliquot into -Trp +galactose medium (1 liter: 3.8 g -Trp drop-out media supplement (US Biological), 6.7 g Yeast Nitrogen Base, 10 mL Pen-Strep (10,000 units/mL stock), 20 g glucose or galactose (glucose for normal growth and galactose for induction of nanobodies), pH 6) followed by shaking for 48 hours, at 25 °C, 220 rpm. For the initial yeast dilution, at least 5 × 1010 yeast cells were used to ensure >10x coverage of the nanobody library during each passage, and thus avoid loss of nanobody clones. [00197] Bead-Based Enrichment (BBE) of nanobodies using yeast surface display DNMT3A- coated beads were prepared by immunoprecipitation as described above. 150 μl washed beads were removed from the magnet and the beads were resuspended in 1000 μl ice-cold selection buffer (20ௗmM HEPES, pH 7.5, 150ௗmM sodium chloride, 2% (w/v) BSA, 1ௗmM EDTA), and placed on ice until needed. 5x1010 induced yeast were used for the first round of selection and 5×108 induced yeast cells were used for subsequent rounds. the yeast cells were washed and resuspended in selection buffer and then incubated with the DNMT3A-coated beads at 4ௗ°C for 2 hours. [00198] Yeast negative selection. Each round of BBE selection began with a negative selection step which involved incubating the yeast with non-antigen-coated beads to remove yeast-expressing nanobodies that bound nonspecifically to the magnetic beads. Specifically, 150^L resuspended Flag-conjugated beads were added to the yeast cells induced with galactose. Cells were incubated on the rotary wheel at 4°C for 2 h. Upon completion of the incubation, the tube was placed on the magnet, taking care to transfer any liquid lodged in the cap of the tube to the bottom portion of the tube. After 2 minutes, the supernatant was carefully removed from the tube and transferred into a fresh 10ml tube. The supernatant served as the input for the next selection step. The beads were resuspended in 1ml ice-cold selection buffer with a pipette and placed on the magnet for 2min. The supernatant was removed from the washed beads and discarded. The beads were resuspended in 1ml ice-cold selection buffer and set aside as negative#1 to enable estimation of the number of cells captured by the negative selection. A second negative selection was performed using the supernatant from the previous step as input, before proceeding to the following step with the resulting depleted supernatant. [00199] Yeast positive selection. After the negative selection, DNMT3A-binding nanobodies were enriched over 3 rounds of BBE selection by staining the yeast with DNMT3A complex- coated beads. Specifically, the yeast cells after negative selection were mixed with the DNMT3A-coated magnetic beads and incubated on the rotary wheel at 4°C for 2h. Upon completion of the incubation, the tube was placed on a magnet, taking care to transfer any liquid lodged in the cap of the tube to the bottom portion of the tube. The cells and the beads were incubated on the magnet for 2min. Then the supernatant was carefully removed from the tube and discarded. The beads were resuspended in 10ml ice-cold selection buffer using a pipette and then placed on the magnet for 2min. The supernatant was removed and the beads contained a population of yeast cells containing nanobodies enriched for binding to the target, in this case DNMT3A. [00200] Rescue the enriched yeast population. Beads from the previous positive selection were resuspended in -Trp4.5 media and transferred to a sterile culture tube containing 4ml - Trp4.5 media (5ml -Trp_media+beads+cells in total). The tube was vortexed gently and then a 5^L sample was collected from the culture. The sample (5ul) was diluted into 995^L - Trp4.5_media (200x dilution) and set aside (tube labeled as positive#1) for a later analysis step. The cells on the beads were grown at 30°C with shaking for 48 hours. [00201] Plate fractions of beads from negative and positive selections to estimate the number of cells recovered in each step. The saved beads from the negative sorts (negative#1) were vortexed and 100^L beads were transferred into 400^L fresh -Trp4.5_media. Take these diluted samples, vortex, and transfer 5^L of each sample into 995^L -Trp4.5_media (200x dilution) (negative #2). Thee 200x dilutions of the negative sort(negative#2) and the positive sorts (positive#1) were vortex and 10^L from each population was transferred into 190^L - Trp4.5_media (4000x dilution). A -Trp4.5_media plate was divided into four regions using a permanent marker. Each dilution was vortex and 20^L was plated. The plate was incubated at 30°C for 3 days and the colonies were counted. One colony in the 200x and 4000x dilutions represents 5x104 and 1x106 cells recovered, respectively. [00202] Expand and induce selected yeast cells for further rounds of selection. The beads from the first sort are removed, the yeast cells are expanded, and a portion of them are induced as follows. After overnight growth, measure the OD600 of the cells. If the OD600 was still low, the cells were allowed to grow another day. Once the culture approaches saturation, the cells were pelleted (at 900xg for 5 minutes) and the supernatant was aspirated. The pellet was resuspended in 1ml Trp4.5_media and the cells were transferred to a 2ml tube. The supernatant was recovered following magnetic precipitation and the cells were diluted into two cultures for further expansion - 2.5x108 cells into 25ml -Trp4.5_media for growth and induction and remaining cells into 25ml -Trp4.5_media for overnight growth and temporary storage at 4 degree in case the first selected population needs to be induced and selected again. Once the yeast culture containing ~2.5x108 cells reached an OD600 between 2 and 5, 5x108 cells were pelleted and resuspended in 50ml -Trp4.5_media before incubation at 25°C with shaking for 48h to induce. [00203] Confirm the binding of DNMT3A complex with enriched nanobody library. 3xFLAG tagged DNMT3A was expressed in HEK-293T cells, and the resulting cell lysis containing the DNMT3A complex mixture of proteins was used as the selection antigen. After each round of BBE selection, following galactose induction of nanobodies, nanobody-expressing yeast were incubated with the DNMT3A complex, washed, and then stained with Anti-DYKDDDDK Tag (DYKDDDDK (SEQ ID NO: 5931) tag) Mouse Monoclonal antibody (FITC (Fluorescein)) (GenScript, A01632, 1:50 dilution), and HA-Tag (6E2) Mouse mAb (Alexa Fluor® 647 Conjugate) (Cell Signaling Technology, 3444S, 1:50 dilution). DNMT3A binding was confirmed and analyzed by flow cytometry (ZE5) to verify the enrichment for nanobody binders compared to the naive yeast library. [00204] After three round BBEs selections, the library of nanobody plasmids was extracted from the enriched yeast library by Zymoprep Yeast Plasmid Miniprep II (Cat# D2004). High-throughput screening of nanobodies capable of silencing in human cells [00205] Pooled library cloning of selected nanobodies into a lentiviral construct The library of nanobody plasmids was extracted after three rounds of yeast display enrichment, and then PCR amplified. 8x 50 ^L reactions were set up in a clean PCR hood to avoid amplifying contaminating DNA. For each reaction, 10 ng of template were used, 23 μl H2O, 1 μl of each 10 μM primer, and 25 μl of Q5 Hot Start High-Fidelity 2X Master Mix (NEB). The thermocycling protocol was 3 minutes at 98°C, then cycles of 98°C for 10s, 55°C for 30s, 72°C for 50s, and then a final step of 72°C for 10 minutes. The default cycle number was 29x, and this was optimized for the library to find the lowest cycle that resulted in a clean visible product for gel extraction (in practice, 25 cycles was the minimum). After PCR, the resulting dsDNA libraries were gel extracted by loading ^ 4 lanes of a 2% TAE gel, excising the band at the expected length (around 400 bp), and using a QIAgen gel extraction kit. The libraries were cloned into a lentiviral recruitment vector pWJ036 with 4x10 μl GoldenGate reactions (75 ng of pre-digested and gel-extracted backbone plasmid, 5 ng of library (2:1 molar ratio of insert:backbone), 0.13 ml of T4 DNA ligase (NEB, 20000 U/ml), 0.75 μl of Esp3I-HF (NEB), and 1 μl of 10x T4 DNA ligase buffer) with 30 cycles of digestion at 37°C and ligation at 16°C for 5 minutes each, followed by a final 5 minute digestion at 37°C and then 20 minutes of heat inactivation at 70°C. The reactions were then pooled and purified with MinElute columns (QIAgen), eluting in 6 ^L of ddH2O. 2 ml per tube was transformed into two tubes of 50 ml of Endura electrocompetent cells (Lucigen, Cat#60242-2) following the manufacturer’s instructions. After recovery, the cells were plated on 3-7 large 10’’ x 10’’ LB plates with carbenicillin. After overnight growth at 37°C, the bacterial colonies were scraped into a collection bottle and plasmid pools were extracted with a HiSpeed Plasmid Maxiprep kit (QIAgen). 2-3 small plates were prepared in parallel with diluted transformed cells in order to count colonies and confirm the transformation efficiency was sufficient to maintain at least 30x library coverage. To determine the quality of the libraries, the domains were amplified from the plasmid pool and from the original oligo pool by PCR with primers with extensions that include Illumina adapters and sequenced. The PCR and sequencing protocol were the same as described below for sequencing from genomic DNA, except these PCRs use 10 ng of input DNA and 17 cycles. These sequencing datasets were analyzed as described below to determine the uniformity of coverage and synthesis quality of the libraries. In addition, 20-30 colonies from the transformations were Sanger sequenced (Quintara) to estimate the cloning efficiency and the proportion of empty backbone plasmids in the pools. [00206] High-throughput recruitment to measure nanobody silencing activity Large scale lentivirus production and infection of HEK-293 cells were performed as follows: To generate sufficient lentivirus to infect the libraries into HEK-293 cells, HEK293T cells were plated on four 10-cm tissue culture plates. On each plate, 5ௗ×ௗ106 HEK293T cells were plated in 10 mL of DMEM, grown overnight, and then transfected with a mixture of the three third-generation packaging plasmids (6.5μg pMDLG/pRRE, 5μg Rev, 3.5μg VSVG) and 10 μg of rTetR- Nanobody library vectors using the calcium phosphate method.^Lentivirus was harvested at 48 hours and 72 hours. The pooled lentivirus was filtered through a 0.45-mm PVDF filter (Millipore) to remove any cellular debris. For the third round enriched Nanobody library targeting the DNMT3A complex screen, 8x10cm plates with HEK-293 reporter cells at the density of 250 cells/mm2 were infected with the lentiviral library for two separate biological replicates of the infection. Infected cells grew for 3 days and then the cells were selected with 2 μg/mL puromycin. Infection and selection efficiency were monitored every other day using flow cytometry to measure mScarlet (and thus nanobody) positive cells (ZE5). After 3 days of selection with puromycin, cells in each 10cm plate were transferred to 15cm plate to make the maintenance coverage > 25,000 x cells per library element (a very high coverage level that compensates for losses due to incomplete puromycin selection, library preparation, and library synthesis errors). On day 3 post-infection, nanobody recruitment at the reporter was induced by treating the cells with 1 μg/ml doxycycline (Fisher Scientific) for 5 days. Cells were split every other day and measured for maintenance coverage on ZE5. [00207] Magnetic separation of reporter cells The reporter included a synthetic surface marker, consisting of the human IgG1 Fc region linked to an Igk leader and PDGFRb transmembrane domain, to enable magnetic separation of OFF from ON cells. At each time point, HEK-293 cells were trypsinized and spun down at 300 x g for 5 minutes. Cells were then resuspended in the 15ml PBS (GIBCO) and the spin down and aspiration was repeated, to wash the cells and remove any IgG from serum. Dynabeads M-280 Protein G (ThermoFisher, 10003D) were resuspended by pipetting gently. 50 mL of blocking buffer was prepared per 2 x 108 cells by adding 1 g of biotin-free BSA (Sigma Aldrich) and 200 μL of 0.5 M pH 8.0 EDTA (Thermo Fisher, 15575020) into DPBS (GIBCO), and then kept on ice. Beads (60 ^L) were prepared for every 1 x 107 cells, by adding 1 mL of buffer per 200 μL of beads, vortexing for 5 s, placing on a magnetic tube rack, waiting one minute, removing supernatant, and finally removing the beads from the magnet and resuspending in 100-600 μl of blocking buffer per initial 60 μL of beads. After incubation, the bead and cell mixture were placed on the magnetic rack for > 2 minutes. The unbound supernatant was transferred to a new tube, placed on the magnet again for > 2 minutes to remove any remaining beads, and then the supernatant was transferred and saved as the unbound fraction. Then, the beads were resuspended in the same volume of blocking buffer, magnetically separated again, the supernatant was discarded, and the tube with the beads was kept as the bound fraction. The bound fraction was resuspended in a blocking buffer or PBS to dilute the cells (the unbound fraction is already dilute). Flow cytometry (ZE5) was performed using a small portion of each fraction to estimate the number of cells in each fraction (to ensure library coverage was maintained) and to confirm separation based on citrine reporter levels (the bound fraction should be > 90% citrine positive, while the unbound fraction is more variable depending on the initial distribution of reporter levels). Finally, the samples were spun down and the pellets were frozen at -80 °C until genomic DNA extraction. [00208] Genomic library preparation and next generation sequencing Genomic DNA was extracted with the QIAgen Blood Maxi Kit following the manufacturer’s instructions with up to 1.25 x 108 cells per column. DNA was eluted in EB and not AE to avoid subsequence PCR inhibition. The domain sequences were amplified by PCR with primers containing Illumina adapters as extensions. A test PCR was performed using 400 ng of genomic DNA in a 50 ^L (half size) reaction to verify if the PCR conditions would result in a visible band at the expected size for each sample. Then, 25x 50 ^L reactions were set up on ice (in a clean PCR hood to avoid amplifying contaminating DNA), with the number of reactions depending on the amount of genomic DNA available in each experiment. 400 ng of genomic DNA, 23 ^L H2O, 1 of each 10 uM primer, and 25 ^L of Q5 Hot Start High-Fidelity 2X Master Mix (NEB) was used in each reaction. The thermocycling protocol was to preheat the thermocycler to 98 °C, then add samples for 3 minutes at 98 °C, then 32x cycles of 98 °C for 10 s, 55 °C for 30 s, 72 °C for 50 s, and then a final step of 72 °C for 10 minutes. The PCR reactions were pooled and ^140 ^L were run in at least three lanes of a 2% TAE gel alongside a 100-bp ladder for at least one hour, the library band around 400 bp was cut out, and DNA was purified using the QIAquick Gel Extraction kit (QIAgen) with a 30 ^L elution into non-stick tubes (Ambion). A confirmatory gel was run to verify that small products were removed. These libraries were then quantified with a Qubit HS kit (Thermo Fisher) and sequenced on an Illumina NextSeq with a High output kit using a paired end (forward read 200 and reversed read 100) and 8 cycle index reads. [00209] High-throughput sequencing data analysis Sequencing reads were demultiplexed using bcl2fastq (Illumina). Individual CDR sequences were extracted from read pairs and CDR sequence combination instances were counted using the Python script ‘make_nanobody_counts.py’. Briefly, the script uses portions of the nanobody constant sequences that bookend each CDR to define the boundaries of and extract CDR sequences along with their per-nucleotide quality scores. CDR1 and CDR2 information were extracted from the R1 read while CDR3 information was extracted from the reverse complement of the corresponding R2 read. CDR-wide mean quality scores were computed from the per-nucleotide quality scores and read sequence and quality information were compiled into a dataframe. Reads with one or more undetected CDR and/or with mean quality scores less than 30 were filtered out. Reads with identical CDR combinations at the DNA-sequence level were grouped and counted. This process was repeated for each sample sequenced. The enrichments for each nanobody (CDR combination) between OFF and ON samples were computed using the script ‘makeRhos.py’. In this script, nanobodies with fewer than 5 reads in both samples for a given replicate were filtered out, whereas nanobodies with fewer than 5 reads in one sample would have those reads adjusted to 5 to avoid inflating enrichment values due to low sequencing depth. Counts were normalized to the sum of counts in that sample to account for differences in sequencing depth (in effect, frequencies were computed) prior to computing log2(OFF:ON) enrichment scores. Individual validations of nanobody function in human cells [00210] Interaction assay between nanobodies and DNMT3A Individual nanobodies were synthesized (gBlock, IDT) and cloned as fusions with rTetR(SE-G72P) with a 3xFLAG, upstream of a P2A-mScarlet and puromycin selection marker using Gibson assembly cloning into the lentivirus backbones pWJ036. Full length DNMT3A with 3xHA was cloned into the pRetro-CMV2-TO-puromycin vector using Gibson assembly. 5ௗ×ௗ106 HEK-293T cells were seeded in 10ௗcm cell culture dishes and grown for 24ௗh. The plasmid expressing 3xFLAG- rTetR(SE-G72P)-Nanobody (12.5ug) and the plasmid expressing 3XHA-DNMT3A(12.5ug) were co-transfected into HEK-293T cells using Calcium phosphate method. 36 hours after transfection, cells were scraped in lysis buffer (50ௗmM Tris–HCl, pH 8.0, 1ௗmM EDTA, 150ௗmM NaCl, 1% NP-40, 1xNEM and 1× PMSF), and cell lysates kept on ice for 30ௗmin. Cell lysates were cleared by centrifugation at 10,000ௗ×ௗg for 10ௗmin. Immunoprecipitations were performed with Anti-Flag Magnetic Beads (No. B26101, Bimake) for 5ௗh at 4ௗ°C. Samples were washed 5ௗ×ௗ30ௗmin in lysis buffer. Proteins from the immunoprecipitates and from whole-cell lysates were separated by 12% SDS-PAGE, transferred to nitrocellulose membranes, blocked with 5% milk in TBST and analyzed by immunoblotting with mouse anti-FLAG M2 (Sigma, 1:1000) and mouse anti-HA (#901501, Biolegend, 1:1000). Staining of tubulin from whole-cell lysates with mouse anti-tubulin (12G10, Developmental Studies Hybridoma Bank, 1:5000) was used as the loading control. [00211] Silencing assay measurement by flow cytometry For each individual nanobody tested, a lentivirus expressing 3xFLAG-rTetR(SE-G72P)-Nanobody was produced in the pWJ036 backbone. K562, Hela and HEK-293 cells expressing pJT055-pEF-citrine reporter with TetO sites upstream of the pEF were then infected with each of these lentiviruses. One day after infection, selection for the nanobody constructs was started using puromycin (2 ug/mL), and continued until > 90% of the cells were mScarlet positive (~2 days). Cells were split into separate wells of a 24-well plate and either treated with doxycycline (1ug/ml) to recruit rTetR- Nanobody to the reporter or left untreated. After 5 days of treatment, doxycycline was removed by spinning down the cells, replacing media with DPBS (GIBCO) to dilute any remaining doxycycline, and then spinning down the cells again and transferring them to fresh media. Time points were measured every 2-3 days by flow cytometry analysis of > 30,000 cells on a ZE5 flow cytometer (BioRad). Data was analyzed using FlowJo. Events were gated for viability and for mScarlet as a delivery marker. Example 1 Nanobodies against GFP-CRs are used to control gene expression [00212] To test whether nanobodies can be used to recruit CRs for efficient gene expression control, a nanobody against green fluorescent protein (GFP) was fused to a reverse tetracycline repressor (rTetR) DNA-binding domain and used to recruit various GFP-tagged CRs (GFP-CR) to a TagRFP fluorescent reporter gene located at the AAVS1 locus in HEK293T cells (FIG. 1A). The reporter contained five Tet operator (TetO)-binding sites upstream of a constitutive pEF promoter driving the expression of the fluorescent gene. Upon the addition of doxycycline (dox) to the cell media, rTetR bound to these sites, allowing recruitment control and release of the rTetR-nanobody fusion at the reporter. [00213] Using this system, time-lapse microscopy was used to measure the localization dynamics of GFP-HP1Į and GFPHDAC5 during transient expression, and their connection to gene expression in a cell population stably expressing the rTetR-antiGFP nanobody fusion and the TagRFP reporter. Both GFP-tagged HP1Į and HDAC5 mediated fast silencing of the reporter, as indicated by the decrease in TagRFP signal as early as 24 h after dox addition (FIG. 1B). Furthermore, although HP1Į localizes mostly in the nucleus (FIG. 11A), HDAC5 was seen translocating in and out of the nucleus (FIG. 11B), as seen previously. In cells where there was dilution of the GFP expression vector and thus the expression of the CR was lost, the TagRFP reporter reactivated after ~50 h (FIG. 1B). At the end of 5 days of recruitment (dox treatment), the TagRFP reporter was still silent in cells that were still expressing GFP (FIG. 1C). [00214] Based on these results, flow cytometry was used to measure reporter silencing after 5 days of recruitment in cells ectopically expressing GFP-HP1Į, GFP-HP1ȕ, GFP-HP1Ȗ, or GFP- HDAC5. By the end of 5 days of transient expression and recruitment, all CRs tested were able to silence the reporter in a dox-dependent manner in the majority of the cells still expressing the GFP-CR (FIG. 1D 11C). In addition, recruitment of GFP alone did not lead to silencing of the reporter, which suggested the silencing observed was dependent on the activity of the recruited CR (FIG. 1D). HP1Į, HP1ȕ, and HDAC5 led to a higher fraction of cells silenced compared to HP1Ȗ, consistent with their reported roles in silencing and association with heterochromatin. Nevertheless, HP1Ȗ, which can associate with either heterochromatin or actively transcribed regions, still led to silencing in a majority of cells (FIG. 1D). Surprisingly, lower levels of GFP- CRs led to a higher fraction of cells silenced after 4 days of recruitment for all four CRs (FIG. 11D). Moreover, increasing the number of anti-GFP nanobodies fused to a single rTetR to 8 did not increase the fraction of cells silenced (FIG. 11E), suggestive that a single nanobody is sufficient for silencing in the reporter system. Example 2 Nanobodies against DNMT1 and HP1 can silence a Reporter Gene and Confer Epigenetic Memory [00215] Two existing nanobodies against endogenous CRs, antiHP1 and antiDNMT1, were tested for their capacity to silence and induce epigenetic memory. The antiHP1 nanobody was shown to bind to all three isoforms of HP1 in cell lysate by western blotting and in cells by immunofluorescence. The antiDNMT1 nanobody has been used to immunoprecipitate endogenous DNMT1 from whole cell lysate and visualize endogenous DNMT1 in live cells. Fusions between rTetR and either antiHP1 or antiDNMT1 were cloned and stably integrated into HEK293T cells containing the TagRFP reporter with TetO sites at the AAVS1 locus (FIG. 2A). Both nanobodies mediated dox-dependent silencing upon recruitment at the reporter for 5 days, albeit weaker than KRAB (FIG. 2B), a known strong repressor used in CRISPRi. Although KRAB can strongly silence nearly all (97.9%) of the cells, antiHP1 and antiDNMT1-mediated silencing were weaker, resulting in 16.8% and 28.5% of cells silenced, respectively (FIG. 2B). Silencing by antiHP1was weaker than GFP-tagged HP1 proteins when mediated by the anti-GFP nanobody (16.8% vs. 64% to 87% depending on the HP1 isoform recruited, FIG. 1D). This difference in silencing was not due to low levels of endogenous HP1 in the cell, as overexpression of HP1Į did not enhance silencing by the antiHP1 nanobody (FIG. 11F). [00216] To study the effects of antiDNMT1 recruitment on DNA methylation, the CpG methylation state of the reporter was analyzed via targeted bisulfite sequencing. Due to antiDNMT1’s weak silencing of the reporter after 5 days of dox, silenced cells (TagRFP- negative; +dox OFF) were sorted from non-silenced (TagRFP-positive, +dox ON) cells (FIG. 2A) to measure any differences in methylation between these two cell populations. Recruitment of antiDNMT1 resulted in higher levels of methylated CpGs at the reporter in the sorted OFF population when compared to the ON population in the dox-treated cells, to the no dox cells (FIG. 2C), and to the negative controls at the actin locus and unmethylated DNA (FIG. 12B). The DNA methylation levels with antiDNMT1 were comparable to direct DNMT1 recruitment (FIG. 2C), but lower than those obtained after recruitment of DNMT3B. Overall, these results show that silencing of the reporter by antiDNMT1 is associated with methylation of CpG DNA. In addition, treatment of the cells with 5-aza-2’- deoxycytidine (5-aza-2’), a DNA methyltransferase inhibitor, during recruitment abolished antiDNMT1 mediated silencing but not KRAB or antiHP1 (FIG. 12C), indicating that DNA methylation is required for antiDNMT1- mediated silencing. On the other hand, treatment with chaetocin, a broad-spectrum inhibitor of lysine histone methyltransferases, reduced all three effectors’ ability to silence (FIG. 12D), suggesting that histone methylation is involved in both antiHP1- and antiDNMT1-mediated silencing. [00217] After recruiting the effectors for 5 days with dox, the cells were released from dox and the epigenetic memory was assessed by measuring the percentage of cells still silenced over 30 days with flow cytometry (FIG. 2D). Silencing mediated by antiHP1 was almost completely reversible (FIG. 2E; orange line), consistent with previous reports that silencing by HP1 is associated with limited and partial memory, with the majority of the cells reactivating gene expression over time. In contrast, KRAB and antiDNMT1 led to more permanent memory, with about 50% and 65% cells irreversibly silenced out of the total initially silenced, respectively (FIG. 2E; blue and purple lines). These results demonstrate use of a nanobody that can mediate gene silencing and impart epigenetic memory. [00218] In addition to testing the nanobodies individually, the two nanobodies were fused together with a short flexible linker to see if this would enhance gene silencing and memory at the reporter gene (FIG. 12E). Recruitment of rTetR-antiDNMT1-antiHP1 led to about 80% of the cells being silenced, which was more than each nanobody individually (FIG. 2B). Moreover, the antiDNMT1-antiHP1 fusion had improved epigenetic memory over KRAB, with 61.4% cells still silent at 30 days after dox removal vs. 48.6% cells, respectively (FIG. 2E; green vs. blue line). These results show that combining two nanobodies that bind different CRs can be used to enhance gene silencing as well as epigenetic memory and demonstrate the use of nanobodies in combination with other CRs to improve transcriptional control. [00219] The effectiveness of these nanobodies at silencing, when transiently expressed, was tested in the TagRFP reporter cell line. AntiHP1 and antiDNMT1 led to weak silencing of the reporter after 5 days of dox treatment (FIG. 13A). Recruitment of antiDNMT1-antiHP1 led to about 55% of the cells being silenced, which is more than each nanobody individually (FIG. 13A similar to the stable expression results (FIG. 2B). To measure epigenetic memory independent of silencing efficiency, the silenced (TagRFP-negative) cells were sorted at the end of 5 days of dox treatment and measure their persistence of silencing for 30 days. Similar to stable expression, the antiDNMT1-antiHP1 fusion had improved epigenetic memory over KRAB, with 35.4% cells still silent at 30 days post sorting vs. 15% cells, respectively (FIG. 13B). Transient expression of nanobodies can also lead to reporter silencing and impart epigenetic memory, but less efficiently than stable expression. Example 3 Recruitment of antiDNMT1 improves silencing speed and epigenetic memory of other CRs at the reporter. [00220] The antiDNMT1 nanobody was tested in combination with KRAB (FIG. 14A). Transient expression and recruitment of rTetR-KRAB-antiDNMT1 to the reporter for 5 days resulted in strong silencing (87.8%), on the same level as KRAB alone (88.9%) (FIG. 3A) but demonstrated improved memory (FIG. 3B; pink vs. blue line). Epigenetic memory of KRAB when transiently expressed (FIG. 3B; blue line, 15%) was lower than in cell lines stably expressing KRAB (FIG. 2E; blue line, 48.6%), presumably due to the dilution of the plasmid. The further addition of the antiHP1 nanobody to the KRAB-antiDNMT1 fusion did not increase silencing or memory further (FIGS. 3A and 3B; brown). [00221] Moreover, when targeting rTetR-KRAB and rTetR-antiDNMT1 simultaneously at the reporter as stably expressed separate fusions, epigenetic memory was even lower than with KRAB alone (FIG. 14B). A similar decrease in memory and silencing was measured upon co- recruitment of separate fusions of rTetR with antiHP1 and antiDNMT1 (FIG. 14B vs. FIG. 2E). The observed reductions in epigenetic memory and silencing may be due to effector binding competition at the five TetO sites and suggest that direct fusions of multiple CRs might be preferable to independent co-recruitment at a locus for gene transcriptional control. [00222] The KRAB-antiDNMT1 fusion was tested with the dCas9 system at the reporter gene (FIG. 14C). Single-guide RNAs (sgRNAs) were designed to target the 5× TetO-binding site upstream of the reporter and a genomic site with no annotated function to serve as a negative control (called safe-targeting guide). When the programmable dCas9-KRAB-antiDNMT1 was targeted to the reporter gene for 5 days, there was strong silencing (FIG. 3C) and improvement in memory over dCas9-KRAB (FIG. 3D; 27.2% vs. 18.1%). Recruitment of dCas9-KRAB- antiDNMT1 demonstrated improved memory over dCas9-KRAB and over a combined repressor, dCas9-KRABMeCP2, (FIG. 3D; 27.2% vs. 17.5%). KRABMeCP2 had the same memory as KRAB alone (FIG. 3D; 18.1% vs. 17.5%) and the addition of antiDNMT1 to this fusion resulted in a similar improvement in memory as when added to KRAB (FIG. 3D; 33% for KRAB- MeCP2-antiDNMT1 vs. 27.2% for KRAB-antiDNMT1). These results show that the antiDNMT1 nanobody can be used to enhance the permanent silencing ability of CRs. When the dCas9-KRABantiDNMT1 fusion was targeted to the CXCR4 endogenous gene, there was no observed increase of epigenetic memory compared to dCas9-KRAB alone (FIGS. 14D and 14E). Although the level of memory seen after rTetR-KRAB-antiDNMT1 recruitment at the reporter is smaller than previously observed with the triple combination KRAB-DNMT3A-Dnmt3L5,7, KRAB-antiDNMT1 is about three times smaller in size (FIG. 14A; ~580 bp vs. ~1770 bp) and thus may be a more suitable tool for viral-based methods. [00223] Realizing the potential of the antiDNMT1 nanobody in improving the gene repressive effects of KRAB, combining the nanobody with the catalytic domain of DNMT3A was tested to determine if it would enhance it as well. When the rTetR-antiDNMT1-DNMT3A fusion was recruited to the reporter gene via rTetR for 5 days, it led to stronger and faster silencing when compared to DNMT3A alone (FIGS. 3E and 3F; dark green vs. light green). In addition, it has been shown that DNMT3L can enhance the catalytic activity of DNMT3A. Consistent with previous work, the addition of the C-terminal domain of mouse Dnmt3L and the catalytic domain of DNMT3A enhanced silencing of the reporter from 35.6 to 76 percent (FIG. 3E; light blue). Surprisingly, the addition of the smaller antiDNMT1 nanobody to DNMT3A led to a similar improvement in silencing as the larger Dnmt3L domain (FIGS. 3E and 3F; dark green vs. light blue). The antiDNMT1 nanobody further improved silencing when added to the DNMT3A- 3L fusion (FIGS. 3E and 3F; dark blue vs. light blue). In fact, of the different rTetR fusion combinations tested, the antiDNMT1-DNMT3A-3L triple fusion was by far the strongest (FIG. 3E; dark blue) resulting in about 87% of the cells being silenced at 5 days of dox. In summary, the antiDNMT1 nanobody improved the speed of silencing in all combinations with DNMT3A (FIG. 3F). All fusions containing rTetR-DNMT3A, including the ones containing antiDNMT1, led to permanent epigenetic memory at the reporter gene (FIG. 15A). A similar increase in the speed of silencing of the reporter gene was seen when antiDNMT1 was fused to the HDAC enzyme HDAC4 (FIG. 15B). Example 4 Nanobody-mediated recruitment of CRs for synthetic circuit control. [00224] These nanobody-based tools for controlling gene expression and epigenetic memory may be suitable to serve as devices in synthetic circuits for detecting and recording signals. Cellular stopwatches and recording devices are important components of synthetic biology circuits. The response of the antiDNMT1 nanobody presents a unique opportunity of implementing a very compact stopwatch that can both measure and record the duration of a signal. The desired signal can be coupled to the expression of rTetR-antiDNMT1, which in turn can be recruited upstream of an output gene encoding for fluorescence, signaling molecules, or proteins involved in cell death or survival (FIG. 4A). The addition of dox starts the time recording session, while removal of dox ends it. As silencing by antiDNMT1 recruitment is quite slow, it leads to a linear response in the percentage of cells silent as a function of signal duration (FIG. 4B). Moreover, as antiDNMT1 recruitment leads to permanent memory in the majority of cells silenced, the signal duration can be recorded as a percentage of cells in the population that have the output gene off (FIG. 4C). Coupling antiDNMT1 expression or recruitment to another cellular signal (such as a cytokine or a hormone) allows one to measure and permanently record the total duration of that signal as the fraction of cells with the reporter silenced. [00225] The linear response to signal duration and its recording in the fraction of irreversibly silent cells can be described using a 3-state model of gene control (FIG. 4D). In this model, active cells (A) silence at a slow rate (kS) during recruitment by antiDNMT1. They first reach a reversible silent state (R) and can transition from this to an irreversibly silent state (I) with a rate kI. After release of the nanobody, the reversibly silent population reactivates at a rate kA, while the irreversibly silent cells remain silent. Experimentally, a delay was observed between the addition of dox and the onset of silencing and was incorporated in the model as an additional parameter: Tlag (“Methods”). By fitting the experimental data for antiDNMT1 silencing and reactivation of the 14- day time course in FIG. 4C (“Methods”), the three rates, the lag time before onset of silencing, and their 95% confidence intervals were extracted: kS = 0.11/day (0.1– 0.12), kA = 0.29/day (0.12–0.45), kI = 0.37/day (0.31–0.43), Tlag = 1.7 days (1.3–2.0). As the exponential rate of silencing was quite low, it resulted in an approximately linear fraction of cells silenced as the duration of signal (dox recruitment) increases (FIGS. 4B and 4C, red dots). Similarly, the fraction of irreversibly silent cells that recorded signal duration looked approximately linear with signal duration for a large range of signals (FIG. 4E). [00226] Once calibrated, the three-state phenomenological model can be used to predict the fraction of cells silent over time for different types of signals without changing the three transition rates and initial lag time. For example, the model predicted that the fraction of cells silenced at the end of a 5-day pulsed signal (3 days of dox, 2 days of no dox, and then 2 days of dox) was approximately the same as at the end of a continuous 5-day signal (5 days of dox and then 2 days of no dox), matching experimental data (FIG. 4F). This model also predicted that continuous signals result in a small but systematically higher level of cells permanently silenced compared to interrupted signals of the same total duration, which was also consistent with experimental data (FIG. 4G). For pulsed signals there are two start times of dox addition, so there are two lag time values. For all the model predictions in FIGS. 4F and 4G, the first lag time was set to the value obtained from fitting the 14-day continuous time course in FIG. 4C (Tlag1 = 1.7), but it was assumed that there is no delay between dox addition and silencing on the second dox pulse (Tlag2 = 0). Although this assumption slightly overestimates the percentage of cells silent in the pulsed experiments, it led to predictions that are closer to experimental data than setting the two lag times equal (FIG. 16). The reduction in the second lag time suggests an intricate interplay between DNA methylation and silencing: it is possible that residual methylation from the first dox pulse allows onset of silencing to happen faster on the second pulse (Tlag2 < Tlag1). This type of modeling finds use to program the fraction of cells active or silenced for synthetic biology applications. Example 5 High-throughput development and characterization of new functional nanobodies for gene regulation and epigenetic control in human cells [00227] A high-throughput method for selecting nanobodies that can silence gene expression in human cells was developed. This method (FIG. 5) combines yeast display with HT-recruit in mammalian cells (Tycko J. et al., Cell. 2020 Dec 23;183(7):2020-2035, incorporated herein by reference in its entirety). A yeast display against a chromatin regulator (CR) that is known to act as a repressor was used to reduce library size to about ~104 sequences. This library was recloned into a lentiviral vector, and lentiviral infection was used to deliver the pooled enriched library to human cells. [00228] A nanobody sequence was identified (antiDNMT3A NB#8). antiDNMT3A NB#8 acted as a strong gene silencer when fused to rTetR in multiple cell types (FIG. 7), silenced a reporter gene when fused to dCas9 (FIG. 8), and imparted epigenetic memory (FIG. 9). [00229] The silencing ability of nanobodies can be measured in a high-throughput manner in human cells (FIG. 10), and this method was used to select for nanobodies against the DNMT3A complex that can silence gene expression. From the high-throughput measurement in human cells, of the approximately 3000 nanobodies obtained at the end of three rounds of yeast display against DNMT3A and lentiviral delivery in human cells, only about ~60 (2%) had a strong effect on gene expression. Besides NB#8, several other individual nanobodies (NB#25, #26, #27, #28) that were identified in the high-throughput screening were validated as strong gene silencers (FIG. 10). [00230] This method allowed selection of nanobodies that can perform the desired silencing function in human cells. While many nanobodies can bind an endogenous repressive complex, few silence in human cells. For example, in a small-scale test, only 1/24 nanobodies that bound DNMT3A silenced a reporter gene in the majority of cells (Table 2, FIG. 7). In contrast, 5/5 nanobodies chosen from the top hits from the functional silencing screen in human cells (Table 3, FIG. 10A) silenced >90% of the cells (FIG. 10C). Table 2: antiDNMT3A nanobodies
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Table 3: Exemplary individual anti-DNMT3A nanobodies recovered from the disclosed high-throughput screen, as shown by SEQ ID NO encoding each of 3 CDRs Each CDR is separated by ‘ ’
Figure imgf000071_0002
Figure imgf000072_0001
Figure imgf000072_0002
Figure imgf000072_0003
Figure imgf000073_0001
Figure imgf000073_0002
Figure imgf000073_0003
Figure imgf000074_0001
Figure imgf000074_0002
Figure imgf000074_0003
Figure imgf000075_0001
Figure imgf000075_0002
Figure imgf000075_0003
Figure imgf000076_0001
Figure imgf000076_0002
Figure imgf000076_0003
Figure imgf000077_0005
Figure imgf000077_0003
Figure imgf000077_0004
Figure imgf000077_0001
Table 5: Nanobodies selected against TET1/2/3 complexes
Figure imgf000077_0002
Figure imgf000078_0001
Figure imgf000079_0001
[00231] All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein. [00232] Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

Claims

CLAIMS What is claimed is: 1. A method for identifying and selecting nanobodies for a target chromatin regulator comprising: a) preparing a yeast nanobody library comprising yeast cells each displaying a single nanobody on its surface; b) mixing the yeast nanobody library with a target chromatin regulator wherein the target chromatin regulator is configured to bind to a solid surface; c) separating yeast cells bound to target chromatin regulator from unbound yeast cells; d) amplifying nanobody DNA sequences from the yeast cells bound to target chromatin regulator; e) cloning the nanobody sequences into a human lentiviral vector to prepare a nanobody lentiviral library, wherein each nanobody DNA sequence is expressed as a fusion protein with a DNA binding domain; f) transforming the nanobody lentiviral library into reporter cells, wherein a reporter cell comprises a two-part reporter gene comprising a surface marker and a fluorescent protein, wherein the two-part reporter gene is capable of being silenced or induced by the target chromatin regulator; g) separating the reporter cells based on presence or absence of the surface marker, the fluorescent protein, or a combination thereof; and h) identifying the nanobody as a binding protein for the target chromatin regulator and capable of regulating gene expression.
2. The method of claim 1, further comprising repeating steps a-d one or more times.
3. The method of claim 1 or 2, wherein the chromatin regulator comprises a methylase, a demethylase, an acetylase, or ATP-dependent chromatin remodeling complex.
4. The method of any of claims 1-3, wherein the DNA binding domain is an inducible DNA binding domain and the method further comprising treating the reporter cells with an agent configured to induce the inducible DNA binding domain for a length of time.
5. The method of any of claims 1-4, wherein the two-part reporter gene is under the control of a strong promoter and capable of being silenced by a functional chromatin repressor.
6. The method of claim 5, wherein the chromatin regulator is a functional chromatin repressor when recruited by the nanobody due to enrichment of cells with an absence of the surface marker, the fluorescent protein, or a combination thereof.
7. The method of any of claims 1-4, wherein the two-part reporter gene is under the control of a weak promoter and capable of being induced by a functional chromatin activator.
8. The method of claim 7, wherein the chromatin regulator is a functional chromatin activator when recruited by the nanobody due to enrichment of cells with a presence of the surface marker, the fluorescent protein, or a combination thereof.
9. A fusion protein comprising a DNA binding protein, DNA binding domain, or a functional fragment thereof covalently linked to at least one nanobody configured to bind a chromatin regulator.
10. The fusion protein of claim 9, wherein one or more of the at least one nanobody is identified by a method as disclosed of any of claims 1-8.
11. The fusion protein of claim 9 or 10, wherein the at least one nanobody is configured to bind DNA (cytosine-5)-methyltransferase 3A (DNMT3A) complexes.
12. The fusion protein of claim 11, wherein the at least one nanobody comprises at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFXXXXM (SEQ ID NO: 13), EZVAXIXXGXXTNY (SEQ ID NO: 14), and AAXXXXXYYXXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid.
13. The fusion protein of claim 11 or 12, wherein the at least one nanobody comprises a first CDR comprising an amino acid sequence of GTIFXXXXM (SEQ ID NO: 13), a second CDR comprising an amino acid sequence of EZVAXIXXGXXTNY (SEQ ID NO: 14), and a third CDR comprising an amino acid sequence of AAXXXXXYYXXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid.
14. The fusion protein of any of claims 11-13, wherein the at least one nanobody comprises at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), GNIFDGASM (SEQ ID NO: 4), EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), AAGRYYYPGHGY (SEQ ID NO: 8), AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 12).
15. The fusion protein of any of claims 11-14, wherein the at least one nanobody comprises: a first CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), and GNIFDGASM (SEQ ID NO: 4); a second CDR comprising an amino acid sequence selected from the group consisting of EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), and AAGRYYYPGHGY (SEQ ID NO: 8); a third CDR comprising an amino acid sequence selected from the group consisting of AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 12); or a combination thereof.
16. The fusion protein of any of claims 11-15, wherein the nanobody comprises an amino acid sequence at least 70% similar to any of SEQ ID NOs: 16-20.
17. The fusion protein of claim 9 or 10, wherein the at least one nanobody is configured to bind ten-eleven translocation methylcytosine 1/2/3 (TET1/2/3) complexes.
18. The fusion protein of claim 17, wherein the at least one nanobody comprises at least one CDR having an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933- 5959.
19. The fusion protein of claim 17 or 18, wherein the at least one nanobody comprises: a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933-5941; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5942-5950; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5951-5959; or a combination thereof.
20. The fusion protein of any of claims 17-19, wherein the nanobody comprises an amino acid sequence at least 70% similar to any of SEQ ID NOs: 5960-5968.
21. The fusion protein of any of claims 9-20, further comprising a transcription factor or regulator.
22. The fusion protein of claim 21, wherein the transcription factor or regulator comprises Krüppel-associated box (KRAB).
23. A nucleic acid encoding the fusion protein of any of claims 9-22.
24. A DNA (cytosine-5)-methyltransferase 3A (DNMT3A) binding protein comprising at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFXXXXM (SEQ ID NO: 13), EZVAXIXXGXXTNY (SEQ ID NO: 14), and AAXXXXXYYXXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid.
25. The DNMT3A binding protein of claim 24, wherein the DNMT3A binding protein comprises a first CDR comprising an amino acid sequence of GTIFXXXXM (SEQ ID NO: 13), a second CDR comprising an amino acid sequence of EZVAXIXXGXXTNY (SEQ ID NO: 14), and a third CDR comprising an amino acid sequence of AAXXXXXYYXXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid.
26. The DNMT3A binding protein of claim 24, wherein the at least one CDR comprises an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), GNIFDGASM (SEQ ID NO: 4), EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), AAGRYYYPGHGY (SEQ ID NO: 8), AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), AND AAKPSRVYWRDYRFFY (SEQ ID NO: 12).
27. The DNMT3A binding protein of claim 26, wherein the DNMT3A binding protein comprises: a first CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), and GNIFDGASM (SEQ ID NO: 4); a second CDR comprising an amino acid sequence selected from the group consisting of EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), and AAGRYYYPGHGY (SEQ ID NO: 8); a third CDR comprising an amino acid sequence selected from the group consisting of AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 12); or a combination thereof.
28. The DNMT3A binding protein of any of claims 24-27, wherein the DNMT3A binding protein comprises an amino acid sequence at least 70% similar to SEQ ID NOs: 16-20.
29. The DNMT3A binding protein of any of claims 24-28, wherein the DNMT3A binding protein is an antibody or a fragment thereof.
30. The DNMT3A binding protein of any of claims 24-29, wherein the DNMT3A is a nanobody.
31. A nucleic acid encoding the DNMT3A binding protein of any of claims 24-30.
32. The nucleic acid of claim 31, wherein at least one CDR is encoded by a nucleic acid sequence of any of those disclosed in SEQ ID NOs: 21-5928.
33. A fusion protein comprising the DNMT3A binding protein of any of claims 24-30 covalently attached to a DNA binding protein, DNA binding domain, or a functional fragment thereof.
34. A TET1/2/3 complex binding protein comprising at least one CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs 5933-5959.
35. The TET1/2/3 complex binding protein of claim 34, wherein the TET1/2/3 complex binding protein comprises: a first CDR comprising an amino acid sequence selected from the group consisting of GSISYYSVM (SEQ ID NO: 5933), GNIFYEYNM (SEQ ID NO: 5934), GNISGPRGM (SEQ ID NO: 5935), GTIFIYSYM (SEQ ID NO: 5936), GYIFGYNSM (SEQ ID NO: 5937), GSIFSYSDM (SEQ ID NO: 5938), GSISYWWDM (SEQ ID NO: 5939), GSIFYYYEM (SEQ ID NO: 5940), and GTIFDEYYM (SEQ ID NO: 5941); a second CDR comprising an amino acid sequence selected from the group consisting of EFVAAISPGGITNY (SEQ ID NO: 5942), ELVATIDAGASTYY (SEQ ID NO: 5943), ELVAGIAYGSSTYY (SEQ ID NO: 5944), EFVAGISPGGSTNY (SEQ ID NO: 5945), ELVASIDGGGSTYY (SEQ ID NO: 5946), EFVAAINYGGNTNY (SEQ ID NO: 5947), EFVASISLGGNTNY (SEQ ID NO: 5948), EFVAGIDYGSTTYY (SEQ ID NO: 5949), EFVAAIARGTSTYY (SEQ ID NO: 5950); a third CDR comprising an amino acid sequence selected from the group consisting of AVRGIDSYYDWGYYYY (SEQ ID NO: 5951), AAYAYYWYGYIY (SEQ ID NO: 5952), AAAYDEPDYKDEVYEY (SEQ ID NO: 5953), AAENDFYPYYGADYYSSLYY (SEQ ID NO: 5954), AVDSYYWYHLY (SEQ ID NO: 5955), AAAGYDTTGWYDKRSYSYWY (SEQ ID NO: 5956), AVDPYSIGDYGYSYIADYLY (SEQ ID NO: 5957), AVWSDDDDVPYEDYYDYHAY (SEQ ID NO: 5958), and AAWDGSDYKFDY (SEQ ID NO: 5959); or a combination thereof.
36. The TET1/2/3 complex binding protein of claim 34 or 35, wherein the TET1/2/3 complex binding protein comprises an amino acid sequence at least 70% similar to any of SEQ ID NOs: 5960-5968.
37. The TET1/2/3 complex binding protein of any of claims 34-36, wherein the TET1/2/3 binding protein is an antibody or a fragment thereof.
38. The TET1/2/3 complex binding protein of any of claims 34-37, wherein the TET1/2/3 is a nanobody.
39. A nucleic acid encoding the TET1/2/3 complex binding protein of any of claims 34-38.
40. The nucleic acid of claim 39, wherein the nucleic acid comprises a sequence having at least 70% similar to any of SEQ ID NOs: 5969-5977.
41. A fusion protein comprising the TET1/2/3 complex binding protein of any of claims 34-38 covalently attached to a DNA binding protein, DNA binding domain, or a functional fragment thereof.
42. A nucleic acid encoding the fusion protein of claim 33 or 41.
43. The nucleic acid of claim 42, wherein the nucleic acid sequence comprises one or more of any of SEQ ID NOs: 21-5928.
44. The nucleic acid of claim 42, wherein the nucleic acid comprises a sequence having at least 70% similar to any of SEQ ID NOs: 5969-5977.
45. A vector encoding the nucleic acid of any of claims 23, 31-32, 39-40, and 42-44.
46. A cell comprising a fusion protein of any of claims 9-22, a nucleic acid of claim 23, 31-32, 39-40, and 42-44, or vector of claim 45.
47. The cell of claim 46, wherein the cell comprises two or more fusion proteins of any of claims 9-22, nucleic acids of claim 23, 31-32, 39-40, and 42-44, or vectors of claim 45.
48. A composition or system comprising one or more of: a fusion protein of any of claims 9-22, a nucleic acid of claim 23, 31-32, 39-40, and 42-44, a vector of claim 45, or a cell of claim 46 or 47.
49. The composition or system of claim 48, wherein the composition or system comprises two or more fusion proteins, nucleic acids, vectors, or cells.
50. The composition of claim 48 or 49, further comprising a transcription factor or regulator.
51. The composition of claim 50, wherein the transcription factor or regulator comprises a transcriptional repressor.
52. The composition of claim 50, wherein the transcription factor comprises Krüppel-associated box (KRAB).
53. The composition of any of claims 48-52, further comprising a chromatin regulator.
54. The composition of claim 53, wherein the chromatin regulator comprises a methylase, a demethylase, an acetylase, or ATP-dependent chromatin remodeling complex.
55. The composition of any of claims 48-54, further comprising a guide RNA or a nucleic acid encoding a guide RNA.
56. A method for modifying gene expression, epigenetic memory, or a combination thereof of at least one target nucleic acid comprising: contacting the target nucleic acid with at least one fusion protein comprising a DNA binding protein, DNA binding domain, or a functional fragment thereof covalently linked to at least one nanobody configured to bind a chromatin regulator.
57. The method of claim 56, wherein the at least one target nucleic acid is in a cell and the chromatin regulator is endogenous to the cell.
58. The method of claim 56 or 57, wherein the contacting comprises introducing into the cell the fusion protein, or a nucleic acid encoding thereof.
59. The method of claim 58, wherein the introducing comprises administering to a subject.
60. The method of any of claims 56-59, further comprising contacting the target nucleic acid with an exogenous chromatin regulator.
61. The method of any of claims 56-60, wherein the nanobody is identified or selected by a method as disclosed in any of claims 1-8.
62. The method of any of claims 56-67, wherein the endogenous or exogenous chromatin regulator comprises a methylase, a demethylase, an acetylase, or ATP-dependent chromatin remodeling complex.
63. The method of any of claims 56-62, wherein the chromatin regulator comprises DNA (cytosine-5)-methyltransferase 3A (DNMT3A).
64. The method of claim 63, wherein the at least one nanobody comprises at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFXXXXM (SEQ ID NO: 13), EZVAXIXXGXXTNY (SEQ ID NO: 14), and AAXXXXXYYXXXXXY (SEQ ID NO: 15), wherein Z is L or F and each X is any amino acid.
65. The method of claim 63 or 64, wherein the nanobody comprises at least one CDR comprising an amino acid sequence selected from the group consisting of GTIFAHSRM (SEQ ID NO: 1), GTISSDGYM (SEQ ID NO: 2), GTIFYFFGM (SEQ ID NO: 3), GNIFDGASM (SEQ ID NO: 4), EFVASIAYGGNTNY (SEQ ID NO: 5), ELVAAIAGGTITNY (SEQ ID NO: 6), ELVAGITPGAITNY (SEQ ID NO: 7), AAGRYYYPGHGY (SEQ ID NO: 8), AAGRYYYPGNGY (SEQ ID NO: 9), AATKYGFYYYSSHFY (SEQ ID NO: 10), AVVDFYDSVYYY (SEQ ID NO: 11), and AAKPSRVYWRDYRFFY (SEQ ID NO: 12).
66. The method of any of claims 63-65, wherein the nanobody comprises an amino acid sequence at least 70% similar to SEQ ID NOs: 16-20.
67. The method of any of claims 56-62, wherein the chromatin regulator comprises ten-eleven translocation methylcytosine 1/2/3 (TET1/2/3) complexes.
68. The method of claim 67, wherein the at least one nanobody comprises at least one CDR having an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933-5959.
69. The fusion protein of claim 67 or 68, wherein the at least one nanobody comprises: a first CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5933-5941; a second CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5942-5950; a third CDR comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 5951-5959; or a combination thereof.
70. The method of any of claims 67-69, wherein the nanobody comprises an amino acid sequence at least 70% similar to any of SEQ ID NOs: 5960-5968.
71. The method of any of claims 56-70, further comprising contacting the target nucleic acid with a transcription factor or regulator.
72. The method of claim 71, wherein the fusion protein further comprises the transcription factor or regulator.
73. The method of claim 71 or 72, wherein the transcription factor or regulator comprises a transcriptional repressor.
74. The method of any of claims 71-73, wherein the transcription factor comprises Krüppel- associated box (KRAB).
75. The method of any of claims 56-74, further comprising contacting the target nucleic acid with a chromatin regulator.
76. The method of any of claims 56-75, wherein the gene expression, epigenetic memory, or a combination thereof of at least two genes are modulated.
77. Use of the fusion protein of any of claims 9-22, or a composition or nucleic acid encoding thereof for modifying gene expression, epigenetic memory, or a combination thereof of at least one target nucleic acid.
78. The use of claim 77, wherein the at least one target nucleic acid is in a cell and the chromatin regulator is endogenous to the cell.
79. The method of claim 78, wherein the contacting comprises introducing into the cell the fusion protein, or composition or a nucleic acid encoding thereof.
PCT/US2022/076208 2021-09-10 2022-09-09 Nanobody-mediated control of gene expression and epigenetic memory WO2023039528A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163242898P 2021-09-10 2021-09-10
US63/242,898 2021-09-10

Publications (1)

Publication Number Publication Date
WO2023039528A1 true WO2023039528A1 (en) 2023-03-16

Family

ID=85507729

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/076208 WO2023039528A1 (en) 2021-09-10 2022-09-09 Nanobody-mediated control of gene expression and epigenetic memory

Country Status (1)

Country Link
WO (1) WO2023039528A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110182897A1 (en) * 2008-06-05 2011-07-28 Ablynx N.V. Amino acid sequences directed against envelope proteins of a virus and polypeptides comprising the same for the treatment of viral diseases
WO2020033585A1 (en) * 2018-08-07 2020-02-13 The Broad Institute, Inc. Methods for combinatorial screening and use of therapeutic targets thereof
US20200282011A1 (en) * 2015-03-02 2020-09-10 Inserm (Institut National De La Sante Et De La Recherche Medicale) Method for treating and prognosing cancer
US20210163923A1 (en) * 2017-12-06 2021-06-03 Iontas Limited Selecting for developability of polypeptide drugs in eukaryotic cell display systems
US20210246494A1 (en) * 2008-09-26 2021-08-12 The Children's Medical Center Corporation Methods of Epigenetic Analysis

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110182897A1 (en) * 2008-06-05 2011-07-28 Ablynx N.V. Amino acid sequences directed against envelope proteins of a virus and polypeptides comprising the same for the treatment of viral diseases
US20210246494A1 (en) * 2008-09-26 2021-08-12 The Children's Medical Center Corporation Methods of Epigenetic Analysis
US20200282011A1 (en) * 2015-03-02 2020-09-10 Inserm (Institut National De La Sante Et De La Recherche Medicale) Method for treating and prognosing cancer
US20210163923A1 (en) * 2017-12-06 2021-06-03 Iontas Limited Selecting for developability of polypeptide drugs in eukaryotic cell display systems
WO2020033585A1 (en) * 2018-08-07 2020-02-13 The Broad Institute, Inc. Methods for combinatorial screening and use of therapeutic targets thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
VAN MIKE V., FUJIMORI TAIHEI, BINTU LACRAMIOARA: "Nanobody-mediated control of gene expression and epigenetic memory", NATURE COMMUNICATIONS, vol. 12, no. 1, XP093047116, DOI: 10.1038/s41467-020-20757-1 *

Similar Documents

Publication Publication Date Title
AU2021203547B2 (en) Human mesothelin chimeric antigen receptors and uses thereof
JP2023524733A (en) Compositions, systems and methods for creating, identifying and characterizing effector domains for activating and silencing gene expression
Gottesman et al. The molecular basis of multidrug resistance in cancer: the early years of P-glycoprotein research
DK2971098T3 (en) QUANTITATIVE ASSESSMENT FOR CAPE EFFECTIVENESS OF MESSENGER RNA
CN107794267B (en) Bispecific aptamer targeting PD1-PDL1 and derivative thereof
CN105924519A (en) Comprehensive Monoclonal Antibody Generation
US10895569B2 (en) Genome-scale T cell activity array and methods of use thereof
WO2018071576A1 (en) Treatment of tumors by inhibition of cd300f
CN116234558A (en) Engineered T cells conditionally expressing recombinant receptors, related polynucleotides and methods
US20190111114A1 (en) Methods for identifying and treating hemoglobinopathies
Lim et al. Engineering high affinity protein–protein interactions using a high-throughput microcapillary array platform
KR20220152227A (en) BCA-directed chimeric antigen receptor T cell composition and methods and uses thereof
WO2023023553A2 (en) Compositions, systems, and methods for activating and silencing gene expression
WO2023039528A1 (en) Nanobody-mediated control of gene expression and epigenetic memory
Bosso et al. IFI16 knockdown in primary HIV-1 target cells
Feldman et al. Steering tumor progression through the transcriptional response to growth factors and stroma
Zhang et al. Efficient genetic engineering of murine cochlear organoids
Luteijn et al. SLC19A1 is a cyclic dinucleotide transporter
WO1999006592A9 (en) Separation of nucleic acids by antibodies to halogenated nucleotides
Šimonová Interaction of a surface marker of immune cells with low-molecular weight ligands and their polymer conjugates
JP2023526278A (en) Methods for Producing Donor Batch Cells Expressing Recombinant Receptors
Ajayi Reprogramming of Sterol Biosynthesis in Chinese Hamster Ovary Cells for Enhanced Recombinant Protein Production
Minguet et al. Harnessing CD3 diversity to optimise CAR T cells
WO2022165267A1 (en) Method for making car-t libraries
Hsu Co-opted Functions of the SIX1 Homeobox Transcription Factor Maintain Myoblastic Programs in Fusion-Negative Rhabdomyosarcoma

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22868341

Country of ref document: EP

Kind code of ref document: A1