CRISPR-Cas9 editing of synaptic genes in human embryonic stem cells for functional analysis in induced human neurons (2024)

  • Journal List
  • STAR Protoc
  • v.5(2); 2024 Jun 21
  • PMC11152723

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (1)

Link to Publisher's site

STAR Protoc. 2024 Jun 21; 5(2): 103089.

Published online 2024 May 24. doi:10.1016/j.xpro.2024.103089

PMCID: PMC11152723

PMID: 38795356

Aiden Houcek,1,2,3,4,5 Z. Zack Ma,1,2,4, Brent Trauterman,1,2 Burak Uzay,1,2 Lisa M. Monteggia,1,2 and Ege T. Kavalali1,2,6,∗∗

Author information Copyright and License information PMC Disclaimer

Associated Data

Data Availability Statement

Summary

Generating stable human embryonic stem cells (hESCs) with targeted genetic mutations allows for the interrogation of protein function in numerous cellular contexts while maintaining a relatively high degree of isogenicity. We describe a step-by-step protocol for generating knockout hESC lines with mutations in genes involved in synaptic transmission using CRISPR-Cas9. We describe steps for gRNA design, cloning, stem cell transfection, and clone isolation. We then detail procedures for gene knockout validation and differentiation of stem cells into functional induced neurons.

Subject areas: CRISPR, neuroscience, stem cells

Graphical abstract

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (2)

Open in a separate window

Highlights

  • Protocol for custom gRNA design and assembly into LentiCRISPR v2

  • Transient transfection and single-cell cloning of hESCs

  • Validation of gene editing and individual allele screening of hESCs

  • Steps for the induction of WT and edited hESCs into functional neurons using neurogenin-2

Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.

Generating stable human embryonic stem cells (hESCs) with targeted genetic mutations allows for the interrogation of protein function in numerous cellular contexts while maintaining a relatively high degree of isogenicity. We describe a step-by-step protocol for generating knockout hESC lines with mutations in genes involved in synaptic transmission using CRISPR-Cas9. We describe steps for gRNA design, cloning, stem cell transfection, and clone isolation. We then detail procedures for gene knockout validation and differentiation of stem cells into functional induced neurons.

Before you begin

The protocol detailed below describes a methodology to generate gene knockout hESCs that can be differentiated into induced human neurons (iNs) for functional studies of synapses. While hESCs are used in this protocol, users may also apply this gene editing strategy to alternative pluripotent cell lines such as iPSCs. The targeted genes of interest to knockout using this protocol are ideally expressed in induced neurons and therefore have minimal effects on hESC survival, proliferation, and differentiation. The use of CRISPR/Cas9 provides a means to generate precise genomic edits in mammalian cells.1 Wedescribe here the transient plasmid expression of Cas9 and gRNAs to almost eliminate the possibility of constitutive expression due to transgene integration, which can introduce unspecific off-target editing of the genome. Subsequent induction of edited hESCs into iNs also preserves the functional deficits associated with the hESC gene knockout of protein production in human neurons,2 demonstrating the variety of downstream applications this technology may be employed to investigate.

Institutional permissions

Users of this protocol will require institutional permission to use human embryonic stem cells and laboratory animals. All animal procedures were performed in accordance with the guide for the care and use of laboratory animals and approved by the Institutional Animal Care and Use Committee at Vanderbilt University.

gRNA design

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (3)Timing: 1 h

Proper gRNA design is crucial for genome targeting specificity and subsequent Cas9 cleavage of the desired gene target. This section explains how to design gRNAs that can be rapidly assembled into a LentiCRISPR v2 plasmid.

  • 1.

    Import mRNA sequence (GenBank: NM_XXXX), and each exon FASTA seq (GenBank: NM_XXXX exons) of a gene of interest from the NCBI database to Benchling.com.

    • a.

      Use Benchling integrated gRNA design program to design gRNAs targeting the exon containing the coding sequence (CDS).

Note: gRNAs should target regions immediately downstream of the CDS containing exon.

  • 2.

    Select up to 4 gRNAs (20 nucleotide length) based on the on-target and off-target scores,3 targeting different regions downstream of the CDS.

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (4)CRITICAL: Selecting gRNAs with on-target and off-target scores of 80 or higher in the program is essential for target specificity and successful genome editing.

  • 3.

    For gRNA cloning into LentiCRISPR v2, the sense oligonucleotide sequence is CACC (overhang for golden gate assembly), G (transcription start site), and N20 (gRNA target).4

    Note: Example: CACCG (CCCTCGGGGCGCTGTGCTGT)

    • a.

      The antisense oligo is AAAC (overhang for golden gate assembly), N20 (gRNA reverse complement), and C (antisense of the transcription start site).

      Note: Example: AAAC (ACAGCACAGCGCCCCGAGGG) C.4

      Alternatives: Using Benchling for gRNA design is a very effective approach in our practice. Besides Benchling, we do not have experience working with other sequence analysis and gRNA design tools. Still likely, using those tools could also work as an alternative approach for this purpose. Users can make their own decisions.

Cell culture

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (5)Timing: 1month

These steps are general instructions for thawing and passaging human embryonic kidney (HEK) cells and hESCs and do not need to be maintained throughout this protocol. Ideally, hESCs or HEK cells are thawed and passaged at least two times before subsequent transfection.

hESC culture

  • 4.

    Coat a 6-well plate with Matrigel dissolved in DMEM/F12 (100ng/mL) and incubate at 37°C for a minimum of 2 h, but ideally 12h for optimal coating.

  • 5.

    Thaw hESCs rapidly in a 37°C water bath and gently transfer cells to a 15mL conical tube.

  • 6.

    Centrifuge the thawed hESCs (∼ 1×106 cells) for 5min at 300×g.

  • 7.

    Aspirate freezing media and resuspend the pellet in 6mL of mTeSR Plus.

  • 8.

    Aspirate Matrigel solution from 6-well plates and plate all cells in three wells of a 6-well plate.

Note: Thawing hESCs with ROCK pathway inhibitor (ROCKi) will increase single-cell survival following thawing and plating. Using ROCKi is highly recommended for enhancing hESC survival but is not essential for hESC survival during thawing and plating.

Note: WiCell Stem Cell Bank provides a good resource for the detailed protocols for hESC culture and passaging (https://www.wicell.org/stem-cell-protocols.cmsx).

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (6)CRITICAL: Incomplete thawing of hESCs at 37°C before plating will decrease cell viability.

  • 9.

    Change media to fresh mTeSR Plus every 48h and passage hESCs at approximately 70%–80% confluence using ReLesR dissociation reagent.

  • 10.

    To passage hESCs, aspirate mTeSR Plus from each well and rinse with 1mL of PBS.

  • 11.

    Add 1mL of ReLesR to one well and let sit for 1min at 20°C.

  • 12.

    Aspirate approximately 0.9mL of the ReLesR from the well and incubate at 37°C for 7–10min.

  • 13.

    Add 2mL mTeSR Plus to each well of a 6-well plate to collect dissociated cells. Add 10mL mTeSR Plus to 2mL cell suspension (1: 6 dilution).

  • 14.

    Transfer 12mL of cells to a new Matrigel-coated 6-well plate (2mL per well).

HEK293T cell culture

  • 15.

    Culture HEK293T cells in DMEM+ media and passage at 80% confluence using 0.25% Trypsin EDTA.

  • 16.

    To passage HEKs using 0.25% Trypsin-EDTA, aspirate the culture media and rinse the cells with PBS.

  • 17.

    Add 5mL of Trypsin-EDTA to a T75 culture flask or 1mL to one well of a 6-well plate and incubate for 7–10min at 37°C.

  • 18.

    Collect dissociated cells in Trypsin-EDTA and centrifuge for 5min at 300×g.

  • 19.

    Aspirate the Trypsin-EDTA and resuspend the pellet in 1mL of fresh DMEM+. Plate approximately 0.1mL of this cell suspension with 10mL of fresh media in a T75 flask.

Key resources table

REAGENT or RESOURCESOURCEIDENTIFIER
Bacterial and virus strains
NEB DH5α competent E.coliNEBCat# C2987
Chemicals, peptides, and recombinant proteins
Y-23579 (ROCKi)STEMCELLCat# 72304
Cytosine arabinoside (Ara-C)SigmaCat# C1768
MatrigelCorningCat# 354234
Recombinant human/murine/rat BDNFPeproTechCat# 45002
Recombinant human NT-3PeproTechCat# 45003
Mouse lamininInvitrogenCat# 23017015
B27 supplementGibcoCat# 17504-010
MEM non-essential amino acid solutionGibcoCat# 11140050
GlutaMAX supplementGibcoCat# 35050061
PuromycinSigmaCat# P8833
DoxycyclineSigmaCat# D9891
Penicillin-StreptomycinGibcoCat# 15140122
Neurobasal Plus mediaGibcoCat# A3582901
PBSCorningCat# 21031-CV
DMEM, high glucoseGibcoCat# 10569044
DMEM/F12 mediaInvitrogenCat# 11330057
Opti-MEMThermo Fisher ScientificCat# 31985062
CloneR2STEMCELLCat# 100–0691
AccutaseGibcoCat# A1110501
N-2 supplementGibcoCat# 17502048
NEAA supplementThermo Fisher ScientificCat# 11140076
Polybrene infection/Transfection reagentSigmaCat# TR-1003-G
ReLeSRSTEMCELLCat# 100–0483
mFreSRSTEMCELLCat# 05854
Fetal bovine serum (FBS)Fisher ScientificCat# SH30070.03 Hyclone
mTeSR Plus basal mediumSTEMCELLCat# 100–0276
Trypsin-EDTA (0.25%), Phenol redGibcoCat# 25200072
Cellartis DEF-CS 500 culture systemTakaraCat# Y30010
LB Agar+ Ampicillin platesGibcoCat# J63197.EQF
AgaroseThermo Fisher ScientificCat# 16500500
dNTP MixThermo Fisher ScientificCat# R0192
Critical commercial assays
DNeasy Blood and Tissue KitQIAGENCat# 69504
PCR cleanup systemPromegaCat# A9281
Plasmid Mini KitQIAGENCat# 12123
Plasmid Midi KitQIAGENCat# 12143
Lipofectamine 3000 transfection reagentThermo Fisher ScientificCat# L3000001
Fugene6 transfection reagentPromegaCat# E2691
T4 polynucleotide kinaseNEBCat# M0201S
T4 ligaseNEBCat# M0202S
ATP (10mM)NEBCat# P0756S
Q5 DNA polymeraseNEBCat# M0491
T4 endonuclease INEBCat# M0302
T7 endonuclease INEBCat# M0302S
NEBuffer 2NEBCat# B7002S
rCutSmart bufferNEBCat# B6004S
EcoRVNEBCat# R0195
Esp3INEBCat# R0734S
HiFi DNA assembly reaction kitNEBCat# E2621
10× Klentaq1 reaction bufferKlentaqCat# RB20
Klentaq LAKlentaqCat# 110
DNA polymeraseKlentaqCat# 110
QIAquick Gel Extraction KitQIAGENCat# 28704
Exo-CIP Rapid PCR Cleanup kitNEBCat# E1050
Experimental models: Cell lines
Human embryonic stem cellsWiCellCat# WA01
HEK293T cellsATCCCat# CRL-3216
Experimental models: Organisms/strains
CD1 miceCharles RiverStrain code: 022
Oligonucleotides
LentiCRISPR v2 primer: FWD
GAGCCAATTCCCACTCCTTTCAAG
This paperN/A
pBluscript EcoRV overhang sequence FWD
CGGTATCGATAAGCTTGAT
This paperN/A
pBluscript EcoRV overhang sequence REV
GCTGCAGGAATTCGAT
This paperN/A
Recombinant DNA
pRSV-REV (lentiviral packaging)AddgeneCat# 12253
pMD2.G (lentiviral packaging)AddgeneCat# 12259
pMDLg/pRRE (lentiviral packaging)AddgeneCat# 12251
pFUW-TetO-hNgn2-EGFP-PuroRAddgeneCat# 79823
lentiCRISPR v2AddgeneCat# 52961
pBlueScript SK(+)NovoProCat# V011755
Software and algorithms
BenchlingBenchling.comN/A

Open in a separate window

Materials and equipment

Prepare cell culture media and aliquot reagents.

  • mTesR Plus: Add 100mL 5X supplement to 400mL basal media). Store at 4°C for up to 1month.

  • mTesR Plus+ CloneR2: Dilute 100X stock to 1X. Store at 4°C for up to 1month.

  • DMEM+ (Supplemented with FBS and Pen-strep at a final concentration of 10% and 1%, respectively). Store at 4°C for up to 1month.

  • Matrigel. Aliquot as supplied and stored at −80°C for up to 3months.

  • Doxycycline, BDNF, NT-3, Laminin, and AraC stock solutions.

    • Doxycycline: Dissolve in sterile water to make 1mg/mL (500X), and store at −20°C for up to 6months. Protect from light.

    • BDNF and NT3: Dissolve in sterile 0.1% bovine serum albumin in PBS to make 10μg/mL (1000X). Store at −80°C for up to 3months.

    • Laminin: Aliquot as supplied (1mg/mL, 5000X). Store at −80°C for up to 3months.

    • AraC: Dissolve in sterile water to make 4mM stock (4000X). Aliquot and store at −20°C for up to 6months.

Alternatives: Cellartis DEF-CS 500 culture system (Takara) may also be used as an alternative to mTesR Plus+ CloneR2 for single-cell isolation and expansion, but CloneR2 appears optimal in our experience for successful single-cell cloning of hESCs.

Neuronal induction media

ReagentFinal concentrationVolume
DMEM-F12N/A500mL
N21X5mL (100X)
NEAA1X5mL (100X)
Doxycycline2μg/μL1mL
BDNF10ng/μL0.5mL
NT-310ng/μL0.5mL
Laminin0.2μg/μL100μL

Open in a separate window

Store neuronal induction media at 4°C for up to 1month.

Neuronal selection media

ReagentFinal concentrationVolume
DMEM-F12N/A500mL
N21X5mL (100X)
NEAA1X5mL (100X)
Doxycycline2μg/μL1mL
BDNF10ng/μL0.5mL
NT-310ng/μL0.5mL
Laminin0.2μg/μL100μL
Puromycin1μg/μL0.5mL

Open in a separate window

Store neuronal selection media at 4°C for up to 1month.

Neurobasal media

ReagentFinal concentrationVolume
Neurobasal plus mediaN/A500mL
GlutaMAX1X5mL (100X)
B27 Supplement1X10mL (50X)
BDNF10ng/μL0.5mL
NT-310ng/μL0.5mL
Laminin0.2μg/μL100μL
Doxycycline2μg/μL0.5mL

Open in a separate window

Note: Add fresh recombinant proteins and Doxycycline/Puromycin to an aliquot of neural induction, selection, or neurobasal media directly before use.

Neuronal growth media

ReagentFinal concentrationVolume
Neurobasal plus mediaN/A500mL
GlutaMAX1X5mL (100X)
FBS5%25mL
B27 Supplement1X10mL (50X)
AraC1μM125μL (4mM)

Open in a separate window

Store neuronal growth media at 4°C for up to 1month.

Step-by-step method details

gRNA phosphorylation, annealing, and golden gate assembly

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (7)Timing: 1–2days

These steps describe the process of assembling the gRNA oligonucleotides designed using Benchling.com with the added overhangs into the LentiCRISPR v2 expression vector using golden gate assembly. We detail the steps to complete phosphorylation and annealing of oligonucleotides, which facilitate ligation into LentiCRISPR v2.

  • 1.

    Phosphorylation and annealing of gRNA oligonucleotides:

Reaction mix on ice

ReagentAmount
Oligo 1 (sense) (100μM)1μL
Oligo 2 (antisense) (100μM)1μL
10X T4 PNK buffer1μL
T4 Polynucleotide Kinase (PNK)0.5μL
ATP (10mM)1μL
ddH2O5.5μL
Total10μL

Open in a separate window

Cycling conditions

StepsTemperatureTimeCycles
phosphorylation37°C30min1
Annealing95°C5min1
Ramp down to 25°C5°C/min1
25°C1min1
Hold4°C

Open in a separate window

Note: 10X T4 PNK buffer is from a commercial kit (see key resources table for catalog information).

  • 2.

    Add 90μL ddH2O to each reaction to bring the final volume to 100μL after phosphorylation and annealing.

Note: The following reaction mix utilizes the restriction enzyme Esp3I. Two Esp3I restriction sites are located on the LentiCRISPR v2 plasmid and generate complementary overhang sequences to those in the previously design gRNA sequences.

  • 3.

    Clone gRNAs into LentiCRISPR v2 using golden gate assembly:

Reaction mix on ice

ReagentAmount
LentiCRISPR v275ng
Annealed oligos (from step 1)1μL
Esp3I1μL
T4 ligase buffer2μL
T4 ligase1μL
ddH2O14μL
Total20μL

Open in a separate window

Note: T4 ligase buffer is from a commercial kit (see key resources table for catalog information).

Cycling conditions

StepsTemperatureTimeCycles
Cloning42°C5min10
16°C5min
37°C for Esp3I10min1
Inactivation80°C20min1
Hold4°C

Open in a separate window

Alternatives: BsmBI-v2 may be used as an alternative enzyme to Esp3I. However, the cloning must be done at 55°C as opposed to 37°C for Esp3I.

Note: Following golden gate assembly, plasmids may be stored at 4°C or at −20°C for long-term storage.

Testing gRNA editing efficiency using T7 endonuclease assay in HEK cells

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (8)Timing: 4days

This section explains the steps involved in determining the relative editing efficiency of multiple gRNAs targeting a single gene of interest using a rapid T7 endonuclease assay in HEK cells. T7 endonuclease recognizes and cleaves mismatched DNA, allowing users to determine the relative efficiency of gRNA editing using the relative amount of cleaved DNA following gRNA editing and T7 endonuclease digestion. Pre-determining gRNA efficiency enhances the probability of successful genome editing in hESCs.

  • 4.

    Transform 2μL of the assembled gRNAs– LentiCRISPR v2 plasmid from step 3 into DH5α Competent E.coli by following the company instructions without any modifications to the provided commercial protocol. Brief steps are described below.

    • a.

      Thaw DH5α on ice for 10min and add 100ng of LentiCRISPR v2 plasmid to the tube. Carefully flick the tube and place it on ice for 30min.

    • b.

      Heat shock at exactly 42°C for 30 s. Rest on ice for 5min.

    • c.

      Add 1mL of outgrowth medium (in the kit) and allow cells to recover at 37°C for 1h while rotating at 250rpm.

    • d.

      Spread 100μL onto pre-warmed LB agar+Ampicillin (100μg/mL) plates and incubate at 37°C O/N.

  • 5.

    Pick two bacterial colonies for each transformed gRNA and use a Mini-prep kit to grow and purify the plasmid DNA. Follow the company-provided protocol without any modifications. Brief steps are described below.

    • a.

      Pellet 1.2mL bacterial culture by centrifuging at 7,000×g for 3min.

    • b.

      Resuspend pelleted bacterial cells in 250μL Buffer P1. Add 250μL Buffer P2 and mix thoroughly by inverting the tube until the solution becomes clear.

    • c.

      Add 350μL Buffer N3 and mix thoroughly by inverting the tube.

    • d.

      Centrifuge for 10min at ∼17,000×g and transfer supernatant to the spin column provided by the kit. Centrifuge for 1min and discard the flow-through.

    • e.

      Add 750μL Buffer PE to wash. Centrifuge for 1min and discard the flow-through.

    • f.

      Elute DNA in 50μL Buffer EB by centrifuging for 1min.

  • 6.

    Sanger-sequence the plasmid with the LentiCRISPR v2 primer (see key resources table) to confirm gRNA sequence insertion into the vector.

  • 7.

    Transfect one well of a 6-well plate of HEK cells using 6μL FuGENE 6 transfection reagent mixed with 1.5μg LentiCRISPR v2 plasmid with validated gRNA sequence from step 6.

    • a.

      Incubate the mixture of FuGENE6 and plasmid in 100μL Opti-MEM for 15min at 20°C.

    • b.

      Add the mixture drop-wise to HEK cells and incubate at 37°C.

    • c.

      Change media to fresh DMEM+ after 24h of incubation with transfection complexes and incubate at 37°C for an additional 48 h.

  • 8.

    Extract the genomic DNA of transfected HEK cells using a DNeasy blood and tissue kit for each gRNA transfection. Follow the company-provided protocol without any modifications. Brief steps are described below.

    • a.

      Centrifuge at 300×g for 5min to collect cells and resuspend in 200μL PBS (add 20μL proteinase K from the kit).

    • b.

      Add 200μL Buffer AL and mix thoroughly by vortexing.

    • c.

      Add 200μL 100% ethanol and mix thoroughly by vortexing.

    • d.

      Pipet the mixture into a DNeasy Mini spin column and centrifuge at 6,000×g for 1min. Discard the flow-through.

    • e.

      Add 500μL Buffer AW1 and centrifuge at 6,000×g for 1min. Discard the flow-through.

    • f.

      Add 500μL Buffer AW2 and centrifuge at 20,000×g for 3min. Discard the flow-through.

    • g.

      Elute the DNA by adding 200μL buffer AE to the center of the spin column membrane. Incubate for 1min and centrifuge at 6,000×g for 1min.

Note: Alternative transfection reagents such as calcium phosphate may be used for HEK transfection, but FuGENE 6 is recommended for optimal transfection efficiency.

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (9)CRITICAL: Passage cells 1 day before transfection so that they are approximately 75% confluent at the time of transfection.

  • 9.

    PCR amplify the target CDS region in the gene of interest.

Note: Ideal primers for this reaction should generate a 600bp amplicon with the gRNA target toward the middle of the PCR product so that T7 digestion generates a 200–300bp fragment (see troubleshooting problems 1 and 4).

PCR Reaction mix on ice

ReagentAmount
5X Q5 reaction buffer10μL
dNTPs (10mM)1μL
Genomic DNA4μL
Gene-specific CDS primer FWD (10μM)2.5μL
Gene-specific CDS primer REV (10μM)2.5μL
Q5 DNA polymerase0.5μL
ddH2O29.5μL
Total50μL

Open in a separate window

PCR cycling conditions

StepsTemperatureTimeCycles
Initial Denaturation98°C30 s1
Denaturing
Annealing
Extension
98°C10 s35
70°C–72°C30 s
72°C15 s
Final extension72°C2min1
Hold4°C

Open in a separate window

Note: The 5X Q5 reaction buffer is from a commercial kit (see key resources table for catalog information).

Note: The Q5 DNA polymerase rate is 30 s/kb. Users may need to adjust the extension time accordingly based on the size of the PCR product.

  • 10.

    Run a 1% agarose gel (dissolve 1g of agarose in 100mL TAE buffer) with 10μL PCR product for 30min at 120V to confirm the PCR product is the right size.

  • 11.

    As purified PCR products are required to achieve ideal results in the T7 reaction, we purify the remaining PCR product with a Promega PCR cleanup system by following the company-provided protocol without any modifications. Brief steps are described below.

    • a.

      Add an equal volume of membrane binding solution to the PCR product.

    • b.

      Transfer the mixture to the mini-column assembly (in the kit) and centrifuge for 1min at 16,000×g.

    • c.

      Add 700μL membrane wash solution to the column and centrifuge for 1min at 16,000×g.

    • d.

      Repeat the previous step with 500μL membrane wash solution and centrifuge for 5min at 16,000×g.

    • e.

      Elute PCR product in 50μL nuclease-free water by centrifuging for 1min at 16,000×g.

  • 12.

    Quantify the concentration using a NanoDrop 2000 spectrophotometer (Thermo Fisher).

  • 13.

    Run a T7 endonuclease I assay to determine genome targeting efficiency by cutting mismatched double strands.

Reaction mix

ReagentAmount
PCR product200ng
10X NEBuffer 22μL
Add ddH2O to 19μL

Open in a separate window

Hybridization

TemperatureTime
95°C5min
95°C - 85°CDecrease 2°C per sec, 5 s
85°C - 25°CDecrease 0.1°C per sec, 10min
25°C30 s
4°C Hold

Open in a separate window

  • 14.

    Add 1μL T7 endonuclease to 19μL annealed PCR product and incubate at 37°C for 15min.

  • 15.

    Run the T7 digested PCR products on a 1% agarose gel for approximately 30min at 120 V. Select the plasmid that generates a relatively high cleavage product following T7 digestion (Figure1).

    Open in a separate window

    Figure1

    Relative editing efficiency of different gRNAs following T7 assay in HEK cells

    Visualization of gRNA efficiency following T7 assay in HEK cells. Lane 1: 1 Kb DNA ladder; Lane 2–3: two replicates of gRNA #1 transfected HEK cells; Lane 4–5: two replicates of gRNA #2; Lane 6–7: two replicates of gRNA #3, Lane 8–9: non-transfected HEK cells, Lane 10: 1 Kb DNA ladder. gRNA #2 produced the highest intensity of the smaller T7 product compared to two other gRNAs, so gRNA #2 may have the highest editing efficiency compared to the other gRNAs.

  • 16.

    Choose the LentiCRISPR v2 plasmid that resulted in the highest percentage of T7 cleavage product and transform this plasmid into DH5α Competent E.coli as described in step 4.

    • a.

      Use a Midi-prep kit to purify the desired LentiCRISPR v2 plasmid for downstream transfection of hESCs. Refer to step 5 for general kit instructions.

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (11)Pause point: The prepared gRNA plasmids can be stored at −20°C while preparing hESC cultures.

hESC transient transfection and clone isolation

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (12)Timing: 4weeks

These steps describe how to transfect hESCs with the assembled and T7-tested LentiCRISPR v2 plasmid. This section also details selecting for transient plasmid expression using puromycin and subsequent single-cell cloning and re-expansion of edited hESC lines.

  • 17.

    Maintain hESCs in mTeSR Plus media using 6-well plates (see hESC culture in the “cell culture” section).

  • 18.

    Passage hESCs with Accutase one day before transfection. Brief steps are described below.

    • a.

      Aspirate media and rinse wells with 1mL of PBS.

    • b.

      Add 1mL of Accutase to the well and incubate at 37°C for 10min.

    • c.

      Collect and centrifuge cells for 5min at 300×g. Aspirate Accutase from the pellet and resuspend in fresh mTesR Plus+ ROCKi.

    • d.

      Seed hESCs as single cells at 70%–80% confluence in a 6-well plate (add 10μM ROCKi to support single-cell survival).

Note: Accutase is a gentle dissociation reagent that promotes the dissociation of hESCs as single cells unlike ReLesR, which promotes the dissociation of hESCs in colonies.

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (13)CRITICAL: Passaging hESCs at approximately 70% confluence and maintaining hESCs with less than 5% visible differentiation (Figure2C) are ideal for transfection and downstream cell line propagation.

Open in a separate window

Figure2

Example images of hESC colonies after selection and expansion for 10days in a 10cm dish

(A) Example of an optimal colony to be isolated and analyzed for gene editing with distinct borders and minimal to no contact with neighboring colonies.

(B and C) Example colonies to avoid isolating with visible signs of differentiation (C)or contact with neighboring colonies (B). Scale bar= 267.8μm.

  • 19.

    Transfect hESCs with 2.5μg of the LentiCRISPR v2 plasmid mixed with 7.5μL of Lipofectamine per well of a 6-well plate using Lipofectamine 3000 for optimal transfection efficiency by following the company instructions. Brief steps are described below.

    • a.

      Incubate the mixture of Lipofectamine and plasmid in 250μL Opti-MEM for 15min at 20°C.

    • b.

      Add the mixture drop-wise to hESCs and incubate at 37°C.

  • 20.

    Change media to fresh mTesR Plus after 24h and incubate cells at 37°C for an additional 24 h.

  • 21.

    Change media to selection media (mTESR Plus with 1μg/mL puromycin) for 24 h.

  • 22.

    Replace selection media with mTesR Plus+ CloneR2 supplement to support the growth of surviving single hESCs after selection.

  • 23.

    Allow surviving hESCs to recover for 3–5days in mTesR Plus+ CloneR2.

Note: If hESCs begin to proliferate after 3days, it is not necessary to wait until 5days before proceeding to clone isolation.

  • 24.

    Change media to fresh mTeSR Plus+ CloneR2 every two days.

  • 25.

    Coat one 10cm dish with Matrigel (100ng/mL) for 24h at 37°C.

  • 26.

    Dissociate recovered hESCs using Accutase (refer to step 18).

    • a.

      Pellet and resuspend hESCs in 1mL mTesR Plus+ CloneR2.

  • 27.

    Determine the volume equivalent to 1000 cells using a hemocytometer or automated cell counter.

Note: Users may find the volume equivalent to 1000 cells too small to work with comfortably. Diluting the resuspended pellet by a factor of 5 or 10 will increase the volume equivalent to 1000 cells added to a 10cm dish.

  • 28.

    Aspirate Matrigel from a 10cm dish, seed 1000 single cells to the 10cm dish, and incubate at 37°C.

  • 29.

    Change media to fresh mTeSR Plus+ CloneR2 on day 3 and day 6 post-seeding.

  • 30.

    Pick colonies that grow to a considerable size of approximately 0.5–1mm in diameter (usually one week after seeding single cells) (Figure2A). Use a sterile 20-gauge needle to cut the colony in a grid pattern. Use a p200 pipette tip to pick up the pieces and transfer them to a single well of a Matrigel-coated 24-well plate.

  • 31.

    Isolate at least 12 candidate clones and ensure each clone is seeded in one different well of a 24-well plate.

Note: Large colonies or colonies with irregular shapes may indicate cell growth from multiple hESCs in close proximity (see troubleshooting problem 2).

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (15)CRITICAL: Avoid picking colonies with visible differentiation or colonies that have merged with neighboring colonies to ensure hESC stemness and clonal purity (Figures2B and 2C).

  • 32.

    After reaching 70% confluence in a 24-well plate (usually one week post-seeding), use Accutase to collect all cells of each clone and passage them to two wells of a Matrigel-coated 6-well plate.

Note: One well is used for cryopreservation, and the other is used for DNA extraction and gene editing validation.

  • 33.

    After one week of the culture in mTeSR Plus+ CloneR2, or when the candidate clone has reached 70%–80% confluence, cryopreserve one well of confluent hESCs using mFreSR. The other well of hESCs will be used for the screening procedure beginning at step 36.

  • 34.

    Cryopreserve hESCs in mFreSR by dissociating hESCs using ReLesR and centrifuging for 5min at 300×g. Aspirate media from the pellet and resuspend the pellet in 1mL of mFreSR.

  • 35.

    Transfer 1mL of hESCs in mFreSR to a cryopreservation vial and place in −80°C.

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (16)CRITICAL: Gentle but thorough trituration of hESCs in mFreSR is essential to ensure hESC colonies are dissociated and exposed to the freezing media.

Note: Screening a minimum of 12 candidate hESC clones for each gRNA will result in a higher probability of identifying at least one successfully edited cell line.

hESC clone screening and validation of gene knockout

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (17)Timing: 1–2weeks

This step details the process of isolating genomic DNA from candidate hESC clones and subsequent verification of genome editing in hESCs.

  • 36.

    Rinse hESCs with 1mL of PBS.

  • 37.

    Add 1mL of Accutase to each well and incubate for 10min at 37°C.

  • 38.

    Collect hESCs in Accutase and centrifuge at 300×g for 5min to pellet cells. Aspirate the media from the cell pellet.

  • 39.

    Extract genomic DNA from the cell pellet using DNeasy blood and tissue kit (refer to step 8).

  • 40.

    PCR amplify the gRNA target site from hESC genomic DNA following the same procedure performed in HEK cells (refer to step 9).

Note: Ensure the use of CDS-specific primers with pBlueScript overhang sequences (key resources table) for the subsequent subcloning of the PCR product into pBlueScript at EcoRV sites.

  • 41.

    Purify PCR product using the Promega PCR cleanup system (Refer to step 11).

  • 42.

    Sequence the purified PCR products using Sanger sequencing.

  • 43.

    Perform alignment of sequencing results to reference genome by uploading the DNA sequencing to Benchling and aligning them to a reference WT genome sequence from the NCBI gene.

    • a.

      Insertions or deletions in the user’s DNA sequence will appear in red, while aligned nucleotides will correspond to the reference genome sequence (Figure3).

      Open in a separate window

      Figure3

      Candidate clone sequencing and individual allele screen of CRISPR KO hESCs

      Sanger sequencing of the candidate clone shows a high degree of DNA base uncertainty around the Cas9 cut site, indicating the potential occurrence of gene editing. Further sequencing of individual PCR products cloned into pBlueScript from this candidate hESC clone reveals a 1 (PTC generating) and 12bp (non-PTC generating) deletion.

  • 44.

    Select candidate clones with edits that potentially generate a nonsense mutation leading to premature stop codons in the coding sequence.

    • a.

      Indels not divisible by 3 are ideal candidates for downstream validation, as these types of indels create frameshift mutations in the mRNA sequence, leading to a truncated peptide and non-functional protein. (See troubleshooting problem 3).

  • 45.

    To identify the genotypes after gene editing, subclone individual PCR products from promising candidate clones into EcoRV-digested pBlueScript vector described in steps 46–48.

  • 46.

    To digest pBlueScript with EcoRV, set up digests with no more than 1μg of pBlueScript per 20μL in 1X NEB rCutSmart Buffer. Add 0.5μL EcoRV per 1μg of pBlueScript and digest for 2–3h at 37°C.

  • 47.

    Run all 20μL of the digested plasmid on a 1% agarose gel (refer to step 10) and extract and purify the 3 kb band of EcoRV-digested pBlueScript vector using a DNA gel extraction kit. Brief steps are described below.

    • a.

      Excise the DNA fragment from the gel and add 3 volumes of Buffer QG to the gel fragment.

    • b.

      Incubate at 50°C for 10min and add 1 gel volume of isopropanol to the mixture.

    • c.

      Transfer the mixture to a spin column and centrifuge for 1min at 16,000×g.

    • d.

      Add 750μL Buffer PE and centrifuge for 1min at 16,000×g.

    • e.

      Elute DNA by adding 50μL Buffer EB and centrifuge for 1min at 16,000×g.

  • 48.

    Ligate the PCR product from candidate hESCs into the digested and purified pBlueScript prepared in steps 46–47:

Ligation reaction master mix

ReagentAmount
PCR product0.12 pmol
pBlueScript0.06 pmol
2X HiFi DNA assembly mix5μL
ddH2OTo 10μL
Total10μL

Open in a separate window

Reaction conditions

StepsTemperatureTimeCycles
Ligation50°C1 h1
Store−20°CIf not used immediately for the transformation

Open in a separate window

Note: The 2X HiFi DNA assembly mix is from a commercial kit (see key resources table).

  • 49.

    Transform 2μL of reaction to DH5α Competent E.coli. and plate on LB+ Ampicillin plates and incubate at 37°C for 18h (refer to step 4).

  • 50.

    Pick 6 colonies for each candidate hESC clone and directly add to PCR tubes described below:

PCR reaction mix on ice

ReagentAmount
10X KLA buffer2μL
dNTP (10mM)0.2μL
KlentaqLA0.04μL
Gene-specific CDS primer FWD (10μM)0.4μL
Gene-specific CDS primer REV (10μM)0.4μL
ddH2O16.96μL
Total20μL

Open in a separate window

PCR cycling conditions

StepsTemperatureTimeCycles
Initial denaturation94°C3min1
Extension94°C40 s35
68°C2:30min
Hold4°C

Open in a separate window

Reaction mix

ReagentAmount
PCR product5μL
Exo clipA1μL
Exo clipB1μL
Total7μL

Open in a separate window

  • 52.

    Sequence PCR products and align each (6 sequences per clone) to the wild-type reference genome (see troubleshooting problem 5).

    • a.

      Evidence of hom*ozygous or heterozygous indels can be assessed through the distribution of insertions or deletions in multiple sequence alignments for a single hESC clone.

Direct reprogramming of hESCs into mature human neurons

CRISPR-Cas9 editing of synaptic genes inhumanembryonic stem cells forfunctionalanalysisininduced human neurons (19)Timing: 5weeks

To study the role of synaptic genes in neurotransmission of mature neurons, the verified hESC knockout will be differentiated into induced human neurons (iNs). This step describes how to directly reprogram hESCs into functional neurons using over-expression of human neurogenin-2 (hNgn2) (Figure4). This induced human neuron platform will allow users to investigate the functional consequences of the knockout generated in hESCs and employ a variety of assays in human neurons to uncover the functional changes in neural signaling as a consequence of the gene knockout.

  • 53.

    (Day-1) Use Accutase to collect confluent hESCs (refer to step 18).

  • 54.

    Generate lentiviral particles by transfecting HEK-293T cells in a T-75 flask with three lentiviral packaging constructs (pRSV-REV; pMD2.G; pMDLg/pRRE; each 5μg in Opti-MEM) and hNgn2 construct (pFUW-TetO-hNgn2-EGFP-PuroR; 10μg in DMEM) using FuGENE 6 (refer to step 7).

    • a.

      Change DMEM+ media to mTeSR plus media 16h after transfection.

    • b.

      Wait for another 48h and collect lentivirus-containing media for transduction. The typical titer from the supernatant is ∼ 1×106 IU/mL.

  • 55.

    Seed ∼3×106 hESCs as single cells to one Matrigel-coated 6-well plate in mTeSR Plus culture media containing hNgn2 expressing lentiviral particles with polybrene (final concentration 8μg/mL) and ROCK inhibitor (Y-27632, final concentration 10μM).

    • a.

      In each well of a 6-well plate, add 1.5mL fresh mTeSR Plus media and 0.5mL of lentivirus-containing media collected from HEK cell culture so that the desired multiplicity of infection is between 1 and 2.

  • 56.

    (Day 0) After 16–24 h, replace mTesR plus media with neuronal induction media.

  • 57.

    (Day 1) 24h later, replace the induction medium with neuronal selection media for 48 h.

Note: The duration of puromycin selection varies with lentiviral infection efficiency and can be optimized. However, the 48-h selection is recommended to obtain pure iN populations.

  • 58.

    Isolate mouse astrocytes by dissecting mouse pup forebrains and culture mouse primary astrocytes with DMEM+ media in a T-75 flask by following an existing detailed protocol.5

    • a.

      Passage astrocytes at least once with 0.25% Trypsin-EDTA before using them for induced neuron co-culture.

Note: Astrocytes are essential for the development and maturation of functional neural networks and are an essential cell type to co-culture with induced neurons to achieve long-term functional viability.

  • 59.

    (Day 2) Seed ∼1×105 mouse astrocytes to each well of a 24-well plate in Neurobasal plus media supplemented with FBS (final FBS concentration is 10%).

Note: Neurotrophic supplements are not needed for astrocyte growth in 24-well plates prior to seeding iN cells.

  • 60.

    (Day 3) Aspirate the selection medium and collect iN cells using Accutase from one 6-well plate (refer to step 18).

    • a.

      Resuspend the cell pellet in Neurobasal media and use a cell counter to determine the total number of iNs.

    • b.

      After counting iNs, plate approximately 1×105 cells per well in a 24-well plate with mouse astrocytes added on day 1 in neurobasal media with supplements.

  • 61.

    (On day 6& 8) replace half of the media in each well with fresh neurobasal media.

  • 62.

    (On day 10& 13) Change half of the media to neuronal growth media.

  • 63.

    Change half of the growth media in each well weekly (days 20, 27, and 34). iNs are assayed for electrophysiology and imaging between 35-50days post-transduction (Figure5).6

    Open in a separate window

    Figure5

    Representative images of human neural progenitors at Day 13 and mature neurons at Day 40 after lentiviral transduction of hNGN2-P2A-EGFP

    EGFP fluorescence shows the morphology of immature human neurons at Day 13 and mature neurons at Day 40 after hNGN2 overexpression in hESCs. Scale bar= 200μm.

Open in a separate window

Figure4

Timeline of hESC differentiation to induced neurons and culture protocol

This timeline highlights the daily procedure from hESCs to mature iNs for functional assays.

Validating gene knockout and function in mature iNs

Following neural induction and confirmation of Ngn2 expression through visualization of GFP fluorescence, users can employ assays in mature iNs to validate the loss of synaptic protein function. As shown below, we used immunostaining to verify the loss of the synaptic vesicle protein (Syt1) and employed patch-clamp electrophysiology to validate the deficit in neurotransmission in Syt1 KO iNs (Figure6).

Open in a separate window

Figure6

Immunostaining and functional assays in mature iNs validate gene knockout of synaptic protein in hESCs

Immunolabeling for MAP2, Synapsin1, and Synaptotagmin1 in WT and Syt1 KO iNs reveal a loss of Syt1 expression and a loss of synchronous neurotransmission with respect to WT iNs. Scale bar= 20μm and 5μm for zoomed inset of synapses.

Expected outcomes

Expandable human embryonic stem cells combined with generating stable cell lines using targeted gene editing strategies allow for precise interrogation of protein function in multiple cellular contexts and conditions. Targeting synaptic proteins using the protocol described here preserves the expected functional deficits in neurotransmission following neuronal induction. This strategy represents a proof of principle for the validity and use of this methodology in uncovering enigmatic protein function in differentiated cell types, including but not limited to induced neurons.

Limitations

Generating gene knockout hESCs using the precision of CRISPR/Cas9 is a valuable tool in maintaining highly controlled experimental conditions. However, the genome editing process detailed here cannot be described as isogenic. Off-target effects of gRNAs and epigenetic changes following single-cell cloning may contribute to possible confounding variables in induced human neuron function. Isolating multiple independent hESC lines derived from different gRNAs for a given gene target can help reduce the probability that observed phenotypes are due to off-target clonal variation. Also, experiments using shRNA knockdown of the same gene target could help validate the loss-of-function phenotypes due to the gene KO in edited human neurons generated by this approach.

Troubleshooting

Problem 1

Low gRNA efficiency in T7 assay.

Potential solution

  • Ensure designed gRNAs have the highest relative efficiency and on-target scores. Ideally, on-target and off-target scores should be approximately 80 or higher (in Benchling) for ideal genome specificity and successful editing.

  • Design new gRNAs to target different exons.

  • Transfect HEK cells at lower confluency to increase the probability of amplifying mutant HEK cell DNA.

  • Alternative transfection reagents such as Lipofectamine 3000 may enhance the transfection efficiency.

Problem 2

Stem cells start to differentiate following transient transfection.

Potential solution

  • Change media to fresh mTeSR Plus+ CLoneR2 every 24 h.

  • Off-target effects of gRNAs may contribute to hESC differentiation.

  • Selecting alternative gRNAs with high specificity scores may reduce hESC differentiation.

Problem 3

Lack of genome editing in hESCs with high gRNA transfection efficiency.

Potential solution

  • Increase the selection window with puromycin from 24 to 48 h.

  • Isolate >12 candidate hESCs following single-cell cloning.

  • Change media with fresh mTeSR Plus+ CLoneR2 every 24h to ensure mutant single cells can grow.

Problem 4

No PCR amplification of target site from genomic DNA.

Potential solution

  • Depending on the size of the PCR product, increase the extension time accordingly based on the Q5 DNA polymerase extension at the speed of 30 s/kb.

  • Ensure primers have low off-target binding and adequate CG content for Tm in cycling conditions.

Problem 5

Sequencing of individual PCR products from pBlueScript colonies.

Potential solution

  • Make sure to pick larger colonies on LB selection plates following plasmid transformation.

  • Using primers targeting the PCR insert as opposed to a region of the vector may increase the success of insert amplification.

Resource availability

Lead contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Ege T. Kavalali (ude.tlibrednav@ilalavak.ege).

Technical contact

Questions regarding the technical specifics of performing this protocol should be directed to and will be answered by the technical contact, Aiden Houcek (ude.tlibrednav@kecuoh.nedia).

Materials availability

The materials used and generated in this study are available from the lead contact upon reasonable request with a completed Materials Transfer Agreement. However, there are restrictions to the availability of plasmid reagents generated for this study due to the individualized cloning of gRNAs into expression vectors and gene targets of interest.

Data and code availability

No original code or any dataset has been generated in this study.

Acknowledgments

This work was supported by National Institutes of Health grants (R01 MH066198 to E.T.K. and R01 MH070727 to L.M.M.). Cartoon figures were generated using BioRender.com. We thank Natalie J. Guzikowski for comments and feedback on the protocol.

Author contributions

Conceptualization, A.H., Z.Z.M., and E.T.K.; methodology and investigation, A.H., Z.Z.M., B.T., and B.U.; validation, A.H.; writing– original draft, A.H. and Z.Z.M.; writing– review and editing, A.H., Z.Z.M., B.T., B.U., L.M.M., and E.T.K.; resources and funding acquisition, L.M.M. and E.T.K.; supervision, Z.Z.M. and E.T.K.

Declaration of interests

The authors declare no competing interests.

References

1. Veres A., Gosis B.S., Ding Q., Collins R., Ragavendran A., Brand H., Erdin S., Cowan C.A., Talkowski M.E., Musunuru K., Musunuru K. Low incidence of off-target mutations in individual CRISPR-Cas9 and TALEN targeted human stem cell clones detected by whole-genome sequencing. Cell Stem Cell. 2014;15:27–30. [PMC free article] [PubMed] [Google Scholar]

2. Zhang Y., Pak C., Han Y., Ahlenius H., Zhang Z., Chanda S., Südhof T.C., Patzke C., Acuna C., Covy J., et al. Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron. 2013;78:785–798. [PMC free article] [PubMed] [Google Scholar]

3. Hsu P.D., Scott D.A., Weinstein J.A., Ran F.A., Konermann S., Agarwala V., Li Y., Fine E.J., Wu X., Shalem O., et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat. Biotechnol. 2013;31:827–832. [PMC free article] [PubMed] [Google Scholar]

4. Shalem O., Sanjana N.E., Hartenian E., Shi X., Scott D.A., Mikkelson T., Heckl D., Ebert B.L., Root D.E., Doench J.G., Zhang F. Genome-scale CRISPR-Cas9 knockoutscreening in human cells. Science. 2014;343:84–87. [PMC free article] [PubMed] [Google Scholar]

5. Schildge S., Bohrer C., Beck K., Schachtrup C. Isolation and Culture of Mouse Cortical Astrocytes. J.Vis. Exp. 2013;71 doi:10.3791/50079. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

6. Uzay B., Houcek A., Ma Z.Z., Konradi C., Monteggia L.M., Kavalali E.T. Neurotransmitter release progressively desynchronizes in induced human neurons during synapse maturation and aging. Cell Rep. 2023;42 [PMC free article] [PubMed] [Google Scholar]

Articles from STAR Protocols are provided here courtesy of Elsevier

CRISPR-Cas9 editing of synaptic genes in human embryonic stem cells for functional analysis in induced human neurons (2024)
Top Articles
Latest Posts
Article information

Author: Jamar Nader

Last Updated:

Views: 6706

Rating: 4.4 / 5 (75 voted)

Reviews: 90% of readers found this page helpful

Author information

Name: Jamar Nader

Birthday: 1995-02-28

Address: Apt. 536 6162 Reichel Greens, Port Zackaryside, CT 22682-9804

Phone: +9958384818317

Job: IT Representative

Hobby: Scrapbooking, Hiking, Hunting, Kite flying, Blacksmithing, Video gaming, Foraging

Introduction: My name is Jamar Nader, I am a fine, shiny, colorful, bright, nice, perfect, curious person who loves writing and wants to share my knowledge and understanding with you.