[go: up one dir, main page]

Academia.eduAcademia.edu
                                                                                                                                                                                        !"#$       %  "  &'  ' (  ($     )*+,$ # -   .*/#%0 1 (     *23)   )   !%4*+)* $   !   #           5,1 6   5 *+*7 %   "   & '    ' (   ($    Laboratory Techniques in Genetic Engineering Dr. H.K. Garg Professor Department of Biotechnology Institute for Excellence in Higher Education, Bhopal 1 Contents 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. RNA Purification from Blood Affinity Purification of Total RNA Gene Isolation Isolation of Cellular DNA 3.1 DNA Extraction from Strawberry Plant 3.2 Isolation of DNA from Banana 3.3 Isolation of Genomic DNA from Plant Source by CTAB Method 3.4 Isolation of Chromosomal DNA by Lysozyme Method 3.5 Isolation of Plasmid DNA from Bacteria 3.6 DNA Extraction: Organic Method 3.7 Silica Absorption Method 3.8 Inorganic Method 3.9 Chelex Method 3.10 Differential Method Qualification of Nucleic Acids 4.1 Spectrophotometry 4.2 Characterization of DNA by Spectrophotometric Assay and Melting Temperature (Tm) 4.3 Fast Technology Analysis Recombinant DNA Technology 5.1 Designing DNA Probes Bergs Terminal Transferase - Boyer Cohen Chang Experiment Preparation of Competent Cells for Efficient DNA Uptake in E. coli Bacterial Transformation in vitro Using Electroporation 8.1 CaCl2 mediated Transformation Agarose Gel Electrophoresis Protein Analysis by SDS-PAGE Blue-White Colony Selection employing X-Gal / IPTG Genomes and Interrelatedness In situ Hybridization Amplification of DNA using Polymerase Chain Reaction 05 09 13 22 24 26 28 31 34 36 39 41 44 46 48 50 54 60 64 66 69 72 77 80 83 88 92 96 101 110 2 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 14.1 Real-time PCR 14.1.1 Optimization of Annealing Temperature 14.1.2 Reverse Transcription PCR | RT-PCR Digestion of DNA with RE 15.1 Digestion of DNA with RE in Bacteriophage Southern Blotting of DNA Northern Blotting of RNA Western Blotting Dot Blot Immunoprecipitation Sanger Sequencing Maxam-Gilbert Sequencing Pyrosequencing Multiplex DNA Sequencing Automated Sequencing Construction of Molecular Maps Restriction Mapping Molecular Markers 28.1 Restriction Fragment Length Polymorphism 28.2 Random Amplified Polymorphic DNA 28.3 Amplified Fragment Length Polymorphism 28.4 DNA Chip and Microarrays Genomic Library cDNA Library DNA Fingerprinting Genetic Selection and Screening Method 32.1 Use of Chromogenic Substrates 32.2 Antibiotic Sensitivity Test for Detection of Recombinants 32.3 Insertional Inactivation 32.4 Complementation of Defined Mutations Protein Engineering 33.1 Rational Design Stem Cell Therapy Reverse Genetics Transgenic Technology 36.1 Transgenic Plants 116 117 123 130 133 140 143 146 150 154 157 161 164 168 171 174 177 180 180 182 184 186 189 192 196 199 199 203 206 209 213 213 217 220 223 223 3 36.2 Transgenic Animals Buffers and Reagents Some Common Lexes References 224 227 231 239 4 RNA Purification from Blood RNA extraction is a technique used to isolate RNA from biological samples. This is a challenging process due to the presence of ribonuclease enzymes, which can quickly break down RNA. The most common method used is TRIzol extraction, which starts with one full Yellow-Top (type A) BD Vacutainer tube of human blood, roughly equal to 8 milliliters, to yield around 30 micrograms of RNA. It's essential to use RNAse-free tubes and solutions and maintain a clean environment as RNA is highly sensitive to RNases, and autoclaving does not inactivate them. Principle The TRIzol method isothiocyanate, a is based on powerful protein the use of denaturant, guanidinium and acidic phenol/chloroform to separate RNA from the other components in the sample. Keep the pH low so that only RNA will be separated in the aqueous phase otherwise, at neutral pH, DNA will be separated. Stock 1. 10x RBC Lysis Buffer 10.0 g KHCO3 2.0 ml 0.5 M EDTA 89.9 g NH4Cl • In a large container, dissolve 10.0 g of KHCO3, 2.0 ml of 0.5 M EDTA, and 89.9 g of NH4Cl in approximately 800 ml of ddHO. • Adjust the pH of the solution to 7.3 by adding or removing ddHO, as necessary. 5 • Mix the solution thoroughly. • Transfer the solution to a tightly closed bottle and store at 2-8°C for up to 6 months. 2. 1x RBC Lysis Buffer • Take the 10x stock solution and dilute it 1:10 with ddHO. • Store the solution at room temperature up to 1 week. 3. TRIzol Reagent or RNA STAT-60 Reagent: Purchase either TRIzol Reagent (Invitrogen Life Technologies, Cat No. 15596018) or RNA STAT-60 Reagent (Tel-Test, Cat No. CS-111). 4. Other Reagents  Phosphate Buffered Saline (PBS)  Isopropanol (2-propanol)  Ethanol  RNAse-free water  RNAse-Away (a cleaning solution to neutralize RNAses on laboratory equipment). Procedure 1. Take a blood sample and transfer it to a 50 ml polypropylene conical centrifuge tube. Adjust the volume of the sample to 45 ml by adding RBC Lysis Buffer. 2. Let the sample stand at room temperature for 10 minutes. 3. Pellet the cells at 600 xg for 10 minutes using a centrifuge. Remove the supernatant and gently resuspend the pellet in 1 ml of RBC Lysis Buffer. 4. Transfer the resuspended pellet to a 1.5 ml microcentrifuge tube. 6 5. Pellet the cells for 2 minutes at room temperature using a microcentrifuge. Remove the supernatant and resuspend the pellet in 1 ml of sterile DPBS. 6. Pellet the cells again and remove the supernatant. 7. Add 1200 μl of TRIzol solution to each tube and resuspend the cells. Add 0.2 ml of Chloroform (CHCl3) and vortex each tube for 15 seconds. 8. Centrifuge the samples at 13,000 rpm for 10 minutes at 4°C. Transfer the upper phase to a clean microcentrifuge tube. 9. Remove ~20% of the upper phase for future collection of micro-RNA. Add an equal volume of cold isopropanol to the remaining upper phase and mix by inverting. 10. Store the samples in a -20°C freezer for precipitation. 11. Centrifuge the samples again at 13,000 rpm for 10 minutes at 4°C. Remove the supernatant and rinse the pellet with 0.5 ml of ice-cold 75% ethanol. 12. Centrifuge the samples again. Remove the supernatant along with any remaining liquid in the bottom of the tube with a pipette. 13. Allow the pellet to dry for 5 to 10 minutes. Dissolve the RNA pellet by adding 20 μl of RNAse-free H2O to each sample. 14. Measure the RNA concentration within 2 hours of elution and store at -80°C. Note: The stock solution of RBC Lysis Buffer is stable for 1 week at room temperature and the RNA solution is stable for 6 months at 2– 8°C in a tightly closed bottle. Advantages TRIZol or Tri is a popular reagent used for RNA extraction. The advantages of using this reagent include the combination of phenol and guanidine isothiocyanate, which provides benefits from both 7 reagents. This combination effectively removes protein and DNA, but the success of the extraction largely depends on the user's pipetting skills. Also, the RNA is protected during the extraction process. Disadvantages However, there are also some disadvantages of using TRIzol or Tri. The extraction process is heavily dependent on the user's pipetting skills and any disturbance to the phases can result in contamination. Furthermore, phenol and chloroform are harmful reagents that should be handled with caution, such as under a hood. Finally, this method of RNA isolation may not be suitable for some samples such as low-input or degraded RNA. 8 Affinity Purification of Total RNA The Affinity purification of total RNA involves isolating high-quality mRNA for further use in molecular biology techniques. The principle behind this technique is the affinity selection of polyadenylated mRNA using oligodeoxythymidylate [Oligo (dT)]. In simple terms, this means that the process uses a specific type of molecule, oligodeoxythymidylate, to selectively target and purify the mRNA that has a polyadenylated tail. This allows for the isolation of highquality mRNA for further use in molecular biology techniques. The first step in this process is to extract the total RNA from the sample of interest. This RNA contains both mRNA and non-coding RNA. Thereafter, the extracted RNA is mixed with oligodeoxythymidylate, which will specifically bind to the polyadenylated mRNA. The mixture is then passed through a column that separates the mRNA bound to the oligodeoxythymidylate from the rest of the RNA. Finally, the column is washed to remove any contaminants and the purified mRNA is eluted, or released, from the column. This highquality mRNA is then ready for use in molecular biology techniques, such as cDNA library construction. Stock To perform this procedure, several materials are required. It's crucial that all materials used in this process are sterile and of molecular biology grade, including:  RNase-free water  SDS  Oligodeoxythymidylate-cellulose [oligo (dT)]  RNase-free glass wool 9  Pasteur pipettes  5 M NaCl  3 M Sodium acetate pH-6.0  Absolute alcohol, 70% ethanol  Loading buffer  Elution buffer  Recycling buffer. All Tris-containing solutions should be prepared using RNase-free water and autoclaved, while all other solutions should be treated with diethyl pyrocarbonate (DEPC) and autoclaved, but for Tris, which inactivates DEPC. To prepare RNase-free water, add 0.1% DEPC to water, let it stand overnight at 37°C, and then autoclave it to destroy residual DEPC activity. SDS should be weighed in a fume hood, as it is dangerous, if inhaled. A 10% stock solution is usually prepared, but it is unstable if autoclaved, so it should be heated at 65°C for 2 hours to destroy any residual RNase activity. Oligo (dT) cellulose should be purchased commercially and suspended in loading buffer at a concentration of 5 mg/1 ml. It should be stored either dry at 4°C or in suspended condition in loading buffer at -20°C. The glass wool and Pasteur pipettes should be wrapped in aluminum foil and baked at 200°C for 2 to 4 hours to remove any RNase activity. The rest of the materials should be stored as follows:  5 M NaCl (Store at room temperature).  3 M Sodium acetate pH-6 (Store at room temperature).  Absolute alcohol (Store at -20°C).  70% ethanol (Prepare this solution using DEPC-treated water. Store at 4°C).  Loading buffer (0.5 M NaCl in 0.5% SDS, 1 mM EDTA, 10 mM Tris-HCl - pH 7.5 Store at room temperature). 10  Elution buffer (1 mM EDTA, 10 mM Tris-HCl - pH 7.5. The buffer can be stored at room temperature but should be preheated to 65°C prior to use).  Recycling buffer (0.1 M NaOH, which should be prepared immediately before use and used fresh). Note that this method is dependent on the purity of the samples, and it may not work on low-input or degraded RNA. Also, when handling DEPC, gloves should be worn, as hands are a major source of RNase activity, and DEPC is a carcinogen that should be handled in a fume hood with extreme care. Procedure Step 1: Preparing an Oligo (dT) Column: 1. Take a syringe and remove the plunger. 2. Fill the base of the syringe with glass wool. 3. Take a sterile, RNase-free Pasteur pipette and add the oligo(dT) cellulose on top of the glass wool. 4. The oligo(dT) cellulose will form a column above the glass wool. 5. Oligo (dT) column is now ready for use. Step 2: Isolating Poly (A+) RNA 1. Resuspend the RNA pellet in loading buffer. 2. Heat the RNA pellet to denature it. 3. Load the denatured RNA onto the oligo (dT) column. 4. Wash the column to remove any unbound RNA. 5. Add elution buffer to the column to recover the bound poly (A+) mRNA. 6. Precipitate and wash the mRNA to obtain pure and intact mRNA. 11 Advantages 1. Elimination of organic solvents. 2. Compatible with a variety of sample types. 3. DNase treatment to eliminate contaminating genomic DNA. Disadvantages 1. May be less efficient in obtaining RNA. 2. More expensive compared to other methods. 12 Gene Isolation Gene isolation is a process in molecular biology and genetics that involves separating a single gene from its surrounding DNA sequence in the genome. The goal of gene isolation is to obtain a pure, functional copy of a gene, which can then be studied, manipulated, or expressed in vitro. The process of gene isolation can involve a variety of techniques, including restriction enzyme analysis, PCR, and hybridization, and can be used to study the function of specific genes or to develop new therapies for genetic diseases. DNA Extraction and Purification DNA is a crucial element in molecular biology and can be found in various sources including human tissues, blood, hair, leaves, bacteria, insects, and more. DNA extraction is a process of taking DNA from cells or tissues of interest. This technique is the first step in the study of specific DNA sequences, genomic structure, DNA fingerprinting, restriction fragment length polymorphism (RFLP), and PCR analysis. DNA extraction is done through different methods such as mechanical methods where the cells are blended to release the DNA, chemical methods where detergents or enzymes are used to break down the cell membrane or physical methods where centrifugation is used to separate the DNA. Apart from these, there are various methods that can be used to perform DNA extraction, such as organic extraction, salting out, magnetic separation and silica-based technology. The choice of method depends on factors such as tissue type, DNA concentration, sample number, safety, and cost. 13 After extraction, the DNA is purified to get rid of any contaminants that may still be present. This is done through methods such as ethanol precipitation, column-based purification, or size-exclusion chromatography. The final purified DNA is then stored in a buffer solution at low temperature for future use. There are various techniques used for DNA extraction, but all of them include the following fundamental processes: 1. Cell lysis: This is the breakdown of the cellular structure, which releases long strands of DNA. Depending on the source, cell lysis can be done chemically, physically, or a combination of both. For example, plant and bacterial samples' cell walls are broken by applying physical force, while chemical agents such as lysozyme, EDTA, and detergents are used for lysis in other sources. 2. Elimination of membrane lipids: After lysis, DNA is processed to remove further contaminants before the membrane lipids are extracted. This is done by washing the DNA. 3. Protein denaturation and removal: Proteins can interfere with molecular biology experiments, so they are denatured and removed using an enzyme called protease. 4. Removal of additional cellular elements: Through repeated washing processes, other biological components are separated from the DNA. 5. Denaturation and removal of RNA: RNA is a significant DNA contaminant, so it is denatured and removed using an enzyme called RNase. 14 6. DNA elution and storage: The purified DNA is eluted in an alkaline buffer solution or in double distilled water and stored at -20°C for future use. Stock Chemical Ethylene diamine tetra acetate (EDTA), NaOH, Tris-HCl, sucrose, MgCl2, Triton X100, Sodium dodecyl sulphate (SDS), NaCl, Sodium perchlorate, TE buffer or double distilled water, cold chloroform, cold ethanol. Preparation of solutions 1. 0.5 M EDTA, pH 8.0 Add 146.1 g of anhydrous EDTA to 800 ml of distilled water. Adjust pH to 8.0 with NaOH (about 20 g). Make up the volume to 1 L with distilled water. 2. 1 M Tris-HCl, pH 7.6 Dissolve 121.1 g of Tris base in 800 ml of distilled water. Adjust pH with concentrated HCl (about 60 ml). Make up the volume to 1 L with distilled water. 3. Reagent A (Red Blood Cell Lysis Solution) 0.01M Tris-HCl (pH 7.4), 320 mM Sucrose, 5 mM MgCl2, and 1% Triton X100. Add 10 ml of 1 M Tris to 109.54 g of sucrose, 0.47 g MgCl2 and 10 ml Triton X100 to 800 of distilled water. Adjust pH to 8.0; make up the volume to 1 L with distilled water. 4. Reagent B (White blood Cell Lysis Solution) 15 0.4 M Tris-HCl, 150 mM NaCl, 0.06 M EDTA, 1% SDS, pH 8.0. Take 400 ml of 1 M Tris (pH 7.6), 120 ml of 0.5 M EDTA (pH 8.0), 8.75 g of NaCl, adjust pH to 8.0 with NaOH. Make up the volume to 1 L with distilled water. Autoclave at 15 psi for 15 min. After autoclaving the mixture, add 10 g of SDS. Procedure 1. Obtain 3 ml of whole blood and place it in a 15-ml falcon tube. 2. Add 12 ml of reagent A to the tube and mix for 4 minutes using a rolling or rotating blood mixer at room temperature. 3. Centrifuge the mixture at 3000g for 5 minutes at room temperature. 4. Discard the supernatant without disturbing the cell pellet and remove any remaining moisture by blotting onto tissue paper. 5. Add reagent B and 250 μL of 5 M NaCl to the tube and mix by inverting several times. 6. Place the mixture in a water bath at 65°C for 15 to 20 minutes. 7. Add 2 ml of ice-cold chloroform and mix on a shaker for 20 minutes. 8. Centrifuge the mixture at 2400g for 2 minutes. 9. Transfer the upper phase to a clean falcon tube using a sterile pipette. 10. Add 2 to 3 ml of ice-cold ethanol to the tube and gently invert to allow DNA to precipitate. If necessary, add more ethanol. 11. Using a clean Pasteur pipette, spool the DNA onto the hooked end and transfer to a 1.5-ml microcentrifuge tube. 16 12. Centrifuge the microcentrifuge tube at 6000 rpm for 5 minutes. 13. Gently remove the supernatant (ethanol layer) without disrupting the DNA pellet and leave to dry. 14. Resuspend the DNA in 200 μL of TE buffer or doubled distal water and label. 15. Determine the yield and purity of the extracted nucleic acid if necessary. The results should show a cloudy precipitation, representing the isolated genomic DNA, visible to the naked eye. Alternative Method Step 1: Lysis  Add lysis buffer to the blood sample  The lysis buffer will break open the red blood cells and release the DNA into solution Step 2: Precipitation  Add alcohol (such as ethanol or isopropanol) to the lysed blood sample  The DNA will precipitate out of solution and can be collected by centrifugation Step 3: Wash  Wash the DNA pellet with a buffer to remove remaining contaminants Step 4: Resuspension  Resuspend the purified DNA in a buffer of your choice 17 Step 5: Quantification  Quantify the purified DNA using spectrophotometry or fluorometry. Alternate method to extract genomic DNA from lizard Stock 1. SE Sodium chloride (75 mM)1M 37.5 ml EDTA (25 mM) 500 Mm 25.0 ml Distilled water 2. 437.5 ml Sodium acetate 3M Sodium acetate 24.6 gm Distilled water 100.0 ml (adjust pH to 5.2 with glacial acetic acid) 3. TEN Tris (pH 8.0) (10 mM) 1M 5.0 ml Sodium chloride (100 mM) 1 M 50 ml EDTA (pH 8.0) (25 mM) 500 mM 25 ml Distilled water 4. 420 ml TE Tris (pH 8.0) (10 mM) 1 M 1ml EDTA (1 mM) 500 mM 0.2 ml Distilled water 98.8 ml 5. SDS 10% and Proteinase K (10 mg/ml) 6. Distilled and buffered saturated phenol 7. Phenol–Chloroform-Isoamyl alcohol (25:24:1) 8. Absolute alcohol 9. Ethanol 70%, prechilled at - 20oC 18 Laboratory ware 1. Homogenizer 2. Micropipettes 3. Pipette tips 4. Centrifuge tubes 5. Eppendorf tubes 6. Cheese clothe Procedure 1. Homogenize a Calotes embryo at 4°C in SE (salt-EDTA buffer). 2. Centrifuge the homogenized sample at 4000 rpm for 10 minutes at 4°C, then discard the excess liquid. 3. Redissolve the remaining particle in 20 ml of TEN (TrisEDTA-NaCl) buffer at room temperature. 4. Add SDS (sodium dodecyl sulfate) to the mixture to a final concentration of 1% and proteinase K to a final concentration of 100 ug/ml. 5. Incubate the mixture at 37°C for an overnight period. 6. Add an equal amount of phenol to the mixture, stir in a vortex mixer for 15 minutes, then centrifuge for 10 minutes at 12000 rpm. 7. Transfer the aqueous phase from the previous step to a new tube, and add an equal volume of phenol, chloroform, and isoamyl alcohol. Centrifuge and discard the organic phase. 8. 9. Repeat the above step to further purify the DNA. Add an equal volume of chloroform-isoamyl alcohol to the aqueous phase, transfer the aqueous phase to a new tube after centrifugation. 10. Combine 1/30 volume of sodium acetate (3M) with 2 volumes of 100% ethanol and maintain at -20°C for 1 hour. 19 11. Transfer the precipitated DNA to a new Eppendorf tube containing 70% ethanol (chilled at -20°C). 12. Centrifuge the tube briefly, extract as much of the remaining alcohol as possible, lyophilize the DNA, and dissolve in TE (Tris-EDTA) buffer. Alternative Method Step 1: Tissue Collection  Obtain fresh or frozen tissue samples from the lizard, such as blood, muscle, or liver.  Store the tissue samples in a clean and sterile container. Step 2: Lysis  Homogenize the tissue samples using a mechanical  Ensure that the tissue is fully lysed to release the cellular homogenizer or a chemical lysis buffer. contents, including the DNA. Step 3: Cell Debris Removal  Centrifuge the lysate to separate the cell debris from the DNA-containing supernatant.  Carefully pour off the supernatant, leaving behind the cell debris. Step 4: DNA Purification  Use a DNA purification kit or ethanol precipitation to purify the DNA from the supernatant.  Follow the instructions provided with the DNA purification kit or refer to the appropriate protocols for ethanol precipitation. 20 Step 5: DNA Quality and Quantity  Check the quality and quantity of the purified DNA using methods such as spectrophotometry, gel electrophoresis, or PCR.  Ensure that the purified DNA is of sufficient quality and quantity for your intended use. Notes The specific protocol may vary based on the source of the tissue and the DNA purification kit used. To avoid cross-contamination, take proper precautions and maintain sterile conditions throughout the extraction process. 21 Isolation of Cellular DNA Stock  Coconut endosperm  Sodium chloride  Sodium citrate  Mortar and pestle  Centrifuge tubes  Absolute alcohol Procedure 1. Grind 200mg of tissue (coconut endosperm, spleen, heart, testis, or kidney) in a saline citrate solution (85ml 0.9% sodium chloride solution and 15ml 0.5% sodium citrate solution with pH 7.4). 2. Transfer the homogenate to a centrifuge tube, bring the volume to 10ml with saline citrate solution. 3. Centrifuge at 3000 rpm for 8 minutes, discard the supernatant. 4. Re-homogenize the pellet with 5 ml saline citrate solution, adjust the volume to 10 ml, repeat the centrifugation process for 8 minutes and discard the supernatant. 5. Suspend the pellet in 12% sodium chloride and centrifuge at 10,000 rpm for 15 minutes in a refrigerated centrifuge. 6. Transfer the supernatant to a 30 ml test tube, add 2-3 volumes of absolute alcohol, mix gently. 7. Collect the fibrous white DNA by winding it around a clean glass rod. 22 8. Transfer the DNA into a 1.5 ml Eppendorf tube, add 1 ml 70% alcohol, centrifuge at 1000 rpm for 5 minutes, discard the supernatant. 9. Dry the DNA pellet; dissolve in 2 ml distilled water, measure optical density at 260 nm wavelength in a spectrophotometer. 23 DNA Extraction from Strawberry Plant The process of isolating pure genomic DNA from plant tissue is crucial for studying plant genetics and making changes to plant genes and metabolic pathways. This process is different from extracting DNA from animal sources due to the presence of a tough cellulose cell wall and a larger DNA molecule. The goal of the isolation process is to obtain pure and high-quality DNA without any cellular material or degradation. To achieve this, the cell wall and cellular membranes need to be broken down through either mechanical or non-mechanical methods. Mechanical methods use physical force to open the cell wall, while nonmechanical methods use enzymes or chemicals along with physical force to break down the cell wall components. Once the cell wall is broken, the cell membrane forms small cracks, which allows for the use of detergents to break down the cell membrane. The DNA is then separated from the protein by using isopropanol or ethanol. The final product is a clean DNA suspended in a buffer or distilled water. Stock Chemical 1. Strawberry 2. Extraction solution 3. 96% cold ethanol or isopropanol 24 4. TE buffer or double distilled water. 5. Preparation of extraction solution Add 100 ml detergent to 750 ml of distilled water and then add 11 g NaCl. Make up the volume to 1 L with distilled water. Equipment and Glassware Microfuge centrifuge, electronic balance, razor blade, mortar and pestle, cheese cloth, funnel, graduated cylinder 25 ml, beaker 50 ml, test tube, centrifuge tube, Pasteur pipette, micropipette, and tips. Procedure 1. Remove the green leaves from the strawberry and weigh the plant using a sensitive balance. 2. Chop the plant into small pieces using a clean razor blade. 3. Mix the chopped plant with the extraction solution for 5 minutes using a pestle. 4. Pour the mixture through cheese cloth into a clean beaker. 5. Pipette a small amount of the mixture into a test tube and add cold ethanol or isopropanol. 6. The DNA will appear as a clear white thread, spool it onto the hooked end of a Pasteur pipette. 7. Transfer the DNA to a centrifuge tube and spin it at 6000 rpm for 5 minutes. 8. Remove the supernatant (ethanol layer) gently, leaving the DNA pellets to dry. 9. Suspend the DNA pellets in TE buffer or double distilled water. Result Cloudy precipitation will be obtained, and it represents the isolated DNA. 25 Isolation of DNA from Banana Stock  Fleshy berry fruits like banana, grapes, etc.  Liquid soap  Distilled water  Salt (NaCl)  Ice-cold isopropyl alcohol (IPA)  Measuring spoons  Glass stirring rod  Test tubes  Glass beakers  Plastic cups  Strainer or coffee filter  Funnel Principle The DNA extraction protocol involves disrupting the cell wall, cell membrane, and nuclear membrane of plant cells to release the DNA into solution, followed by precipitation and removal of contaminating biomolecules. Procedure 1. Grind a piece of banana in a mortar and pestle to form a mash or blend. 2. Add 1/2 cup of distilled water to the banana mash and transfer the mixture into a glass beaker. 26 3. In a plastic cup or zip lock bag, mix 1 teaspoon of liquid soap and 1 teaspoon of salt with 2 tablespoons of distilled water. Stir gently until the salt and soap are dissolved. 4. Add 2 tablespoons of the banana mash mixture to the cup containing the salt and soap solution. Stir the mixture for 1015 minutes using a glass stirring rod. 5. Filter the fruit mixture through a fine sieve or coffee filter. 6. Chill a test tube of IPA by placing it in a beaker containing ice cubes and water. 7. Using a dropper, slowly add the filtrate to the chilled IPA in the test tube. 8. 9. Place the test tube undisturbed for 5-6 minutes. The isolated DNA will appear as a white precipitate in the alcohol layer. 10. Gently spool out the DNA using a hook, bent paperclip, or glass stirring rod. Observation The DNA will precipitate out into the alcohol layer, appearing as white stringy mucus. Result The experiment yielded a good quantity of DNA, making it suitable for use in biological experiments and biotechnology applications. 27 Isolation of Genomic DNA from Plant Source by CTAB method Stock  Young and tender leaves (Tulsi and Bryophyllum)  Mortar and pestle  Conical flask  Measuring cylinder  Distilled water  Ethidium Bromide  Ethanol  Micropipette  Centrifuge  Electrophoresis unit  CTAB buffer  Water bath  Eppendorf tube  CTAB buffer: CTAB - 2 g Tris HCl (1 M)10 ml EDTA (0.5 M) 4 ml NaCl (5 M) 28 ml H2O 40 ml PVP 40 1 g 28 Principle The CTAB method uses a detergent to break open plant cells and solubilize their contents. The DNA is then extracted from the cell homogenate, with RNA removed by RNAase, and DNA precipitated and washed in organic solvents before being redissolved in aqueous solutions. Procedure 1. Take young and tender leaves, wash them with distilled water, and grind them using a mortar and pestle. 2. Transfer the powdered leaves into an Eppendorf tube and add 50 ml of CTAB buffer. 3. Incubate the plant homogenate for 15 minutes at 55°C in a 4. After incubation, spin the tube at 12,000 rpm for 5 minutes water bath. at 4°C. Transfer supernatant to another Eppendorf tube and discard the pellet. 5. Add 250 ml of phenol: chloroform: isomyl alcohol (25: 24: 1) and mix the solution. 6. Spin the tube at 12,000 rpm for 1 minute at room temperature. Transfer the aqueous phase to another tube. 7. Add 500 ml of isopropanol and mix by inverting the centrifuged tube at 10,000 rpm for 5 minutes at room temperature. Discard the supernatant. 8. Add 1 ml of 70% ethanol and mix with the pellet by inverting it. 9. Centrifuge the tube at 12,000 rpm for 1 minute at room temperature. 10. Discard the supernatant and let the pellet dry on ice for 15 30 minutes. 11. Add distilled water to dissolve the DNA. 29 12. Warm the DNA solution at 65°C for 20 minutes. 13. Quantitate the DNA and check its purity. 14. Run the DNA on a gel. Results DNA was successfully isolated from the leaves using the CTAB method. Precautions  Use soft leaves.  Avoid over-drying the DNA as it may become difficult to suspend in TE.  Handle the water bath carefully.  Pre-chill the CTAB buffer and wash it in 70% ethanol before use. 30 Isolation of Chromosomal DNA from E. coli using the Lysozyme Method To isolate chromosomal DNA, mechanical barriers in bacterial cells need to be disrupted, including the plasma membrane, cell wall, and outer membrane in gram-negative bacteria. In this experiment, Trisbuffer containing EDTA is used to make the cells isotonic and prevent them from bursting. Lysozyme treatment is used to attack N-Acetalglucosamine residues of bacterial cell walls, making the weakened cell wall porous and exposing periplasmic spaces. SDS treatment is used to dissociate the cell membrane, followed by treatment with phenol-chloroform to denature proteins and separate aqueous organic phases. Chilled isopropanol is used to precipitate DNA from the aqueous phase, and reprecipitation with 70% ethanol is performed to eliminate divalent cations. The resulting DNA palate is relatively pure and is suspended in Tris-EDTA buffer. Stock  E. coli cells  Saline-EDTA  SDS (sodium dodecyl sulphate)  Chloroform: isoamyl alcohol  Ethanol  Phenol  Tris-HCl  NaCl 31  RNase Chemical preparation  TE buffer: Dissolve Tris-HCl and EDTA together at their respective molarities to create a single solution.  TGE buffer: Mix 25 mM Tris-HCl pH 8.0, 50 mM glucose, and 10 mM EDTA.  10% SDS of pH 6.8-7.2.  10 mg/ml RNase: Weigh 10 mg pancreatic ribonuclease and dissolve in 1 ml of 100 mM Tris-HCl of pH 7.5 containing 150 mM NaCl. Boil in a water bath for 10 minutes.  Prepare 70% (v/v) ethanol in autoclaved double-distilled water.  Chill isopropanol by refrigeration.  Lysozyme: Dispense 20 mg lysozyme in 5 ml TGE buffer and keep on ice until use.  Solutions preparation:  Solution A: Saturated phenol with 0.1 g of α- hydroxyquinoline and 100 ml melted phenol, followed by transfer into a separating funnel to allow layers to form. Remove the aqueous layer containing Tris-HCl and impurities. Collect the phenol layer in an amber-colored bottle.  Solution B: Mix chloroform and isoamyl alcohol in the ratio 24:1.  Solution C: Mix solution A and B in the ratio 1:1. Procedure 1. Grow E. coli cells in LB broth until they reach the log phase (indicated by an absorbance of 0.4). Harvest 50 ml of bacterial suspension by centrifuging at 10,000 rpm for 10 minutes. Gently discard the supernatant. 32 2. Resuspend the pellet in 1 ml of TGE buffer. Spin the mixture at 5,000 rpm for 5 minutes and carefully remove the supernatant. 3. Add 150 μL of lysozyme solution, vortex, and let it sit at room temperature for 30 minutes. 4. Slowly add 30 μL of RNase solution to the walls of the Eppendorf tubes, and incubate them undisturbed at 37°C for 30 minutes. 5. Slowly add 50 μL of 10% SDS to the walls of the tubes, mix gently to prevent the formation of froth, and incubate at 37°C for 2 hours. 6. Add 230 μL of solution C and vortex for 2 minutes to mix well. Centrifuge the mixture at 10,000 rpm for 15 minutes at 4°C. 7. Using a micropipette, carefully transfer the supernatant aqueous phase to another tube. Add an equal volume of icecold isopropanol and mix well. 8. Spin the mixture at 10,000 rpm at 4°C for 10 minutes and carefully discard the supernatant. 9. Add 0.5 ml of chilled 70% ethanol to the pellet to precipitate the DNA. Recover the DNA by centrifuging at 10,000 rpm for 10 minutes and discard the supernatant. Dry the pellet in air. 10. Dissolve the pellet in 50-100 μL of 10 mM TE buffer and keep at 50°C for 5 minutes for better dissolution. Electrophorese the isolated DNA on an agarose gel. Result The lysozyme method successfully isolated DNA from bacteria. Precautions  Wear gloves while performing the experiment.  Handle phenol with care. 33 Isolation of Plasmid DNA from Bacteria Plasmids are circular DNA molecules found in bacteria that carry genetic information separate from the chromosomal DNA. Their isolation is significant for genetic engineering as plasmids can confer resistance to various substances, act as vectors in recombinant DNA technology, and play a role in bacteria's survival and adaptation. In order to isolate plasmid DNA, bacteria are grown in a suitable medium overnight or until reaching an optical density of 0.6-1.0 at 600 nm, and stored, if necessary, at 4°C. The bacteria are then lysed with a lysis buffer, lysozyme, Tris-HCl, EDTA, and NaCl, and SDS. The lysozyme degrades the bacterial cell wall, while the Tris-HCl, EDTA, and NaCl neutralize the charge and protect the DNA from degradation by nucleases. The SDS denatures the chromosomal DNA and makes it more accessible to degradation by proteases. The plasmid DNA can be extracted through phenol-chloroform extraction and ethanol precipitation, followed by washing with 70% ethanol and resuspending in TE buffer. The purified plasmid DNA can be used for further experiments like restriction digestion. Stock 1. Bacterial culture 2. Lysis buffer (such as lysozyme, Tris-HCl, EDTA, NaCl) 3. Sodium dodecyl sulfate (SDS) 4. Phenol-chloroform 34 5. Isopropanol Equipment 1. Centrifuge 2. Microcentrifuge tubes Procedure 1. Grow the bacterial culture in a suitable medium, such as Luria-Bertani (LB) broth, overnight to reach an optimal density of bacteria. 2. Lyse the cells by adding a lysis buffer such as a solution of NaOH, EDTA, and SDS and incubating the mixture at 65°C for 10 minutes. This will break open the bacterial cell walls and release the plasmid DNA into the solution. 3. Neutralize the lysis solution by adding a neutralization buffer, such as Tris-HCl, to restore the pH to 7.0. 4. Centrifuge the lysis solution at high speed to separate the bacterial debris (protein and other contaminants) from the supernatant containing the plasmid DNA. 5. Precipitate the plasmid DNA from the supernatant by adding an equal volume of isopropanol. The isopropanol will cause the plasmid DNA to form a dense band, which can be collected and purified. 6. Dialyze the plasmid DNA to remove the excess salts and impurities. 7. Precipitate the plasmid DNA from the dialysis solution by adding ethanol and sodium acetate. The plasmid DNA will form a pellet, which can be washed with 70% ethanol to remove any remaining impurities. 8. Dry the plasmid DNA pellet, and dissolve it in a suitable buffer, such as TE buffer, for downstream applications. 35 DNA Extraction Organic Method The process of extracting DNA from a material involves several important factors, including the efficiency of the extraction, the amount of DNA obtained, the removal of impurities, and the quality and purity of the DNA. One common technique for DNA extraction is the organic method, also known as the phenol-chloroform method. This method is particularly effective for extracting large amounts of high molecular weight DNA, which was necessary for early DNA fingerprinting techniques. Principle The principle behind this method is to mix a watery sample with a solution of phenol and chloroform in equal parts. Upon mixing and centrifugation, the mixture separates into two distinct layers - an aqueous phase on top and an organic phase at the bottom. The nucleic acids and other impurities stay in the aqueous phase, while proteins move into the organic phase. Stock (Reagents) 1. Chloroform 2. Ethanol  Ether (optional)  Nucleic acid solution to be purified  Phenol:Chloroform (1:1)  Tris EDTA(pH7.8) (optional)  3 M sodium acetate pH 5.2 or5 M ammonium acetate 36  100% ethanol Equipment 1. Automatic pipette fitted with a disposable tip 2. Pipettes, large-bore (optional) 3. Polypropylene tube 4. Rotating wheel(optional) Procedure The step-wise method for the organic method is as follows: 1. Add an equal volume of phenol: chloroform to the nucleic acid sample. Mix the tube's contents to create an emulsion. 2. Centrifuge the mixture for one minute at room temperature at 80% of the tubes' maximum speed. Transfer the aqueous phase to a new tube, discarding the organic phase and interface. 3. Repeat steps 1-4 until there are no longer any proteins visible at the organic / aqueous phase interface. 4. Repeat steps 2-4 with an equal volume of chloroform. Measure the aqueous phase's volume in order to retrieve DNA. 5. Add 0.1 volume of pH 5.2 sodium acetate with a 0.3 M final concentration. 6. Add two to three litres of cold 100% ethanol. Keep at -20 degrees Celsius for longer than 20 minutes. 7. Spin a microfuge for 10-15 minutes at its fastest speed. Decant the supernatant carefully. 8. Add 1 cc of 70% ethanol and mix. Spin quickly and decant the supernatant with care. 37 9. Dry the pellet with air or briefly vacuum. Resuspend the pellet in the required volume of double-distilled water or Tris EDTA buffer. 10. Store and carry out quantification and intended usage The approach discussed has various advantages, one being its versatility in being used on different samples. However, it also has some limitations, such as being a time-consuming process, being susceptible to contamination and posing a risk to the scientist as it involves handling hazardous substances. 38 Silica Absorption Method The silica absorption method is a widely used process for purifying DNA from different sources like blood, saliva, or plant tissue. This process involves several steps to isolate the DNA from impurities like proteins, lipids, and polysaccharides. Firstly, the sample is lysed using a buffer containing detergents to release the DNA. Then, the lysate is mixed with a solution containing silica particles and high salt concentrations, (which cause the DNA to bind to the silica under specific conditions, such as the use of specific salts and pH levels) and the impurities remain in the solution. The silica-DNA complex is then washed with a buffer containing high salt to remove impurities. The purified DNA is finally eluted from the silica particles by reducing the salt concentration. The purified DNA can then be checked for quality and quantity. There are variations of this method, such as using chaotropic agents to enhance the efficiency of purification, and the choice of buffer, salt concentration, and elution method can impact the purity and yield of the purified DNA. The protocol may also need to be adjusted for different types of samples, such as FFPE samples. It's essential to be careful of cross-contamination and to take appropriate measures to reduce the risk of contamination. Although the exact mechanism of the silica DNA extraction method is not entirely understood, it remains a widely used technique in many laboratories. 39 Stock To perform this technique, kits are available that include all necessary materials. Procedure 1. Prepare the DNA sample for the extraction process. 2. Run the sample through a microchannel. 3. Allow the DNA to bind to the silica in the channel. 4. Wash away any other molecules in the buffer solution. 5. Purify the channel to remove any contaminants. 6. Dry the silica. 7. Extract the DNA by using either water or a buffer with low salt concentration. 8. Collect the DNA at the end of the channel. Advantages  Quick  Dependable  High-quality DNA yield Disadvantages  Expensive  Interference from certain sample sources like chewing gum. 40 Inorganic method The non-organic method is a way to clean up DNA or RNA without using any chemicals derived from organic materials. The key to this method is adding a specific enzyme, Proteinase K, to the mix. This enzyme helps to protect the DNA or RNA from other enzymes, known as nucleases that can break down the nucleic acids during the purification process. Stock Reagents  Digestion Buffer (10 mM NaCl, 10 mM TRIS (pH 8.0), 10 mM EDTA (pH 8.0),  0.5%SDS  Proteinase K (20 mg/ml)  Sodium Acetate pH 5.2 (3M)  Ice-cold 98% ethanol  Ice-cold 70% ethanol  1X TE  Water  Tissue Equipment 1. Incubator 2. Centrifuge 3. Sterile 1.5-ml micro-centrifuge tubes 41 Procedure The non-organic method includes: Step 1: Tissue Digestion 1. Obtain a clean micro-centrifuge tube and add 1.5 ml of the digestion buffer. 2. Calculate the appropriate amount of proteinase K to add to the buffer based on the formula: 5 μl of proteinase K for every ml of digestion buffer. 3. Homogenize the tissue in the solution by mixing. 4. Incubate the mixture for 1-12 hours at 55°C. This can be done overnight. 5. Vortex the mixture for a short period of time. 6. Centrifuge the mixture at high speed for two minutes at 4°C, discarding the top layer as you go. 7. Pour the top aqueous layer into a new sterile microcentrifuge tube. 8. Discard the bottom layer. Step 2: Precipitation of Protein and Cell Debris 1. Fill a sterile 1.5 ml micro-centrifuge tube with 0.1 volume of sodium acetate (pH 5.2). 2. Close the tube and invert it to gently stir the contents. 3. Incubate the mixture for 15 minutes at -20°C. 4. Centrifuge the mixture at high speed for 20 minutes at 4°C, removing the top layer. 5. Transfer the top aqueous layer into a new sterile microcentrifuge tube. 6. Discard the bottom layer. 42 Step 3: Precipitation of Nucleic Acids 1. Fill a sterile 1.5 ml micro-centrifuge tube with 2 volumes of ice-cold 98% ethanol. 2. Close the tube and invert it to gently stir the contents. 3. Incubate the mixture for 15 minutes at -20°C. 4. Centrifuge the mixture at maximum speed for 20 minutes at 4°C and discard the supernatant. 5. Add 1 ml of ice-cold 98% ethanol and vortex the mixture briefly. 6. Centrifuge the mixture at maximum speed for five minutes at room temperature and discard the supernatant. 7. Add 1 cc of ice-cold 70% ethanol and vortex the mixture 8. Centrifuge the mixture at maximum speed for five minutes briefly. at room temperature and discard the supernatant. 9. Repeat steps 7-10 with 1 ml of ice-cold 70% ethanol (optional). 10. Use air to dry the pellet. 11. Add 10 μl of 1XTE (Option 1) or 10 μl of water (Option 2). 12. Resuspend the pellet by shaking or vortexing. The advantage of this method includes the ability to use it in the presence of substances that break down proteins, such as SDS and urea, as well as with agents that bind to metal ions, like EDTA, and reagents that interact with sulfhydryl groups and enzymes like trypsin or chymotrypsin. 43 Chelex Method The Chelex method is a process for preparing DNA for PCR, which is a commonly used method for amplifying DNA. This technique uses a special resin, called Chelex, to protect the DNA from being damaged by enzymes called DNAases. These enzymes can break apart DNA, making it impossible to use for PCR. The Chelex resin binds to essential elements for DNAases, like magnesium ions, and disables them, thus safeguarding the DNA from harm. Stock  Chelex 300 μL  Heating Block  ddH2O  Sterile Forceps  Vortex  Centrifuge tubes Procedure The method for using Chelex to extract DNA is as follows: 1. Obtain 1 spare tube for a control and 3 pre-made tubes filled with 300 L 10% Chelex from the refrigerator. Handle the container while wearing gloves, and shake the desired number of tubes into your gloved hand. 2. Label each Chelex tube with the sample number it belongs 3. Activate the heating block and preset it at 95 °C. Water is to and the date and initials. poured into the openings. 44 4. Sterilize forceps by dipping them in ethanol and then waving them across an alcohol burner's flame to ignite. Repeat this process twice more. 5. Remove a small piece of tissue from the sample using sterile forceps. Place the tissue in the Chelex tube with the correct label, and screw the cap back on. 6. Repeat step 5 for each sample, sterilizing the forceps three times between samples. Create a negative Chelex control by dipping sterilized forceps into a tube of Chelex slurry. 7. Vortex the samples in the Chelex slurry for 10 to 15 seconds. 8. Spin the samples rapidly in a microcentrifuge for 10-15 seconds. 9. Heat the samples to 95 °C for 20 minutes. While the tubes are incubating, check to make sure the covers have not fallen off. 10. Vortex the samples once again for 10 to 15 seconds. 11. Spin the tubes rapidly once more to make sure all the contents are at the bottom of the microcentrifuge tube. 12. The samples are now usable for PCR. Advantages and Disadvantages The Chelex method has benefits such as being cost-effective, fast, and safe as it does not involve any dangerous chemicals. However, it has some drawbacks like not working well with blood samples, producing DNA samples of poor quality, and not being suitable for DNA analysis using restriction fragment length polymorphism. 45 Differential Method The differential extraction technique is a method used to separate sperm cells from other cell types for DNA extraction. This technique, also known as differential lysis, is commonly used in sexual assault cases to compare the DNA profiles of the perpetrator and the victim. The sperm cells are more challenging to lyse compared to other cells due to the presence of protein disulfide links in their outer membrane. The differential method includes several steps, including an optional wash phase, non-sperm cell lysis, and sperm cell lysis. Principle Differential extraction, also known as differential lysis, is a method used to separate DNA from two different types of cells without blending their contents. It is commonly used in forensic investigations of sexual assault cases, where the DNA from sperm cells and vaginal epithelial cells is extracted to compare the DNA profiles of the attacker and the victim. The reason why sperm cells can survive the extraction process better than epithelial cells is because of the presence of protein disulfide links in the outer membrane of sperm cells. These links protect the DNA from being damaged during the extraction process. Procedure Differential Extraction can be carried out as follows: 1. This is an optional step to remove pollutants and cellular waste. To carry out this step, add a buffer and detergent to the sample and incubate it either at room temperature or in 46 a refrigerator. Discard the supernatant (wash fraction) after the sample has been cleaned. 2. Mix the sample with an extraction buffer that contains a buffer, detergent, and proteinase K. Incubate the mixture to lyse all cells but for spermatozoa. Remove the supernatant (fraction 1) containing the DNA from the lysed cells and wash the sperm pellet with a buffer several times to remove extra DNA. 3. Incubate the pelleted sperm cells with a buffer, detergent, DTT, and a stronger dose of proteinase K to lyse the sperm cells. This will result in fraction 2. 4. Extract each fraction, including the wash fraction if necessary, with a phenol/chloroform/isoamyl alcohol mixture to purify the DNA. Advantage and Disadvantage Differential extraction is a method used in forensic science to separate the DNA from multiple contributors in a sample, such as a male culprit and a female victim. This process is important because it helps eliminate confusion and improve accuracy when identifying the sources of DNA. By following quality assurance standards, the process ensures the quality of the DNA extraction. However, there is a potential drawback to differential extraction as the sperm head must be strong enough to withstand the extraction process or the results may be unreliable. 47 Qualification of Nucleic Acids Qualification of nucleic acids refers to the process of characterizing and verifying the purity and integrity of nucleic acid samples, such as DNA or RNA. It is an important step in many molecular biology and biotechnology applications, such as cloning, sequencing, and gene expression analysis. There are several methods that can be used to qualify nucleic acids, including: Spectrophotometry This method uses a spectrophotometer to measure the absorbance of nucleic acids at different wavelengths. The ratio of absorbance at 260 nm to 280 nm (A260/A280) is used to calculate the purity of the nucleic acids. A ratio of around 1.8 is considered to indicate a pure sample of DNA, while a ratio of around 2.0 is considered to indicate a pure sample of RNA. Gel electrophoresis This method uses agarose or polyacrylamide gel electrophoresis to separate nucleic acids based on size and charge. The separated nucleic acids can then be visualized under UV light after staining with ethidium bromide or a similar dye. This method can be used to confirm the size and integrity of the nucleic acids, as well as to detect contaminants such as proteins or salts. 48 Fluorometry Fluorometry uses a fluorometer to detect nucleic acids by measuring their fluorescence. It is a very sensitive method, which can quantitate the amount of nucleic acid present in a sample. It can also be used to determine the ratio of double-stranded to single-stranded nucleic acids, providing information about the integrity and quality of the sample. Quantitative PCR (qPCR) QPCR is a method that uses fluorescent dyes and PCR amplification to quantify nucleic acids. It can be used to measure the copy number of specific sequences in a sample, and also to determine the quality and integrity of the nucleic acids. Other Methods Other methods like gel filtration, centrifugation, electrophoretic mobility shift assay (EMSA) and others can also be used to qualify nucleic acids. It's important to note that, depending on the downstream application, different methods might be more appropriate for different samples. Therefore, it's important to carefully consider the sample type and downstream application when choosing which method(s) to use for nucleic acid qualification. 49 Spectrophotometry To measure the amount of DNA or RNA in a sample, scientists use a process called quantitation of nucleic acids. This is important because reactions that use these types of molecules require a specific amount and purity for best results. There are different methods to determine the concentration of nucleic acids, including spectrophotometry and UV fluorescence. Spectrophotometry works by exposing the sample to ultraviolet light and measuring the amount of light that passes through the sample. The sample absorbs the ultraviolet light in a specific pattern, and the more light that is absorbed, the higher the concentration of nucleic acids in the sample. This measurement is done using a machine called a spectrophotometer and it detects the light absorbed at a specific wavelength of 260 nm. Principle Using the Beer-Lambert Law, it is possible to relate the amount of light absorbed to the concentration of the absorbing molecule. The extinction coefficient, which is the amount of light absorbed, is different for different types of molecules. For example, doublestranded DNA has an extinction coefficient of 0.020 (μg/ml) −1cm−1 at a wavelength of 260 nm, while single-stranded RNA has an extinction coefficient of 0.025 (μg/ml) −1 cm−1. This means that the concentration of the absorbing molecule can be calculated based on the amount of light absorbed. A value of 1 Absorbance (A) corresponds to a concentration of 50 μg/ml for double-stranded DNA. However, this method is only valid for up to an A of 2. For 50 more accuracy, a prediction of the extinction coefficient can be made using the nearest-neighbor model, especially for short singlestranded oligonucleotides which are dependent on the length and base composition. Stock  UV/VIS Spectrophotometer  1 ml quartz cuvette  DNA sample(s)  TE Buffer  Disposable 1 ml polyethylene Transfer Pipettes(Berol) [2 per group]  Eppendorf tubes (1.5 ml) [2 per group]  Ruler  Kim wipes Procedure Step 1: Setting up the Spectrophotometer (BeckmanDU64) 1. Turn on the spectrophotometer at the power strip and ensure that the printer is connected and ready. 2. Turn on the UV lamp source and allow it to warm up for 5 minutes. 3. Select the absorbance reading mode (ABS key). Press the SCAN key, "Edit" will be displayed. 4. Enter the starting wavelength as 280 nanometers (nm) and press enter. Enter the ending wavelength as 260 nm and press enter. The speed for the scan of the sample will be displayed. It should read 750 nm/min. If it does not, press the STEP key and scroll through the options until 750 nm/min is displayed. Press enter. 51 5. Upper limit will be displayed. Set the upper limit at 2,000 absorbance. Press enter. 6. Lower limit will be displayed. Set the lower limit at 0.000 absorbance. Press enter. The starting wavelength will reappear. The instrument is now ready to be calibrated against a control solution. The purpose of the calibration is to measure and then subtract from the sample absorbance any absorbance from the buffer solution. 7. Place 200 microliters (μL) of the TE Buffer into the quartz cuvette. This is the solution you will use to calibrate the instrument. 8. Open the sample compartment lid on the instrument. Carefully wipe the cuvette with a Kimwipe and be careful not to get fingerprints on the quartz panels. Place the cuvette into the cuvette holder so that the quartz sides are in the path of the light source (left to right). 9. Close the sample compartment lid. Press CALB. The absorbance of the TE Buffer solution will now be recorded in memory as the "background" and "Bkg" will be displayed. 10. Press READ. Calibration is complete when "Scan" is displayed. The instrument is now ready to measure DNA samples. 11. Open the sample compartment lid. Discard 200 microliters of TE Buffer from the cuvette by pouring it out and disposing of it properly. 12. Thoroughly rinse the cuvette twice with the TE Buffer solution and then carefully drain the cuvette onto a Kimwipe to remove any remaining droplets. 52 Step 2: Sample Preparation 1. Carefully add 200 microliters of the DNA sample to the cuvette and place the cuvette in the sample holder of the spectrophotometer. 2. Initiate the measurement process by pressing the READ button. The spectrophotometer will measure and record the absorbance of the sample between 260 and 280 nanometers and will then plot the results as a graph on the printer. 3. Repeat steps 2 and 3 for any other DNA samples that you have been assigned to analyze. Advantages 1. It is easy to perform. 2. It is cost-effective. 3. It provides reliable results if done correctly. Disadvantages 1. It cannot reliably assess protein contamination. 2. It does not contribute much error to DNA quantity estimation. 3. This method requires a spectrophotometer and cuvettes. 53 Characterization of DNA by Spectrophotometric Assay and Melting Temperature (Tm) To make sure that the extracted DNA samples are suitable for further experiments, it's important to check their quantity and quality. This is called characterization and can be done using different methods. In this experiment, two methods will be used: spectrophotometry and DNA melting temperature analysis. Spectrophotometry is a way to figure out how much and how pure the DNA is. The DNA absorbs light at 260 nm and 234 nm in ultraviolet light, and the 260 nm is the most important for measuring the amount of DNA. The ratio of the light absorbed at 260 nm and 280 nm can tell us if there's any protein contamination, since proteins absorb more light at 280 nm. The ratio of light absorbed at 260 nm and 230 nm can also help us make sure that the sample is pure and free from other contaminants like carbohydrates, peptides, and ethanol. DNA melting temperature analysis is used to see how stable the DNA is and what its GC content is. This method involves heating up a diluted DNA solution and measuring the light absorbed at 260 nm as the two strands of the double helix slowly separate. 1. To determine the concentration and purity of extracted DNA using UV spectrophotometer. 54 2. To determine the DNA melting temperature and GC content percentage. Principle The melting temperature (Tm) is a crucial characteristic of DNA that determines its stability. It is defined as the temperature at which half of the DNA molecules are no longer paired. The Tm value is determined by the length and the GC content of the DNA, which is the proportion of guanine and cytosine nucleotides. The GC content is significant for the stability of the DNA and can be measured using a Tm profile. To ensure that the extracted DNA samples are suitable for further use, spectrophotometry and DNA melting temperature analysis are performed. These techniques are used to determine the concentration, purity, and stability of the DNA, ensuring that it meets the requirements for downstream applications. Stock  The extracted DNA  0.1 X SSC buffer Preparation of 20X SSC buffer Dissolve 175.3 g of NaCl, 88.2 g of sodium citrate dehydrate in 800 ml distilled water. Adjust pH to 7.0 with diluted HCl. Make up the final volume to 1 L with distilled water. Procedure 1. Dilute the extracted DNA in 0.1 X SSC buffer. To do this, take 1 ml of the stock DNA and mix it with 10 ml of the buffer, resulting in a 1:10 ratio of DNA to buffer. 55 2. Measure the concentration and purity of the DNA sample using a spectrophotometer. Place the sample in a quartz cuvette, and use a second cuvette filled with distilled water as a blank. Set the spectrophotometer to measure the absorbance of nucleic acids at a wavelength of 10 mm. The results will be given in μg/ml. 3. Alternatively, you can measure the absorbance at three specific wavelengths (230, 260, and 280 nm) to determine the concentration and purity of the DNA. 4. Determine the melting temperature of the DNA. Dilute the stock DNA to a concentration of 10 μg/ml in 0.1 X SSC buffer and place it in a quartz cuvette. Fill a separate test tube with 1 ml of distilled water as a blank. Cover both tubes with aluminum foil and place them in a water bath at 25°C for 4 minutes to equilibrate. 5. Transfer the sample and blank to quartz cuvettes and place them back in the water bath for 1 minute to equilibrate. Read and record the absorbance at 260 nm. 6. Increase the temperature of the water bath to 50°C, 60°C, 70°C, and boiling, and repeat the steps of equilibrating the sample and blank and reading the absorbance at 260 nm. Observe the change in absorbance to determine the melting temperature of the DNA. Wavelength (nm) 230 Absorbance of DNA R P a l 260 280 56 Results A. Characterization of DNA by Spectrophotometric Assay (concentration and purity): Find out the concentration of the DNA samples using the following equation: Concentration of DNA (μg/ml) = (A260 / ε L)× Dilution factor (DF) Determine the purity of the DNA samples by calculating A260/A280 and A260/A230 ratios. B. Melting Temperature Temperature (°C ) 25 DNA Absorbance at 260 nm R P a l 50 60 70 Boiling Plot the value of absorbance vs. temperature and calculate the Tm for sample DNA. Find out the GC content of your sample using the following formula: (G + C)% = (Tm - 69.3) x 2.44. 57 Alternative Method for Characterization Step 1: Extraction of DNA Sample 1. Obtain the sample to be tested, such as blood, saliva, or tissue. 2. Follow the appropriate DNA extraction protocol to isolate the DNA. Step 2: Concentration and Purity Analysis 1. Transfer a known volume of the extracted DNA solution to a cuvette. 2. Load the cuvette into a UV spectrophotometer and run the analysis. 3. The spectrophotometer will measure the absorbance of the DNA sample at 260 nm, which is proportional to the concentration of DNA. 4. The ratio of the absorbance at 260 nm to 280 nm (A260/A280) is used to determine the purity of the DNA. A ratio of 1.8 or higher is considered pure. Step 3: Determination of DNA Melting Temperature 1. Load the DNA sample into a thermocycler and run a melting temperature (Tm) analysis. 2. The Tm is the temperature at which half of the DNA is melted. It can be used as an indicator of DNA quality and purity. Step 4: Analysis of GC Content Percentage 1. Transfer a known volume of the extracted DNA sample to a 2. Add reagents to the PCR tube, including Taq polymerase PCR tube. and primers. 58 3. Run a PCR reaction to amplify the DNA. 4. Load the amplified DNA product into an electrophoresis gel and run the analysis. 5. The GC content of the DNA can be estimated based on the position of the DNA fragments on the gel. Note: The steps outlined above are general and may vary depending on the specific protocols used in the laboratory. 59 Fast Technology Analysis Fast technology analysis method is not a specific method that is used in DNA extraction or purification. It could refer to any technology or method that allows for faster analysis or processing of DNA samples. For example, it could refer to the use of high-throughput sequencing technologies such as Illumina or PacBio, which can rapidly generate large amounts of DNA sequencing data. Or it could refer to automation of lab processes such as using robotic liquid handlers for DNA extraction and PCR set-up. In general, fast technology analysis method can be used in various stages of the DNA analysis process, such as sample preparation, DNA isolation, amplification, sequencing, and data analysis. The use of these methods can increase the speed and efficiency of DNA analysis, resulting in faster and more accurate results. It's important to note that, while fast technology analysis method can be very powerful, it's also important to consider the possibility of errors and artifacts that may occur with these technologies. Therefore, quality control measures and proper validation are essential to ensure the accuracy and reliability of the results obtained. Paper extraction The fast technology analysis (FTA) paper extraction method is a simple technique used to extract DNA, initially in forensic science, but now widely used in other fields as well. The process involves 60 smearing a sample, typically blood, onto a piece of paper, allowing it to dry. Then, circles are punched out from the dried paper, and put into a test tube. The extracted DNA is then cleaned using a solvent, before finally being added to a polymerase chain reaction (PCR) mixture. This method is known for its ease and simplicity, which is why it has gained popularity in various fields. Principle The FTA (fluid transfer assay) method is a process used to preserve and protect DNA in biological samples such as blood and saliva. The idea behind this method is that when these samples are applied to a special type of paper, the biological material will stick to the paper while the chemical mixture used in the process will break down the cells and change the proteins. Once the sample has been dried and stored properly, the nucleases, which are enzymes that break down DNA, are no longer active. This leaves the DNA stable and protected from damage caused by factors such as oxidation, ultraviolet light, bacteria, and fungus. The FTA method helps to minimize the damage to nucleic acids and ensures that the DNA remains intact for further analysis. Stock FTA Purification Reagent  TE Buffer (10mM Tris-HCL, 0.1 mM EDTA, pH 8.0)  Solution 1:0.1N NaOH, 0.3mM EDTA, pH13.0  Solution 2:0.1M Tris-HCL, pH 7.0 Procedure Step 1: Washing 61 1. Obtain a 6 mm punch from a dry area using a standard single-hole paper punch. 2. Place the punch in a 1.5 ml microtube and rinse the cutting end with ethanol to prevent cross-contamination. 3. Pour 1000 μl of the FTA purification reagent into the microtube and mix the mixture by either manual stirring or flash vortexing five times. 4. Allow the mixture to sit at room temperature for 5 minutes, and mix it again if desired. 5. Using a pipette, remove and discard all used FTA purification reagent. 6. 7. Repeat steps 2-4 a total of three times. Add 1000 μl of TE buffer to the microtube and incubate at room temperature for 5 minutes. 8. Using a pipette, remove and discard all used TE Buffer. 9. Repeat steps 6-8 twice more for a total of three washes with TE buffer. The punch should be white or pale in color after this process. Step 2: pH Treatment 1. Obtain a 6 mm punch that has been previously rinsed and add 140 μl of Solution 1. 2. Incubate the punch at 65°C for 5 minutes (this is different from the typical room temperature protocol used by the FTA company). 3. Add 260 μl of Solution 2 and mix the mixture by flashing the 4. Allow the mixture to sit at room temperature for an vortex five times. additional 10 minutes. 5. Vortex the mixture again for 10 flashes. 6. Remove the punch and squeeze it to recover the most elute volume possible. 62 7. Use a clean pipette tip to remove the punch if needed. 8. The elute carries 66 mM Tris-HCl, 0.1 mM EDTA. For a 25 μl PCR reaction, use 0.5 μl of the elute. Advantage The FTA method has several benefits that make it an attractive option for storing and preserving DNA. Firstly, it does not require refrigeration and can be kept at room temperature, making it convenient for storage and transportation. Secondly, it is effective in killing harmful bacteria while preserving the DNA, ensuring its quality and integrity. The small size of the discs makes it easy to store and ship, and the process of obtaining the DNA is simple and can be repeated multiple times without having to measure the amount of DNA beforehand. Disadvantage However, there is also one disadvantage to the FTA method - the small discs of DNA can be easily contaminated by static electricity. This means that there is a risk of contamination during handling, which could impact the quality of the DNA obtained using this method. 63 Recombinant DNA Technology Recombinant DNA technology, also known as genetic engineering, is the process of manipulating the genetic makeup of an organism by inserting, deleting or replacing specific genes. This technology allows for the production of genetically modified organisms (GMOs) with desired traits, such as resistance to disease or improved nutritional content. The technology comprises the extraction of a specific gene or DNA segment from an organism, which is then inserted into a vector like a plasmid. The vector is then introduced into a host organism, which can be a plant, animal, yeast, or bacteria. Once inside the host organism, the vector replicates and expresses the inserted DNA. This technology has numerous applications across various fields such as agriculture, medicine, and biotechnology. For instance, in agriculture, GMOs with pest and disease resistance can be produced, leading to increased crop yields. In medicine, it is utilized to produce human insulin and other therapeutic proteins, while in biotechnology, it is used for enzyme and industrial product production. However, the use of recombinant DNA technology also raises safety and ethical concerns, such as potential unintended effects on the environment and human health from the modification of food crops. There are also ethical concerns regarding the use of recombinant DNA technology in medical treatments, like gene therapy. Hence, it's crucial that any application of recombinant DNA technology 64 undergoes thorough evaluation for safety and ethical implications and is continuously monitored and regulated for the future. 65 Designing DNA Probes DNA probe designing is the process of creating a DNA fragment that can selectively hybridize to a complementary DNA target sequence. DNA probes are used to detect and identify specific DNA sequences in a sample. The design of a DNA probe involves selecting a sequence of DNA that is complementary to the target sequence, as well as optimizing the length, specificity, and sensitivity of the probe. Different types of probes can be designed, including fluorescent, biotinylated, or radioactive probes, depending on the intended use. DNA probe designing is an essential step in many molecular biology techniques, such as polymerase chain reaction (PCR), DNA sequencing, and gene expression analysis. Principle The principle of DNA probe design is to create a short, singlestranded piece of DNA that is complementary to a specific sequence of the target DNA. This complementary sequence allows the probe to hybridize or bind with the target DNA, allowing for detection or identification of the target sequence. The design of the DNA probe must also consider factors such as length, specificity, and sensitivity to ensure accurate and reliable results. Procedure 1. Determine the target DNA sequence by searching databases or sequencing DNA of interest. 2. Decide on the length of the DNA probe, aiming for 18-25 nucleotides. The length of the DNA probe is an important 66 consideration as it can affect the specificity and sensitivity of the probe. 3. Choose a labeling method such as fluorescent dyes, biotin, or radioactive isotopes. The choice of labeling method will depend on the intended use of the probe and the detection method being used. 4. Design the probe sequence to complement the target DNA sequence and include the chosen labeling moiety. The probe sequence should be complementary to the target DNA sequence and should contain the labeling moiety at the appropriate location. It is important to avoid secondary structures and self-complementarity in the probe sequence. 5. Test the probe for specificity and sensitivity by hybridizing it to the target DNA sequence. 6. Optimize the probe by adjusting its length, labeling method, or concentration. 7. Validate the probe by testing it on a variety of samples and verifying consistent and reproducible results. Advantages 1. DNA probes are designed to be complementary to a specific DNA sequence, so they are highly specific in their detection of the target DNA. 2. DNA probes can detect even small amounts of the target DNA, making them a powerful tool in molecular biology. 3. DNA probes can provide rapid results, with detection often 4. DNA probes can be designed to detect any DNA sequence, taking only a few hours. making them a versatile tool for a range of applications. 67 Disadvantages 1. DNA probes can be expensive to design and manufacture, particularly if multiple probes are required for a single experiment. 2. Designing a DNA probe requires a high level of expertise in molecular biology, and the process can be complex and time-consuming. 3. DNA probes can sometimes cross-react with other DNA sequences, leading to false positive results. 4. DNA probes can degrade over time, so they must be stored carefully and used quickly to ensure accuracy. 68 Bergs Terminal Transferase - Boyer Cohen Chang Experiment Berg's Terminal Transferase (TdT) is an enzyme that catalyzes the addition of deoxy-nucleotides to the 3' end of DNA strands. The Boyer-Cohen-Chang experiment was conducted in 1973 to demonstrate the activity of TdT. The experiment involved incubating a partially double-stranded DNA template with TdT, deoxynucleotides, and radioactive ATP. The results showed that TdT added radioactive nucleotides to the 3' end of the DNA strand, proving its ability to extend DNA chains. This experiment was critical in establishing TdT's role in DNA synthesis and repair processes. Principle The principle of Berg's Terminal Transferase – Boyer Cohen Chang experiment is to use a bacterial enzyme called terminal transferase to add homopolymeric tails to the 3' ends of DNA molecules. This process is important for many molecular biology techniques, such as cloning and sequencing, as it allows for specific and efficient hybridization of DNA molecules. The experiment demonstrated that the terminal transferase enzyme could add homopolymeric tails to double-stranded DNA, and that the length and composition of the tails could be controlled by adjusting the reaction conditions. The discovery of this enzyme and its ability to modify DNA has greatly facilitated the development of many molecular biology techniques. 69 Procedure 1. Prepare a reaction mixture containing the following components - DNA template, terminal deoxynucleotidyl transferase (TdT), dGTP, dATP, dTTP, and MnCl2. 2. Mix the reaction mixture and incubate it at 37°C for 15-60 minutes. 3. Stop the reaction by adding EDTA. 4. Denature the DNA by heating the reaction mixture at 95°C for 5 minutes. 5. Analyze the reaction products using polyacrylamide gel electrophoresis. 6. Visualize the reaction products by staining the gel with 7. Compare the size and intensity of the reaction products with ethidium bromide. the control samples. 8. Interpret the results and draw conclusions based on the findings. 9. Repeat the experiment with different variations to confirm the results. Advantages 1. Berg's terminal transferase (TdT) is a highly specific enzyme that can incorporate nucleotides onto the 3' end of DNA fragments. 2. The Boyer Cohen Chang experiment provided a new and efficient method for introducing DNA into bacterial cells, which revolutionized the field of genetic engineering. 3. TdT has low error rates, making it an ideal tool for constructing cDNA libraries. 70 4. The Boyer Cohen Chang experiment paved the way for recombinant DNA technology, which has since been used to produce a wide range of useful products. Disadvantages 1. TdT is highly sensitive to temperature, pH, and salt concentration, which can lead to variable results and limit its use in certain applications. 2. The Boyer Cohen Chang method requires the use of antibiotics to select for transformed cells, which can lead to the development of antibiotic-resistant strains. 3. The process of creating recombinant DNA can be timeconsuming and complex, requiring specialized equipment and expertise. 4. There are ethical concerns surrounding the use of recombinant DNA technology, particularly with regards to genetically modified organisms (GMOs). 71 Preparation of Competent Cells for Efficient DNA Uptake in E. coli Competent cells are a type of laboratory-grown bacterial cells that have been artificially modified to accept foreign DNA into their genomes. The process of modifying the cells to be competent involves making small changes to their cell membranes and cellular components, which allows them to efficiently uptake and integrate foreign DNA. Competent cells are an essential tool in molecular biology, biotechnology, and genetic engineering. They are used in a variety of applications including gene cloning, genetic modification, and the production of recombinant proteins. The ease and efficiency of DNA transfer into competent cells makes them a crucial tool in the field of biotechnology and genetic engineering. One of the most common methods for making competent cells is by transforming them with calcium chloride. In this process, the bacteria are treated with a high concentration of calcium chloride, which temporarily permeates the cell membrane, allowing foreign DNA to enter. Once the DNA has been taken up by the cells, they return to their normal state, and the DNA becomes integrated into the genome. Another method for making competent cells is by electroporation, which involves applying a high-voltage electrical pulse to the cells, which creates temporary pores in the cell membrane, allowing the foreign DNA to enter. This method is often faster and more efficient 72 than the calcium chloride method, but it also involves a higher risk of cell death. Competent cells are also classified into two types: chemically competent cells and electro-competent cells. Chemically competent cells are made by using a chemical agent, such as calcium chloride, to permeate the cell membrane and allow DNA to enter. Electrocompetent cells are made by using an electrical pulse to create temporary pores in the cell membrane, allowing the DNA to enter. In addition to their use in biotechnology and genetic engineering, competent cells also have applications in medical research. They can be used to produce vaccines, produce recombinant proteins for use in diagnostic tests, and to study the interactions between bacteria and their hosts. Procedure 1. Choose a bacteria that is susceptible to transformation, such as E. coli, and grow it in a culture medium until it reaches logarithmic phase. 2. Centrifuge the bacterial culture to harvest the cells. Discard the supernatant and resuspend the pellet in cold sterile buffer. 3. Slowly freeze the cells in a solution of glycerol and buffer. This will protect the cells from damage and make them more competent for transformation. 4. Thaw the cells quickly in a water bath at 37°C. The cells should be kept at 37°C for no more than 30 seconds. 5. Add calcium chloride to the cells to increase their competence. Incubate the cells at 37°C for 15 minutes. 73 6. Add the DNA to be transformed to the competent cells. Incubate the cells at 37°C for 1 hour. 7. Transfer the transformed cells to a nutrient agar plate and incubate overnight at 37°C. 8. Screen the colonies that have grown on the agar plate for the desired phenotype. Confirm the transformation by conducting further tests, such as PCR or sequencing. Alternative Method Principle Competent cells in bacteria are necessary for efficient DNA uptake. The bacteria's plasma membrane must be permeable to foreign DNA, which can be achieved by artificially making the bacterial cells competent. However, since both bacteria and DNA are negatively charged, it is challenging for DNA to enter bacterial cells. To overcome this, divalent cations are used, which shield charges by coordinating phosphate groups and other negative charges, thereby changing the electrical charge on the bacterial cell surface and weakening the bacterial cell membrane. This promotes the entry of foreign DNA into bacterial cells. Stock  Bacteria  Spectrophotometer  LB Agar Plate  LB Broth  Glycerol  CaCl2  Tubes  Centrifuge 74 Preparation of LB Medium Prepare Luria Bertani medium by mixing 10 g tryptone, 5 g yeast extract, and 10 g of NaCl in 1 litre of distilled water. Adjust the pH to 7.0 with 1N NaOH and autoclave the mixture for 25 minutes at 120°C. Procedure 1. Grow E. coli in 5 ml LB Broth overnight. 2. Dilute overnight culture 100-fold and grow bacteria in 250 ml of LB in 1L conical flask at 37°C, over a shaker. 3. Check the OD of the culture at 600 nm until it reaches 0.250.30. 4. Transfer culture into 50 ml centrifuge tubes and centrifuge at 5. Discard supernatant and resuspend cells in 30 ml of chilled 3,000 rpm for 10 minutes at 4°C. 100 mM CaCl2 for 30 minutes on ice with intermittent hand shaking. 6. Centrifuge cells at 3,000 rpm for 10 minutes at 4°C, discard supernatant, and resuspend bacterial pellet in 5-10 ml of 100 mM CaCl2 plus 15% glycerol. 7. Prepare 100 ul aliquots of this bacterial cell suspension and store at -70°C for further use. Observation Competent cells are prepared successfully and are frozen at -70°C for further use. 75 Precautions 1. Perform all procedures in cold conditions. 2. After CaCl2 treatment, bacterial cells become fragile, so avoid vortexing them. 3. Do not freeze and thaw competent cells repeatedly. 76 Bacterial Transformation in vitro using Electroporation Principle Plasmid DNA is a circular, double-stranded DNA molecule that exists independently from the chromosomal DNA of bacterial cells. Plasmids often contain genes that can provide an advantage to the bacterial cell, such as antibiotic resistance genes. Introducing plasmid DNA into a bacterial cell provides the cell with new genetic material that can give it a new function or advantage. Bacterial transformation is the process of introducing foreign DNA into bacterial cells, which can be achieved through various methods such as electroporation, heat shock, and chemical transformation. In this experiment, electroporation is used to introduce plasmid DNA into bacterial cells by creating transient pores in the cell membrane using a high voltage electric pulse. Stock  Bacterial culture  Plasmid DNA  Restriction enzymes  DNA ligase  LB agar plates  Ampicillin  Sterile pipettes and tips  Microcentrifuge tubes  Heat block 77  Incubator  Electroporator Procedure 1. Prepare bacterial cells for electroporation by centrifuging the culture and resuspending the cells in ice-cold sterile water. 2. Wash the cells twice to remove any residual media. 3. In a microcentrifuge tube, mix 50 μl of the washed cells with 2 μl of linearized plasmid DNA. 4. Transfer the cell and plasmid DNA mixture to a sterile electroporation cuvette. 5. Apply an electric pulse to the cells using the electroporator according to the manufacturer's instructions. 6. Immediately add 1 ml of LB broth to the cuvette and transfer the mixture to a sterile microcentrifuge tube. 7. Incubate the cells at 37°C for 1 hour to allow for expression of the plasmid genes. 8. Plate the transformed cells on LB agar plates containing ampicillin and incubate overnight at 37°C. 9. Observe bacterial growth and look for colonies that have grown on the ampicillin-containing plates. Results Successful transformation will result in colonies growing on the ampicillin-containing plates, which are resistant to the antibiotic and carry the plasmid DNA. The presence of the plasmid DNA can be confirmed by performing a plasmid isolation and restriction digest, or by sequencing the plasmid. 78 Precautions 1. Maintain aseptic techniques throughout the experiment to prevent contamination of the bacterial culture with other microorganisms or foreign DNA. 2. Use sterile equipment and solutions to avoid introducing unwanted DNA or bacteria. 3. Ensure the bacterial culture is healthy and in the exponential growth phase to increase the likelihood of transformation. 4. Use appropriate controls, including an untransformed control, to verify that any observed changes in bacterial phenotype are due to the introduction of plasmid DNA and not an artifact of the experiment. 5. Optimize electroporation parameters (such as voltage, time constant, and pulse duration) to maximize the efficiency of transformation while minimizing cell damage. 6. Use the appropriate antibiotic concentration to select for transformed cells without causing any adverse effects on the growth of the bacterial culture. 7. Store and handle the plasmid DNA properly to prevent degradation or contamination. 79 CaCl2 Mediated Transformation CaCl2 mediated transformation is a method of introducing foreign DNA into a host organism, often with the aim of expressing a new trait or function. This transformation method is widely used in molecular biology, biotechnology, and genetic engineering for the study and manipulation of genes and genomes. The transformation process includes the preparation of the DNA to be introduced into the host organism. The DNA is first extracted from the source organism, often using a restriction enzyme to cut the DNA at specific sites. The DNA is then ligated to a plasmid, a circular piece of DNA that is used as a vector for transformation. The plasmid contains a gene of interest and the origin of replication, which allows the plasmid to replicate in the host organism. Once the plasmid is ready, the next step is to introduce it into the host organism. This is where CaCl2 comes into play. CaCl2 is used as a mediator in the transformation process to help the plasmid enter the host organism. CaCl2 acts as a permeater, making the cell membrane of the host organism temporarily permeable, allowing the plasmid to enter the cell. The transformation process can be carried out in various ways, such as electroporation, where an electrical field is applied to the host organism, or chemical transformation, where chemicals such as CaCl 2 are used to create temporary holes in the cell membrane. In both methods, the permeated cells are then incubated with the plasmid solution, allowing the plasmid to enter the cell. 80 Once the plasmid is inside the cell, it can integrate into the host organism’s genome or remain as a separate piece of DNA. If the plasmid integrates into the host organism’s genome, the new gene will be expressed and the host organism will display the new trait or function. If the plasmid remains as a separate piece of DNA, it can still express the new gene and the host organism will still display the new trait or function. Procedure 1. Grow bacterial cells in liquid medium until they reach midlog phase, then chill the culture on ice for 10 minutes. Transfer the cells to 50 ml tubes containing ice-cold 50% glycerol and mix gently by inverting the tubes. Store the competent cells at -80°C until use. 2. Isolate the plasmid DNA and purify it using a plasmid isolation kit. 3. Dissolve 0.1 M CaCl2 in distilled water and sterilize the solution by autoclaving. 4. Thaw the competent cells on ice and add the purified plasmid DNA to the cells. Incubate the mixture on ice for 30 minutes. 5. Add the sterilized CaCl2 solution to the cell-DNA mixture, keeping the volume of CaCl2 solution equal to that of the cell suspension. Incubate the mixture on ice for another 2-5 minutes. 6. Transfer the mixture to a 42°C water bath and incubate it for exactly 90 seconds. Quickly transfer the mixture back to the ice and incubate it for 2 minutes. 81 7. Add 1 ml of LB medium to the mixture and incubate it at 37°C for 1 hour. Spread the mixture onto an LB agar plate containing the appropriate antibiotics and incubate it overnight at 37°C. 8. Incubate the plate for an additional 16-20 hours at 37°C to allow for the growth of the transformed cells. Check for the presence of colonies and pick the desired colonies for further analysis. 82 Agarose Gel Electrophoresis Agarose gel electrophoresis is a laboratory technique used to separate and analyze different sizes of DNA fragments. The process includes the preparation of a gel from agarose, a sugar found in seaweed. This gel is poured into an electrophoresis chamber and solidified. Then, the DNA samples are loaded into wells in the gel. An electric field is then applied to the mixture, causing the negatively charged DNA molecules to move towards the positively charged anode. The speed at which the DNA moves through the gel is dependent on the size of the DNA molecules, allowing for separation and quantification of different sized fragments. The DNA bands can be visualized by staining the gel with a fluorescent dye – ethidium bromide, and observing it under ultraviolet light. By using a transluminator and a UV light source with a wavelength of 254 nm, 310 nm, or 354 nm, electrophoresis enables the separation and localization of DNA in agarose gel. Various buffers, such as TAE and TBE, can be used in the electrophoresis process, with different buffers affecting the migration rate of the DNA. The molecular weight of the DNA, the concentration of agarose, the conformation of the DNA, and the applied current all have an impact on the electrophoretic mobility of DNA through agarose gel. The size of the DNA fragments that can be separated depends on the concentration of agarose in the gel, with different concentrations allowing for the separation of different sized fragments. For example, a 0.5% gel can separate DNA fragments between 1 kb and 30 kb, 83 while a 2.0% gel can separate fragments between 50 bp and 2 kb (refer table below): w/v % Gel type Size of DNA fragments (1 Kb = 1000 bp) 0.5 % 1 kb to 30 kb 0.7 % 800 bp to 12 kb 1.0 % 500 bp to 10 kb 1.2 % 400 bp to 07 kb 1.5 % 200 bp to 03 kb 2.0 % 50 bp to 02 kb Agarose gel electrophoresis is widely used in molecular biology and genetics and has several applications, including the analysis of PCR products, the separation of DNA fragments after restriction digestion, and the detection of genetic variations such as mutations and polymorphisms. It is important to note that for the separation of fragments smaller than 100 bp, a different technique called Polyacrylamide gel electrophoresis is used. 1. To assess the purity of the extracted DNA by agarose gel electrophoresis. 2. To separate and calculate the molecular size of DNA fragment by comparing the separated bands with known standard molecular weight marker. 3. To quantify DNA fragment by comparing the separated band with known quantity of DNA. Principle Electrophoresis is a laboratory method used to separate charged molecules, like DNA, based on their size and charge. An electric current is applied to the DNA sample, previously broken down by restriction enzymes, driving the negatively charged DNA towards the positive electrode. The agarose gel acts as a semi-solid matrix that 84 allows the DNA fragments to separate based on their size. Larger molecules travel further, while smaller ones travel less distance. This enables effective separation of the biomolecules. The separated fragments can then be analyzed and visualized. The equipment typically includes a power source, a casting tray for the gel, and a container for the sample. Stock 1. Tris-Borate-EDTA (TBE) stock solution (5X) Tris base 54.0 gm Boric acid 27.5 gm EDTA (pH 8.0) 0.5 M 20.0 ml Distilled water to make 1000.0 ml 2. Working Buffer: 1X or 0.5X TBE 3. Loading Buffer (10X) Bromophenol blue 0.25% Xylene cyanol 0.25% Ficoll (type 400) 25.0% in distilled water 4. DNA Sample DNA 150-200 ng Distilled water 18 μl 10X loading buffer 2 μl Before conducting an electrophoresis experiment, it is important to prepare a stock solution of ethidium bromide. To do this, prepare a stock solution of 10 mg/ml ethidium bromide and keep it stored at 4oC in a coloured glass tube or container. 85 Agarose powder, 1X TBE buffer (89 mM Tris-base, 89 mM boric acid and 2 mM EDTA) prepared from 10X TBE, Ethidium Bromide (5 mg/ml), Gel loading dye (Glycerol and orange dye), 1 kb and 100 bp DNA ladder, horizontal electrophoresis apparatus and power supply. Procedure Step 1: Preparation of Agarose Gel 1. Weigh out the required amount of agarose powder to make a 1% gel using a scale. 2. Heat the agarose powder in a microwave to dissolve it and create a homogeneous mixture. 3. Add 4 microliters of ethidium bromide to the mixture and mix it in carefully. This will stain the DNA for visualization under UV light. 4. Prepare the gel plate and comb by placing the comb in the slots on each side of the gel plate and pour the melted agarose onto the gel plate in the electrophoresis tray. 5. Let the gel cool to room temperature and remove the comb. 6. Place the gel in the electrophoresis chamber and add enough electrophoresis buffer (1X TBE) to cover the gel. Step 2: Loading of DNA 1. Mix 300 ng of DNA with 3-4 microliters of loading dye. 2. Add a DNA ladder (3 microliters) to the first well using a micro-pipette. 3. Add the prepared DNA samples to adjacent wells. 4. Run the electrophoresis at 95 volts for 45 minutes and 5. Once the electrophoresis is complete, place the gel on a UV periodically check the gel. light box to take a picture. 86 Step 3: Observing the Gel 1. Check the gel under UV light using a transluminator. 2. Wear a perspex shield or safety glasses to avoid the damaging effects of UV light. Note: A lambda DNA digested with Hind III restriction enzyme is typically used to determine the molecular weight of the experimental DNA in this procedure. This results in 8 bands of different sizes. However, similar amounts of degraded genomic DNA will exhibit a smear instead of sharp bands. Advantage and Disadvantage The method of using Ethidium bromide is simple and easy to follow. It is also known for its fast results and efficient performance. However, Ethidium bromide is recognized as a carcinogen and thus, it needs to be used with caution and proper disposal. 87 Protein Analysis by SDS-PAGE Sodium Dodecyl Sulfate Polyacrylamide Gel Electrophoresis (SDSPAGE), is a widely used method in biochemistry and molecular biology for the separation and analysis of proteins based on their size and charge. The process comprises the denaturation of proteins using SDS, a detergent that uniformly coats and unfolds the protein, creating a negative charge proportional to its size. The SDS-protein mixture is then loaded onto a polyacrylamide gel, which serves as a sieve to separate the proteins based on their size. An electric field is applied, causing the negatively charged proteins to migrate towards the positive electrode, resulting in a separation of the proteins by size. One of the key advantages of SDS-PAGE is its ability to separate proteins based on their molecular weight, allowing for easy identification of specific proteins and the estimation of their relative molecular weight. This technique is commonly used in the analysis of protein samples from tissues, cells, and bodily fluids, as well as for purifying and isolating specific proteins. SDS-PAGE is also widely used for quality control in biotechnology and pharmaceutical industries, where it is used to ensure the purity and consistency of protein-based products such as enzymes, vaccines, and antibodies. The technique is also used in protein purification, as well as in the detection of protein modifications such as phosphorylation, glycosylation, and proteolytic cleavage. To enhance the resolution of SDS-PAGE, variations such as twodimensional gel electrophoresis (2D-PAGE) can be used. This 88 technique separates proteins based on both size and charge, providing a more comprehensive view of the protein profile in a sample. Stock  Ammonium per sulfate (10%)  Coomassie brilliant blue (0.3%)  Destaining mixture  Gel staining dish  Electrophoresis apparatus with power supply  Running buffer SDS (10%) Tris-HCl (1.5 M, pH 8.8) Tris-HCl (0.5 M, pH 6.8)  Acrylamide-bis-acrylamide stock solution Reagent Preparation  Acrylamide-bis-acrylamide stock solution: Dissolve 29.2 g acrylamide and 0.8 g bis-acrylamide in distilled water, then raise the final volume to 100 ml.  Tris-HCl (1.5 M, pH 8.8): Dissolve 18.15 g of Tris in 50 ml distilled water, adjust the pH to 8.8 with HCl, then raise the final volume to 100 ml.  Tris-HCl (0.5 M, pH 6.8): Dissolve 6 g Tris in 60 ml distilled water, adjust the pH to 6.8 with HCl, then raise the final volume to 100 ml.  SDS (10%): Dissolve 1 g SDS in 5 ml distilled water, then raise the final volume to 10 ml. 89  Gel running buffer: Dissolve 14.4 g glycine and 1 g SDS in 1 L distilled water, adjust the pH to 8.3 with Tris, then raise the final volume to 1 L.  Ammonium per sulfate (APS) (10%): Dissolve 500 mg solid APS in 5 ml distilled water. Use freshly prepared APS solution only.  Coomassie Brilliant Blue R 250: Dissolve 600 mg CBBR-250 in 80 ml methanol, add 20 ml glacial acetic acid, then raise the final volume to 200 ml with distilled water.  Destaining solution: Mix 400 ml methanol, 100 ml glacial acetic acid, and 500 ml distilled water to obtain 1 L of solution.  Laemmli buffer: 62.5 mM Tris-HCl, pH 6.8 (diluted from 0.5 M Tris-HCl, pH 6.8), 10% glycerol, 5% mercaptoethanol, 2% SDS. Procedure SDS-PAGE is a commonly used technique for separation and analysis of proteins based on their molecular weight. Here is a step-by-step procedure to perform SDS-PAGE of a protein sample: 1. To ensure efficient separation, it is important to denature and reduce the protein sample. This is typically done by adding a detergent like SDS to the sample and heating it to 95-100°C for 5-10 minutes. In addition, a reducing agent like DTT or beta-mercaptoethanol can be added to reduce any disulfide bonds present in the protein sample. 2. The denatured and reduced protein sample is mixed with a loading buffer which contains SDS, glycerol, and a tracking dye to monitor protein migration during electrophoresis. 90 3. An electrophoresis apparatus, such as a vertical or horizontal gel apparatus, is set up and the acrylamide gel is prepared and poured into the apparatus. The gel is then allowed to polymerize. 4. Using a micropipette, load the denatured protein solution into the wells. The protein sample should be denatured in Laemmli buffer by boiling for 5 minutes. Add standard molecular weight marker proteins in one lane. For detection by CBB dye, 20 to 50 g protein is generally sufficient. 5. Connect the electrodes of the apparatus tightly to the power supply and run the gel at a constant current of 20 mA. Larger proteins move slower through the gel compared to smaller proteins. Track the mobility of the sample in the matrix using dye (bromophenol blue is generally added to the Laemmli buffer). The tracking dye provides a visual representation of the migration of proteins. Once the run is complete, switch off the button and disconnect the apparatus. 6. Transfer the gel to a staining tray containing a protein stain like Coomassie blue or silver stain to visualize the separated protein bands. Stain the gel for at least 2 hours or overnight under shaking conditions on a rocking shaker. The entire gel should turn blue. 7. Carefully transfer the gel to a de-staining solution and shake on a rocker shaker for 30 minutes. Add fresh de-staining solution, repeating these steps until the bands are clearly visible in the gel. At this stage, take a photograph of the gel. 8. Analyze the photographed gel to observe several distinct blue-colored bands. Each band represents one or multiple bands in the lane, with the intensity of these bands varying depending on the amount of polypeptide present in the protein solution loaded in the gel. 91 Blue-White Colony Selection employing X-Gal / IPTG Blue-white colony selection is a widely used method in molecular biology to select transformants, which are cells that have taken up a plasmid with a desired gene or sequence. This method is based on the use of X-Gal, a blue chromogenic substrate, and IPTG, an inducer of gene expression, to visualize and select colonies that contain the desired sequence. The process of blue-white colony selection involves the transformation of bacteria with a plasmid that contains a gene of interest. The plasmid also contains a lacZ gene, which encodes for βgalactosidase - an enzyme that hydrolyzes X-Gal to produce a blue color. To ensure that the lacZ gene is expressed only when the gene of interest is present, the plasmid also contains a promoter that is regulated by IPTG. After the transformation, bacteria are plated on agar plates that contain X-Gal and IPTG. Bacteria that have taken up the plasmid will express the lacZ gene and produce β-galactosidase, which will hydrolyze X-Gal to produce blue colonies. Bacteria that have not taken up the plasmid will not express the lacZ gene, and therefore will not produce β-galactosidase, resulting in white colonies. The presence of IPTG in the agar medium will induce the expression of the lacZ gene, leading to the production of β-galactosidase and the hydrolysis of X-Gal to produce blue colonies. The concentration of IPTG used in the agar medium can be adjusted to control the level 92 of gene expression and ensure that only the desired colonies are selected. The blue-white colony selection method has several advantages over other selection methods. Firstly, it is simple and easy to perform, requiring only the addition of X-Gal and IPTG to the agar medium. Secondly, it is rapid, allowing the selection of colonies within a few hours. Finally, it is highly specific, as only colonies that contain the desired gene or sequence will be blue. Its simplicity, rapidity, and specificity make it an ideal choice for a variety of applications, including cloning, gene expression, and protein production. Stock  X-Gal  Dimethylformamide (DMF)  dH2O  Isopropyl β-D-1-thiogalactopyranoside (IPTG)  Screening Antibiotic  Agar Media  Plates. Procedure 1. Preparation of X-Gal and IPTG: • To incorporate X-Gal and IPTG, prepare a 20 mg/ml X-Gal solution in DMF and a 100 mM IPTG solution in dH2O (see IPTG Stock Solution Procedure) or dilute from a 1M IPTG solution. • Integrate X-Gal and IPTG into the agar media by adding 10 μl of 20 mg/ml X-Gal solution per 1 ml of media or 2 μl of 100 mg/ml X-Gal solution per 1 ml of media. • To obtain a final concentration of 1 mM IPTG, add 10 μl of 100 mM IPTG solution per 1 ml of media. 93 Notes To enhance the screening process, a higher concentration of X-Gal may be used. It increases blue color intensity, reduces blue color development time, refrigeration time, and decreases the number of ambiguous colonies requiring rescreening. 2. Screening on agar media containing IPTG and X-Gal: • Autoclave the growth media agar and cool to 50°C. • Add the screening antibiotic and pour plates. Allow them to cool to room temperature before use, which usually takes at least 30 minutes. Spread transformed competent cells as desired. • 3. Screening on pre-made agar plates lacking IPTG and X-Gal: • Pour autoclaved growth media containing screening antibiotic on media plates and dry in a laminar flow hood. • Add 40 μl of 100 mM IPTG and 120 μl of X-Gal (20 mg/ml) to the surface of each plate and spread over the entire surface. • Dry X-Gal/IPTG-coated media in a laminar flow hood for approxi-mately 30 minutes before use. • Spread transformed competent cells and incubate inverted at 37°C until blue colonies form (usually ~24 hours). Result The bacterial cell that is transformed with a vector containing recombinant DNA will produce white colonies whereas bacterial cell that is transformed with the vector without recombinant DNA will produce blue colonies. 94 Notes In this, foreign DNA is inserted to interrupt the beta-galactosidase coding sequence, which otherwise reacts with x-gal substrate to produce a blue color. Due to the defective enzyme produced by insertion of foreign DNA in gal gene, white colored colonies are produced. 95 Genomes and Interrelatedness Genomes refer to the complete set of genetic material that exists within an organism's cells. It is the blueprint of life, and it carries the information required for an organism to develop and function. The genome is made up of DNA, which is a long chain of nucleotides. These nucleotides store the genetic information in the form of four letters: A, T, C, and G, which represent the four types of nitrogenous bases in the DNA molecule. The concept of genomic interrelatedness is crucial in genetics and genomics, as it refers to the tight connections between species and their genomes. This interrelatedness provides insight into the evolutionary history of life on earth and the relationships between species. One of the primary ways that interrelatedness is studied is through comparative genomics. This involves comparing the genomes of different species to identify similarities and differences. This approach provides insight into the evolutionary relationships between species, including when species evolved and how they are related. For example, comparing the genomes of humans and chimpanzees provides valuable information about their evolutionary history. The genomes of these two species are nearly identical, with differences in only about 1-2% of their DNA. This suggests that humans and chimpanzees are closely related, and that they have evolved from a common ancestor. 96 Interrelatedness also has practical applications in medicine and biotechnology. By comparing genomes, scientists can uncover genetic causes of diseases and develop new treatments. Three types of genomes can be compared: 1. Nuclear genome: This refers to the genetic material found in the nucleus of a cell, which includes both the chromosomes (structures that carry the genetic information) and the nonchromosomal DNA. In most organisms, the nuclear genome is the largest and most complex component of the genome. 2. Mitochondrial genome: This refers to the genetic material found in the mitochondria, the organelles that produce energy for the cell. The mitochondrial genome is much smaller than the nuclear genome and typically contains only a few genes that are important for energy production. 3. Chloroplast genome: This refers to the genetic material found in the chloroplasts, the organelles that carry out photosynthesis in plants and algae. The chloroplast genome is also smaller than the nuclear genome and contains genes involved in photosynthesis and the maintenance of the chloroplast. The degree of relatedness between genomes can be determined by comparing their genetic sequences. This can be done at several levels, including: 1. DNA sequence: This refers to the specific order of nucleotides (A, C, G, and T) in a DNA molecule. Comparing DNA sequences can reveal similarities and differences between genomes and can provide information about evolutionary relationships. 97 2. Gene content: This refers to the number and types of genes present in a genome. Comparing gene content can reveal similarities and differences between genomes and can provide information about the functions and adaptations of different organisms. 3. Chromosomal structure: This refers to the organization and arrangement of chromosomes within a genome. Comparing chromosomal structure can reveal similarities and differences between genomes and can provide information about the evolutionary history of different organisms. Overall, the degree of relatedness can vary greatly between genomes, with some organisms being very closely related (such as different species within the same genus) and others being more distantly related (such as different phyla or kingdoms). Principle The process of measuring the similarity of DNA sequences from different species is known as hybridization. This technique is used to estimate the extent to which organisms from different species have common DNA sequences. The process of hybridization is carried out in a series of steps. First, a solution of denatured DNA from one species is filtered through a membrane. The single-stranded DNA sticks to the filter and becomes irreversibly bound to it when the filter is air dried. Next, the filter is incubated in a solution of denatured heterologous DNA. The incubation is done at a temperature 25°C below the melting temperature of the denatured DNA in the solution. The incubation causes duplexes to form between the filter-bound DNA and the DNA in the solution. If the DNA in the solution is isotopically labeled, the hybrids will bind the label to the filter. However, free 98 hybrid molecules are formed in the solution but they are not detected as they are not bound to the filter. The experiment measures the differential ability of some heterologous DNA to form DNA / DNA hybrids, thus estimating the similarities of the different DNA sequences. This is done by comparing the amount of hybrids formed when no competitor is present versus the amounts of hybrids formed in the presence of different quantities of competitor. Before starting the experiment, the DNA solutions are denatured by placing them in a boiling water bath for 10 minutes. Procedure Step-1: Preparing the Tubes for DNA Analysis 1. Collect tubes for each competitor's DNA. 2. Add a homologous and heterologous DNA filter to each tube. 3. Gently tap the tubes to mix the contents. 4. Add 0.5 ml of fluid to cover the top of the tubes. 5. Cover the tubes and place them in a water bath set at 61°C for 18 hours. Step-2: Analyzing the Filters for Hybrid Formation 1. An hour before the incubation period ends, place a rack containing l XSSC in the water bath. 2. Remove the filters from the tubes and transfer them to l XSSC. 3. Rapidly stir the filters in l XSSC. 4. Transfer the filters from one row of tubes to the next, until they are all in the third row. 5. Blot the filters dry on a sheet of paper towel. 99 6. Pin the filters to the towel and incubate at 60°C for 20 minutes. 7. Place the filters in a scintillating vial and count for 10 minutes. 8. Discard the filters into a container for radioactive waste. Step-3: Data Treatment and Analysis 1. Subtract the counts per minute of the heterologous filter from the homologous filter. 2. Express the results as percentages. 3. Plot the results graphically to show the percent of hybrids formed against the amount of competitor DNA added. 100 In situ Hybridization In situ hybridization studies on chromosomes provide an approach to genetic mapping of the sequence of interest. When one of the hybridization partner remains in situ, using a given labeled polynucleotide (DNA or RNA) probe, location of the homologous sequences in cells can be determined. The pattern of functional organization or its expression can also be studied conveniently by this technique at cellular or at organ level. In situ hybridization (ISH) is a laboratory technique used to detect and map the location of specific RNA or DNA sequences within cells or tissues. The process involves labeling a specific RNA or DNA probe with a fluorescent or radioactive tag, and then hybridizing the probe to its complementary target sequence within the sample. The basic steps of ISH include: 1. Sample preparation: The tissue or cells are fixed and embedded in a paraffin block or frozen section. 2. Probe preparation: The specific RNA or DNA probe is labeled with a fluorescent or radioactive tag, such as digoxigenin or biotin. 3. Hybridization: The labeled probe is added to the sample and allowed to hybridize to its target sequence. The probe will only bind to its specific target sequence, allowing for the detection and localization of that specific sequence within the sample. 101 4. Detection: The hybridized probe is detected using an appropriate detection method, such as a fluorescent microscope or a auto-radiography. 5. ISH is a powerful technique that allows for the visualization of specific RNA or DNA sequences within cells or tissues, and can be used to study gene expression patterns, chromosomal abnormalities, and viral infections, among other applications. Stock 1. 2. 20X SSC Sodium chloride 3M Sodium citrate 0.3 M Gelatin solution 0.1% Gelatin 100 mg Distilled water 100 ml (warm at 70 C for 1 hr) o 3. Sodium acetate 3M (pH 5.2 with the help of glacial acetic acid) 4. RNase 10 mg /10 ml 5. Alcohol 70/90/100 % 6. Tris (pH 7.5, pH 8.0, pH 9.5) 1 M Sodium chloride EDTA 5M 0.05 M TE 7. Tris 10 mM (pH 8.0) 102 EDTA 1 mM 8. Digoxigenin-dUTP labeled DNA probe 9. Salmon sperm DNA 10 mg/ml 10. Hybridization mix 11. Buffers I 12. - Tris 100 mM, pH 7.5; NaCl 150 mM II - Blocking reagent in Buffer - I 0.5% w/v III - Tris 100 mM, pH 9.5 ; NaCl 100 mM, MgCl2 50 mM IV - Tris 10 mM, pH 8.0 ; EDTA 1mM Developer (to be prepared fresh) Nitroblue Tetrazolium 4.5 μl (NBT 75 mg/ml in Dimethyl formamide) 5-Bromo-4-Chloro-3-Indolyl Phosphate 3.5 μl (50 mg/ml in Dimethyl formamide) Buffer - III to make 1 ml 13. Stains Safranin 100 mg Distilled water 100 mg (Dissolve Safranin powder in water at room temperature. The prepared stain can be stored and used later on). 14. Entellan mountant Laboratory ware 1. Three incubators, preset at 37o, 42o and 60oC. 2. Sterilized glass slides and cover glasses 3. Slide racks 4. Slide trays 5. Couplin jars 103 6. Magnetic stirrer 7. Micropipettes 8. Pipette tips 9. Plastic box 10. Forceps Procedure Step 1: To label the probe DNA Chromosome in situ hybridization is a method used for genetically mapping a specific sequence of DNA. This method uses a labeled polynucleotide (DNA or RNA) probe to identify the position of homologous sequences in cells when one of the hybridization partners is still in place. This technique allows for the study of the pattern of functional organization or expression at the cellular or organ level. One way to label the probe DNA is through the Random Priming Method, which involves the following steps: 1. Denature a necessary quantity of linear DNA by heating it in boiling water for 10 minutes, then quickly chilling it on ice. 2. Add 2 μl of hexanucleotide mix, 2 μl of dUTP labeled mix, 19 μl of distilled water, and 1 μl of Klenow enzyme (3-5 units) to the chilled DNA. 3. Incubate the mixture at 37°C for an hour, then add 0.8 ml of 0.5 M EDTA to stop the process (final concentration 20m M) 4. Add 2.0 μl of salmon sperm DNA (10 mg/ml), 2.5 μl of lithium chloride (4 M), and 75 μl of prechilled (-20oC) ethanol to the labeled DNA to precipitate it. 5. Incubate the mixture at -70oC for two hours, then centrifuge the tubes for 30 minutes at 4°C at 12,000 rpm. 104 6. Separate the supernatant from the pellet and wash it in 70% ethanol. Dry in a warm environment. 7. Dissolve the labeled probe in the necessary volume of TE. The labeled probe can be stored at -20oC for more than two years. Step 2: To test the effectiveness of DIG labeling 1. Take a small piece of nylon membrane and moisten it with 2X SSC while under vacuum. Allow it to dry at room temperature for about 30 minutes. 2. Cross-link the probe DNA with the membrane by either heating the filter for two hours at 70°C or by exposing it to UV for three to four minutes on a transluminator. 3. Clean the filter briefly with Buffer I. 4. Block the membrane surface for 30 minutes at room temperature in Buffer II to facilitate the subsequent use of an antibody that will bind specifically to it. Use Buffer-I to rinse. 5. Incubate in Anti-DIG Antibody-enzyme conjugate (1 μl in 4 ml of buffer- I) for 30 mins at room temperature. 6. Wash twice in Buffer I with a 15-minute gap between each wash. Rinse briefly with Buffer- III 7. Place the blot in a tiny polythene bag, add the color developer, and then seal the bag in a dimly lit area. 8. Wrap the blot with aluminum foil and incubate it inside a sealed bag in a dark closet until the desired level of color signal appears. 9. Take the blot out of the bag and keep it in buffer IV to stop the reaction once a sufficient signal arises. The blot should be kept dry or in buffer IV. 105 Notes Under ideal probe labeling circumstances, 0.1 pg of probe produces a measurable signal in less than 30 minutes. Step 3: To take care of prepared slides 1. First submerge the slides in a freshly made 0.1% solution of gelatin for 3-5 seconds. This is done to apply a coat of gelatin on the slides which helps to keep the cells in a good state by preventing the binding of the probe. After that, airdry the slides. 2. Place 100 ml of RNase (100 g/ml in 2 X SSC) over the material on the slides. Then, cover each preparation with 22 mm2 cover glasses and place the slides in a moist chamber with filter papers soaked in 2 X SSC. The slides should be incubated for 2 hours at room temperature. This step helps to remove any RNA from the preparations. 3. After the 2-hour incubation, gently dip the slides into a beaker containing 2 X SSC and allow the cover glasses to fall into the solution. Clean the slides three times in 2 X SSC for 5 minutes each, twice in 70% ethanol for 10 minutes each, and once in 95% ethanol for 5 minutes. If necessary, air dry and store the slides in that manner. This step helps to remove any remaining RNase and clean the slides. 4. Now, immerse the slides in 0.07 N-NaOH for 3 minutes to denature chromosomal DNA. This step helps to break down any DNA present on the slides. 5. Finally, wash the preparations twice in 95% ethanol for 5 minutes each and three times in 70% ethanol for 10 minutes each. Air dry the slides. This step helps to remove any remaining NaOH and clean the slides. The prepared slides are now ready for further use. 106 Hybridization mix 1. Formamide 500 μl 2. 20 X SSC 250 μl 3. DIG labeled probe (per slide) 10-20 4. H2O to make 1000 μl ng *For one slide, 15 to 20 μl of hybridization mix is sufficient. Step 4: Hybridization Hybridization is a process used to identify specific regions of DNA on a chromosome. The following steps outline the procedure for performing hybridization using a tagged probe DNA and a hybridization mixture. 1. To denature the tagged probe DNA, place the tubes containing the probe in a boiling water bath for 10 minutes. This will cause the double-stranded DNA to separate into single strands. After denaturation, add the desired quantity of denatured probe to the hybridization mixture. 2. Mix 20 μl of the hybridization mixture with 10-20 ng of the labeled probe. Place a cover glass over the mixture and seal the edges using DPX. 3. Incubate the slides for 12-14 hours at 37°C in a humid, closed environment. This will allow the probe to bind to its complementary sequence on the chromosome. 4. After the incubation period, remove the DPX sealing with forceps and dip the slides in 2 X SSC to remove the cover glass. 5. The slides should be washed three times in one SSC for 15 minutes each at 60°C. This step is important to remove any unbound probe and other contaminants. 107 Step 5: Detection of Color 1. Rinse the slides for one minute in Buffer I and 30 minutes in Buffer II. This step is necessary to remove any remaining contaminants from the previous washing step. 2. Rewash the slides for one minute in Buffer I. 3. Incubate the slides in anti-Digoxigenin antibody alkaline phosphatase conjugate for 30 minutes after diluting it 1:5000 in buffer I. This step is necessary to bind the probe to the chromosome. 4. Wash the slides twice for two minutes each in Buffer I, followed by a rinse in Buffer III. 5. Depending on the desired signal, place 20-30 μl of freshly made color reagent on the slide, cover it with a cover glass, and seal it with DPX. Leave the slide in a dark room at room temperature for 1-12 hours. 6. Examine the slides under a microscope before placing them in Buffer IV to stop the reaction. 7. After counterstaining with safranin for 5-10 seconds and airdrying, wash the surface twice with distilled water. 8. Allow the finishing touches to air dry before mounting with Entellan (E. Merk). Result The hybridized probe binds to specific regions of the chromosomes, causing a purplish-blue color deposit. By using polytene chromosome maps, it's possible to identify the exact location of the hybridization signal. Notes Several factors must be considered to ensure that the hybridization signal is strong and accurate. 108 1. The quality of the chromosomal preparation is decisive. If the chromosomes are not prepared properly, the hybridization signal may be weak or not visible at all. 2. The process of denaturing the chromosomes must be precisely regulated to avoid ruining the structural details. The probe must also be denatured right before use. 3. Inadequate washing after hybridization and antibody binding may result in unwanted background. It is important to properly wash the slides to remove excess probe and antibody. 4. The use of safranin can improve the shape and contrast of the chromosomes, making it easier to see the hybridization signal. If safranin is not available, an alternative stain 2% aceto-orcein can be used. However, care must be taken to prevent the staining from masking the hybridization signal. 5. It is important to avoid air bubbles while mounting the cover glass. Air bubbles can impede the local response and obstruct the hybridization signal, leading to inaccurate results. 109 Amplification of DNA using Polymerase Chain Reaction Polymerase chain reaction, commonly known as PCR, is a laboratory technique used to amplify specific DNA sequences. It involves multiple steps, starting from identifying the target sequence, designing and checking the specificity of the primers, optimizing the PCR conditions, analyzing the results, and finally, starting the reaction and visualizing the results. Primer Design Primer design is an important step in the PCR process. Effective primers must be designed to ensure accurate amplification of the target sequence. The primer sequence must be complementary to the flanking sequences of the target region and should not contain any repeat or run sequences that could cause mispriming. The primer should match the target sequence at the 3’ end, and there should not be any complementary sequences between the primers to prevent primer dimers. The primer length should be between 1825 base pairs and the optimal GC content should be between 4060%. The GC content is calculated by dividing the number of G and C bases by the total number of A, T, G, and C bases and multiplying by 100. Melting temperature (Tm) is a measure of the temperature at which the double-stranded DNA molecule will separate into two single strands. It is important for the Tm to be within the range of 50-60°C and for the Tm difference between the forward and reverse primer 110 not to exceed 5°C. The Tm can be calculated using the formula [(G + C) * 4] + [(A + T) * 2]. Annealing temperature (Ta) is the temperature at which the primers bind to the target DNA. It is important that the Ta is not too high or too low, as this can lead to low product yield or non-specific products. The presence of G or C bases within the last five bases from the 3' end of primers is known as GC clamp, and it promotes specific binding at the 3' end. The GC clamp should not be more than 2 Gs or Cs. GC clamp refers to the presence of G or C bases within the last five bases from the 3' end of primers. It promotes specific binding at the 3' end and should not be more than 2 Gs or Cs. There are various tools available for primer design, such as the NCBI Primer design tool and Primer3 or primer3Plus, and it is important to check for specificity by running a BLAST on NCBI. PCR optimization is the process of finding the most efficient set of conditions for a PCR reaction. Different reactions may require different conditions to yield the best results, and it is important to optimize the conditions for the best results. Post-PCR analysis refers to the analysis of the products of a PCR reaction, known as amplicons. One common method of analysis is to separate the DNA by size using an agarose gel. The gel provides visual evidence of the success or failure of the reaction and the concentration of agarose used is determined by the size of the amplicon. PCR has several advantages, including simplicity, ease of use, sensitivity, and availability of reagents and equipment. The standard operating procedure for the technique has been extensively validated. 111 PCR has many different applications, including genotyping, cloning, mutation detection, sequencing, microarrays, RT-PCR, forensics, and paternity testing. Its versatility makes it a valuable tool in many different fields of research and analysis. 1. To amplify a specific region of DNA. 2. To prepare right primers. 3. To determine the parameters that may affect the specificity, fidelity, and efficiency of PCR. Stock 1. Purified genomic DNA of male and female Calotes 2. Stock solution of each primer 10 pM/μl 3. 10X Taq Polymerase Buffer KCl 500 mM Tris-HCl (pH 8.4 at 26oC) 100 mM MgCl2 15 mM 4. Gelatin 1 mg/ml 5. DNTPs mix (each of the 4 dNTPs) 1.25 mM 6. Mineral Oil 7. 1% Agarose gel Laboratory ware Thermal cycler/ 3 water baths set at 92oC, 72oC and 60oC. Procedure The process of amplifying DNA through Polymerase Chain Reaction (PCR) requires careful selection of DNA using two primers. These primers are designed to match two regions of potential sequence conservation that are separated by a DNA segment of around 60 to 112 1000 nucleotides. The design of the PCR primers is a crucial aspect of the amplification process. To create the primers, the first step is to identify conserved protein regions. Then, degenerate oligonucleotide primers are synthesized to match the potential DNA sequences that could encode the amino acids. To match the DNA sequences that could encode the Nterminal protein patch, a mixture of forward primers is synthesized. These primers are positioned more towards the 5’ end of the gene. On the other hand, reverse primers are positioned more towards the 3’ end of the gene, and are made to match the inverse complement of DNA sequences that could encode the C-terminal protein patch. When designing the primers, the following guidelines should be followed: 1. Each degenerate primer should roughly match the potential encoding DNA sequences for at least 15 to 20 nucleotides. 2. Degeneracy should be introduced into the 3’ half of the primer sequence (at 6-10 nucleotide positions) in such a way that all possible codons that could code for the conserved amino acids are represented. 3. The 3’ terminal base of the forward primer should match either the second codon position for an amino acid encoded by 2, 3 or 4 codons (excluding Arg, Leu, Ser) or the third position of a Met or Trp codon. The 3’ terminal base of the reverse primer should compliment the first base of a codon specifying an amino acid encoded by few alternative codons (e.g, Met, Cys, Trp, His....). 4. Degeneracy should be kept low (up to 1024 fold) by proper selection of priming sites. 113 5. Primer sequences should not have mono or dinucleotide polymers and should not end on three G or C residues at the 3’ end. 6. If the genes you're trying to clone are more varied than the species you have data for, design primers around regions that are rich in Cys, His, and Pro. These amino acids are less likely to change. Avoid regions rich in Ser, Ala, Asp, Glu, Phe, Tyr, and Arg because they are more likely to change........ 7. If the amplified DNA fragment becomes longer than 600 base pairs, the 5’ end of each primer should be altered in such a way that a six-cutter restriction site is present. 8. Using a compatible software, ensure that the designed primer sequences do not form stable hybrids with each other or amongst degenerate primer mixes. Avoid complementarity of more than three contiguous bases if these lie at the extreme 3’ end of a primer. Proper primer design is crucial for successful PCR amplification, and the guidelines provided above can be used as a reference to ensure that the primers will match the desired DNA sequences and produce accurate results. Procedure 1. Take two clean Eppendorf tubes of 1.5 ml each (one for male and another for female genomic DNA) and add ……. 10x Polymerase Buffer 5 μl dNTPs mix 8 μl Primers (10 pM/ul) 3 μl each Genomic DNA (male and female separately) 100 ng each Taq polymerase 1 U Distilled water to make 50 μl 114 Mix and overlay with 50 ul of mineral oil. 2. Set up the PCR machine with appropriate cycling conditions (temperature, time, and number of cycles). For example, an initial denaturation step at 95°C for 3 minutes, followed by 35 cycles of denaturation at 95°C for 30 seconds, annealing at 55°C for 30 seconds, and extension at 72°C for 1 minute. The final extension is usually carried out at 72°C for 10 minutes. 3. To visualize the amplified DNA bands, load the PCR products onto an agarose gel and run gel electrophoresis. Capture an image of the gel using a gel imaging system. Result The polymerase chain reaction (PCR) amplifies the target DNA sequence through a series of repeated cycles that involve denaturation, annealing, and extension. The resulting PCR product is a mixture of DNA fragments of varying sizes, which can be analyzed using gel electrophoresis. The detection of a specific DNA band on the gel confirms the successful amplification of the target sequence. Advantages and Disadvantages PCR is a powerful tool for isolating and amplifying specific DNA sequences, making it ideal for disease diagnosis through genetic mutation detection. However, there are two main drawbacks associated with PCR: 1. The use of DNA polymerase can sometimes lead to errors and mutations in the amplified DNA. 2. The primers used in PCR can sometimes bind incorrectly to the template DNA, leading to the production of non-specific PCR fragments. 115 Real time PCR Procedure Prepare the qPCR reaction mix in a sterile microcentrifuge tube: • Add 10 μl of qPCR buffer. • Add 0.5 μl each of primers (10 μM stock solution). • Add 0.5 μl of fluorescent probe (10 μM stock solution). • Add 0.25 μl of DNA polymerase with a fluorescent probe (5 U/μl stock solution). • Add 4.25 μl of sterile water. • Add 10 μl of DNA sample (concentration varies depending on the source and quality of DNA). Thoroughly mix the contents of the tube using a pipette. Transfer the reaction mix to a qPCR tube or plate that is compatible with the qPCR machine. Set up the qPCR machine with appropriate cycling conditions (temperature, time, and number of cycles), such as:  An initial denaturation step at 95°C for 3 minutes.  Followed by 40 cycles of denaturation at 95°C for 15 seconds, annealing/extension at 60°C for 60 seconds, and plate read at 60°C.  Analyze the qPCR data using suitable software to obtain a quantification cycle (Cq) value, which indicates the point at which the fluorescence signal of the amplified DNA reaches a threshold level. The Cq value is used to calculate the amount of DNA present in the sample. 116 Result Real-time PCR, also known as quantitative PCR (qPCR), enables realtime detection and quantification of amplified DNA during the amplification process. This technique utilizes fluorescent dyes or probes that specifically bind to the amplified DNA product, allowing for real-time monitoring of the reaction. The results are usually presented as a graph of fluorescence intensity over time, which enables determination of the amount of starting DNA template and the efficiency of the amplification reaction. Note  Prior to conducting the experiment, it is essential to take necessary precautions to ensure safety in the laboratory, maintain the quality and purity of the starting DNA material, optimize primer design and reaction conditions, and minimize potential sources of contamination.  Accurate and reliable results can be ensured by incorporating appropriate positive and negative controls.  In real-time PCR, calculating the threshold cycle (Ct) value with precision is critical. The Ct value indicates the point at which the fluorescence signal reaches a set threshold level. Using suitable software for data analysis is also crucial. 117 Optimization of Annealing Temperature • To refine the parameters that impact PCR results. • To fine-tune the annealing temperature for optimal PCR results. • To gain proficiency in the PCR process and the use of a thermal cycler. Principle To achieve optimal PCR results, familiarity with both the PCR technique and thermal cycler device is essential. This involves a comprehensive understanding of the basic concepts of PCR, including the functions of each reagent, the PCR reaction mechanism, and the thermal cycling steps. Familiarity with the thermal cycler device's specific features such as temperature range, ramp rate, and programmable settings is also necessary to optimize the PCR reaction. Procedure PCR Optimization In order to optimize a PCR reaction, it's important to adjust and find the best concentration of each of the components involved in the reaction. Each component has a typical or optimal concentration range, which needs to be determined through a series of experiments. The goal is to find the optimal concentration for each component to achieve the most efficient and specific amplification of the target DNA. It's important to note that while optimizing one 118 component, the concentrations of other components should remain constant to avoid interference. These components include the Taq polymerase, deoxyribonucleotides (dNTPs), magnesium, forward and reverse primers, and the DNA template. To determine the ideal concentration, optimization should be done one component at a time, while keeping the other components constant. The optimal concentrations of these components are generally within the following ranges: Taq polymerase at 1.25 units dNTPs at 200 μM each magnesium at 1.5-2.0 mM primers at 0.1-0.5 μM each DNA template at 1ng-1μg The aim of optimizing the component concentrations is to find the combination that provides the best reaction conditions for a successful PCR. Thermal Cycle Thermal cycling optimization is the process of finding the best temperature and duration for each step in the Polymerase Chain Reaction (PCR) process. The objective is to obtain the optimal results by determining the best temperature, duration and number of cycles for each step. There are three stages in the thermal cycling process. The first stage is the initial denaturation which takes place at a temperature range of 94-97°C and lasts for 3 minutes. The purpose of this step is to denature the template and activate the DNA polymerase. The second stage is where the PCR process is repeated 25 to 35 cycles. This stage consists of three steps, denaturation, annealing, 119 and elongation. Denaturation occurs at 94-97oC and lasts for 30 seconds. Annealing takes place at a temperature range of 50-65oC and lasts for 30 seconds. The elongation step occurs at 72-80 oC and lasts for 30 seconds to 1 minute. The final stage is the final elongation phase which lasts for 5-7 minutes. This step is crucial as it allows for the synthesis of many uncompleted amplicons to finish. Annealing Temperature The optimal annealing temperature refers to the temperature that allows the primers to bind best to the target DNA. Finding the optimal temperature is crucial to ensure that only specific products are produced and non-specific products are avoided. A common approach to optimize the annealing temperature is to incrementally raise or lower the temperature in small steps and measure the amount of amplified product produced at each step, until the optimal temperature is found. The temperature gradient PCR technique is often used to find the optimal annealing temperature. This technique involves performing the PCR reaction at different temperatures starting from 5°C below the calculated melting temperature of the primer pair. For instance, if the melting temperature of the primer pair is 58°C, the annealing temperature will start from 53°C and will be increased by 8 degrees, typically ranging from 53-60°C. Stock 1. PCR buffer 2. DNA Taq polymerase 3. dNTPs 4. MgCl2 5. Primers 120 6. DNA template 7. Nuclease free water Procedure To carry out a PCR reaction Step 1: Determine the standard concentration of PCR components: 1. Prepare a table to calculate the volume of each component needed. 2. Components include PCR buffer, Taq polymerase, dNTPs, MgCl2, forward primer, reverse primer, DNA template, and water Step 2: Prepare the master mix 1. 2. Mix all the components except for the DNA template. Multiply the volume of each component by the number of desired reactions plus one to account for pipetting error. 3. Distribute the master mix into special PCR tubes using pipettes. 4. Add the DNA template to each tube. Step 3: Centrifuge the tubes  Briefly spin the tubes to ensure that the components are well mixed Step 4: Set the thermal cycling conditions 1. Temperature, time, and number of cycles must be set 2. Different annealing temperatures may be tried based on the primer pair Tm Step 5: Start the PCR reaction  The final volume in the thermal cycler must be set to 50 μl 121 Result 1. Use a 2% agarose gel to analyze the results 2. Determine the optimum Ta. 122 Reverse Transcription PCR | RT-PCR Reverse transcription polymerase chain reaction (RT-PCR) is a technique that combines the principles of reverse transcription (RT) and PCR to amplify and detect specific RNA sequences. Reverse transcription is a process that converts RNA into complementary DNA (cDNA) using an enzyme called reverse transcriptase. PCR is a technique that amplifies specific DNA sequences using the enzyme polymerase and a set of specific primers. The basic steps of RT-PCR are as follows: 1. Reverse transcription: The RNA sample is mixed with reverse transcriptase, a primer (often called an oligo-dT primer) that binds to the poly-A tail of mRNA, and other reagents such as dNTPs (deoxynucleoside triphosphates) and MgCl2. This mixture is then heated to allow the reverse transcriptase to synthesize cDNA from the RNA template. 2. PCR amplification: The cDNA is then used as a template for PCR amplification. The PCR reaction mixture contains the cDNA, a set of primers that specifically bind to the target sequence of interest, and the polymerase enzyme. The PCR reaction is typically performed in a thermal cycler, where the sample is repeatedly heated and cooled to allow the primers to bind to the template and the polymerase to synthesize new DNA strands. 123 3. Detection: The amplified PCR product can then be detected by various methods, such as gel electrophoresis, fluorescence, or sequencing. RT-PCR is a highly effective method that enables the detection and quantification of specific RNA sequences, even in very small amounts. This technique is used across various fields such as molecular biology, genetics, and medicine. It serves various purposes such as analyzing gene expression, identifying infectious diseases, and discovering cancer markers. The technique is frequently utilized in gene expression profiling, to evaluate the level of gene expression and determine the sequence of an RNA transcript. It can also be used to identify the position of exons and introns when the genomic DNA sequence of a gene is known. To identify the 5' end of a gene, which represents the starting point of transcription, RACE-PCR (Rapid Amplification of cDNA ends) is performed. When studying gene expression during development, two main aspects are considered, whether a specific gene is expressed in an embryo and where is the gene expressed in the embryo. Techniques such as RNA extraction and northern blotting can demonstrate expression, but in situ hybridization is needed to determine the specific location of expression. However, low levels of gene expression can make these techniques difficult to perform, so RTPCR is used to amplify transcripts for analysis and cDNA cloning. One of the main advantages of RT-PCR is its sensitivity, which allows for the detection of low-abundance RNA molecules. Besides, RT-PCR can be used to detect both known and unknown sequences, making it a valuable tool for discovery-based research. However, RT-PCR also has some limitations, such as the potential for false positive results, the need for specific primers, and the potential for contamination. 124 Stock 1. Stock solution - D Guanidium isothiocyanate 10 gm Water 11.72 ml Sodium citrate (pH 7.4) 0.704 ml 10% Sacrocyl 1.05 ml 2. Working solution - D Stock solution - D 1 ml 2 – mercaptoethanol 7.5 ml Sodium citrate (pH 7.0) 0.75 M 3. 10% N-lauryl sarcosine 4. Water saturated phenol 5. Chloroform 6. Isopropanol 7. Ethanol 8. 10 X DNase I reaction buffer Sodium acetate 1 M MgSO4 1 M DNase I (RNase free) 9. RNase inhibitor EDTA 20 mM Oligo dT 0.5 μg/ml dNTP mix 10 mM DTT 0.1 M 10. 10 X Synthesis buffer 11. Reverse transcriptase 12. Primers 13. Taq DNA polymerase 125 14. Agarose (Solutions for RNA extraction should be DEPC treated) *Glassware should be acid treated autoclaved and baked at 300oC for 4 hrs and plastic wares should be rinsed with chloroform to inactivate RNase. Procedure Step 1: Isolation of RNA by AGPC Technique 1. Take out 10 mg of tissue in chilled PBS. 2. Mince the tissue on an ice-slab and homogenize it with 100 μl of solution D. Place the contents in a test tube. 3. Then, add 10 μl of 2M Sodium acetate (pH 4.0), 100 μl of phenol, and 200 μl of chloroform to the test tube. Mix well in a cyclomixer and store the mixture on ice for 15 minutes. 4. After 15 minutes, centrifuge the sample at 10 K for 20 minutes. 5. Carefully remove the aqueous phase (RNA), avoiding the organic and interphase (proteins and DNA). 6. Mix an equal volume of isopropanol with the RNA and store it at -70oC for 4 hours. 7. After 4 hours, centrifuge the RNA at 10 K for 20 minutes. Discard the supernatant and dissolve the pellet in 25 μl of solution D (1/4th volume). 8. Transfer the suspension to an Eppendorf tube and precipitate the RNA with 1 volume of isopropanol or 2 volumes of ethanol at -20oC for 4 hours. 9. Centrifuge the RNA at 15 K for 10 minutes, while maintaining the temperature at 4oC. 10. Wash the pellet in 80% ethanol, sediment it and let it dry. 11. Treat the extract in DEPC water at 65oC for 10 minutes, if needed. If not, dissolve it in 0.5% SDS. 126 12. Finally, store the RNA at -70oC for future use. Step 2: DNase I Treatment to RNA samples Prepare a work area with an ice slab. Gather the following materials: 1 μg of RNA, 1 RNase free microcentrifuge tube (0.5 ml), 1 μl of 10 X DNase I reaction buffer, 1 μl of RNase inhibitor, 1 unit of amplification grade DNase I, DEPC treated water, 1 μl of 20 mM EDTA. 1. Take 1 μg of RNA and transfer it to the RNase free microcentrifuge tube. 2. Add 1 μl of 10 X DNase I reaction buffer and 1 μl of RNase inhibitor to the tube with RNA. 3. Add 1 unit of amplification grade DNase I to the reaction 4. Bring the volume to 10 μl with DEPC treated water. 5. Place the tube with the reaction mixture on the ice slab and mixture. incubate it at 37°C for 15 minutes. 6. After 15 minutes, inactivate DNase I by adding 1 μl of 20 mM EDTA to the reaction mixture and heating it at 65°C for 10 minutes. 7. After 10 minutes, extract the mixture with phenol:chloroform and then with chloroform only. 8. Precipitate the mixture by adding 2 volumes of alcohol. 9. Wash the mixture with 80% alcohol. 10. Dry the mixture and dissolve it in DEPC treated water. Step 3: Preparation of First cDNA Strand 1. Take an autoclaved microcentrifuge tube and add 1-5 ug of total RNA in 13 μl of DEPC treated water. 2. Now, add 1 μl of oligo dT (0.5 mg/ml) to the tube and mix gently. 127 3. Heat the mixture to 70oC for 10 minutes and incubate on ice for a minute. 4. To the mixture, add the following components in order: 10 X synthesis buffer (2 μl) dNTP mix (10 mM, 1 μl) DTT (0.1 M, 2 μl) RTase (200 U/μl, 1 μl) 5. Mix all the components gently and collect the reaction mixture after a brief spinning. 6. Incubate the mixture at room temperature for 10 minutes. 7. Transfer the tube to a water bath preset at 42oC and let it stand for 50 minutes. 8. Finally, terminate the reaction by increasing the temperature to 70oC for 15 minutes and then cooling on ice. Step 4: Polymerase chain reaction 1. Prepare the reaction mixture. Take a small sample of 1st strand cDNA (approximately 1 μl). Add 8 μl of 10X synthesis buffer to the sample. Add 1 μl of Primer 1 (10 μM) to the mixture. Add 1 μl of Primer 2 (10 μM) to the mixture. Add 1 μl of Taq DNA polymerase (5 U/μl) to the mixture. Add water to the mixture to make a total volume of 80 μl. 2. Overlay with mineral oil: Gently stir the reaction mixture and add 2 drops (approximately 100 μl) of mineral oil on top to prevent evaporation during heating. 3. Initial denaturation: Place the reaction mixture in a thermocycler and heat it at 94oC for 5 minutes to denature the DNA. 4. Repeat the PCR cycle 15-30 times: 128 Denature the DNA at 94oC for 1 minute. Anneal the primers to the DNA at 50oC for ½ minute. Synthesize the new DNA strand using Taq polymerase at 72oC for 1 minute. 5. Gel electrophoresis: Remove 10-12 μl of the amplified DNA from the reaction mixture and analyze it on an agarose gel to confirm successful amplification. Notes The concentration of Mg++ may affect the PCR results and may need to be optimized for different PCR conditions. 129 Digestion of DNA with RE Restriction endonucleases are a type of enzyme found in bacteria and other prokaryotes that have the ability to recognize and cut specific sequences of nucleotides in double-stranded DNA. These enzymes play a role in defending the cell against invading viral bacteriophages by cleaving their DNA and preventing replication. There are over 300 known restriction enzymes, and each is named based on the organism it was isolated from. When restriction enzymes cut DNA, they produce fragments called restriction fragments that can have either a blunt end or a sticky end. Both types of cuts are useful in molecular genetics and can be used to join DNA fragments. Restriction enzymes play a significant role in several areas of genetic research. These enzymes are designed to help create new DNA molecules by cutting and splicing existing DNA sequences through a process called recombinant DNA technology. This allows scientists to produce genetic material with specific traits. Restriction enzymes are also used to study the genetic structure of DNA fragments and entire genomes. By cutting DNA into smaller pieces, they can map out the genetic makeup of an organism and provide valuable information for scientific research. This process is known as Restriction Fragment Length Polymorphism (RFLP) and is used to detect variations in the sequence of nucleotides in the similar fragments. 130 Principle The process of cleavage by restriction endonucleases (RE) involves the incubation of genomic DNA or DNA fragments that have been amplified using PCR. The RE enzyme is used to restrict the DNA at specific sequences that it recognizes. This process results in the production of fragments of different sizes, which can then be separated using agarose gel electrophoresis. For instance, the RE enzyme - MstII used here, cuts the DNA at the sequence '5CCTNAGG-3'. To ensure successful cleavage, the appropriate conditions of temperature, pH, and ionic strength must be maintained during the incubation process. Stock 1. DNA solution (0.5 μg/μl) 2. MstII (3U/ μl) 3. 10X restriction buffer 4. NaCl solution 5. Nuclease free water 6. 0.5 M EDTA Procedure 1. Gather all necessary materials including a clean microcentrifuge tube, a DNA solution with a concentration of 1 μg/μl, 10X restriction buffer, a solution of NaCl, and water. 2. Label the micro-centrifuge tube. 3. Add the following components to the tube:  1 μl of the DNA solution  2 μl of the 10X restriction buffer  1 μl of the NaCl solution  15 μl of water 131 4. Add MstII to the reaction mixture. The amount of MstII should be 3 units for each microgram of DNA. 5. Incubate the reaction mixture for 20 minutes at 37°C in an incubator. 6. Stop the reaction by adding 0.5 μl of 0.5 M EDTA. 7. Add 5 μl of gel loading buffer to the reaction mixture. This will allow the mixture to be loaded onto the gel and run through electrophoresis to visualize the restriction enzyme's effects on the DNA. 132 Digestion of DNA with RE in Bacteriophage Restriction enzymes are specialized proteins that are found in various bacteria and single-celled organisms. These enzymes are designed to search a length of DNA for a specific base sequence. This sequence is usually between 4 and 6 base pairs long and is referred to as the recognition site. When the enzyme finds this recognition site, it binds to the DNA molecule and cleaves each of the double helix strands, resulting in the fragmentation of the DNA molecule. In this experiment, we will be working with a virus called Bacteriophage λ. This virus is specially designed to infect bacteria and has been the subject of many studies in molecular biology. We will take the DNA from this small virus and cut it into smaller pieces using restriction enzymes. The genome of Bacteriophage λ is quite small, only containing 48,502 base pairs, which is much shorter than the human genome, which contains about 3 billion base pairs. After cutting the DNA into smaller pieces, we use electrophoresis to separate the fragmented DNA on an agarose gel. This is accomplished by adding a loading buffer to the DNA sample to inhibit the restriction enzyme, and then exposing the sample to an electric field overnight. This drives the DNA fragments to migrate into the gel. Once the migration is complete, the gel is dyed with methylene blue, which allows the DNA bands to become visible. The gel can then be photographed and the pattern so received can then 133 be compared to a predicted result to figure out the location of specific genes and regions within the genome. 1. To learn about the nature and operation of a DNA restriction enzyme. 2. To gain proficiency in using micropipettes 3. To become familiar with DNA electrophoresis 4. To identify a DNA sample using a restriction digestion map 5. To compare the λ DNA bands on a gel with a known λ DNA restriction map Stock Equipment 1. Electrophoresis chamber 2. Container with TBE solution (1X) 3. Cooler with crushed ice 4. Freezer (frosty, if possible) 5. Microtube rack 6. Four microtubes 7. Camera, if desired 8. Gloves 9. 37°C water bath w/ floating rack 10. 60°C water bath or saucepan on a hot plate 11. 20-μl micropipette (or 10-μl micropipette) and sterile tips 12. Waterproof pen 13. 500-ml beaker Reagents 1. Beaker or foam cup with crushed ice for the following: 20 μl of 0.4 μg/μl λ DNA 2.5 μl BamHI restriction enzyme 134 2.5 μl EcoRI restriction enzyme 2.5 μl HindIII restriction enzyme 10 l distilled water 2. 1.0% agarose gel 3. 20 μl 10X loading dye 4. 0.002% methylene blue stain 5. Distilled water Procedure Step 1: Preparation for Gel Electrophoresis Advance Preparations 1. Check if the 1X TBE solution from the previous Gel Electrophoresis with Dyes activity is still available for reuse. 2. Collect enough ice cubes and foam cups for each lab group to use as containers for keeping the restriction digests cool during the lab. 3. Heat a pan of water to 55°C on a hot plate to be used for heating the restriction digests. 4. Fill another pan with water and heat it to 37°C on a hot plate to be used for heating the lambda DNA. 5. To reconstitute the lambda DNA, add it to sterile distilled water to reach a concentration of 0.4 g/l. 6. For each group, measure out the necessary amounts of lambda DNA, enzymes, and loading dye, and store them in the freezer until the lab. 7. Prepare the 1.0% agarose gel solution by melting 1.0 g of agarose in 100 ml of 1X TBE buffer either in a microwave or on a hot plate, and store it in the refrigerator if not in use within the next 30 minutes. 135 To prepare gel for electrophoresis, follow these steps: 1. Before starting the procedure, put on gloves and keep all the necessary enzymes and DNA aliquots on ice to maintain their integrity. 2. Label 4 microtubes with the following labels - 10X buffer, DNA, BamHI, EcoRI, HindIII and Water. Place these tubes in a tube rack for ease of access. 3. Using a micropipette set to 4 μl, add 4 μl of 10X buffer to each of the four tubes. Make sure to use a new tip for each buffer to avoid contamination. 4. Using the same micropipette, add 4.0 μl of DNA to each of the four tubes, again using a new tip for each sample. 5. In the control tube, add 32.0 μl of distilled water and in the other reaction tubes, add 30.0 μl. 6. Close the microtubes and place them in a 55°C waterbath for 10 minutes to heat the samples. Immediately after, place the tubes on ice for 2 minutes. 7. Add 2 μl of the relevant restriction enzyme to each of the reaction tubes. Make sure to use a new tip for each enzyme to prevent contamination. 8. Close the microtube caps and tap the bottom of the tubes gently on the desktop to ensure all the liquid has settled at the bottom. Finally, incubate the tubes overnight at 37°C. The tubes will be frozen until use, and can be used within 60 days. Step 2: Setting Gel on the Tray 1. Heat a pan of water to 60°C. 2. Pour enough agarose gels into the pan to warm and liquefy them. 3. Secure the ends of the gel tray with labeling tape and place the plastic comb in the slots. 136 4. Pour approximately 35-40 ml of agarose into each gel tray to create a thick gel. 5. Allow the gel to cool and solidify (about 15 minutes). 6. Store the gel trays overnight in a container or ziploc baggie with 0.5X TBE solution to prevent drying out. Procedure for Setting Gel on the Tray and Loading buffer: 1. Put on gloves. Fill a styrofoam cup with ice and place your DNA digestion tubes on it. 2. Take the 1.0% agarose gel and place it in the gel box with the wells at the negative end. 3. Carefully add 150 ml of 1X TBE solution to the gel box, making sure the gel is covered with 2 mm of buffer. 4. Gently remove the comb and make sure the buffer covers the gel. 5. Heat the microtubes in a 60°C water bath for 3 minutes to ensure the DNA is in linear form. 6. Use a micropipette set to 4 μl and add 4 μl of loading dye to the bottom of each microtube. 7. Set up the electrophoresis apparatus. Using a micropipette, load 20 μl of each sample into a well. 8. Turn on the power supply for 30-45 minutes and turn off once the purple dye is 1 cm from the end of the gel. 9. Unplug the gel box. To visualize the bands, place the gel in a 0.002% methylene blue solution in 0.1X TBE and stain overnight at 4°C or for 2 hours at room temperature. Step 3: Observation 1. Remove the student gels from the refrigerator. 2. Set up containers for staining near a sink and note that gels can be discarded in the regular trash receptacle. 137 Proceed as follows: 1. Take the gel and place it under white light. 2. Look closely at the gel to see if the bands are visible. 3. If the bands are not visible due to high background staining, take a container filled with 0.1X TBE solution. 4. Place the gel in the solution and agitate it gently. 5. Change the buffer every 30 to 60 minutes. Continue this process until the gel has reached the desired degree of destaining. 6. If you wish, take a picture of the gel. Restriction enzymes are special types of enzymes that can cut DNA at specific locations. These locations, known as restriction sites, are determined by the sequence of bases in the DNA, which often form a repeating pattern that reads the same forwards and backwards. These repeating sequences, called palindromes, are found on both the forward and reverse strands of the DNA. Restriction enzymes recognize and bind to these palindromic sequences and then cut the DNA between specific bases. In the present instance, there are three restriction enzymes used - EcoRI, HindIII, and BamHI, each of which has its own unique recognition sequence. To measure the size of each of the fragments produced when λ DNA is cut with each of these restriction enzymes, the fragments are separated using electrophoresis and compared to a molecular 138 ladder. The molecular ladder has bands of known sizes and is separated at the same time as the digested λ DNA. This allows for the determination of the sizes of the fragments produced by each restriction enzyme. Notes 1. Enzymes, particularly restriction enzymes, should be stored care-fully and at a specific temperature to maintain their activity. 2. When working with Lambda (λ) DNA, it is recommended to heat the sample to break the hydrogen bonds that hold it in a circular form. 3. Methylene blue dye is a less sensitive alternative to ethidium bromide, but it can be used to stain larger quantities of DNA. It should be handled with caution, as it can stain clothes and equipment. 4. When de-staining gels, only use distilled or deionized water and make sure to wash the work area thoroughly. 5. When making 0.1X TBE buffer, only use deionized water to avoid damaging the DNA with high chlorine levels in tap water. 139 Southern Blotting of DNA Southern blotting is a technique used to detect specific DNA sequences in a sample. The technique, named after Prof EM Southern in 1975, involves the separation of DNA fragments by size through electrophoresis, followed by transfer of the separated DNA onto a solid support, such as nitrocellulose or nylon membrane. The transferred DNA is then exposed to a labeled probe that binds to the specific sequence of interest. The probe can be radioactive, fluorescent, or enzyme-linked, and is usually complementary to the target sequence. The probe-bound DNA can then be visualized using autoradiography (for radioactive probes), fluorescence microscopy (for fluorescent probes), or chemoluminescence (for enzyme-linked probes). The Southern blotting process can be divided into three main steps: 1. Restriction enzyme digestion: The DNA sample is first treated with one or more restriction enzymes to cut the DNA at specific sites. This produces fragments of DNA of varying sizes. 2. Electrophoresis: The restricted DNA fragments are then separated by size through electrophoresis. This is typically done by running the fragments through an agarose gel, which acts as a sieve to separate the fragments based on their size. 3. Transfer and hybridization: The separated DNA fragments are then transferred from the gel to a solid support, such as 140 nitrocellulose or nylon membrane. This is done by a process called blotting, which can be done by capillary action or by electro-blotting. The transference of DNA from the gel to the membrane involves:  Depurination: The agarose gel containing DNA is treated with 0.2N HCl to depurinate the fragments and transfer fragments larger than 8kb.  Denaturation: The denaturation solution denatures the double-stranded DNA to single-stranded DNA, allowing hybridization with the probe.  Neutralization: The neutralizing solution adjusts the pH to enable hybridization. Once the DNA is transferred, it is fixed in place on the membrane. The membrane is then exposed to a labeled probe that binds to the specific sequence of interest. The probe can be radioactive, fluorescent, or enzyme-linked, and is usually complementary to the target sequence. The probe-bound DNA can then be visualized using autoradiography (for radioactive probes), fluorescence microscopy (for fluorescent probes), or chemiluminescence (for enzyme-linked probes). Stock Nylon membrane Vacuum transfer apparatus Shaker apparatus UV Transilluminator Micropipette Agarose gel electrophoresis 141 Procedure 1. Cut a nylon membrane slightly larger than the gel and activate it in distilled water for 5-10 minutes. 2. Set up the vacuum transfer apparatus and place the nylon membrane, followed by the gel. 3. Depurinate the DNA strand with 0.25N HCl. 4. Wash the gel with distilled water and treat it with 0.5N NaOH and 1.5N NaCl for 30 minutes. 5. Neutralize the gel after denaturation by treating it with neutralizing solution for 30 minutes. 6. Maintain the vacuum pump and simultaneously pour buffer over the gel using a pipette. Check the transfer carried out in 3 hours. After that, remove the gel and mark the pores on the membrane with an HB-Pencil. 7. Check the completion of transfer by viewing the nylon membrane with a UV transluminator. Wash the membrane in neutralizing solution, air dry, and bake at 80oC for 2 hours to fix the DNA to the nylon membrane. Result The DNA smear on the membrane indicates successful transfer of DNA. When the gel after transfer is viewed under UV light, no bands are seen, indicating that the transfer is complete. Advantages and Disadvantages Southern blotting is a time-consuming and labor-intensive process but it allows for the determination of molecular weight of restriction fragments and the measurement of relative amounts of different fragments in different samples. 142 Northern Blotting of RNA Northern blotting is a technique used to detect specific RNA sequences in a sample. The process is similar to Southern blotting, which is used to detect specific DNA sequences. The main difference is that Northern blotting is used to analyze RNA instead of DNA. The Northern blotting process can be divided into three main steps: 1. The RNA sample is first extracted from the tissue or cells of interest. The RNA is then denatured by heating it in the presence of formaldehyde or a similar chemical, which causes the RNA to become single-stranded. 2. The denatured RNA is then separated by size through electro-phoresis. This is typically done by running the RNA through an agarose gel, which acts as a sieve to separate the RNA fragments based on their size. 3. The separated RNA fragments are then transferred from the gel to a solid support, such as nitrocellulose or nylon membrane. This is done by a process called blotting, which can be done by capillary action or by electroblotting. Once the RNA is transferred, it is fixed in place on the membrane. The membrane is then exposed to a labeled probe that binds to the specific sequence of interest. The probe can be radioactive, fluorescent, or enzyme-linked, and is usually complementary to the target sequence. The probe-bound RNA can then be visualized using autoradiography (for radioactive probes), fluorescence microscopy (for fluorescent probes), or chemo-luminescence (for enzyme-linked probes). 143 The Northern blotting technique is widely used in molecular biology for detecting specific RNA sequences, for example for gene expression analysis, studying alternative splicing, and for identifying non-coding RNA. However, with the advancement of more sensitive techniques like quantitative PCR (qPCR) and microarray, the use of Northern blotting has declined. These newer techniques are more efficient in detecting and quantifying RNA molecules, but Northern blotting still holds its importance as it provides information about the size of RNA, which could be useful in identifying degradation products and determining the presence of specific isoforms. Principle The Northern blotting process involves using electrophoresis to separate RNA samples based on their size and then using a hybridization probe to detect the target sequence. Procedure 1. Obtain either total RNA or mRNA and prepare it for gel electrophoresis. 2. Load the RNA sample onto an agarose gel and run the gel electrophoresis to separate the RNA molecules based on size. 3. Carefully transfer the separated RNA from the gel to a sheet of nitrocellulose or other suitable blotting paper, ensuring that the pattern of separation remains intact. 4. Incubate the blot with a single-stranded DNA probe. This probe will form base pairs with its complementary RNA sequence and bind to form a double-stranded RNA-DNA molecule. The probe can be radioactive or have an enzyme bound to it (e.g. alkaline phosphatase or horseradish peroxidase). 144 5. Incubate the blot with a colorless substrate that the attached enzyme can convert to a colored product that can be seen or gives off light. Alternatively, if the probe was labeled with radioactivity, expose X-ray film directly. 6. Analyze the results to determine the size and quantity of the RNA molecules present in the sample and the location of the RNA-DNA hybrid. Advantages and Disadvantages Northern blotting has many benefits, such as being able to determine the size of RNA, detect alternate splice products, use probes with partial homology, and measure the quality and quantity of RNA before blotting. It can also be used to study gene expression by detecting specific RNA sequences in a mixture of RNA molecules. Moreover, the membranes used in the process can be stored and reused for years. However, there are also some downsides to Northern blotting. Small changes in gene expression may go undetected, and the samples can be degraded by RNases. Compared to RT-PCR, Northern blotting is less sensitive, but it has a higher specificity which reduces the chances of false-positive results. 145 Western Blotting Western blotting is a laboratory technique used to detect and analyze specific proteins in a sample. The basic steps of the procedure include: 1. The sample, typically cells or tissue, is lysed to release the proteins. The lysate is then separated by size using gel electrophoresis. This creates a separation of the proteins based on their molecular weight, with the smaller proteins migrating further than the larger proteins. 2. The separated proteins are then transferred from the gel to a solid support, such as a nitrocellulose or PVDF membrane. This step is called electroblotting. 3. The membrane is then blocked to prevent non-specific binding of the primary antibody. Common blocking agents include bovine serum albumin (BSA) or non-fat dried milk. 4. The primary antibody, which specifically binds to the protein of interest, is then added to the membrane and incubated. 5. A secondary antibody, conjugated to a detection enzyme or fluorophore, is then added to the membrane. This antibody binds to the primary antibody, thereby indirectly detecting the protein of interest. 6. The signal from the detection enzyme or fluorophore is then visualized, typically using X-ray film or a digital imaging system. The intensity of the band on the membrane corresponds to the amount of protein present in the original sample. 146 Western blotting is a common method used in the fields of molecular biology, biochemistry, and medicine to detect and quantify specific proteins in a sample of tissue homogenate or extract. This technique helps researchers understand and describe proteins, and can also be used to see how the expression levels of proteins change when subjected to different stimuli or treatments. Principle The western blot technique involves the separation of proteins through gel electrophoresis. The proteins can either be separated based on their natural 3-D structure or their length if they are denatured. After the separation, the proteins are transferred to a membrane like nitrocellulose or PVDF and then stained with specific antibodies to identify the target protein. The use of gel electrophoresis is important in western blot analysis to eliminate any potential issues with cross-reactivity of the antibodies. Stock Protein / antibody sample SDS-PAGE gel Nitrocellulose or PVDF membrane Transfer buffer (such as Towbin's transfer buffer) Blocking solution (such as 5% non-fat dry milk in TBS-T) Primary antibody Secondary antibody conjugated to a detection enzyme (e.g., HRP-conjugated anti-rabbit IgG), Chemiluminescent substrate (e.g., ECL substrate) X-ray film. 147 Procedure 1. Prepare an SDS-PAGE gel based on the desired molecular weight range. 2. Load the protein/antibody sample onto the gel and run the gel according to the manufacturer's instructions. 3. Transfer the separated proteins to a nitrocellulose or PVDF membrane using a transfer buffer and transfer apparatus. 4. Block the membrane with a blocking solution for 1 hour at room temperature. 5. Incubate the membrane with the primary antibody diluted in blocking solution for 1 hour at room temperature or overnight at 4°C. 6. Wash the membrane with TBS-T buffer to remove any unbound primary antibody. 7. Incubate the membrane with the secondary antibody conjugated to an enzyme for 1 hour at room temperature. 8. Wash the membrane with TBS-T buffer to remove any unbound secondary antibody. 9. Incubate the membrane with a chemiluminescent substrate for the required time. 10. Expose the membrane to X-ray film and develop the film according to the manufacturer's instructions. Results The Western blotting technique results in the appearance of a band on the X-ray film, indicating the presence and amount of the target protein in the sample. The technique is widely used in research and diagnostic laboratories. 148 Notes  Handle the protein/antibody sample carefully to avoid degradation or contamination.  Use appropriate safety precautions when handling chemicals and X-ray film.  Ensure that the primary and secondary antibodies are specific to the target protein and do not cross-react with other proteins in the sample.  Other types of paper or membranes can be used in place of nitrocellulose. Advantages and Disadvantages The western blot is a diagnostic tool that has several advantages and disadvantages. The benefits of using western blot include its high level of sensitivity and specificity in detecting anti-HIV antibodies, its role as the definitive test for mad cow disease, and its use in some forms of Lyme disease and Hepatitis B testing. In veterinary medicine, it is also used to confirm the FIV status in cats. However, the Western Blot is a complex and time-consuming procedure that requires specialized equipment and trained personnel. It can be expensive, especially when multiple tests are required. In addition, the quality of the samples can impact the results, leading to false negatives. 149 Dot Blot Dot blot is a biochemical assay that is used to detect and quantify the presence of specific target molecules in a sample. It is a simple, quick and cost-effective method that can be used to detect a wide range of targets, including DNA, RNA, proteins and small molecules. In a dot blot, the sample is blotted onto a nitrocellulose or nylon membrane and then probed with a labeled reagent, such as a radioactively labeled antibody or DNA probe. The target molecules on the membrane are then detected by autoradiography or chemiluminescence, respectively. Dot blots are often used as a preliminary screen for the presence of specific target molecules, or to quickly test multiple samples for the presence of a specific target. Principle The principle of dot blot in biotechnology is to transfer a small volume of sample onto a solid support such as nitrocellulose or PVDF membrane, and then to detect specific biomolecules such as DNA, RNA, proteins, or antibodies, using various detection methods such as hybridization with labeled probes, immunodetection with specific antibodies, or colorimetric reactions. The Dot Blot method allows for quick and simple analysis of a large number of samples in a small area, making it a useful tool for identifying specific molecules in large numbers of samples. The method is commonly used in applications such as DNA fingerprinting, protein identification, and disease diagnosis. 150 Procedure Dot blot is a method used to transfer nucleic acids or proteins from a gel or liquid onto a nitrocellulose or nylon membrane. Here is a stepby-step guide to perform dot blot: 1. The sample should be in a liquid form and can be extracted from cells, tissues or bacteria. 2. Cut the nitrocellulose or nylon membrane to the desired size and place it on a flat surface. 3. Using a micropipette, load the samples onto the membrane in a small spot or dot form. Ensure that the dots are spaced evenly on the membrane. 4. Place the membrane on top of a vacuum manifold and apply suction. This will cause the samples to be transferred from the dots onto the membrane. 5. Cross-link the samples to the membrane using a UV crosslinker or a chemical cross-linking agent. This helps to prevent the samples from washing away during the subsequent steps. 6. To prevent non-specific binding, block the membrane with a blocking buffer, such as 5% BSA. Incubate the membrane in the blocking buffer for about 30 minutes. 7. Incubate the membrane in the primary antibody for 1-2 hours. The primary antibody should be specific to the protein or nucleic acid of interest. 8. Wash the membrane with a washing buffer to remove any unbound primary antibody. This step should be repeated several times to ensure that all unbound antibody is removed. 9. Incubate the membrane in a secondary antibody that is conjugated to a detection enzyme, such as horseradish peroxidase (HRP). 151 10. Wash the membrane again to remove any unbound secondary antibody. 11. Add a detection reagent, such as ECL or chemiluminescent substrate, to the membrane. Incubate the membrane in the detection reagent for 1-2 minutes. Visualize the result by exposing the membrane to X-ray film or using a chemiluminescent imaging system. 12. The resulting dots will show the presence or absence of the protein or nucleic acid of interest. The intensity of the dots can be used to quantify the amount of the target molecule. Advantages 1. Dot blot is a simple and straightforward technique that does 2. This technique is relatively cheap compared to other not require specialized training or expensive equipment. molecular biology techniques such as PCR, gel electrophoresis, and Southern blotting. 3. Dot blot is highly sensitive and can detect even trace amounts of target molecules, making it ideal for detecting low abundance analytes. 4. Dot blot can be used to detect a variety of different types of molecules such as proteins, DNA, and RNA. 5. The dot blot technique is highly robust and can be performed under a wide range of conditions. Disadvantages 1. One of the main limitations of dot blot is that it provides low resolution compared to other techniques such as gel electrophoresis. 2. Dot blot can provide qualitative data but is not suitable for accurate quantification. 152 3. There is a possibility of interference from non-specific binding or cross-reactivity, which can result in false positive or negative results. 4. In some cases, sample preparation can be complex and time-consuming, requiring special techniques and reagents. 5. Dot blot can only be used to analyze one target at a time, making it unsuitable for multiplexing applications. 153 Immunoprecipitation Immunoprecipitation is a laboratory technique used in molecular biology and biochemistry to isolate specific proteins from a mixture. This technique involves using an antibody that specifically binds to the target protein, and using it to pull out the protein from the mixture. The protein-antibody complex is then precipitated (or pelleted) using a reagent such as protein A or protein G, and the resulting precipitate is washed to remove any non-specific contaminants. The target protein can then be analyzed by techniques such as gel electrophoresis or mass spectrometry. Immunoprecipitation is commonly used in the study of proteinprotein interactions, protein localization, and post-translational modifications. Principle The principle of immunoprecipitation is based on the specific interaction between an antibody and its antigen. In this technique, a specific antibody is used to bind to a target protein in a mixture of proteins. The antibody-antigen complex is then precipitated, or separated, from the other proteins in the mixture, allowing for the purification and analysis of the target protein. This method is commonly used in molecular biology and biochemistry to isolate and study specific proteins in complex mixtures. 154 Procedure 1. Prepare the sample by lysing cells or tissues in a lysis buffer. The sample should contain the target protein and any interacting proteins. 2. Prepare the primary antibody that will be used to specifically bind to the target protein. It is important to choose an antibody with high specificity for the target protein. 3. Mix the lysed sample with the primary antibody and incubate for 1-2 hours at room temperature. 4. Add protein A/G beads to the sample-antibody mixture and incubate for another 1-2 hours. 5. Wash the beads with washing buffer to remove any 6. Elute the target protein from the beads by adding elution unbound proteins. Repeat this step 2-3 times. buffer. The target protein is now ready for further analysis, such as Western blotting or mass spectrometry. 7. Analyze the eluted target protein using the chosen method of analysis. The goal is to identify any interacting proteins that were immunoprecipitated with the target protein. Advantages 1. Immunoprecipitation is based on the binding between an antibody and its target antigen, which provides high specificity in selecting a particular protein from a complex mixture. 2. It can detect low-abundance proteins that might not be 3. It can be used for the study of post-translational detectable by other methods, such as gel electrophoresis. modifications, protein-protein interactions, and the identification of novel proteins. 155 4. It can be used in various research areas, including cellular biology, biochemistry, and proteomics. Disadvantages 1. Immunoprecipitation is a multi-step process that can take several hours to complete, and can be prone to contamination and human error. 2. Antibodies are expensive and must be purchased for each experiment, making the process cost-prohibitive for some labs. 3. Non-specific binding of the antibody to other proteins or contaminants can occur, leading to false-positive results. 4. The quality of the antibody can greatly affect the results of the immunoprecipitation, and antibodies may not be available for all proteins of interest. 156 Sanger Sequencing DNA sequencing is the act of finding out the exact order of the nucleotide bases (As, Ts, Cs, and Gs) in a DNA molecule. This information is significant in various areas like medical diagnosis, biotechnology, and forensic biology. There are several techniques for DNA sequencing, including Maxam-Gilbert sequencing (chemical degradation method), Sanger sequencing (dideoxy chain- termination method), and high-throughput sequencing technologies. Among these methods, Sanger sequencing is the most commonly used and it was developed by Sanger and his team in 1975 due to its simplicity and reliability. The Sanger sequencing method employs the cycle sequencing technique, where dideoxynucleosides are marked with different fluorescent dyes, allowing all four reactions to occur in the same tube and be separated in a single lane on the gel. When the labeled DNA fragments pass through the bottom of the gel, a laser reader detects the fluorescence of each fragment (blue, green, red, or yellow) and compiles the data into an image. The Sanger method is based on the mechanism of DNA synthesis by DNA polymerases and requires the synthesis of a complementary DNA strand to the strand being analyzed. This process uses ddNTPs tagged with fluorescence dye (each nucleotide with a different color). The identity of the added deoxynucleotide is determined by its complementarity through base pairing with a base in the template strand. 157 In the Sanger sequencing reaction, nucleotide analogs called dideoxynucleoside triphosphates (ddNTPs) interrupt the DNA synthesis as they lack the 3'-hydroxyl group required for the next step. For instance, the addition of ddCTP to an otherwise normal reaction system causes some of the synthesized strands to end prematurely at the position where dC would normally be added, opposite a template dG. This results in different colored DNA fragments, which can be separated by size in an electrophoretic gel in a capillary tube. All fragments of a given length move together in a single band through the capillary gel and the color associated with each band is detected with a laser beam. The DNA sequence is read by identifying the color sequences in the bands as they pass the detector, with the amount of fluorescence in each band being represented as a peak in the computer output. Procedure Sanger's Method is a classic DNA sequencing method which involves the following steps: 1. A single-stranded DNA template is prepared for sequencing. This template is usually derived from plasmids, bacteriophages, or genomic DNA. The template can be amplified using PCR (Polymerase Chain Reaction) or other methods. 2. A short complementary primer, about 20-24 nucleotides in length, is annealed to the template. The primer is designed to anneal at the 5’ end of the sequence of interest. 3. A modified form of DNA polymerase (such as Taq polymerase) is used to extend the primer in the presence of four different dNTPs (deoxynucleoside triphosphates). The dNTPs contain one of the four bases (A, C, G, T), each labeled with a different fluorescent dye. 158 4. As the DNA polymerase extends the primer, it encounters a modified dNTP (ddNTP), which terminates the extension reaction. This creates a series of fragments of different lengths, each with a fluorescent label at the end of the sequence. 5. The labeled fragments are separated by electrophoresis on a gel or capillary. The gel or capillary is run at a high voltage, causing the fragments to separate based on their size. 6. The fluorescent labels are detected and the resulting data is analyzed to determine the sequence of the template DNA. The sequence is determined by analyzing the relative positions of the fluorescent labels on the gel or capillary. 7. The individual sequences obtained from the different reactions are assembled to form the final DNA sequence. This assembly is done using specialized software that matches the overlapping regions between the sequences to determine the final DNA sequence. Alternate method 1. Amplify the specific region of DNA using PCR (polymerase chain reaction). This will result in multiple copies of the desired DNA fragment. 2. Purify the PCR product mixture by removing any unwanted primers and dNTPs (deoxynucleoside triphosphates) to obtain a pure sample of the DNA fragment. 3. Perform the sequencing reaction, which uses special dyes to label the different DNA bases (A, C, G, and T). This will determine the sequence of the DNA fragment. 4. Clean up the product after the sequencing reaction to remove any excess dye terminators and unused primer. This is done using an ethanol precipitation protocol to purify the sample. 159 5. Separate the labeled DNA bases and identify them using capillary electrophoresis. The resulting data is then analyzed to produce a final DNA sequence. 6. Analyze the data obtained from capillary electrophoresis to produce a complete and accurate DNA sequence. This sequence will provide valuable information about the DNA fragment being analyzed. Advantages Sanger sequencing has a wide range of applications, including the detection of single nucleotide polymorphisms (SNPs), single-strand conformation polymorphism (SSCP), and mutations. It is a reliable and efficient method for DNA sequencing, making it a popular choice for many genetic research projects. 160 Maxam-Gilbert Sequencing DNA sequencing is the process of determining the precise order of nucleotides within a DNA molecule. The Maxam-Gilbert method, also known as chemical degradation sequencing, is a chemical-based DNA sequencing method that was first introduced in 1977. It is based on the selective chemical modification of DNA strands and the subsequent analysis of the modified strands. The Maxam-Gilbert method uses chemicals to selectively break DNA at specific locations, depending on the type of chemical used. These modifications generate specific fragments of DNA that can be separated and identified through electrophoresis. Electrophoresis is a method that uses an electric field to separate DNA fragments based on size. The first step in the Maxam-Gilbert method is to choose a DNA sample that needs to be sequenced. The sample is then divided into four aliquots, each of which is treated with a different chemical. The chemicals used in the Maxam-Gilbert method are hydroxylamine, which cleaves the DNA at purine bases, and a combination of chemicals called G, A, and C, which cleave the DNA at guanine, adenine, and cytosine bases, respectively. After treatment with the chemicals, the DNA fragments are separated by electrophoresis and the resulting bands are visualized using a method such as autoradiography. Autoradiography uses Xray film to detect radioactive isotopes that have been incorporated into the DNA samples during treatment. 161 Procedure The Maxam-Gilbert method is a chemical cleavage method for DNA sequencing. The steps for DNA sequencing through Maxam-Gilbert's method are as follows: 1. Obtain a sample of DNA from a suitable source such as bacteria or human cells. The DNA should be pure, free from contaminants and suitable for sequencing. 2. Cut the DNA sample into smaller fragments using restriction enzymes. These enzymes recognize specific DNA sequences and cut the DNA at these sites. 3. Heat the DNA sample to separate the double-stranded DNA 4. Add specific chemicals to modify the DNA, such as ethidium into single strands. bromide, for visualization of the fragments under UV light. 5. Label the DNA fragments with radioactive phosphorus or phosphorus-32 (32P) to make them visible under autoradiography. 6. Load the radiolabeled DNA fragments onto a polyacrylamide gel. Apply an electric field to the gel, causing the DNA fragments to move towards the positive electrode. The smaller fragments move faster and separate from the larger fragments. 7. Treat the gel with chemicals, such as hydrazine or nitrous acid, to cleave the DNA at specific points along the strand. 8. Expose the gel to X-ray film to visualize the DNA fragments. The radioactive 32P labels on the DNA produce an image on the film, which shows the position of each DNA fragment on the gel. 162 9. Analyze the autoradiogram to determine the sequence of the DNA fragments. The position of each fragment on the gel indicates the sequence of bases in the DNA. 10. Combine the sequences of the different fragments to obtain the complete DNA sequence. Advantages and Disadvantages The Maxam-Gilbert method has several advantages over other DNA sequencing methods. One of the main advantages is that it provides high-resolution data and can be used to sequence large DNA fragments. Additionally, the Maxam-Gilbert method is relatively quick and easy to perform and can be automated for highthroughput sequencing. However, there are also some limitations of the Maxam-Gilbert method. The method is not as sensitive as other DNA sequencing methods, and it may not work as well on samples that contain large amounts of contaminants. Furthermore, the method requires the use of hazardous chemicals and radioactive isotopes, making it potentially dangerous and requiring proper safety measures. 163 Pyrosequencing Pyrosequencing is a type of DNA sequencing technology that uses bioluminescence to identify the individual nucleotides (A, C, G, T) that make up a DNA strand. It is based on the principle of real-time sequencing by synthesis, where the next nucleotide is incorporated into the growing strand only after the previous one has been identified. The result of the reaction is a series of light signals, which are then converted into a DNA sequence. This technology is highly sensitive, fast, and scalable, making it useful for a wide range of applications, including genomic sequencing, epigenetic analysis, and bacterial identification. Principle Pyrosequencing is a fast and efficient DNA sequencing method that uses bioluminescence to identify and quantify the order of nucleotides in a target DNA sample. The process involves four main steps: template preparation, primer annealing, enzyme addition, and detection of incorporated nucleotides. The DNA sample is first amplified through PCR and mixed with a sequencing primer that serves as the starting point for the sequencing reaction. The addition of a sequencing enzyme triggers bioluminescent reactions for each incorporated nucleotide, producing light that is detected and processed to determine the DNA sequence. This technology is unique in its ability to directly detect the incorporation of nucleotides into a growing DNA strand. 164 Procedure Pyrosequencing is a method of sequencing DNA that involves the sequential release of individual nucleotides. The following are the steps involved in performing pyrosequencing: 1. The first step is to prepare the DNA sample for sequencing. This is done by isolating the DNA from the sample, such as blood or tissue, and then purifying it. 2. The next step is to amplify the target DNA sequence using polymerase chain reaction (PCR). This increases the amount of DNA available for sequencing. 3. The amplified DNA is then mixed with a primer that is complementary to the target sequence. This primer allows the DNA to be attached to a solid support, such as a bead or a well. 4. The next step is to add a sequencing enzyme, such as luciferase or ATP sulfurylase, to the mixture. This enzyme is responsible for releasing nucleotides in response to the incorporation of each new base into the growing DNA strand. 5. As the DNA strand grows, the sequencing enzyme releases individual nucleotides, which are detected and quantified by a luminescence-based assay. 6. The release of nucleotides is detected by a luminometer, which measures the amount of light emitted by each nucleotide. This data is used to determine the sequence of the DNA. 7. The data from the luminometer is then analyzed to determine the sequence of the DNA. This can be done using computer algorithms that match the data with a reference genome. 165 8. Finally, the results of the sequencing analysis are interpreted to determine the identity of the DNA sample and any mutations or variations that may be present. These are the steps involved in performing pyrosequencing. It is a fast and efficient method of sequencing DNA, and it is widely used in a variety of applications, including medical genetics and microbial genomics. Advantages 1. Pyrosequencing can sequence hundreds of thousands of DNA molecules simultaneously, making it a high-throughput method. 2. The results of Pyrosequencing are generated in real-time, 3. It can be used for a wide range of applications, including which makes it a fast method of sequencing. single nucleotide polymorphism (SNP) detection, transcriptomics, metagenomics, and others. 4. It is a highly sensitive method, capable of detecting small amounts of DNA. 5. Pyrosequencing is relatively less expensive compared to other sequencing methods, making it an attractive option for large-scale sequencing projects. Disadvantages 1. The maximum read length of pyrosequencing is around 400450 base pairs, which is shorter compared to other sequencing methods. 2. The accuracy of pyrosequencing is not as high as other sequencing methods, making it unsuitable for certain applications. 166 3. Pyrosequencing does not have the ability to detect complex genomic structures, such as insertions, deletions, or inversions. 4. This method requires large amounts of starting material, which can limit its use in certain applications. 5. The equipment used in pyrosequencing is complex and requires technical expertise to operate, which can limit its use in certain settings. 167 Multiplex DNA Sequencing Multiplex DNA sequencing is a high-throughput sequencing technique that allows the simultaneous analysis of multiple DNA samples in a single run. This method involves the addition of unique barcodes to each sample, which allows for the identification and separation of the individual sequences once the DNA has been sequenced. Multiplex DNA sequencing is commonly used in largescale genomic projects, gene expression analysis, and disease association studies. It offers significant cost savings compared to sequencing each sample separately, as well as increased efficiency in the analysis of large numbers of samples. Principle Multiplex DNA sequencing is a method of sequencing multiple DNA samples simultaneously in a single sequencing run. The principle behind this technique is to distinguish different DNA samples in the sequencing process by assigning them a unique identifier or barcode. This identifier is added to the DNA samples before sequencing, allowing them to be separated and identified later in the process. The multiplex sequencing process results in highthroughput and cost-effective sequencing of multiple DNA samples, making it useful for applications such as large-scale genomic studies and targeted gene sequencing. 168 Procedure Multiplex DNA sequencing is a technique used to sequence multiple DNA samples in a single reaction. Here are the steps to perform multiplex DNA sequencing: 1. Collect the DNA samples you wish to sequence and extract the DNA. You will also need to prepare libraries of the DNA samples, which involves fragmenting the DNA, ligating adapters, and amplifying the fragments. 2. Mix the DNA libraries of different samples in a single reaction to create a pool. 3. Check the quality of the DNA pool using gel electrophoresis to confirm that the DNA fragments are of the correct size and the concentration is adequate for sequencing. 4. Hybridize the DNA pool with a sequencing bead array, which is a bead-based technology that contains millions of individual beads, each with a specific sequence. This allows for parallel sequencing of multiple samples in a single run. 5. Perform a PCR reaction in an oil-based emulsion to amplify the DNA-sequencing bead complexes. This results in many copies of the bead-DNA complexes that are ready for sequencing. 6. Load the bead-DNA complexes into a sequencing instrument and run the sequencing reaction. The instrument will read the DNA sequences and generate data that can be analyzed. 7. Analyze the data generated by the sequencing reaction to determine the DNA sequences. You can use bioinformatics tools to align the sequences and compare them to reference sequences. 8. Interpret the data to identify any mutations or variations in the DNA sequences. This can provide valuable information 169 for a range of applications, including disease diagnosis and drug development. Advantages 1. Multiplex sequencing allows the simultaneous sequencing of multiple samples in a single run, making it an efficient and cost-effective solution for large-scale sequencing projects. 2. By sequencing multiple samples at once, the chances of detecting errors in the data are reduced, leading to improved accuracy of the results. 3. Multiplex sequencing reduces the costs associated with sequencing as a single run can analyze multiple samples at once, reducing the need for multiple sequencing runs. 4. With multiplex sequencing, a wide range of samples can be analyzed, including samples from different organisms, tissue types, and genetic backgrounds. Disadvantages 1. Multiplex sequencing requires specialized equipment and technical skills, which can make it challenging to implement for some researchers. 2. When multiple samples are analyzed in a single run, the signals from different samples can interfere with each other, affecting the quality of the results. 3. Because of the multiplexing, the sensitivity of the sequencing results may be reduced, potentially leading to missed variants or other important information. 4. Multiplex sequencing involves a complex process that can be difficult to manage and troubleshoot, leading to potential errors in the results. 170 Automated Sequencing Automated sequencing is a process of determining the sequence of nucleotides in a DNA sample using automated and computerized methods. The process involves breaking the DNA sample into smaller fragments, preparing the fragments for sequencing, running the sequencing reactions, and analyzing the data to determine the DNA sequence. Automated sequencing has revolutionized the field of genomics by making it faster, more efficient, and cost-effective to sequence large amounts of DNA. Principle The principle of automated sequencing is based on the use of computer-controlled instruments and laboratory methods to analyze the sequence of nucleotides in a DNA molecule. The process involves breaking down the DNA into smaller fragments, determining the sequence of the individual fragments, and then using software algorithms to reassemble the sequence into a complete genome. The automated sequencing process can be performed using various technologies, such as Sanger sequencing, next-generation or highthroughput sequencing, depending on the application and the desired accuracy and speed of the results. The automated sequencing process has revolutionized the field of genetics and molecular biology, enabling researchers to quickly and efficiently analyze DNA sequences for various applications, such as genetic diagnosis, genetic engineering, and drug discovery. Procedure 171 1. Obtain the DNA sample and extract it to purify and concentrate the DNA. 2. Cut the purified DNA into small fragments, usually ranging from 100-600 base pairs. 3. Attach primers to the DNA fragments. Primers are short pieces of complementary DNA that help the sequencing reaction start at a specific point. 4. Mix the DNA fragments, primers, and sequencing reagents, including dNTPs (nucleotides), polymerase, buffer, and fluorescent dyes. 5. Perform a series of thermal cycles to extend the primers and build a complementary strand of DNA using the dNTPs. 6. Add a sequencing terminator mixture that stops the polymerase reaction and incorporates fluorescent dyes into the newly synthesized strand of DNA. 7. Load the samples into a sequencing machine and analyze the images produced by the fluorescent dyes to determine the sequence of the DNA. 8. Use computer software to analyze the data and produce a readout of the DNA sequence. 9. Check the quality of the sequence, including the accuracy and completeness, and make any necessary adjustments. 10. Store the sequence data in a secure, accessible location for future use. Advantages 1. Automated sequencing machines can process hundreds or thousands of samples in a single run, making it faster than manual sequencing methods. 2. Automated sequencing machines are equipped with advanced algorithms and software, which ensure high accuracy and reproducibility of results. 172 3. Automated sequencing machines reduce the cost per sample as they are capable of processing many samples simultaneously, thus reducing the cost per sample. 4. Automated sequencing machines have the ability to process large amounts of samples in parallel, making it ideal for large-scale sequencing projects. 5. Automated sequencing machines are user-friendly and require minimal technical skills, making it accessible to a wide range of users. Disadvantages 1. Automated sequencing machines are expensive to purchase, maintain and repair. 2. Automated sequencing machines may be limited in their ability to process small samples or rare specimens. 3. Automated sequencing machines are complex machines that require maintenance and technical support. 4. Automated sequencing machines generate large amounts of data that requires specialized software and training to interpret accurately. 5. Automated sequencing machines may have limitations in terms of the types of samples they can process, the range of DNA sequences they can detect, and the types of genetic mutations they can identify. 173 Construction of Molecular Maps Construction of molecular maps is the process of creating visual representations of the spatial relationships and interactions between atoms, molecules, and other components of a biological system. This process can involve using different techniques and tools, such as Xray crystallography, nuclear magnetic resonance (NMR) spectroscopy, electron microscopy, and computer simulations, to obtain detailed information about the structure and function of biological macromolecules. The information obtained from these techniques is then used to create two-dimensional or threedimensional molecular maps, which provide insight into the molecular interactions and processes taking place within the biological system. These maps can be used for a variety of purposes, including the design of new drugs, the understanding of biological processes, and the improvement of medical treatments. Principle The principle of construction of molecular maps is to generate a visual representation of the molecular structure of a substance and its spatial distribution. This is achieved through the use of various techniques such as X-ray crystallography, NMR spectroscopy, electron microscopy, and computer simulations. The molecular maps provide information about the location and orientation of atoms, bonds, and functional groups within a molecule, which can be used to understand its properties, interactions, and reactions with other molecules. 174 Procedure 1. Choose a data set - select a set of genetic or molecular markers. 2. Determine the genetic variations in each sample using PCR or sequencing. 3. Calculate pairwise distances or similarities between markers and visualize them as a dendrogram or matrix. 4. Use specialized software to create the molecular map. 5. Choose highly informative markers based on frequency of occurrence or ability to distinguish between genotypes. 6. Add additional markers or re-analyze the data to improve resolution and identify errors. 7. Compare the molecular map to other reference datasets to ensure accuracy and reliability. Alternative method 1. Start by identifying the target molecule for which the molecular map needs to be constructed. 2. Collect all available information on the target molecule including its chemical structure, physical properties, and behavior under different conditions. 3. Use computer software or tools such as ChemDraw or ACD/Chem-Sketch to draw the chemical structure of the target molecule. 4. Identify the functional groups present in the molecule and mark them on the chemical structure. 5. Determine the bond connectivity between different atoms in the molecule and mark them on the structure. 6. Identify the stereochemistry of the molecule, if applicable, and mark it on the structure. 175 7. Using the marked-up structure, create a molecular map that clearly shows the different functional groups, bond connectivity, and stereochemistry of the molecule. 8. Use appropriate software or tools such as MarvinSketch or ChemBioDraw to label and annotate the molecular map, including the chemical name, molecular weight, and any relevant physical properties. 9. Validate the accuracy of the molecular map by crosschecking it with the original chemical structure and any available literature. 10. Save the molecular map in a suitable file format for further analysis or use in research. Advantages 1. It provides a detailed and accurate view of the molecular structure of a particular organism or system. 2. It provides a tool for identifying and studying diseasecausing genes and mutations. 3. It provides a means to compare the genomes of different organisms, leading to a better understanding of evolution and genetic diversity. Disadvantages 1. May be subject to errors and inaccuracies due to limitations in sequencing technology or data analysis methods. 2. The interpretation of molecular map data can be complex and may require advanced computational skills. 176 Restriction Mapping Restriction mapping is a technique used to create a physical map of a DNA molecule by cutting the DNA at specific locations using restriction enzymes. The resulting fragments are separated by size using gel electrophoresis and the pattern of fragments is used to determine the location and distance between the restriction enzyme cutting sites. Restriction mapping is used in molecular biology research to study the organization and structure of DNA, to identify mutations or genetic variations, and to construct recombinant DNA molecules. Principle The principle of Restriction Mapping is to identify the location of specific DNA sequences within a DNA molecule by using restriction enzymes to cleave the molecule at specific recognition sites. The resulting fragments are then separated by gel electrophoresis, allowing the size and location of the fragments to be determined. This technique can be used to create a map of the DNA molecule, which can aid in gene sequencing and identification. Procedure 1. Collect the DNA sample to be analyzed and obtain the restriction enzymes required for the mapping process. 2. Digest the DNA sample using the restriction enzymes. This can be done by adding the enzymes to the sample and incubating at the appropriate temperature and time for the enzymes to cut the DNA at their specific recognition sites. 177 3. Separate the resulting DNA fragments by size using gel electrophoresis. Load the digested DNA onto an agarose gel and run an electric current through it to separate the fragments according to size. 4. Stain the gel with a DNA-binding dye, such as ethidium bromide, to visualize the DNA fragments. 5. Measure the size of the DNA fragments using a DNA ladder as a reference. A DNA ladder is a mixture of fragments of known size used to calibrate the size of the fragments in the sample. 6. Use the fragment sizes to create a restriction map of the DNA. This can be done by comparing the fragment sizes to the expected sizes based on the known recognition sites for the restriction enzymes used. 7. Verify the restriction map by repeating the digestion and gel electrophoresis steps and comparing the resulting fragment sizes to the predicted sizes on the map. Advantages 1. Restriction mapping provides a precise and accurate map of the DNA sequence, allowing for identification of specific genetic sequences. 2. Restriction enzymes can identify and cut specific sequences, allowing for the identification of specific genes or mutations. 3. Restriction mapping allows for the analysis of DNA sequences, such as the identification of deletions or insertions. 4. Restriction mapping can be used in genetic mapping, which is important in understanding hereditary diseases. 178 Disadvantages 1. Restriction mapping can be a time-consuming process, requiring significant amounts of time and resources. 2. The process of restriction mapping can be expensive, as it requires specialized equipment and reagents. 3. Restriction mapping can be complex, and the interpretation of the results may require significant expertise. 4. Restriction mapping is limited to specific sequences recognized by restriction enzymes, which may not be sufficient for certain genetic analyses. 179 Molecular Markers Molecular markers are specific DNA sequences or variations that can be used to identify and differentiate individuals or populations of organisms. They are important tools in genetic research, plant and animal breeding, and forensic science. Restriction Fragment Length Polymorphism Restriction Fragment Length Polymorphism (RFLP) is a technique that involves digesting DNA with restriction enzymes and separating the resulting fragments by gel electrophoresis. The resulting pattern of fragments can be used to identify differences between individuals or populations. Principle Restriction Fragment Length Polymorphism (RFLP) is a molecular biology technique used to detect variations in DNA sequences between different individuals. The principle of RFLP is based on the fact that DNA sequences vary from person to person, and that these variations can be detected by analyzing the patterns of DNA fragments generated by restriction enzymes. Restriction enzymes are enzymes that cut DNA at specific sites, creating fragments of different sizes. By digesting DNA samples from different individuals with the same restriction enzyme, researchers can compare the resulting fragments and identify differences in the DNA sequences between the samples. 180 These differences can be visualized using gel electrophoresis, a technique that separates DNA fragments based on their size. The resulting pattern of DNA fragments, or DNA fingerprint, can be used to identify individuals, establish relationships between individuals, or detect genetic mutations associated with diseases. Overall, the principle of RFLP is based on the idea that variations in DNA sequences can be detected and analyzed by digesting DNA samples with restriction enzymes and visualizing the resulting fragments using gel electrophoresis. Stock DNA sample EcoRI restriction endonuclease enzyme Agarose, ethidium bromide Electrophoresis apparatus Gel documentation system Procedure 1. Start by obtaining a DNA sample from the organism you wish to analyze. This can be done by collecting a tissue sample or blood sample from the organism. 2. Extract the DNA from the sample using a DNA extraction kit. Follow the manufacturer's instructions for the kit. 3. Take the extracted DNA and cut it using a restriction enzyme. This enzyme recognizes a specific sequence of DNA and cuts it at a specific site. This produces fragments of varying sizes. 4. Separate the DNA fragments using gel electrophoresis. Place the DNA fragments in a gel and run an electric current through the gel. This separates the fragments by size. 181 5. Transfer the separated DNA fragments to a membrane, such as a nitrocellulose or nylon membrane. This is done using a technique called Southern blotting. 6. Hybridize the membrane with a labeled probe that will bind to a specific DNA sequence. The probe will bind to the DNA fragments on the membrane that contain the sequence it recognizes. 7. Use an imaging system to visualize the labeled probe and the DNA fragments it has bound to. This will produce a pattern of bands on the membrane. 8. Analyze the band pattern to determine the genotype of the organism. This can be done by comparing the band pattern to known patterns for the organism or by using computer software to analyze the pattern. Random Amplified Polymorphic DNA Random Amplified Polymorphic DNA (RAPD) is a PCR-based technique that amplifies random fragments of DNA using short, random primers. The resulting pattern of amplified fragments can be used to identify differences between individuals or populations. Principle RAPD is a PCR-based technique that uses short, arbitrary primers to amplify DNA fragments that contain polymorphic regions. The principle of RAPD is based on the fact that the primers used in the reaction are random and do not target specific regions of the DNA. This results in the amplification of a large number of fragments that are specific to the genomic DNA of the sample being tested. The amplified fragments are then separated by gel electrophoresis and visualized to identify polymorphic regions in the DNA. RAPD is a useful technique for identifying genetic variability within populations, 182 for fingerprinting organisms, and for studying genetic relationships between different organisms. Procedure Here is the step-wise procedure to perform RAPD: 1. Collect DNA samples from the organisms you want to study. Make sure to extract high-quality DNA using a suitable extraction method. 2. Prepare the PCR reaction mixture by adding the template DNA, primers, Taq polymerase, and other necessary components in the right proportions. Mix the reaction mixture well. 3. Set up the PCR amplification by placing the reaction mixture in a thermal cycler. Run the PCR reaction for the specified number of cycles, with appropriate temperature and time conditions. 4. Once the PCR amplification is completed, visualize the amplified DNA fragments by running the reaction products on an agarose gel. Stain the gel with an appropriate dye and visualize the bands under UV light. 5. Analyze the RAPD banding patterns by comparing the banding patterns of different DNA samples. The presence or absence of particular bands can be used to identify genetic variations among the studied organisms. 6. Interpret the results of RAPD analysis based on the banding patterns obtained. Use appropriate statistical tools to analyze and compare the results. 7. Record the findings of the RAPD analysis and document them in a clear and concise manner. 8. Draw conclusions based on the RAPD analysis results and their significance for the research question or objective. 183 9. Repeat the RAPD experiment if necessary to confirm the results or to address any additional research questions. Amplified Fragment Length Polymorphism Amplified Fragment Length Polymorphism (AFLP) is a PCR-based technique that combines the use of restriction enzymes and selective amplification of fragments using specific primers. The resulting pattern of amplified fragments can be used to identify differences between individuals or populations. Principle Amplified Fragment Length Polymorphism (AFLP) is a molecular genetic technique that is used to study genetic variation in different organisms. The principle of AFLP involves the use of selective PCR amplification of genomic DNA fragments that are flanked by restriction sites, followed by the separation of these fragments by electrophoresis and detection by autoradiography or fluorescence. AFLP allows for the detection of thousands of DNA polymorphisms at a time, providing a high-resolution genetic fingerprint of the individual or population under study. This technique is widely used in population genetics, evolutionary biology, plant breeding, and molecular systematics. Procedure 1. Start by isolating DNA from the tissue or organism of 2. Cut the DNA using restriction enzymes that recognize interest using a suitable protocol. specific sequences, which will generate fragments of varying lengths. 3. Add specific adapter sequences to the ends of the DNA fragments using ligases. 184 4. Amplify the DNA fragments using PCR with adapter-specific primers. 5. Use a set of selective PCR primers to amplify a subset of fragments that contain the restriction enzyme recognition site and the adapter sequence. 6. Separate the amplified fragments using gel electrophoresis, and visualize them using a suitable method, such as staining or autoradiography. 7. Analyze the resulting fragment patterns to determine genetic variation among the samples. This can be done by creating a dendrogram or performing statistical analysis to cluster similar samples together. 185 DNA Ship and Microarrays DNA chip and microarrays are high-throughput methods that involve immobilizing thousands of DNA fragments on a solid surface and hybridizing them with labeled probes. The resulting pattern of hybridization can be used to identify differences in gene expression or genetic variation between individuals or populations. These techniques can be used for genotyping, gene expression profiling, and other applications in genetics and biotechnology. Principle The principle of DNA chip, also known as microarray technology, is based on the ability to simultaneously analyze the expression or sequence of thousands of genes or DNA sequences in a single experiment. A DNA chip is a small glass slide or silicon chip on which thousands of microscopic spots or probes have been immobilized, each of which contains a unique DNA sequence. The samples containing DNA fragments are labeled with fluorescent dyes and hybridized to the probes on the chip. The intensity of the fluorescence signal is then measured, allowing researchers to identify which genes are expressed or which DNA sequences are present in the sample. This technology is widely used in genomics research, diagnosis, and personalized medicine. Procedure Performing DNA and Microarray Procedure 1. The first step in DNA and microarray procedure is to collect the sample. It could be blood, tissue, or any other material 186 that contains DNA. Make sure to collect a sufficient amount of sample for the experiment. 2. After collecting the sample, the next step is to isolate DNA from it. You can use a DNA isolation kit for this purpose. Follow the manufacturer's instructions to extract DNA. 3. Once you have isolated DNA, you need to measure its concentration using a spectrophotometer. The concentration of DNA should be within the required range for the microarray experiment. 4. The next step is to label the DNA using fluorescent dyes. This process will enable the microarray scanner to detect the DNA hybridization on the microarray slide. 5. The microarray slide contains thousands of DNA probes that can hybridize with the labeled DNA. Before adding labeled DNA to the microarray slide, wash the slide with buffer solution to remove any impurities. 6. Add the labeled DNA to the microarray slide and incubate it for several hours. During this time, the labeled DNA will hybridize with the complementary probes on the slide. 7. After incubation, scan the microarray slide using a microarray scanner. The scanner will detect the hybridized DNA probes and provide you with the data. 8. After scanning the microarray slide, you will get a data file that contains information about the hybridization patterns. Analyze this data using bioinformatics tools to determine gene expression levels or genetic variations. 9. Based on the analysis of the microarray data, you can interpret the results and draw conclusions. This information can be used to diagnose diseases, study genetic variations, or develop new drugs. 10. Finally, include all the relevant data and statistical analyses to draw precise conclusions. 187 Advantages 1. The DNA chip and microarray technology allows for the simultaneous analysis of thousands of genes in a single experiment, making it a highly efficient and cost-effective method. 2. DNA chips and microarrays are highly accurate in detecting gene expression levels, enabling researchers to obtain highly precise and reliable data. 3. DNA chips and microarrays allow researchers to perform experiments much faster than traditional methods, saving time and resources. 4. DNA chips and microarrays produce large amounts of data, allowing researchers to analyze the expression of thousands of genes in a single experiment. Disadvantages 1. DNA chips and microarrays only provide information about the expression of genes that are included in the array, so they may not be suitable for studying genes that are not present on the array. 2. The cost of DNA chips and microarrays can be high, especially if custom arrays are required. 3. Interpreting the large amounts of data produced by DNA chips and microarrays can be challenging, and requires advanced data analysis skills. 4. The technology used in DNA chips and microarrays can be complex, and requires specialized equipment and expertise to run experiments and interpret data. 188 Genomic Library A genomic library is a collection of DNA fragments that represent the complete genome of an organism. The fragments are usually cloned into a suitable vector, such as a bacteriophage or plasmid, and transformed into bacteria to create multiple copies. The genomic library serves as a valuable resource for studying the genetic makeup of an organism and is used in various applications such as sequencing the genome, identifying disease-causing genes, and developing new medicines. Principle The principle of Genomic Library is to create a collection of cloned DNA fragments representing the entire genome of an organism. These cloned fragments can then be used to study the genome and identify specific genes, their functions, and their interactions. This is done by randomly fragmenting the genome, cloning each fragment into a vector, and then transforming the vectors into host cells. The resulting library of cloned fragments can be screened to isolate and study specific genes of interest. The principle of genomic library is to provide a comprehensive representation of the genome for functional analysis and is a valuable tool for genetic research. Procedure 1. First of all, isolate high-quality and high molecular weight genomic DNA from the sample. This can be done using commercial kits or by using a protocol such as phenolchloroform extraction or organic extraction. 189 2. Then, cut the isolated genomic DNA into smaller fragments using restriction enzymes. The choice of restriction enzymes will depend on the size and complexity of the genome and the desired size of the library. 3. The cut DNA fragments are then separated by size using gel electrophoresis. The target fragment size for the library should be within the range of the insert size of the cloning vector. 4. The selected fragments are then ligated to a suitable cloning vector. The cloning vector should have the necessary elements for replication in bacteria and contain a selection marker. 5. The ligated vectors are then transformed into a host organism, such as E. coli. The host organism takes up the vector and replicates the DNA fragments, creating a large number of copies. 6. The transformed host organisms are then grown on selective media to identify those that contain the vector with the DNA fragment of interest. These clones are then screened using techniques such as hybridization, PCR, or restriction analysis. 7. The DNA fragment of interest is then sequenced using techniques such as Sanger sequencing, next-generation sequencing (NGS), or genome-wide sequencing. The sequencing process determines the order of the DNA base pairs in the fragment, providing information on the gene sequence. 8. The data generated from the sequencing process is then analyzed to identify the gene and its function. This information can be used to better understand the biology of the organism and its role in various processes. 190 Advantages 1. A genomic library provides a comprehensive representation of an organism's genetic information. 2. It allows the identification of all genes present in the genome of an organism. 3. It facilitates the study of the functions and interactions of individual genes. 4. It can be used for genetic screening and disease diagnosis. 5. It provides a useful tool for genetic engineering and biotechnology. 6. It can be used to study gene expression patterns, gene regulation, and the effects of mutations. Disadvantages 1. The process of creating a genomic library is complex, timeconsuming, and expensive. 2. The genomic library may not be a complete representation of the genome, as some regions may not be accessible for cloning. 3. The genome sequence may contain a large number of repetitive or redundant elements, making it difficult to distinguish between similar sequences. 4. The genomic library may not accurately reflect the functional state of the genome, as the DNA sequences may not be in their normal, active configuration. 5. There is a risk of contamination or mislabeling of the 6. The genomic library may not be suitable for studying specific genomic library, leading to incorrect results. regions of interest, such as those involved in complex biological processes. 191 cDNA Library cDNA library is a collection of complementary DNA (cDNA) molecules that represent the complete set of expressed genes in a particular cell or tissue. The cDNA is synthesized from messenger RNA (mRNA) molecules using the reverse transcription process and is then cloned into a vector for storage and replication in a host organism. The cDNA library can be used to study gene expression patterns, to identify new genes, or to clone specific genes for functional analysis. The library serves as a source of material for molecular biology experiments and is a valuable resource for understanding the molecular mechanisms underlying cellular processes. Principle The principle of cDNA library is based on the reverse transcription of messenger RNA (mRNA) into complementary DNA (cDNA). The cDNA library is a collection of cDNA molecules that represent the complete set of expressed genes in a particular cell, tissue, or organism. The process of creating a cDNA library involves the following steps: 1. Isolation of mRNA: Total mRNA is extracted from the target tissue or cell. 2. Reverse transcription: The mRNA is reverse transcribed into cDNA using reverse transcriptase, an enzyme that synthesizes DNA from RNA. 192 3. Cloning: The cDNA molecules are then cloned into a suitable vector, such as a plasmid or a phage, to generate a library of cDNA clones. 4. Screening: The library is screened for specific cDNA clones representing the genes of interest. The cDNA library can be used for various applications, including gene expression analysis, functional genomics, and the identification of novel genes. Procedure Step 1: Isolation of Total RNA 1. Obtain a sample of the tissue you wish to study (e.g. brain, liver, muscle). 2. Grind the tissue to a fine powder in liquid nitrogen. 3. Extract total RNA using a kit such as Trizol or RNeasy. Step 2: Synthesis of First-Strand cDNA 1. Add a reverse transcriptase enzyme, along with random primers, to the total RNA sample. 2. Incubate the mixture at 37°C for 60 minutes. 3. Stop the reaction by heating the mixture to 70°C for 10 minutes. Step 3: Synthesis of Second-Strand cDNA 1. Add a DNA polymerase, ligase, and buffer to the first-strand cDNA. 2. Incubate the mixture at 16°C for 2 hours. 3. Stop the reaction by heating the mixture to 70°C for 10 minutes. 193 Step 4: Preparation of cDNA Library 1. Purify the cDNA using a column-based purification kit. 2. Add restriction enzymes to the cDNA to cut the DNA into fragments of desired size. 3. Ligate the fragments to vectors (e.g. plasmids) using a DNA ligase. 4. Transform the ligation mixture into bacteria such as E. coli. 5. Screen the bacteria for successful transformation and pick individual colonies for further analysis. Step 5: Validation of cDNA Library 1. Isolate plasmid DNA from the transformed bacteria. 2. Amplify a portion of the cDNA using PCR. 3. Sequence the amplified cDNA to confirm the presence of target genes. 4. Analyze the cDNA library using techniques such as Northern blotting or qPCR to verify the quality of the library. Advantages 1. cDNA libraries are used in sequencing as they provide a high-quality representation of the mRNA in the sample. 2. cDNA libraries are constructed from a mixture of mRNA species and provide comprehensive coverage of the transcriptome. 3. cDNA libraries are relatively easy to handle and can be processed with existing lab equipment and procedures. 4. cDNA libraries provide consistent and reliable data and are less prone to technical errors than other types of libraries. 194 Disadvantages 1. cDNA libraries do not always provide a representative sample of the transcriptome, as only the most abundant transcripts may be included. 2. cDNA libraries may miss rare transcripts, particularly those that are poorly represented in the mRNA pool. 3. The technical limitations of cDNA library construction can impact the quality and representativeness of the data obtained. 4. The construction of cDNA libraries can be expensive and time-consuming, requiring specialized equipment and skills. 195 DNA Fingerprinting DNA fingerprinting, also known as DNA profiling, is a laboratory technique used to identify an individual's unique genetic information. This information is collected from a sample of an individual's DNA, such as blood, saliva, or hair, and compared to other DNA samples to determine the likelihood of a genetic relationship between two individuals. DNA fingerprinting is used in many applications, including forensic investigations, parentage testing, and genealogy research. Principle DNA fingerprinting is based on the principle of genetic variation. Every individual has a unique DNA sequence, except for identical twins, which allows them to be identified by their specific genetic code. This technique uses restriction fragment length polymorphism (RFLP) to compare the DNA samples from different sources. In RFLP, restriction enzymes are used to cut the DNA into fragments and then the fragments are separated by gel electrophoresis. The pattern of the fragments is unique for each individual and can be used to identify them. The principle of DNA fingerprinting is used in forensic science, paternity testing, and other applications where the identification of individuals is required. Procedure DNA fingerprinting, also known as DNA profiling, is a laboratory technique used to identify individuals based on their unique DNA patterns. Here is a step-by-step procedure for DNA fingerprinting: 196 1. Obtain a DNA sample from sources such as blood, saliva, semen, hair follicles, or skin cells. 2. Separate DNA from cellular components, such as proteins and lipids, to obtain pure DNA. 3. Use polymerase chain reaction (PCR) to create many copies of a specific target region in the DNA sample. 4. Cut the amplified DNA into fragments using restriction enzymes, which recognize specific sequences in the DNA. 5. Separate the fragments by size using an electric field in a gel matrix. Smaller fragments will move faster and end up farther from the point of origin. 6. Transfer the separated fragments to a nitrocellulose or nylon membrane. Fix the transferred DNA to the membrane using baking or ultraviolet light. 7. Probe the transferred DNA with a labeled complementary DNA probe to bind specifically to the target DNA. 8. Expose the membrane to X-ray film to detect the radioactive label on the probe. This will create a unique fingerprint for the individual. 9. Compare the DNA fingerprint with other fingerprints to determine if they are from the same individual. Advantages 1. DNA fingerprinting provides a highly accurate method of identifying individuals based on their unique genetic material. This helps in resolving disputes related to identity, such as in criminal investigations and parentage testing. 2. DNA evidence is considered very reliable in court and is often used to determine guilt or innocence. It is difficult to tamper with and is considered as a robust tool in criminal investigations. 197 3. DNA fingerprinting has been instrumental in solving many crimes, including serial killings, sexual assaults, and other violent crimes. It can also be used to eliminate suspects, leaving behind only those who are most likely to be responsible. 4. The process of DNA fingerprinting has become much faster and more efficient over the years, making it easier for investigators to identify criminals quickly. Disadvantages 1. DNA fingerprinting is a relatively expensive process, especially when compared to other forms of evidence. This can make it difficult for law enforcement agencies and other organizations to use it in all cases. 2. DNA fingerprinting requires special equipment and highly trained professionals. Technical errors or incorrect analysis can lead to false results and mistakes in criminal investigations. 3. The use of DNA fingerprinting raises serious privacy concerns as it involves collecting sensitive personal information. There are also concerns about the misuse of DNA samples and the possibility of genetic discrimination. 4. DNA fingerprinting is not always useful in all cases. For example, it may not provide results if the sample size is too small, or if it is degraded due to age or environmental factors. 198 Genetic Selection and Screening Method Use of chromogenic substrates Genetic selection and screening through the use of chromogenic substrates is a technique that is commonly used in molecular biology to identify and select specific genes or genetic traits. Chromogenic substrates are small molecules that can be used to detect the presence of a specific enzyme or protein. In genetic selection and screening, these substrates are used to identify cells or organisms that express a desired gene or trait. For example, a researcher may want to identify cells that produce a specific enzyme that is involved in a metabolic pathway. They can use a chromogenic substrate that is cleaved by the enzyme, producing a colored product. Cells that produce the desired enzyme will show a color change, allowing the researcher to select and isolate these cells. This technique can also be used to screen for genetic mutations or variations. By using chromogenic substrates that are specific for different genetic variants, researchers can identify individuals or organisms with particular genetic traits or mutations. Overall, genetic selection and screening through the use of chromogenic substrates is a powerful tool in molecular biology that allows for the identification and selection of specific genes and traits. 199 Principle The principle of genetic selection and screening by the use of chromogenic substrates involves the use of specific substrates that are designed to interact with enzymes produced by genetically modified organisms (GMOs). These substrates are often color-coded, so that the presence of a particular color indicates the presence of the desired enzyme or genetic trait. In this process, scientists modify the DNA of an organism to produce a specific enzyme that can be detected by the use of a chromogenic substrate. They then introduce the modified organism into a culture or environment that contains the substrate. If the organism produces the desired enzyme, it will interact with the substrate and produce a color change, allowing researchers to identify and select those organisms that possess the desired genetic trait. This process is commonly used in biotechnology to identify and select organisms that have been genetically modified for specific purposes, such as producing pharmaceuticals, enzymes, or other valuable products. It is a powerful tool for genetic engineering and has revolutionized the field of biotechnology. Procedure 1. Prepare a culture of the microorganisms you want to select or screen. Make sure you grow the culture under the conditions that favor the expression of the desired phenotype. 2. Select a suitable chromogenic substrate that produces a colored product when acted upon by the enzyme or protein of interest. Prepare the substrate according to the manufacturer's instructions. 200 3. Add the prepared chromogenic substrate to the culture containing the microorganisms. Make sure you add the substrate at a concentration that is sufficient to detect the desired phenotype. 4. Incubate the culture at the appropriate temperature and for the necessary amount of time to allow for the desired enzymatic reaction to occur. 5. Observe the culture for the presence of colored colonies or areas. The color development will indicate the presence of the enzyme or protein of interest, and thus the phenotype you are selecting for or screening against. 6. Confirm the presence of the desired phenotype by performing further tests such as genetic analysis or biochemical assays. 7. If necessary, repeat the process with different chromogenic substrates or under different conditions to further refine the selection or screening process. Advantages 1. Genetic selection and screening with chromogenic substrates allow for a quick and easy analysis of a large number of samples. 2. It is a cost-effective method, which reduces the time and cost of analysis. 3. Chromogenic substrates are specific and sensitive, which helps to identify the targeted genes with high accuracy. 4. It enables the detection of mutations in DNA or protein sequence that can be difficult to identify using other methods. 5. It is a non-destructive method, which can preserve the sample for further analysis. 201 Disadvantages 1. Chromogenic substrates have limitations in detecting mutations that occur outside of the specific target area. 2. False positives and false negatives can occur due to the specificity of the chromogenic substrate and the method used for screening. 3. The interpretation of results can be subjective, leading to inconsistencies in the analysis. 4. Genetic selection and screening with chromogenic substrates can only detect changes in the DNA or protein sequence, and may not be able to identify epigenetic changes that influence gene expression. 5. It may require specialized equipment and expertise, which can limit the accessibility of the method for some researchers or institutions. 202 Antibiotic Sensitivity Test for Detection of Recombinants Principle To determine the ability of organisms to produce mutants that are resistant, a gradient plate of a particular antibiotic can be used. This method involves growing bacteria on a gradient plate, which consists of two wedge-like layers of media, a layer of plain nutrients, and a top layer of antibiotic with a nutrient layer. The antibiotic is added as a top layer to the bottom layer, which produces an agar gradient of antibiotic concentration from low to high. Streptomycin is used to create the gradient plate. E.Coli, which is normally sensitive to streptomycin, will be spread over the surface of the plates and incubated for 24 to 72 hours. After inoculation, colonies will appear on the gradients. The colonies that develop in the high concentration are resistant to the action of the streptomycin and are considered as streptomycin-resistant mutants. For the isolation of antibiotic-resistant growth, the commonly used antibiotics are Rifampicin, Streptomycin, and Erythromycin. Stock NAM Petri-plates Glass rod Sterile test tube 203 Sterile swab Streptomycin Procedure 1. Pour 50 ml of nutrient agar into a sterile petri-plate and allow the medium to solidify in a standing position by placing a glass rod under one side. 2. Once the agar medium is solidified, remove the glass rod and place the plate in a horizontal position. 3. Pour nutrient agar with streptomycin (100 gm/ml) solution into the petri-plate. 4. Allow the medium to solidify. 5. Label the low and high antibiotic concentration areas on the 6. Pipette out 200 ml of 24-hour culture onto the gradient bottom of the petri-plate. plate. 7. Incubate the plate in an inverted position at 37°C for 48-72 hours. 8. Observe the plate for the appearance of colonies in the area of Low Streptomycin Concentration (LSC) and High Streptomycin Concentration (HSC). Alternative Method 1. Take a loop and touch the tops of 3-5 colonies of the same type of organisms from the primary culture plate to prepare the inoculum. 2. Transfer the growth to a tube containing saline solution. 3. Adjust the density of the test suspension to the standard turbidity by adding either bacteria or sterile saline. Compare the tube's turbidity with the standard. 4. Incubate the plates by dipping a swab into the inoculum and remove excess inoculum. 204 5. Rotate the plates through an angle of 60° after streaking the swab over the medium's surface three times. 6. Pass the swab around the edge of the agar surface. 7. Close the lid and let the inoculum dry at room temperature for a few minutes. 8. Place the antibiotics on the incubated plates using a template, sterile needle tip, or antibiotic dispenser. Result The gradient plate method produced successful results, with lower growth in the region of the plate without antibiotics compared to the high antibiotic concentration area. Notes  Ensure that the slanting position is not disturbed.  Make sure accurate pipetting.  Closely observe the incubated plates. 205 Insertional Inactivation Genetic selection and screening through insertional inactivation is a method of identifying genes that are essential for the growth and survival of an organism. This method involves the insertion of a DNA sequence, usually a transposon, into the genome of the organism. This insertion disrupts the function of the gene in which it lands. By selecting for cells that have lost the ability to grow or survive in a particular environment, researchers can identify the genes that are important for that function. This technique is commonly used in the study of bacteria and other microorganisms, as well as in the genetic engineering of plants and animals. Principle The principle of genetic selection and screening through insertional inactivation involves the use of genetic engineering techniques to insert a DNA sequence (usually a marker gene) into a specific location within the genome of a cell or organism. This DNA sequence disrupts the normal function of the gene(s) at that location, resulting in a phenotype that can be selected or screened for. For example, in bacterial genetics, a plasmid carrying a marker gene (such as antibiotic resistance) is introduced into a population of bacteria. The bacteria that take up the plasmid will have the marker gene integrated into their genome, disrupting the function of one or more essential genes. By growing the bacteria on a selective medium (containing the antibiotic), only those with the marker gene will 206 survive and grow. This allows for the selection of bacteria with specific phenotypes (e.g. antibiotic resistance). In eukaryotic genetics, similar techniques can be used to create knockout mice, where specific genes have been disrupted using marker genes. By breeding these mice and observing the resulting phenotypes, researchers can study the function of these genes in development and disease. Procedure 1. Start by identifying the gene that you want to inactivate using insertional mutagenesis. This could be a gene that is important for a particular biological process or one that is responsible for a particular trait. 2. Design a DNA construct that can be used to insert a selectable marker into the target gene. The construct should include a selectable marker, such as antibiotic resistance, and a promoter that can drive expression of the marker. 3. Introduce the DNA construct into the cells that you want to mutate. This can be done using a variety of techniques, including electroporation, lipofection, or viral transduction. 4. After introducing the DNA construct, select for cells that have incorporated the selectable marker into the target gene. This can be done using a selectable antibiotic or other selectable markers. 5. Once you have selected for cells with the inserted marker, screen for cells that exhibit the desired phenotype. This could be a change in morphology, growth rate, or some other observable characteristic. 6. Once you have identified cells with the desired phenotype, confirm that the mutation is due to the insertional inactivation of the target gene. This can be done using a 207 variety of techniques, including PCR, sequencing, or functional assays. 7. If necessary, repeat the process to identify additional cells with the desired phenotype or to optimize the mutation. 8. Finally, characterize the mutant phenotype by studying the effects of the gene inactivation on the biological process or trait of interest. This may involve additional experiments, such as gene expression analysis, biochemical assays, or animal studies. Advantages 1. Insertional inactivation is an efficient method of genetic screening as it can target a large number of genes at once. 2. Insertional inactivation can be highly specific in targeting particular genes, allowing for precise genetic modifications. 3. The screening process can be relatively quick, taking only a few days to identify which genes have been disrupted. 4. This method can be used in a wide range of organisms, including bacteria, yeast, plants, and animals. Disadvantages 1. Insertional inactivation can have off-target effects (in other genes), which can have unintended consequences. 2. As much of the genome remains uncharacterized, it is difficult to know which genes are important to disrupt. 3. Insertional inactivation may also affect regulatory regions that control gene expression, leading to unpredictable effects. 4. The process of creating and screening a large library of mutants can be complex and require specialized skills and equipment. 208 Complementation of Defined Mutations Genetic selection and screening through complementation of defined mutations is a method used to identify and study specific genes and their functions. This technique involves inducing mutations in a population of cells or organisms, and then selecting or screening for those with a particular phenotype of interest. Complementation is a process by which a functional copy of a gene can restore the phenotype of a mutant with a non-functional copy of the same gene. By introducing a plasmid or other genetic element carrying a functional version of the mutated gene into the mutant, researchers can determine if the phenotype is restored, indicating that the gene is responsible for the phenotype. This method is commonly used in genetic research to identify the functions of unknown genes, study gene interactions, and develop gene therapies. It is also used in plant and animal breeding to select for desired traits. Principle The principle of genetic selection and screening through complementation of defined mutations involves the use of genetically modified organisms to identify and study the functions of specific genes. This approach relies on the ability of cells to compensate for the loss of a gene function by complementing it with a functional copy of the gene. In this method, a mutant strain with a defined mutation in a gene of interest is crossed with another strain that has a second mutation in 209 the same gene, but at a different site. The resulting progeny are screened for complementation, which occurs when the two mutations together restore the wild-type phenotype. This process enables researchers to identify and study the functions of specific genes by analyzing the complementation patterns of the mutant strains. It can also be used to identify potential drug targets, as well as to study genetic diseases and developmental abnormalities. Procedure 1. Pick the set of mutations that need to be screened for genetic selection. This could include a set of randomly generated mutations or a specific set of mutations that are suspected to be linked to a particular phenotype. 2. Generate a complementation library that contains a set of plasmids or genomic fragments that express wild-type copies of the mutated genes. The library should be designed to cover all the mutations that need to be screened. 3. Transform the complementation library into a suitable host strain that is deficient in the specific function being studied. This is usually achieved by using a host strain that has a specific deletion or mutation in the gene of interest. 4. Plate the transformed host strain on a selective media that does not support growth of the host strain. Only the transformed host strains that express the wild-type copies of the mutated genes will be able to grow on the selective media. This is because the wild-type copies complement the mutations in the host strain. 5. Isolate the plasmids that complement the mutation(s) of interest from the complementation library. 210 6. Verify the complementation of the isolated plasmids by retransforming them into the original host strain and retesting for growth on the selective media. 7. Characterize the complementing plasmids by sequencing and mapping them to identify the wild-type genes that complement the mutated genes. 8. Repeat the screening process with the remaining mutations of interest until all the mutations have been screened and complementing plasmids have been identified and characterized. 9. Analyze the results to identify the genes that are responsible for the observed phenotype and gain a better understanding of the genetic basis of the phenotype. Advantages 1. This method allows for very precise selection and screening of specific mutations. This can help researchers identify the effects of particular mutations on gene function and disease. 2. It is very efficient method in terms of selecting the desired mutation. It requires minimal screening and can quickly identify the required mutations. 3. Since this method is based on complementation, it ensures a high degree of reliability in the screening process. 4. The cost of genetic selection and screening through complementation is relatively low compared to other methods. Disadvantages 1. This method is limited in terms of the mutations it can select for. It only works for mutations that can be complemented by a wild-type gene. 2. Although this method is efficient, it can be time-consuming to generate and screen the mutant strains. 211 3. This method may result in the selection of unexpected secondary mutations that can interfere with the interpretation of results. 4. It requires a large number of genetic resources such as mutant strains and wild-type genes. These resources may not always be readily available. 212 Protein Engineering Rational Design Rational design is the process of creating or engineering proteins with a specific function or property by designing or altering their amino acid sequence. This process is based on a thorough understanding of the structure and function of proteins, as well as the principles of molecular biology and genetic engineering. Rational design involves making deliberate changes to a protein's structure and properties using computational and experimental methods to improve its stability, activity, selectivity, and specificity. This approach has many potential applications in biotechnology, medicine, and other fields where proteins play a critical role. Principle Protein engineering is the process of creating new or improved proteins with specific functional properties through the manipulation of amino acid sequences. The principle of rational design in protein engineering involves using computational and experimental methods to design and engineer proteins with specific functions. This process involves understanding the structure and function of the target protein and using this knowledge to design new sequences that can improve or modify its activity, specificity, stability, and other properties. Rational design in protein engineering is an efficient and effective approach that can be used to create proteins with specific properties for a wide range of applications, including biotechnology, medicine, and industry. 213 Procedure 1. Identify the protein that needs to be engineered. This could be a protein that has a specific function or is involved in a disease process. 2. Determine the three-dimensional structure of the protein through various methods such as X-ray crystallography or NMR spectroscopy. This step is important to identify the regions of the protein that can be modified without affecting its overall structure. 3. Identify the specific site(s) on the protein that needs to be modified to achieve the desired function. This could be an active site or a binding site. 4. Choose the appropriate design strategy based on the specific requirements. Rational design strategies can be broadly classified into two types: sequence-based and structure-based. The former involves modifying the amino acid sequence of the protein while the latter involves modifying the protein structure through the introduction of new amino acids or modification of existing ones. 5. Design the protein variant: Use appropriate software tools to design the protein variant. This step involves identifying the amino acid substitutions or modifications that need to be made and predicting their effect on the protein structure and function. 6. Test the protein variant in vitro using various assays to determine its function and efficacy. This step could involve measuring the binding affinity or enzymatic activity of the variant. 7. Optimize the protein variant by fine-tuning the amino acid substitutions or modifications to achieve the desired 214 function. This step could involve iterative rounds of testing and optimization. 8. Validate the protein variant in vivo using appropriate animal models or cell lines. This step is important to determine the safety and efficacy of the protein variant. 9. Once the production protein variant is of the variant validated, scale-up the for clinical use or commercialization. This step could involve developing a production process that is cost-effective and scalable. Advantages 1. Rational design allows for precise control of the amino acid sequence and 3D structure of the protein. 2. It can increase the stability of a protein, allowing it to withstand harsh environmental conditions or chemical treatments. 3. Rational design can enhance the activity of a protein, making it more effective in its intended function. 4. It can tailor the properties of a protein to specific applications, such as drug delivery, biosensors, or industrial processes. 5. Rational design can reduce the time and costs associated with traditional protein engineering methods, such as directed evolution. Disadvantages 1. Rational design relies on a thorough understanding of the structure and function of the protein, which may be limited for some proteins. 2. It can be complex and require advanced knowledge of protein structure and function. 215 3. It may not always result in a successful outcome, as predicting the effects of mutations can be challenging. 4. It can be time-consuming, especially if multiple rounds of design and testing are necessary to achieve the desired properties. 5. It may limit the diversity of protein variants, as it is focused on specific changes to the protein sequence. 216 Stem Cell Therapy DNA Stem Cell Therapy is a revolutionary breakthrough in medical science that has the potential to cure diseases that were once considered incurable. Stem cells are cells that can divide and differentiate into any type of cell in the body. This capability makes stem cells an essential tool for medical research, especially for the treatment of various diseases and conditions. One of the latest advancements in this field is DNA Stem Cell Therapy, which is gaining recognition for its potential to cure diseases that were once considered incurable. DNA Stem Cell Therapy is a process that involves repairing or altering the DNA in stem cells to treat a specific disease or condition. Scientists have discovered that by making changes in the DNA of stem cells, they can alter their function, allowing them to treat a range of diseases. For instance, researchers have developed techniques to convert stem cells into neurons to treat neurological disorders, like Parkinson's disease, or into heart cells to treat heart disease. This new technology has the potential to revolutionize the way we treat diseases, and it has the potential to cure some of the most debilitating conditions that affect humans. One of the most significant advantages of DNA Stem Cell Therapy is its ability to cure diseases at the genetic level. In traditional medical treatments, drugs are used to manage symptoms, but they do not cure the underlying cause of the disease. With DNA Stem Cell Therapy, however, the root cause of the disease can be addressed, offering a cure to many diseases that were once considered incurable. This technology has the potential to cure diseases like 217 cancer, heart disease, and many others, making it a revolutionary breakthrough in medical science. Another advantage of DNA Stem Cell Therapy is its safety and effectiveness. Stem cells are natural cells in the body, and as such, they do not cause any adverse effects. Furthermore, since stem cells are self-renewing, they can provide a long-lasting cure for the disease. This makes DNA Stem Cell Therapy a highly sought-after treatment, as it offers a safe and effective cure for many diseases. With ongoing research and advancements, it is only a matter of time before DNA Stem Cell Therapy becomes a mainstream treatment option for many diseases. Procedure 1. Collect the stem cells from the patient or from a donor. This can be done through a biopsy or from a sample of cord blood. The collected cells are then isolated and cultured in a laboratory under sterile conditions. 2. The isolated stem cells are then multiplied or expanded in the laboratory. This is usually done by culturing the cells in special media with specific growth factors and nutrients. 3. The expanded stem cells are then characterized to determine their type and quality. This is done using various techniques such as flow cytometry, immunophenotyping, and molecular analysis. 4. The characterized stem cells are then prepared for therapeutic use. This involves selecting the appropriate cells, modifying them as needed, and processing them to obtain a suitable dose for transplantation. 5. The prepared therapeutic cells are then administered to the patient either intravenously or directly into the site of injury or disease. 218 6. The patient is then monitored for any adverse reactions or changes in their condition. This is done through regular check-ups and imaging studies. 7. The success of the stem cell therapy is evaluated over time by monitoring the patient's response and comparing it to the baseline. This is done using clinical and imaging assessments, and measuring the patient's quality of life and functional status. 219 Reverse Genetics Reverse genetics is a technique that involves manipulating the DNA or RNA sequence of a gene or organism in order to understand its function or phenotype. This technique involves starting with a known genetic sequence and then modifying it in some way to observe the resulting effects on the organism. This approach can be used to study the function of individual genes or to identify the roles of specific genetic pathways in biological processes. Reverse genetics is commonly used in research to create knockout or knockdown models of genes or to introduce mutations into an organism's genome to study their effects. It is a powerful tool for studying the genetic basis of disease and for developing new treatments and therapies. Principle The principle of reverse genetics is to work backward from a genetic sequence to its functional effects, rather than starting with a phenotype or observable characteristic and trying to identify the underlying genetic cause. This approach allows researchers to identify new genes and pathways involved in disease and other biological processes, and to develop targeted therapies and treatments based on this knowledge. Procedure 1. Identify the gene sequence you want to modify in the genome and obtain its DNA sequence. 220 2. Using molecular biology techniques, clone the target gene sequence into a plasmid or viral vector. 3. Introduce the recombinant DNA molecule into the host cells using transfection or transformation methods. 4. Screen the transfected or transformed cells to identify those with the desired genetic modification. You can use molecular markers, fluorescence, or antibiotic resistance genes to identify positive clones. 5. Culture the positive clones to obtain a sufficient amount of cells for further analysis or experiments. 6. Analyze the genetic modification of the target gene sequence by sequencing, PCR, or other molecular biology techniques. 7. Observe and measure any changes in the phenotype of the modified cells or organisms. This could include changes in growth rate, morphology, or biochemical properties. 8. Verify that the observed phenotype is caused by the genetic modification of the target gene sequence, rather than other factors. 9. Apply the findings to study the biological function of the modified gene sequence, or to develop new therapies or treatments for genetic disorders. Advantages 1. Reverse genetics allows researchers to identify the function of a particular gene. By silencing or knocking out a gene, researchers can determine what its function is in the organism. 2. Reverse genetics allows for targeted gene manipulation. It enables researchers to manipulate specific genes, enabling them to study the effect of genetic changes. 221 3. Reverse genetics is a fast and precise way of analyzing gene function. It allows for the creation of genetically modified organisms (GMOs) in a short time frame. Disadvantages 1. One of the main concerns with reverse genetics is ethical considerations. The manipulation of an organism’s genes can be viewed as tampering with nature and can raise ethical concerns. 2. Reverse genetics is a technically challenging process that requires significant expertise and specialized equipment. This can make it difficult for some researchers to use this method effectively. 3. Manipulating a gene can lead to unintended consequences that are difficult to predict. This can include changes to the organism’s overall physiology, which can have negative effects on its health and wellbeing. 222 Transgenic Technology Transgenic technology, also known as genetic engineering, is a process of modifying an organism's genetic makeup by adding or removing genes. It involves the introduction of foreign genes or DNA sequences into the genome of an organism, thereby altering its genetic traits. This technology has the potential to enhance the production of food, medicines, and other valuable products, as well as to improve the resistance of crops to pests, diseases, and environmental stresses. However, it also raises ethical and safety concerns related to the impact of genetically modified organisms on human health and the environment. Transgenic Plants 1. The first step is to isolate the gene of interest, which will be used for the transformation process. This gene can be isolated from another plant or synthesized in a laboratory. 2. The next step is to construct a vector that will carry the desired gene into the target plant cell. The most commonly used vectors are plasmids, which are small, circular DNA molecules. 3. Once the vector has been constructed, it needs to be introduced into the target plant cell. This is done through a process called transformation, which can be accomplished through several methods, such as Agrobacterium-mediated transformation, electroporation, or particle bombardment. 4. After transformation, the plant cells need to be screened for the presence of the transgene. This is done through a 223 process of selection, in which cells that have incorporated the transgene are identified and isolated. 5. Once the transgenic cells have been isolated, they need to be grown into mature plants. This is done through a process of regeneration, which involves culturing the transgenic cells on nutrient-rich media until they form shoots and roots. 6. Finally, the transgenic plants are characterized to determine if the desired trait has been successfully introduced. This is done through various analytical techniques, such as DNA sequencing, quantitative PCR, and phenotype analysis. 7. Once the transgenic plants have been successfully characterized, they can be tested in field trials to determine their performance under natural conditions. Transgenic Animals Transgenic animals are animals that have had their genetic makeup altered through the insertion, deletion, or replacement of specific genes using recombinant DNA technology. These animals can be created using a variety of methods, such as microinjection, where a gene is directly inserted into an animal's fertilized egg, or somatic cell nuclear transfer, where the nucleus of an adult animal's cell is transferred into an egg that has had its nucleus removed. Transgenic animals have a wide range of applications in various fields, such as agriculture, medicine, and biotechnology. In agriculture, transgenic animals can be created that are more resistant to disease and can produce more milk, meat, or other products. In medicine, transgenic animals can be used as models for human disease and to produce therapeutic proteins. In biotechnology, transgenic animals can be used to produce enzymes and other industrial products. 224 One of the most common applications is the production of transgenic mice, which are widely used as a model organism for human disease. These mice are genetically engineered to carry a specific human gene that is associated with a disease, such as cancer or Alzheimer's disease. These mice can then be used to study the disease and to test potential therapies. Transgenic animals have also been used to produce human proteins such as clotting factors, which are used to treat people with genetic disorders such as Hemophilia. Although transgenic animals have many potential benefits, there are also worries about their safety and ethics. Some people think that modifying animals' genes could lead to unintended harm to the environment and human health. As a result, it's important that we should also keep a close eye on its future and make sure it's properly regulated. 1. The first step in developing a transgenic animal is to identify the specific gene of interest that will be used to modify the animal. This gene can be isolated from a variety of sources including other animals, plants, or bacteria. 2. Once the gene has been isolated, it must be cloned into a plasmid, which is a small circular piece of DNA. This process ensures that the gene is available in large quantities for further use. 3. Then a construct is created, which is a vector that contains the gene of interest along with regulatory elements that ensure the gene is expressed in the animal. This construct is introduced into cells, such as embryonic stem cells. 4. The next step is to introduce the construct into cells that will be used to create the transgenic animal. This can be done by a variety of methods, including microinjection or electroporation. 225 5. The transgenic cells are then used to create transgenic animals by a process known as blastocyst injection. In this process, the cells are injected into an early-stage blastocyst, which is then implanted into a surrogate mother. 6. Then screen the offspring of the surrogate mother for successful transgenic animals. This is done by analyzing the DNA of the animal to confirm that the gene of interest has been integrated into the animal's genome. 7. Finally, analyze the phenotype of the transgenic animals to determine the effects of the transgene on the animal's development and behavior. This process includes monitoring the animal's growth, development, and overall health. 8. The transgenic animals that are produced can be used for further research or they can be bred to create a line of animals that contain the transgene. This requires ongoing maintenance and management of the animals, including regular monitoring of their health and development. 226 Buffers and Reagents ACES Buffer ACES 0.01 M NaCl 137 mM KCl 2.7 mM Na2HPO4 10 mM KH2PO4 1.8 mM Ampicillin (2S,5R,6R)-6-[(R)-(-)-2-Amino-2-phenylacetamido]3,3-dimethyl-7-oxo-4-thia-1azabicyclo[3.2.0]heptane-2-carboxylic acid. Bovine Serum Albumin (BSA) A globular protein composed of 583 amino acids. Ethidium Bromide C20H14N2Br2 A fluorescent intercalating agent used for the detection of nucleic acids in gel electrophoresis. GelRed It’s a proprietary formulation - a fluorescent nucleic acid stain used in gel electrophoresis Glycine Buffer Glycine 0.01 M 227 NaCl 137 mM KCl (2.7 mM) Na2HPO4 (10 mM) KH2PO4 (1.8 mM) HEPES Buffer HEPES 10 mM NaCl 137 mM KCl (2.7 mM Na2HPO4 10 mM KH2PO4 1.8 mM IPTG Isopropyl-beta-D-thiogalactopyranoside - a chemical used to induce the expression of genes in bacteria that contain a lac promoter. Kanamycin (2S,3R,4R,5R,6S)-6-[(2S,3S,4S,5R)-4-amino-5hydroxy-2-(hydroxymethyl)-3-[[(2S,3S,4S,5R,6R)-5amino-2-(hydroxymethyl)-6-[[(2R,3R,4R,5S,6S)-3,4,5trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy]-4carbamoyloxy-3-carboxy-4-carbamoyloxy-4,5dihydroxyoxan-2-yl]oxy-6-carbamoyloxyoxan-3yl]oxy]tetrahydro-2-furanone - an aminoglycoside antibiotic used to select for cells containing a plasmid with a kanamycin resistance gene. LB Broth Peptone yeast extract NaCl in a buffered solution 228 a rich medium used for growing bacteria in culture. MES Buffer MES 0.01 M NaCl 137 mM KCl 2.7 mM Na2HPO4 10 mM KH2PO4 1.8 mM PBS-T Phosphate-buffered saline (PBS) 0.1 M Tween-20 0.05% Phosphate Buffered Saline (PBS) NaCl 137 mM KCl 2.7 mM Na2HPO4 10 mM KH2PO4 1.8 mM Ripa Buffer NaCl 150 mM Tris-base 50 mM Nonidet P-40 1% Sodium deoxycholate 0.5% SDS 0.1% Protease inhibitor cocktail 1 tablet per 50 ml of buffer SDS Buffer Sodium dodecyl sulfate (SDS) 0.1% Sodium chloride (NaCl) 50 mM Tris-HCl 50 mM, pH 8.0 229 Sodium Dodecyl Sulfate (SDS) C12H25NaO4S - a detergent used to denature proteins in gel electrophoresis. TAE Buffer Tris-base 40 mM Acetic Acid 1 M EDTA 0.01 M Tris-HCl Buffer Tris-base 0.01 M HCl 0.1 M X-Gal 5-bromo-4-chloro-3-indolyl β-D-galactopyranoside - a substrate for beta-galactosidase that turns blue when cleaved, used for detecting gene expression. 230 Some Common Lexes Agarose Gel Electrophoresis This is a technique used to separate DNA fragments based on size. An agarose gel matrix is used to support the DNA during the electrophoresis process. Agarose A linear polysaccharide composed of alternating D-galactose and 3, 6anhydro-L-galactopyranose residues. Alkaline Phosphatase A hydrolytic enzyme that cleaves phosphate groups from nucleotides and other phosphorylated compounds. Ammonium Persulfate A strong oxidizing agent used as a catalyst in gel electrophoresis; an initiator in polymerase chain reactions. Ampicillin A penicillin-based antibiotic used to select bacterial transformants and prevent bacterial growth in cultures. Antifoam A chemical agent used to prevent foam formation in cell cultures. Bacterial strain A pure culture of bacteria used in biotechnology. BamHI A restriction endonuclease that cleaves DNA at a specific recognition 231 site. Bovine Serum Albumin (BSA) A protein used as a blocking agent in ELISAs and other immunoassays. Bromophenol Blue A pH indicator used in gel electrophoresis to monitor protein Buffers migration. Buffers are solutions that help maintain a stable pH during various stages of the genetic engineering process. Examples include Tris-HCl and phosphate-buffered saline (PBS). Calcium Chloride A salt commonly used to increase the stability of enzymes during protein purification. Carboxy-X-rhodamine (ROX) A fluorescent dye used as a reference standard in real-time PCR experiments. Cell culture media A nutrient-rich solution used to grow cells in a laboratory. Cellulase A hydrolytic enzyme that breaks down cellulose, used in the production of biofuels. Chloramphenicol An antibiotic used to inhibit bacterial growth in cell cultures and plasmid preparations. Coomassie Brilliant Blue A dye used in protein quantification assays. DAPI (4',6-diamidino-2- A fluorescent dye used to stain DNA 232 phenylindole) in fluorescence microscopy. Dextran A polysaccharide used as a molecular weight marker in electrophoresis. Dithiothreitol (DTT) A reducing agent used to protect proteins from oxidation and to prepare protein samples for analysis. DTT Dithiothreitol - a reducing agent used to break disulfide bonds in proteins. Ethanol A solvent used to inactivate enzymes and sterilize solutions. Ethidium bromide This is a DNA-staining dye that intercalates into the DNA molecule, causing it to fluoresce under UV light. Ethidium bromide is often used to visualize DNA fragments after agarose gel electrophoresis. Gelatin A protein derived from collagen, used as a coating agent for cell cultures. Glucose A sugar used as a carbon source in cell culture media. Glycerol A cryoprotectant used to store yeast and bacteria. Guanidinium Thiocyanate A chaotropic salt used to lyse cells and solubilize proteins in gene cloning and sequencing experiments. HEPES (4-(2-hydroxyethyl)- A buffering agent used in cell culture 1-piperazineethanesulfonic media. acid) 233 Hydrogen peroxide A chemical used as an oxidizing agent in experiments. Isopropyl alcohol An alcohol used for sterilization. Isopropyl β-D-1- A chemical used to induce expression thiogalactopyranoside of cloned genes in bacteria. (IPTG) KCl (Potassium Chloride) An electrolyte commonly used to stabilize enzymes during protein purification. L-glutamine An amino acid commonly used as a supplement in cell culture media. Ligases These are enzymes that join together the ends of DNA molecules. They are used to seal the ends of the cleaved DNA after insertion of the foreign gene. Examples of ligases include T4 DNA ligase and E. coli DNA ligase. Loading buffer A solution containing a tracking dye, a reducing agent, and a stabilizing agent used to load samples into gels in electrophoresis. Luria broth A nutrient-rich broth used to grow bacteria in a laboratory. Lysis buffer A solution used to break open cells for DNA extraction. Lysogeny broth (LB) A rich growth medium used to cultivate bacteria. Lysozyme An enzyme that cleaves the 234 peptidoglycan layer of bacterial cell walls, used in bacterial lysis and purification of bacterial DNA. Magnesium sulfate A chemical used to stabilize cell membranes. Magnesium Chloride An electrolyte commonly used in PCR (MgCl2) (Polymerase Chain Reaction) and other enzyme-based reactions. N, N-Dimethylformamide An organic solvent used in chemical (DMF) synthesis and purification of biopharmaceuticals. Neomycin An antibiotic used to inhibit bacterial growth in cell cultures and plasmid preparations. Nucleic Acid Gel Stains Fluorescent dyes used to stain DNA and RNA in gel electrophoresis. PCR reagents Polymerase chain reaction (PCR) is a method used to amplify specific DNA sequences. Reagents used in PCR include Taq polymerase, dNTPs (deoxynucleoside triphosphates), and primers. Phenol A polar solvent used for extraction of RNA, DNA and other biomolecules from tissues. Phosphate-buffered saline A buffer solution used as a diluent or (PBS) washing solution in many biochemical assays. 235 Plasmids These are circular pieces of DNA that are separate from the chromosomal DNA in a cell. They can be used as vectors to introduce foreign DNA into a target organism. Plasmids often contain origin of replication and antibiotic resistance genes. Polyethylene glycol (PEG) A polymer used to increase the efficiency of transformation. Polyvinyl alcohol (PVA) A polymer used as a gelling agent in electrophoresis. Ponceau S A protein stain used in electrophoresis to visualize protein bands and confirm transfer to nitrocellulose or PVDF membranes. Protease Inhibitors Compounds used to inhibit the activity of proteases in protein purification and analysis. Restriction enzymes These are enzymes that cut DNA at specific sequences, and are often used to cleave DNA in preparation for insertion of a foreign gene. Examples include EcoRI, HindIII, and BamHI. Restriction enzymes are usually composed of proteins. Sodium chloride (NaCl) A salt used to adjust the osmotic pressure of solutions. Sodium Dodecyl Sulfate A detergent used to solubilize (SDS) proteins in electrophoresis and 236 western blotting. Sodium Hydroxide A strong alkaline solution used in the preparation of cell lysates and protein samples. Sodium Pyruvate A metabolic intermediate used as a supplement in cell culture media. Streptomycin An antibiotic used to select for bacterial transformants. Tris-acetate-EDTA (TAE) A buffering solution commonly used Buffer in gel electrophoresis. Taq polymerase A protein composed of 5 subunits - a heat stable DNA polymerase commonly used in PCR reactions Transformation reagents Transformation is the process of introducing foreign DNA into a target organism. Reagents used in transformation include calcium chloride, heat shock, and electroporation. Tris Buffer A buffer solution used as the electrophoresis running buffer in protein SDS-PAGE electrophoresis. Triton X-100 A non-ionic detergent used in cell lysis and protein solubilization. Tryptic Soy Agar A commonly used solid growth medium for bacteria. Tryptone A complex protein used as a nitrogen source in growth media. 237 Yeast extract A source of vitamins and amino acids used to support the growth of yeast. Zymolyase An enzyme used to isolate yeast from its surrounding cell wall. 238 References Surzycki S. 2000. Basic Techniques in Molecular Biology, Springer. Fakruddin M, Mannan KS, Chowdhury A, Mazumdar RM, Hossain MN, Islam S, et al. Nucleic Acid Amplification: Alternative Methods of Polymerase Chain Reaction. J Pharm. Bioallied Sci. 2013. https://www.neb.com/applications/dna- amplification-pcr-and-qpcr Mullis KB, 1990. The unusual origin of the polymerase chain reaction. Sci Am 4: 56-61. http://himedialabs.com/TD/HTBM016.pdf Cox M, Doudna J, O’Donnell M, 2012. Molecular Biology Genes to Proteins. p.226. http://www.bio-rad.com/en-gu/applications- technologies/pcr-poly-merase-chain-reaction Chien A, Edgar DB, Trela JM, 1976. Deoxyribonucleic acid polymerase from the extreme thermophile, Thermus aquaticus. J Bacteriol, 3: 1550-7. https://www.neb.com/tools-and-resources/usage- guidelines/guidelines-for-pcr-optimization-with-onetaq-andonetaq-hot-start-dna-polymerases Mcpherson M, Møller S, 2007. PCR. Garland Science. Roux KH, 1995. Optimization and Troubleshooting in PCR. PCR Methods Appl, 5:185-94. Rychlik W, Spencer WJ, Rhoads R, 1990. Optimization of the Annealing Temperature for DNA Amplification in vitro. Nucleic Acids Res, 21: 6409–12. 239 Cox M, Doudna J, O’Donnell M, 2012. Molecular Biology Genes to Proteins. p.226. Munshi A. DNA sequencing, 2012 – Methods and Applications. InTech. Online Resources  http://www.thermofisher.com/sa/en/home/lifescience/sequen cing/sanger- sequencing/sanger-sequencing-workflow.html  https://www.thermofisher.com/pk/en/home/references/ambio n-tech-support/rna-isolation/tech-notes/total-rna-fromwhole-blood-for-expression-profiling.html  https://www.lerner.ccf.org/gmi/gmb/documents/RNA%20Isola tion.pdf  http://www3.appliedbiosystems.com/sup/URLRedirect/index.h tm?xDoD=4332809  https://www.ncbi.nlm.nih.gov/pubmed/17417019  http://www.nature.com/nprot/journal/v1/n2/full/nprot.2006.83 .html  https://www.ncbi.nlm.nih.gov/pubmed/16028681  http://openwetware.org/wiki/RNA_extraction_using_trizol/tri  http://openwetware.org/wiki/RNA_extraction#TRIzol_or_tri_foll owed_by_chloroform_and_precipitation  https://lifescience.roche.com/wcsstore/RASCatalogAssetStore/ Articles/NAPI_Manual_page_166169.pdfhttp://www.sabiosciences.com/ newsletter/ RNA.html 240  http://www.gelifesciences.com/webapp/wcs/stores/servlet/Cat egoryDisplay?categoryId=11763andcatalogId=10101andprod uctId=andtop=YandstoreId=11765andlangId=-1  http://www.thermofisher.com/pk/en/home/life-science/dnarna-purification-analysis/rna- extraction/rna-types/mrna- extraction.html  http://www.fastbleep.com/biologynotes/41/122/1216http://cshprotocols. cshlp.org/content/2006/1/pdb.prot4455  http://www.med.upenn.edu/lamitinalab/documents/EthanolPr ecipitationofDNA.  http://bitesizebio.com/384/the-basics-how-phenol-extractionworks/http://physiology.med.cornell.edu/faculty/mason/lab/z umbo/files/PHENOL- CHLOROFORM.pdf  http://www.ncbi.nlm.nih.gov/pubmed/9986822  http://bitesizebio.com/13516/how-dna-extraction-rnaminiprep-kits-work/ http://herpesvirus.tripod.com/research/protoDNA.htm  http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3792701  https://www.ncbi.nlm.nih.gov/pubmed/1867860  https://www.eeb.ucla.edu/Faculty/Barber/Web%20Protocols/P rotocol2.pdf http://www.fastbleep.com/biology- notes/41/122/1216  http://www.nature.com/app_notes/nmeth/2012/120805/pdf/a n8507.pdf  http://www.biotype.de/fileadmin/user/Dokumente/20120821_ Biotype_Differential_Lysis_SOP.pdf  http://projects.nfstc.org/pdi/Subject03/pdi_s03_m02_03.htm 241  http://www2.le.ac.uk/departments/emfpu/genetics/explained/ quantification  https://www.ncbi.nlm.nih.gov/pubmed/18201813  https://www.ncbi.nlm.nih.gov/pubmed/7702855  http://www.fastbleep.com/biology-notes/41/122/1216  https://www.promega.com/~/media/files/resources/applicatio n%20notes/genetic%20ident ity/an102%20extraction%20and%20isolation%20of%20dna%2 0from%20blood%20cards%20and%20buccal%20swabs%20in %20a%2096%20well%20format.pdf?la=en  https://www.gelifesciences.com/gehcls_images/GELS/Related %20Content/Files/1392818611307/litdoc28982222_201403110 44843.pdf  https://worldwide.promega.com/resources/pubhub/enotes/ho w-do-i-determine-the-concentration-yield-and-purity-of-adnasample/http://www.ncbi.nlm.nih.gov/pubmed/21638536  https://en.wikipedia.org/wiki/Polymerase_chain_reaction file:///C:/Users/User/Desktop/Polymerase_chain_reaction.svg  http://link.springer.com/protocol/10.1385%2F1-59259-3844%3A3  https://www.ncbi.nlm.nih.gov/pubmed/2999980http://www.no belprize.org/nobel_prizes/chemistry/laureates/1993/mullislecture.html  http://www.ncbi.nlm.nih.gov/tools/epcr/  https://www.thebalance.com/what-are-restriction-enzymes375674  http://medicine.jrank.org/pages/2779/Restriction-EnzymesUse-Restriction-Enzymes-in- Biotechnology.html 242  https://www.thermofisher.com/sa/en/home/lifescience/cloning/cloning-learning- center/invitrogen-school- of-molecular-biology/molecular-cloning/restrictionenzymes/restriction-enzymes-genome-mapping.html  http://www.bio.davidson.edu/genomics/method/Southernblot .html  https://en.wikipedia.org/wiki/Southern_blot  https://askabiologist.asu.edu/southern-blotting  https://www.mun.ca/biology/scarr/Gr12-18.html  http://www.bio.davidson.edu/courses/genomics/method/nort hernblot.html  https://www.thermofisher.com/pk/en/home/life-science/dnarna-purification-analysis/nucleic-acid-gelelectrophoresis/northern-blotting.html  http://www.nature.com/scitable/definition/northern-blot-287  https://en.wikipedia.org/wiki/Western_blothttp://www.ncbi.nl m.nih.gov/pmc/articles/PMC3456489/  https://www.thermofisher.com/pk/en/home/lifescience/protein-biology/protein-biology-learning-  center/protein-biology-resource-library/pierce-proteinmethods/overview-western-blotting.html  https://www.abdserotec.com/western-blotting.html  http://www.bio-rad.com/en-us/applicationstechnologies/introduction-westernblottinghttp://www.nature.com/scitable/definition/westernblot-288http://www.bio.davidson.edu/courses/genomics/ method/westernblot.html. 243 244