Korean, Edit

Chapter 9. DNA Technology

Higher category: 【Biology】 Biology Index 


1. DNA recombination

2. DNA library

3. PCR

4. DNA fingerprinting

5. Hybridization

6. Gene deletion

7. Nuclear substitution

8. DNA-protein interaction studies

9. Protein-protein interaction studies

10. Gene editing


a. Nucleic Acid (DNA, RNA) Extraction and Purification Experiment

b. Western Blotting Experiment

c. The Romanov Dynasty and Forensic Science



1. DNA recombination

⑴ 1st. Target gene generation from mRNA (for eukaryotic DNA)

① mRNA is reverse transcribed to synthesize cDNA (complementary DNA to mRNA).

② RT-PCR: Add DNA primers to the mRNA solution and treat with reverse transcriptase (RT) once.

○ Oligo dT primer: Enables the synthesis of complete cDNA by using the poly A tail, which is present in eukaryotic mRNA, as a template.

○ Random primer (usually hexomer): Can form a variety of cDNA pools.

③ RT-PCR: Amplify the amount of cDNA like a normal PCR.

⑵ 2nd. Recombinant plasmid

① Restriction enzyme: A kind of endonuclease. A scissors that cuts DNA.

○ Restriction enzymes are produced in bacteria to target and degrade foreign DNA.

○ Their own DNA, which could be targeted, is protected by methylation.

○ Blunt ends: Ends that cannot rejoin after being cut by a restriction enzyme.

○ Sticky ends: Ends that can re-form complementary hydrogen bonds after being cut by a restriction enzyme.

○ Restriction enzymes that form sticky ends are used for DNA recombination.

○ Palindrome recognition: When reading from 5’ to 3’, the sequences on the sense strand and the antisense strand are identical.


스크린샷 2025-02-01 오후 3 07 58


○ The above example is a sticky end.

○ Examples of restriction enzymes

○ AgeI: 5’-A▼CCGGT-3’

○ AluI: 5’-AG▼CT-3’

○ BamHI: 5’-G▼GATCC-3’

○ BglII: 5’-A▼GATCT-3’

○ DpnI: 5’-G metA ▼TC-3’

○ DpnII: 5’-▼GATC-3’

○ EcoRI: 5’-G▼AATTC-3’

○ FspEI: 5’-CmC(N)12▼-3’

○ HindIII: 5’-A▼AGCTT-3’

○ HpaII: Cuts at 5’-C▼CGG-3’, but not at 5’-CmC▼GG-3’ where mC represents methylated cytosine.

○ LpnPI: 5’-CmCDG(N)10▼-3’

○ McrBC: 5’-PumC(N40-3000)PumC-3’

○ MspI: 5’-C▼CGG-3’, 5’-C▼mCGG-3’ where mC represents methylated cytosine.

○ MspJI: 5’-mCNNR(n)9▼-3’

○ MSTII: 5’-CCTN▼AGG-3’, where is a nucleotide sequence with n palindromic structures.

○ NcoI: 5’-C▼CATGG-3’

○ PstI: 5’-CTGCA▼G-3’

○ SalI: 5’-G▼TCGAC-3’

○ Sau3AI: 5’-▼GATC-3’

○ SmaI: 5’-GGG▼CCC-3’, forming blunt ends.

○ Recombinant sequence by BamHI is also cleaved by Sau3AI. The opposite holds true only in specific cases.

○ The fragment by BglII and the fragment by BamHⅠ bind to each other and are not cut again by restriction enzymes.

○ When linear DNA is cut once by a restriction enzyme, it becomes two fragments, whereas circular DNA, when cut once by a restriction enzyme, becomes a single fragment.

○ Restriction mapping

② 2nd-1st. Cut the target DNA and the plasmid DNA with the same restriction enzyme to form the same sticky ends.

○ 2nd-1st-1st. To prevent self-ligation when treating with only one restriction enzyme, treat with alkaline phosphatase (e.g., Calf Intestinal Phosphatase (CIP)).

○ 2nd-1st-2nd. Provide directionality to plasmid DNA binding to target DNA by treating with two or more types of restriction enzymes.

③ 2nd-2nd. Insert the truncated gene and plasmid into one test tube → Complementary sticky ends bind to each other.

④ 2nd-3rd. Create a recombinant plasmid by ligating with DNA ligase.

⑶ 3rd. Transformation: Insert the recombinant gene into the host cell (bacteria) to induce transformation.

① Transformation process

○ 3rd-1st. CaCl2: Ca2+ binds to phospholipids and stabilizes cell membranes.

○ 3rd-2nd. Thermal Shock 42 ℃: A temporary pore is formed in the cell membrane, allowing the recombinant plasmid to be inserted into the host.

② Host: Escherichia coli, yeast, insect cells, etc.

○ To obtain the product of a recombinant gene, it must be put into a host cell with metabolic function.

Condition 1. First generation period should be short.

Condition 2. Easy to cultivate and grow well on cheap media.

Condition 3. Not pathogenic.

Condition 4. Antibiotic intolerance for screening process.

③ Vector: Plasmid or bacteriophage DNA that carries and replicates recombinant DNA.

④ Cloning vector: A vector used for gene replication.

○ Four conditions: Origin of replication (ori), promoter, restriction enzyme recognition sequence, selection marker (antibiotic resistance gene)

○ Additional condition: Small size, higy copy number

○ Cloning site

⑤ Expression vector: Vectors for expressing foreign genes in other hosts with different expression systems.

○ Prokaryotic host condition: Promoter, SD sequence, 70S ribosomal binding site, transcription terminator. Note that cloning gene is cDNA.

○ Proteins that process tertiary structures (e.g., disulfide bonds) are also required.

○ Eukaryotic host condition: Promoter, poly A sequence

○ Example: When a promoter is attached after the activator gene of the lac operon.

⑥ Vector examples

Example 1. pBR322 plasmid

○ 4361 bp. ori is present.

○ It has recognition sequences for restriction enzymes such as EcoRI, BamHI, and HindIII.

○ ampr: The ampicillin resistance gene from plasmid pBR322, recognized by PstI.

○ tetr: The tetracycline resistance gene from plasmid pBR322, recognized by HindIII, BamHI, and SalI.

○ Recombinant strain: Select a tetracycline-sensitive, ampicillin-resistant strain.

Example 2. pUC19 plasmid

○ 2686 bp. ori is present.

○ Has restriction enzyme recognition sequences such as ApaLI.

○ lacZ: Produces a blue-colored product using X-gal as a substrate.

○ ampr

Example 3. Ti plasmid

○ Agrobacterium tumefaciens (Rhizobium radiobacter): Pathogens of plant crown gall (benign tumor).


image

Figure 1. Rhizobium radiobacter attached to carrot cells


○ Ti plasmid: Rhizobium radiobacter plasmid, including T-DNA.

○ T-DNA: 20 kb of DNA encodes opines and auxin·cytokinin synthase (forming crown gall).

○ Opine: Nutrient sources of Rhizobium radiobacter.

○ Process: Agrobacterium tumefaciens infects the plant → vir (virulence DNA, 25 bp) cleaves both ends of T-DNA and randomly inserts it into the plant nuclear chromosome.

○ DNA recombination: Inserting target DNA into the T-DNA region after injuring the plant and infecting it with recombinant Rhizobium radiobacter.

○ The recombinant Agrobacterium tumefaciens does not synthesize auxin or cytokinin, preventing the formation of crown gall tumors.

Example 4. pBHA

Example 5. pBIC-A

Example 6. Gene library vector: BAC Library, YAC Library

Example 7. Bacterial plasmid: F plasmid, R plasmid (antibiotic resistance gene inserted). Approximately 1-10 kb.

Example 8. Virus vector

○ Genetic information is inserted into bacterial DNA through bacterial infection, proliferation, lysis, release and cleavage.

○ λ phage (about 25 kb) is mainly used.

○ Sometimes retrovirus is used.

Example 9. Cosmid: Introduces the phage cos end using the in vitro packaging method, approximately 50 kb in size.

Example 10. Beta-lactamidase gene creates beta-lactam ring to confer penicillin resistance.

⑷ 4th. Screening: Screening for cells or populations with recombinant genes

① Antibiotic resistance gene (selection based on proper insertion of recombinant plasmid)

○ Example: Ampicillin resistance gene, tetracycline resistance gene

○ 1st. Replica formation: After incubating the entire colony, imprint the entire culture plate onto filter paper.

○ 2nd. Add antibiotics to a portion of the colonies imprinted on the filter paper.

○ 3rd. Colonies that die due to the antibiotic indicate that they were not transformed.

○ 4th. Select colonies corresponding to those that survived the antibiotic on the filter paper.

② Antibiotic resistance genes (selection according to recombination success)

○ 1st. Replica formation: After incubating the entire colony, imprint the entire culture plate onto filter paper.

○ 2nd. Add antibiotics to a portion of the colonies imprinted on the filter paper.

○ 3rd. Colonies that died due to the antibiotic indicate that the restriction enzyme cut the antibiotic resistance gene.

○ 4th. Select colonies corresponding to those that died due to the antibiotic on the filter paper.

③ X-gal (selection according to recombination success)

○ Cloning gene is inserted into lac Z gene.

○ 1st. X-gal is originally white, but becomes blue due to lac Z.

○ 2nd. If a colony treated with lacZ-bearing plasmid appears white on X-gal, it indicates successful transformation.

④ Reporter Gene

○ Use genes that are not expressed in the host cell.

○ Examples: eGFP, tdTomato.

⑤ Southern blotting

⑸ 5th. Cloning amplification: Large quantities of product are obtained by mass fermentation and purification.

① Reason for using antibiotics in large-scale culture with antibiotic-resistant plasmids (non-selective purpose)

○ 1st. Plasmids within the bacterial strain are randomly distributed to daughter cells during cell division.

○ 2nd. As division continues, some cells end up without any plasmids.

○ 3rd. Cells containing plasmids consume more energy to replicate them, leading to a difference in growth rate compared to plasmid-free cells.

○ 4th. Ultimately, plasmid-free cells become the majority.”

② Inclusion Body

○ Since the gene introduced during transformation is not originally present in the host, the produced protein may not fold properly and aggregates.

⑹ Examples

① Hormones and bioactive substances: Growth hormone, insulin, secretin, etc.

② Therapeutic and diagnostic reagents: Hepatitis B vaccine, diagnosis of HIV infection, interferon, neoendorphin, analgesic, blood coagulation factor, etc.

③ Agriculture: Golden rice, herbicide-tolerant crops, pest-tolerant crops

④ Ti plasmid transgenic neofunctional crop: Herbicide tolerance, anti-anthrax crops (seed potatoes, etc.)

⑤ Recombinant strain development strategy

○ Feedback / antibiotics resistant mutant

○ Auxotrophic mutant

○ Overexpression mutant



2. DNA library

⑴ Vector for DNA library

① Bacterial Artificial Chromosome (BAC)

○ Advantage: No cross occurs.

○ Disadvantage: Must be adapted to the prokaryotic expression system.

○ Used in human genome projects.

○ It is still used today.

② Yeast Artificial Chromosome (YAC; pYAC3)

○ Artificial chromosome into yeast (~1 Mb), contains ARS, and inserts DNA between the centromere and telomere.

○ Advantage: Suitable for producing human proteins as it utilizes a eukaryotic system.

○ Disadvantage: Crossover occurs during prophase I of meiosis.

③ Virus genome library

○ Disadvantage: Small capacity. Difficult to control breeding.

⑵ Required DNA elements

① Centromere: A GC-rich sequence that attaches to the spindle fibers during cell division, ensuring proper distribution of chromosomes to each daughter cell.

② Telomere: A nucleotide sequence located at the end of a chromosome that prevents chromosome shortening due to cell division.

③ Origin of replication: The site where DNA replication begins.

④ Selection marker: A specific DNA sequence used to confirm the proper insertion of artificial chromosomes into cells.

⑤ Others: Promoter, gene expression regulatory mechanisms.

⑶ Types

① gDNA Library

○ 1st. Precipitate histones by treating with phenol.

○ 2nd. Nucleic acids are present in the genomic DNA supernatant.

○ 3rd. Separate the supernatant → Trichloroacetic acid → Nucleic acid precipitate.

○ 4th. After digesting gDNA with a restriction enzyme, clone all fragments and insert them into a BAC vector.

○ 5th. Selectively amplify Escherichia coli containing the target DNA.

○ 6th. Used in intron research.

○ Triton X-100 is amphipathic and does not denature the DNA structure.

○ SDS-PAGE denatures the DNA structure.

② cDNA Library

○ 1st. Affinity chromatography: Attach oligo-dT (poly T) to beads.

○ 2nd. Reverse transcribe the separated mRNA using reverse transcriptase.

○ 3rd. Treat with a low concentration of RNase: Functions as a primer.

○ 4th. Treat with DNA polymerase I.

○ 5th. Treat with ligase.

○ 6th. Treat with restriction enzyme.

○ 7th. Insert into a vector.

○ 8th. Cloning amplification.

○ 9th. Used in the study of gene expression.

③ Library Screening

○ Transfer library clones onto a filter and activate with a probe to identify the locations of clones containing the same sequence as the probe.



3. PCR

⑴ Overview

① Definition: A DNA amplification technique based on the principles of DNA replication.

② Developed by Kerry Mullis in 1983; he was awarded the Nobel Prize in Chemistry in 1993.

⑵ Sample: DNA sample, DNA primer (2 types), dNTP, Taq polymerase, Buffer (pH stabilization), MgCl2 (DNA stabilization)

① DNA Primer

Condition 1: GC content > 50%

○ Example: 10 out of 18 bases (= 18/2 + 1) are G or C

○ Example: 11 out of 20 bases (= 20/2 + 1) are G or C

Condition 2: The 5’ end should end with AT, and the 3’ end should end with GC

○ Reason: Prevents hairpin formation due to self-ligation

○ Example: 5’-ATGCCTATGCG-3’

○ Example: 5’-ATGCAGGCTAAT-3’ can cause self-ligation

Condition 3: The 3’ end should have 2 to 3 overlapping Gs or Cs

○ Reason: A=T bonds are double bonds, whereas G≡C bonds are triple bonds, making GC-rich sequences more stable

○ Example: 5’-ATGCCTATGCG-3’

② dNTP

○ Note that NTPs are the building blocks for RNA polymerization.

○ Reason why an E. coli colony can be used instead of a DNA template in colony PCR: Due to heat-induced denaturation.

○ In other words, the E. coli colony is broken down under denaturation, providing the necessary materials for DNA polymerization.

③ Taq polymerase

○ Taq polymerase is derived from the archaea Thermus aquaticus, which inhabits high-temperature hot springs.

○ It was first discovered in the 1960s in Yellowstone National Park.

○ Taq polymerase has an optimal polymerization temperature of 55°C and denatures at 92–95°C.

○ After completing polymerization to the 3’ end, Taq polymerase adds an extra A nucleotide.

○ Since it does not require a template, it can be applied to recombinant DNA with blunt-ended sequences.

○ Taq polymerase lacks 3’ → 5’ proofreading ability, resulting in an error rate of 1/104.

○ In contrast, typical DNA polymerases have a replication error rate of 1/107.

④ MgCl2

○ DNA has a negatively charged phosphate backbone, leading to structural instability → Na+ or Mg2+ can stabilize DNA.

⑤ IPP (Inorganic Pyrophosphatase)

○ Enhances the binding of NTPs.

⑶ Process

① 1st. The heating step to separate the two DNA strands. 94 ℃. 30-60 seconds.

② 2nd. Annealing: As the mixture cools, the primers bind to the DNA template. 50-55 ℃. 1 minute.

③ 3rd. DNA elongation: Polymerase starts synthesis at primer. 72 ℃. 1 minute per 1000 bp.

④ 4th. Repeating steps ① to ③ theoretically results in the exponential amplification of the DNA sample by a factor of 2.

⑤ 5th. DNA ligase action (40 ℃) and stabilization (4 ℃).

⑥ 6th. Switch to E. coli cloning amplification after a few cycles: In reality, the amount of artificial nucleotides is insufficient, producing much less DNA.

⑷ Tm vs annealing temperature

① DNA Melting Curve: A graphical representation of the degree of DNA denaturation as a function of temperature.

○ It shows the denaturation level of linear double-stranded DNA at different temperatures using absorbance at 260 nm (A260).

○ A260 value comparison: Double-stranded DNA < Single-stranded DNA < Fragmented nucleic acids

○ Reason: Less base stacking leads to lower shielding and higher absorbance.

② Tm (Melting Temperature): The temperature at which 50% of the DNA remains double-stranded, while the other 50% has denatured into single strands.

○ Generally proportional to (A + T) × 2 + (G + C) × 4.

○ Tm value is independent of repetitive sequences.

○ The Tm of dsRNA is about 15 ℃ higher than that of dsDNA with the same sequence due to the -OH group in RNA stabilizing dsRNA.

③ Annealing

○ The annealing temperature is typically set 5–10°C below Tm.

○ If the annealing temperature is too high, hybridization is inefficient; if too low, mis-hybridization increases.

○ The presence of metal ions affects the optimal annealing temperature.

⑸ Ct (Threshold Cycle)

① Definition: The cycle at which the signal exceeds the threshold value.

② Ct and copy number have an inverse correlation.

⑹ Limitation: The nucleotide sequences at both ends of the template DNA must be known.

① Assumption: If there is a known sequence in the middle and the template DNA has blunt ends, this limitation can be overcome.

Solution 1. Inverse-PCR

○ 1st. Form circular DNA from the template DNA using DNA ligase.

○ 2nd. Generate linear DNA by cutting within the known sequence using a restriction enzyme.

○ 3rd. PCR is possible because we know sequence of both ends.

Solution 2. Anchored-PCR

○ 1st. Attach a new primer to the template DNA using DNA ligase and amplify one half.

○ 2nd. The other half goes through the same process.

Solution 3. Oligonucleotide-directed mutagenesis

Case 1: Replicating the template DNA using a primer with only a few mismatched bases.

Case 2: Replicating the template DNA using a primer with an added nucleotide sequence that induces a hairpin structure in the middle.

Case 3: Replicating the template DNA using a primer with a removed nucleotide sequence to allow the template DNA to form a hairpin structure.

⑺ Types

① RT-PCR: Includes the process of synthesizing cDNA from RNA using reverse transcriptase.

② Real-time PCR (qPCR, quantitative PCR): Allows real-time monitoring of amplified DNA quantity.

○ Initially, the given nucleotide fragment does not fluoresce because the distance between the fluorescent reporter and quencher is too short.

○ During polymerization in PCR, the fluorescent reporter and quencher separate, allowing fluorescence to appear.

○ TaqMan qPCR: A TaqMan probe (containing a fluorophore and quencher) binds to the target DNA and is cleaved during DNA synthesis, generating fluorescence.

③ Multiplex PCR: Amplifies multiple types of DNA samples within a single PCR reaction.

④ Allele-Specific PCR (AS-PCR): A method for detecting allele-specific variations using PCR.



4. DNA fingerprinting

⑴ Overview

① Objective: Paternity confirmation, prediction of genetic diseases, forensic perpetrator identification.

② Using the fact that no two individuals, except for identical twins, are genetically identical.

⑵ Electrophoresis: Allows for the separation of substances by size and the estimation of their quantity.

① Stationary phase, mobile phase

○ Stationary phase (compression gel, stacking gel) 

○ Higher gel density results in slower transport of DNA samples or proteins.

○ Reason for high gel density: To keep the moving speed constant regardless of DNA sample or protein size.

○ Mobile phase (running gel)

○ Low gel density allows rapid movement of DNA samples or proteins.

○ Gel density is 6-15%.

② Nucleic acid electrophoresis: Agarose gel

③ Protein electrophoresis: Polyacrylamide gel + Bisacrylamide gel, SDS-PAGE, TEMED

Material 1. Acrylamide: Forms polymers in the gel.

Material 2. RIPA Buffer: Isotonic solution. Used as a buffer for cell lysis.

Material 3. Loading Buffer or Laemmli Sample Buffer: A buffer that breaks disulfide bonds.

○ Does not contain methanol but includes SDS-PAGE.

○ SDS-PAGE: Used for more efficient separation. Applied in denaturing gels.

○ SDS-PAGE consists of SDS (an anionic surfactant) and β-ME (β-mercaptoethanol).

○ SDS: Removes ionic bonds and hydrophobic interactions. Equalizes the charge density per unit mass of proteins.

○ β-ME: Eliminates disulfide bonds.

○ When SDS-PAGE binds to each amino acid, the repulsion between negative charges results in all proteins having the same charge density, forming a secondary structure.

○ DTT: An additive that breaks disulfide bonds between cysteine residues.

Material 4. Transfer Buffer: Used for transferring from gel to membrane.

○ Contains methanol but does not include SDS-PAGE.

○ Native-PAGE: Used in non-denaturing gels.

○ Methanol: Functions to cool the heat generated during the transfer process.

Material 5. Ammonium Persulfate (APS): Acts as an enzyme that forms cross-links between polyacrylamide and bis-acrylamide gels.

Material 6. TEMED (N,N,N’,N’-tetramethylethylenediamine): Stabilizes APS and helps the gel solidify quickly.

④ Typically the top is the anode, the bottom is the cathode.

⑤ Moving distance = Δt × (a × log M + b), a < 0, M: molecular weight

○ When treated with special substances such as SDS, molecular weight is linearly proportional to size, resulting in greater resistance in the mobile phase and a shorter migration distance.

○ Molecular weight has the greatest influence on migration distance.

○ Migration distance is proportional to time (Δt).

○ DNA ladder (size marker): Provides migration distances based on size, allowing estimation of the size of unknown substances.

○ Supercoiled DNA experiences less resistance in electrophoresis.

Type 1. General electrophoresis

○ Nucleic acids and proteins with smaller molecular weights migrate further.

○ **In protein electrophoresis, SDS-PAGE causes proteins to migrate from the (-) electrode to the (+) electrode.

Type 2. Isoelectric electrophoresis: Electrophoresis using isoelectric points, which are unique values of proteins.

○ Electrophoresis with a pH gradient moves proteins to the point where the total charge is zero (isoelectric point).

○ After electrophoresis, proteins are stained using protein dyes such as Coomassie Blue or silver nitrate.


image

Figure 2. Isoelectric electrophoresis


Type 3. 2D electrophoresis

○ O’Farrel law is common.

○ X-axis: Separation based on charge through isoelectric electrophoresis in the presence of urea or electrophoresis under non-denaturing conditions.

○ In other words, separation occurs according to the isoelectric point.

○ Urea: Removes all R-R interactions except disulfide bonds.

○ Y-axis: Separation based on molecular weight through electrophoresis in the presence of SDS.

○ More than 1,000 types of proteins can be separated.

Application 1. Separate both the X-axis and Y-axis using the same method, but add a specific substance during Y-axis separation → Substances deviating from the diagonal indicate interaction with the added substance.

Application 2. For tRNA and small RNA molecules: Use 10% polyacrylamide gel for 1D and 20% for 2D → Effectively separates various types of RNA.

Fingerprinting Method 1. RFLP (restriction fragment length polymorphism): Paternity testing, genetic disease analysis

① Process: Target genes.

○ 1st. DNA isolation from tissue.

○ 2nd. PCR: Amplify the amount of DNA.

○ 3rd. Cutting DNA into fragments using restriction enzymes.

○ 4th. Generate DNA fragments of different sizes with the same restriction enzyme recognition sites.

○ 5th. Electrophoresis: Separates fragments based on size differences for visualization.

② Cause: Single nucleotide polymorphism (SNP)

○ Defition: Difference between people due to one base being different by point mutation.

○ In humans, approximately one SNP appears per 1,000 base pairs (bp).

○ SNPs can occur in both exons and introns.

○ If an SNP occurs in a restriction enzyme recognition site, RFLP may occur.

○ Difference between mutation and polymorphism: If it occurs in less than 1% of the entire population, it is considered a mutation; otherwise, it is classified as polymorphism.

○ Number of SNPs: As of 2017, there are approximately 10,000 SNPs.


image

Figure 3. Number of SNPs


③ Limitation: Since SNPs do not necessarily occur at restriction enzyme recognition sites, RFLP variations among individuals are not significant.

○ Difficult to use for criminal identification and similar applications.

Fingerprinting Method 2. Tandem repeat: Used for suspect identification. A more powerful genetic fingerprinting method than RFLP, targeting repeat sequences.

① VNTR (variable number tandem repeat): 15-100 repetitions

○ Principle: The number of repeats in repeat sequences varies among individuals, resulting in fragments of different sizes.

○ Cause: Uneven crossover of homologous chromosome

○ 1st. DNA fragments are separated and amplified based on length using electrophoresis.

○ 2nd. Separated DNA fragments are transferred to filter paper.

○ 3rd. After transferring to filter paper, a chemical treatment (alkaline) is applied to break hydrogen bonds → Results in single-stranded DNA.

○ 4th. Compare the location of the VNTR fragments with a VNTR probe labeled with radioactive material.

○ 5th. Band appears when exposing filter paper to X-ray film.

② STR (Short Tandem Repeat): Repeats of 2-4 times.

○ Not significantly different from VNTR.

Fingerprinting Method 3. Other Fingerprinting Methods

① CNV (Copy Number Variation)

② LOH (Loss of Heterozygosity)

③ Genomic Rearrangement

④ Rare Variant



5. Hybridization

⑴ Southern blotting: Nucleic acid probe hybridization to DNA.

① Overview

○ A kind of RFLP.

○ Developed by Edward M. Southern in 1975.

② 1st. Restriction enzyme treatment → PCR amplification → Electrophoresis

○ Electrophoresis: Agarose gel is used.

○ A master plate can be used instead of electrophoresis: Colonies with recombinant plasmids are arranged on a solid medium.

③ 2nd. The gel is shaken in a 0.5 M HCl solution.

○ HCl treatment accelerates the transfer of DNA from the agarose gel to the membrane.

④ 3rd. The gel is shaken in a 1.5 M NaCl, 0.5 M NaOH solution (DNA denaturation).

○ The NaOH solution, sponge, and gel are stacked in order to separate DNA into single strands.

○ The sponge absorbs the NaOH solution and supplies it to the gel.

○ To prevent partial reformation of double-stranded DNA, an additional nucleic acid denaturant is added.

○ Nucleic acid denaturants: Metal ions, formamide, formaldehyde, etc. The form series weakens DNA binding strength.

⑤ 4th. The gel is shaken in a 1.5 M NaCl, 1 mM EDTA, 0.5 M Tris-HCl (pH 7.2) solution.

○ Tris-HCl: Buffer solution.

⑥ 5th. Capillary phenomenon + blotting

○ 5th-1st. Place a positively charged nylon filter and a paper towel on the gel.

○ Nitrocellulose paper can be used instead of nylon filter.

○ 5th-2nd. The NaOH solution that moves into the gel carries single-stranded DNA and travels to the paper towel stack, where it is absorbed.

○ 5th-3rd. Negatively charged single-stranded DNA sticks to nylon membrane.

⑦ 6th. Autoclave: Nylon membrane combined with DNA is treated under high pressure and high temperature to fix the bond.

○ DNA can also bind to the membrane when exposed to ultraviolet (UV) light.

○ In general, sterilization at 121°C for 15 minutes kills most microorganisms.

⑧ 7th. Blocking: A blocking agent is added to enhance clarity.

○ 7th-1st. Identify DNA that is considered insignificant through repeated experiments or electrophoresis for each allele.

○ 7th-2nd. Add salmon sperm DNA fragments to the DNA.

○ Purpose: To prevent probe DNA from non-specifically binding to the membrane during the 8th hybridization process (enhancing clarity).

○ Note: The membrane strongly interacts with DNA or proteins, requiring a blocking process.

⑨ 8th. Hybridization

○ 8th-1st. The nylon membrane with bound DNA is placed in a Seal-a-Meal bag.

○ 8th-2nd. At 62 °C, a radioactively labeled DNA probe is added to the Seal-a-Meal bag.

○ The labeled probe has a complementary sequence to the gene of interest.

○ Probe DNA: 100–500 bp, single-stranded DNA.

○ Probe DNA is typically labeled with the radioactive isotope P32.

○ Probe DNA can also be labeled with dyes such as EtBr or SYBR Green.

○ 8th-3rd. Probes bind to complementary DNA.

○ Through repeated experiments, other DNA considered insignificant is blocked and covered with salmon sperm DNA.

⑩ 9th. Eliminate unhybridized radioactive probes in seal-a-meal bags.

⑪ 10th. Autoradiogram: Irradiation on nylon membrane.

⑫ 11th. Identify the radioactive positions on the electrophoresis gel (or colony positions).

⑬ Housekeeping Gene

○ A gene that is present in all cells and continuously expressed.

○ Used as a control in Southern blotting.

○ Examples: β-actin (ACTB), tubulin, GAPDH, B2M, RPL11, 18S rRNA (most reliable).

⑭ Stringency

○ Conditions that lower stringency: Create an environment where probe binding is easier.

○ Low temperature or high salt concentration facilitates nucleic acid hybridization, leading to lower stringency.

⑵ Northern blotting: Nucleic acid probe hybridization for RNA.

① Very similar to Southern blotting but with the following differences.

Difference 1. Extraction of samples

○ Southern blotting uses DNA extraction.

○ Northern blotting effectively isolates mRNA from extracts by affinity chromatography using oligo dT.

Difference 2. Use of restriction enzyme

○ DNA dealt by Southern blotting is very long and must be used with restriction enzymes.

○ Northern blotting uses RNA, not DNA, so restriction enzymes cannot be used.

Difference 3. Type of solution in capillary action

○ Southern blotting uses basic solutions to denature single-stranded DNA.

○ In Northern blotting, RNA is degraded when exposed to an alkaline solution.

○ Northern blotting uses a salt solution to stabilize RNA.

Difference 4. Types of probe

○ Southern blotting uses gDNA probe.

○ Northern blotting uses cDNA probe.

⑶ Western blotting: Antibody probe hybridization for proteins.

① Numerous differences with nucleic acid blotting.

Difference 1. Type of gel

○ Nucleic acid blotting uses agarose gel.

○ Western blotting uses polyacrylamide gel, SDS-PAGE, etc.

Difference 2. Electrophoresis voltage condition

○ In nucleic acid blotting, nucleic acid transfers well to electrophoresis, so a low voltage must be applied.

○ In Western blotting, proteins are not well transported by electrophoresis, so high voltage must be applied.

Difference 3. Blotting Process

○ Nucleic acid blotting uses capillary phenomenon during blotting.

○ Western blotting uses an electric field during blotting.

Difference 4. Nucleic acid pretreatment

○ Nucleic acid blotting uses salmon sperm DNA fragments.

○ Western blotting uses casein, skim milk, bovine serum albumin (BSA), etc.

○ Tween: Surfactants such as Tween-20 are used.

○ A brand name developed by the American company Uniqema.

○ A detergent composed of a hydrophilic group (polyethylene glycol) and a hydrophobic hydrocarbon group.

○ Uses: Helps mix water and oil, disrupts cell membranes.

○ The numbers 20, 40, 60, 80 indicate increasing carbon chain length, making the detergent more hydrophobic as the value increases.

○ BSA can also be used for nucleic acid blotting.

Difference 5. Types of probe

○ Nucleic acid blotting uses DNA probes.

○ Western blotting uses antibody probe.

○ Primary antibody: An antibody that specifically binds to the target protein.

○ To ensure the specificity of the primary antibody, an enzyme extracted from a different species than the target animal must be used.

○ Secondary antibody: An antibody that specifically binds to the primary antibody. It is typically conjugated with an enzyme that breaks down a chromogenic substrate.

○ To ensure the specificity of the secondary antibody, an enzyme extracted from a species different from both the target animal and the primary antibody host must be used.

○ In Western blotting, the primary and secondary antibodies originate from different species.

Difference 6. Imaging Method

○ Nucleic acid blotting uses EtBr + self-radiation.

○ Western blotting uses the Coomassie Blue staining method.

⑷ DNA chip (microarray)

① Characteristics

○ Simultaneously detects the expression patterns of multiple genes.

○ Each spot contains a single cDNA probe.

○ Allows identification of tissue-specific gene expression using cDNA DNA chips.

○ Microarray data consists of continuous raw data, whereas RNA-seq raw data is count-based.

○ RNA-seq is more robust in detecting genes with extremely low or high expression levels compared to microarray data.

○ Becoming an obsolete technology due to advancements in NGS (Next-Generation Sequencing).

② Process

○ 1st. A cDNA library of human genes is spotted onto a glass slide to create a cDNA chip.

○ 2nd. The cDNA chip is treated with a 1% BSA solution.

○ BSA prevents nonspecific binding between the chip and DNA, allowing only complementary DNA to bind.

○ 3rd. mRNA X (from normal tissue) and mRNA Y (from abnormal tissue, e.g., cancer) are extracted separately.

○ 4th</sup. Oligo-dT is added to X and Y, binding complementarily to poly-A sequences in mRNA.

○ 5th. RT-PCR (Reverse Transcription PCR)

○ X: dNTP + Cy3 (green fluorescent dye)-dTTP.

○ Y: dNTP + Cy5 (red fluorescent dye)-dTTP.

○ Reverse transcription reaction is performed to generate fluorescently labeled cDNA.

○ 6th. 0.1 N NaOH is added to X and Y, incubated at 70°C for 10 minutes, then neutralized with 0.1 N HCl.

○ Template RNA is degraded due to basic condition.

○ 7th. Synthesized cDNA from X and Y is purified and mixed in equal amounts, then hybridized onto the cDNA chip.

○ 8th. The cDNA chip is washed with buffer solution to remove unbound sequences.

○ 9th. Fluorescence intensity of Cy3 and Cy5 is measured and corrected.

○ Black: Neither X nor Y is expressed.

○ Green: Only X is expressed.

○ Red: Only Y is expressed.

○ Yellow: Both X and Y are expressed.

⑸ ISH (in situ hybridization)

① Overview

○ An experimental technique that hybridizes a specific DNA sequence with a chromosome.

○ Used not only to determine the location of specific nucleotide sequences but also to identify the location of RNA.

○ Rapidly diagnoses chromosomal abnormalities but cannot diagnose DNA mutations.

Type 1. Fluorescence in situ hybridization (FISH)

○ Hybridize a specific nucleotide sequence with a fluorescent probe complementary to the nucleotide sequence, then observe using a fluorescence microscope.

○ 1st. Prepare epithelial tissue sections on slides.

○ 2nd. Treat with RNase, incubate at 37°C for 1 hour, then wash.

○ Probes that bind to DNA may bind to RNA, so there is a process to remove RNA.

○ 3rd. Remove the culture medium, add a 2% formaldehyde solution, and incubate for 15 minutes.

○ This step fixes the cells.

○ 4th. Remove the formaldehyde solution, add a 0.2% Triton X-100 solution, and incubate for 5 minutes.

○ Triton X-100 is a surfactant.

○ 5th. Remove the Triton X-100 solution, add 2 M HCl, then treat with pepsin and incubate at 37°C for 10 minutes.

○ Pepsin functions in acidic conditions, so the pH is adjusted accordingly.

○ Pepsin degrades intracellular proteins, facilitating probe penetration in subsequent steps.

○ 6th. Wash the sample.

○ 7th. Prepare a probe that binds to the centromere.

○ This probe contains biotin-labeled dTTP.

○ 8th. Hybridize the sample from step 4 with the probe from step 5.

○ 9th. Wash the sample.

○ 10th. After sufficiently treating with fluorescently labeled avidin, wash the sample.

○ 11th. Stain with DAPI.

○ DAPI (4’-6-diamidino-2-phenylindole) is a nuclear stain.

○ DAPI strongly binds to AT-rich regions of DNA minor groove.

○ It is also used as an apoptosis marker.

○ Since DAPI is cell-membrane-permeable, it can be used to stain both live and fixed cells.

○ When UV light is applied to DNA bound with DAPI, it emits blue fluorescence.

○ 12th. The centromere region will display avidin fluorescence, while DNA will appear as blue fluorescence.

Application 1. RNA ISH (In Situ Hybridization)

○ rRNA molecules are observed in situ rather than being isolated from individual cells.

○ A labeled riboprobe (complementary RNA sequence) hybridizes with the transcript.

○ Visualization is achieved through a probe that binds to the complementary transcript.

Application 2. WM ISH (Whole-Mount ISH) (1989)

○ Requires thousands of animals to create an atlas, making it less commonly used after 2010.

Type 2. Radioactive ISH: The first ISH method, developed in 1969.

⑹ Sequencing technology: Determines DNA nucleotide sequences.

⑺ Gene-trap screen



6. Gene deletion

⑴ Knock-out mouse: Study the function of specific genes.


image

Figure 4. Knockout process


① 1st. Knockout induction of gene X in embryonic stem (ES) cells

○ 1st-1st. Construction of a plasmid vector (targeting vector): Includes gene X which is inactivated by the insertion of neor (neomycin-resistant gene), along with a TK (thymidine kinase) gene located at a distance from the gene X.


image

Figure 5. The structure of plasmid vector


○ 1st-2nd. Insert targeting vector inside ES cell.

○ 1st-3rd. Some ES cells autonomously replace the existing gene X with the inactivated gene X.

○ To enable genetic recombination, the same gene must be included next to gene X.

○ 1st-4th. Selection: Cells are cultured in medium containing G418 (geneticin, neomycin derivative) and gancyclovir.

○ 1st-4th-1st. Untransformed ES Cells: Killed by G418.

○ 1st-4th-2nd. ES cells in which regions other than gene X have been replaced: These cells contain the TK gene, making them susceptible to ganciclovir-induced cell death (TK gene metabolizes ganciclovir into a toxic compound).

○ 1st-4th-3rd. Only ES cells in which only gene X has been replaced survive.

② 2nd. Selection of knockout ES cells.

③ 3rd. Inject knockout ES cells into the embryo.

④ 4th. The germ cells of the chimera mouse consist of both normal germ cells and germ cells derived from knockout ES cells.

⑤ 5th. F1 mice include both (+/+) individuals and (+/-) individuals.

○ Reason: Because the chimera mouse can be considered a heterozygous mutant.

⑥ 6th. Select (+/-) individuals among the F1 mice through genetic testing and perform self-crossing.

○ There are limitations in selection based on mouse fur color.

⑦ 7th. Among the F2 mice, 25% are knockout mice (homozygous mutants) and must be identified through genetic testing.

⑵ Cre-Lox

① The Cre gene encodes recombinase.

② Cre-mediated DNA recombination occurs only between identical lox sequences.

③ If the paired lox sequences are in the same orientation, DNA deletion occurs by Cre; if they are in opposite orientations, DNA inversion occurs by Cre.

⑶ miRNA, siRNA: Gene expression can be suppressed for all genes using RNA interference (RNAi).



7. Nuclear substitution

⑴ GMO (Genetically Modified Organism): Genetically altered substances, food, organisms.

⑵ Methods

① Nuclear Transplantation

○ Utilizes techniques such as microinjection.

○ Applicable to trait transformation and animal cloning.

② Trait transformation without using a vector

○ Can induce trait transformation using gene guns, etc.

Example 1. Formation of recombinant plant cells: Particles coated with recombinant genes are “shot” into plant cells.

Example 2. Recombinant plant cells → Callus → Whole plant formation (in nutrient medium).

○ Plant cells exhibit totipotency.

Example 3. Maintenance and propagation of purebred

Example 4. Proliferation of useful plants

Example 5. Tissue culture with meristematic regions

Example 6. Induction of differentiation

⑶ Examples

Example 1. Recombinant animals: Mass production of beneficial gene products in animals.

Example 2. Edible vaccines

Example 3. Genetically Modified Food (GM Food)

○ Past: Gene-modified crops obtained by increasing the frequency of specific alleles through selective breeding (artificial selection).

○ Present: Genetic recombination technology → Increased shelf life, production rate (resistance to pests, weeds, diseases, drought, and cold).

Example 3-1. Golden rice: Genetically modified to produce beta-carotene (increases nutritional value of rice).

GMO Debate and Principles of Safety Assessment



8. DNA-protein interaction studies

⑴ Gene footprinting assay (DNA footprinting technology)


image

Figure 6. Application of gene footprinting


① Genes binding with proteins appear to be missing on the gel electrophoresis.

② It allows the identification of transcription factor binding sites.

⑵ Electrophoretic mobility shift assay (EMSA; gel shift assay, GMSA)

① Confirms the binding between the transcription factor and the promoter.

② 1st. Probe production: Labels DNA with radioactive isotopes.

③ 2nd. Electrophorese the protein-DNA hybrid, followed by radiation sensitivity analysis.

④ 3rd. Result analysis


image

Figure 7. Example of EMSA results


○ Premise: Since the top is the cathode and the bottom is the anode, DNA will move from top to bottom. (-) refers to the case where only the probe is moved.

○ Interpretation of a: Protein A and Protein B bind to the probe, and A and B bind together with the probe (they could also bind exclusively with each other…).

○ Interpretation of b: Protein C and Protein D bind to the probe, and C and D do not bind to each other.

○ Interpretation of c: Protein E binds to the probe, while protein F does not bind to the probe but binds to protein E.

⑤ Generally, protein means transcription factor and it can be applied to measure transcription activity.

ChIP (Chromatin Immunoprecipitation)

⑷ South-Western Blotting


image

Figure 8. South-Western Blotting


① 1st step: Proteins are initially subjected to Western blotting.

② 2nd step: Subsequently, DNA labeled with fluorescence is subjected to Southern blotting.

⑸ Yeast One-Hybrid Assay

⑹ Filter Binding Assay: Only transcription factors bind to the filter.

⑺ DNA Affinity Chromatography



9. Protein-protein interaction studies

⑴ Dual Hybrid System: A method for identifying proteins that interact with protein X.

① 1st. Transcription factor gene is divided into two parts: DNA binding domain and transcription activation domain.

② 2nd. Bait: Protein X + DNA binding domain.

③ 3rd. Prey: Protein of interest for determining binding + transcription activation domain.

④ 4th. When the two hybrid proteins interact and bind, the prey induces the expression of a reporter gene.

○ Example of a reporter gene: GFP protein.

⑵ Yeast Two-Hybrid: Protein-protein interactions are used to identify protein binding relationships.

⑶ Protein separation and purification using GST-tagged fusion proteins.

⑷ Phage Display Method: Used to obtain monoclonal antibodies.

① Objective: To obtain a monoclonal antibody that binds to a specific protein.

② Method for obtaining monoclonal antibodies in vitro.

③ Conventional monoclonal antibodies: B lymphocytes are fused with myeloma cells (hybridoma), becoming stem cells that proliferate and produce a large amount of antibodies.

④ 1st step. By introducing mutagens and altering the base sequence, a vast phage library is created.

⑤ 2nd step. Screen for phages with desired activity and later confirm the nucleotide sequence.



10. Gene editing

⑴ CRISPR-Cas9 Gene Editing Technology

① Overview

○ Gene Scissors: A mechanism of gene cleavage in bacteria (e.g., E. coli) to eliminate viral DNA.

○ CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)

○ Derived from DNA fragments of bacteriophages that infected prokaryotic cells.

○ Found in 50% of bacterial genomes and 90% of archaeal genomes.

○ CRISPR arrays consist of repetitive DNA sequences and spacers located between each repetition.

○ Bacteria or archaea record spacers, which are fragments of external DNA such as viruses, and add repeat sequences.

○ Subsequently, the CRISPR-Cas9 system detects and cleaves viral DNA that matches the spacer.

○ Cas9 (CRISPR-associated protein 9)

○ Acquired from organisms like Streptococcus pyogenes. It has two nuclease domains, HNH and RuVC9.

○ dCas9 (nuclease-deficient Cas9, dead Cas9): Unable to cut DNA but capable of binding to DNA through sgRNA.

Step 1. CRISPR Processing: The following describes the CRISPR processing of the subtype I-C/Dvulg Cas5d system in Bacillus halodurans.

○ 1st. Cas5d recognizes hairpin structures and 3’ single-stranded sequences in the CRISPR repeat region, cleaving pre-crRNA into unit-length fragments.

○ 2nd. Pre-crRNA processing: Further processing of pre-crRNA into smaller crRNAs.

○ 3rd. Cas5d forms a complex with crRNA, Csd1, and Csd2 proteins.

○ 4th. The crRNA portion in this complex detects and removes viral DNA.


image

Figure 9. CRISPR Processing


Step 2. Reaction to External Nucleic Acids

○ 1st. Cas9 creates a bubble in the double-stranded DNA, and the complementary sgRNA (small guide RNA) binds to the target RNA next to the PAM site within that bubble.

○ The bubble is referred to as DNA double-strand breaks (DSBs).

○ sgRNA: Fragments of viral DNA that remain in the genomic DNA of bacteria (e.g., E. coli) surviving viral infection. Also known as Spacer or gRNA (guide RNA).

○ PAM (Protospacer Adjacent Motif) site: A 5’-NGG-3’ nucleotide sequence that distinguishes self from non-self.


image

Figure 10. Location of sgRNA in dsDNA


○ 2nd. The sgRNA-Cas9 complex cleaves the template DNA, non-template DNA, and sgRNA at the same location.

○ 3rd. Cas9 and sgRNA separate.

○ 4th. The DNA that formed the bubble reanneals through hydrogen bonding.

○ 5th. The cleaved template and non-template DNA undergo DNA repair mechanisms.

○ 5th-1st. Viruses without DNA repair mechanisms are eliminated by the CRISPR/Cas9 mechanism.

○ 5th-2nd. Non-homologous End Joining (NHEJ): Allows for the joining of completely different chromosomes, following the Holliday model.

○ 5th-3rd. Homology Directed Repair (HDR): Effects such as the substitution of specific bases or the addition of repeat sequences occur.

○ 6th. In genetic recombination, research is conducted to replace the unnecessary parts of VNTR genes with useful DNA.

○ Utilizes homology-directed repair mechanisms.

○ Can be applied not only to DNA but also to RNA, epigenomes, and other single nucleotides for editing.

○ Types of gene editing: OE (overexpression), KD (knock-down).

④ Off-Target Issues

○ Definition: The problem of editing regions other than the target gene when applying the CRISPR/Cas9 system to gene therapy.

○ Streptococcus pyogenes Cas9 (SpCas9) nuclease is commonly used due to its efficiency but has a high off-target ratio.

Solutions

○ Nuclease mutation

○ PAM sequence modification

○ gRNA truncation


image

Figure 11. CRISPR/Cas9 Technology Diagram


⑤ Applications

○ For signal transduction research purposes: Multiplexing guide RNAs enable perturbation screening.

○ For imaging research purposes.

○ For drug research purposes: Studying changes in drug uptake when specific genes are suppressed.

○ Gene therapy: Editing genes responsible for rare diseases.

○ Temporal sequencing (e.g., Record-seq).

⑵ Lysogenic viruses: The viruses primarily used in gene therapy are adenoviruses and retroviruses.

⑶ siRNA, miRNA Therapeutics: Current market situation is not favorable

⑷ Nucleic Acid Delivery Systems

① Overview

○ Adopted commercially with great success by Moderna and Pfizer during the COVID-19 pandemic.

○ Since mRNA is unstable in the body, a delivery vehicle is necessary to ensure stability until it reaches the target tissue or cell.

Type 1. mRNA Drug Delivery Systems

○ Solid Lipid Nanoparticle (SLN): The most popular mRNA delivery vehicle adopted by Moderna and Pfizer.

○ A single SLN of 80-100 nm can encapsulate around 100 mRNA molecules.

○ Examples: ALC-0315 (Pfizer/BioNTech), SM-102 (Moderna), ALC-0159 (Pfizer/BioNTech), PEG-DMG (Moderna).

○ Cationic liposome

○ Polymer and polymer/lipid hybrid particle

○ Micelle

○ Emulsion

Type 2. DNA Drug Delivery Systems

Type 3. AAV(adeno-associated virus)

○ Utilized as a formulation for treating Alzheimer’s disease.


image

Figure 12. Comparison between AAV and LNP


⑹ Mega Nuclease

⑺ TALEN (Transcription Activator-Like Effector Nuclease)

⑻ ZFN (Zinc Finger Nuclease)



Input: 2015.7.03 21:57

Modify: 2019.1.25 00:18

results matching ""

    No results matching ""