Target Gene Expression

Target gene expression refers to the process of inducing or evaluating the expression of a specific gene of interest within a biological system, such as cultured cells or an organism. This can be achieved through various methods, including overexpression, gene silencing, or gene editing. To analyze the expression levels of your target gene after performing a transfection or other genetic manipulation, you can use the following techniques:

  1. Quantitative PCR (qPCR) or reverse transcription qPCR (RT-qPCR): These methods are used to quantify the levels of a specific target gene’s mRNA in a sample. qPCR is used for DNA, while RT-qPCR is used for RNA. These techniques are sensitive, specific, and provide a quantitative measure of gene expression levels.
  2. Western blot: This technique allows for the detection and quantification of a specific protein of interest within a sample. By using antibodies specific to the target protein, you can evaluate the expression levels of your target gene at the protein level.
  3. Enzyme-linked immunosorbent assay (ELISA): ELISA is a sensitive and specific technique for detecting and quantifying a target protein in a sample. It uses antibodies and an enzyme-linked detection system to measure the levels of the target protein.
  4. Immunofluorescence microscopy: This technique uses fluorescently-labeled antibodies specific to the target protein to visualize its localization and expression levels within cells. Immunofluorescence can provide information on the subcellular localization of your target gene’s protein product.
  5. Flow cytometry: This method allows for the quantification of target protein expression in individual cells within a population. By using fluorescently-labeled antibodies specific to the target protein or fluorescent reporter genes (e.g., GFP), you can analyze the expression levels of your target gene in a heterogeneous cell population.
  6. RNA sequencing (RNA-seq): This high-throughput technique enables the analysis of gene expression profiles across the entire transcriptome. It provides a comprehensive view of gene expression levels, including the expression of your target gene, in a given sample.

The choice of technique depends on the goals of your experiment and the type of information you wish to obtain, such as mRNA or protein expression, localization, or quantification. In many cases, using a combination of these methods can provide a more comprehensive understanding of your target gene’s expression and function.

Incubate Cells

Incubating cells refers to maintaining the cells under controlled and optimal environmental conditions, allowing them to grow, divide, and carry out their normal cellular functions. To incubate cells, follow these general guidelines:

  1. Choose the appropriate cell culture medium: Use a suitable medium for your specific cell type, which usually contains essential nutrients, salts, and buffering agents. Common media for mammalian cells include Dulbecco’s Modified Eagle Medium (DMEM) or Roswell Park Memorial Institute (RPMI) medium. The medium should also be supplemented with additives such as 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin to provide additional nutrients and prevent bacterial contamination.
  2. Prepare the cell culture flask or plate: Cell culture vessels, such as flasks or multi-well plates, should be treated with a cell attachment factor (e.g., poly-L-lysine or collagen) if necessary, and pre-warmed medium should be added.
  3. Seed the cells: Harvest the cells from a previous culture, count them using a hemocytometer or automated cell counter, and dilute them to the desired concentration. Then, seed the cells in the prepared culture vessel.
  4. Incubation conditions: Place the cell culture vessel in a humidified incubator set at 37°C and 5% CO2. These conditions are standard for most mammalian cells but may vary for other cell types or specific experimental requirements.
  5. Monitor cell growth: Regularly observe the cells under an inverted microscope to check their morphology, confluency, and potential contamination. Depending on the cell type and growth rate, cells may need to be passaged (split) every 2-4 days to prevent overcrowding and maintain healthy growth.
  6. Maintain sterility: Follow aseptic techniques and use a laminar flow hood when handling cell cultures to minimize the risk of contamination.

Note that the specific incubation conditions, cell culture medium, and seeding density can vary depending on the cell type, experimental goals, and the protocol being followed. Always refer to the literature or supplier guidelines for the specific requirements of your c

Complex Condenser

A complex condenser, in the context of transfection, is an agent that promotes the formation of stable and compact complexes between the nucleic acid (DNA or RNA) and the transfection reagent. This condensation can improve the efficiency of cellular uptake and the overall transfection process. The formation of stable complexes is particularly important when using cationic lipids or polymers as transfection reagents, as these agents rely on electrostatic interactions with the negatively charged nucleic acids.

Polyethylenimine (PEI) is one of the most commonly used complex condensers in transfection. It is a cationic polymer that can form stable, positively charged complexes with nucleic acids through electrostatic interactions. The high charge density and multiple amine groups in PEI promote the condensation of the nucleic acids, resulting in the formation of compact, nano-sized particles that can be efficiently taken up by cells.

When using complex condensers like PEI, it is essential to optimize the ratio of the condenser to the nucleic acid (known as the N/P ratio) to achieve the desired enhancement in transfection efficiency without causing significant cytotoxicity. The optimal N/P ratio may vary depending on the cell type, nucleic acid, and transfection method used, so it is crucial to test different ratios and conditions to identify the best approach for your specific experimental system.

Transfection Enhancer

Transfection enhancers are additives or modifications to standard transfection protocols that can improve the efficiency of gene delivery into cells. They work by increasing the cellular uptake of transfection complexes or by promoting their release from endosomes after internalization. Some common transfection enhancers include:

  1. Chloroquine: Chloroquine is a lysosomotropic agent that can raise the pH of acidic intracellular compartments, such as endosomes and lysosomes. By inhibiting the acidification of these organelles, chloroquine can prevent the degradation of the transfection complexes and promote their release into the cytosol.
  2. Polybrene: Polybrene is a cationic polymer that can enhance transfection efficiency by neutralizing the negative charges on the cell surface and promoting the binding and internalization of the transfection complexes.
  3. Sodium butyrate: Sodium butyrate is a histone deacetylase (HDAC) inhibitor that can enhance transfection efficiency by promoting the transcription of the transfected genes. It is particularly useful for increasing the expression of exogenous genes when using viral vectors, such as lentiviruses.
  4. Proton-sponge effect reagents: Compounds such as 25 kDa branched polyethylenimine (PEI) and 2,3-dimethylmaleic anhydride (DMA) can act as “proton sponges” in endosomes, buffering the acidic environment and promoting the release of the transfection complexes into the cytosol.
  5. Dextran sulfate: Dextran sulfate is an anionic polysaccharide that can enhance transfection efficiency by promoting the interaction of the transfection complexes with the cell surface.

When using transfection enhancers, it is essential to optimize their concentrations and incubation times to achieve the desired enhancement in transfection efficiency without causing significant cytotoxicity. Additionally, the effectiveness of transfection enhancers may vary depending on the cell type and transfection method used, so it is crucial to test different enhancers and conditions to identify the optimal approach for your specific experimental system.

HeLa Transfection Reagent

HeLa cells are a widely used human cell line in biological research, and there are several transfection reagents available that have been optimized for these cells. Some popular transfection reagents for HeLa cells include:

  1. Lipofectamine 2000 (Thermo Fisher Scientific): A versatile, cationic lipid-based reagent that has been successfully used to transfect a wide range of cell types, including HeLa cells.
  2. Lipofectamine 3000 (Thermo Fisher Scientific): An advanced, cationic lipid-based reagent that has been designed for improved transfection efficiency, especially in difficult-to-transfect cell types. It is also suitable for HeLa cells.
  3. FuGENE HD (Promega): A non-liposomal, multi-component reagent that has been optimized for high transfection efficiency in a variety of mammalian cell types, including HeLa cells.
  4. JetPRIME (Polyplus-transfection): A versatile reagent that provides high transfection efficiency and low cellular toxicity for a broad range of cell types, including HeLa cells.
  5. X-tremeGENE HP DNA Transfection Reagent (Sigma-Aldrich): A reagent that provides high transfection efficiency for a variety of cell lines, including HeLa cells, with low cytotoxicity.

When selecting a transfection reagent for HeLa cells, consider factors such as transfection efficiency, cytotoxicity, ease of use, and cost. It is also crucial to optimize transfection conditions, including cell density, the amount of plasmid DNA, and the volume of transfection reagent, for the best results in your specific experimental setup.

Transfection Protocol

Transfection is a process by which foreign genetic material, such as DNA or RNA, is introduced into eukaryotic cells to study gene function, protein expression, or to produce recombinant proteins. There are several methods for transfecting cells, including chemical, physical, and viral methods. The choice of method depends on factors such as the cell type, transfection efficiency, and experimental goals. Here is a general transfection protocol using the chemical method with a liposome-based reagent, which is commonly used for transfecting mammalian cells:

  1. Cell preparation: a. Culture cells in appropriate growth medium (e.g., DMEM, RPMI) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin. b. Plate cells in a multi-well plate (e.g., 6-well, 12-well, or 24-well) 24 hours before transfection, ensuring they reach 70-90% confluency at the time of transfection.
  2. Plasmid DNA preparation: a. Prepare the plasmid DNA containing the gene of interest using a plasmid purification kit or commercial source. b. Measure the DNA concentration and quality using a spectrophotometer or a fluorometer.
  3. Transfection reagent preparation: a. Dilute an appropriate amount of the liposome-based transfection reagent (e.g., Lipofectamine 2000, Lipofectamine 3000, or FuGENE HD) in serum-free medium according to the manufacturer’s instructions. b. In a separate tube, dilute the desired amount of plasmid DNA in serum-free medium.
  4. Complex formation: a. Mix the diluted transfection reagent and plasmid DNA by gently pipetting. b. Incubate the mixture at room temperature for 15-20 minutes to allow complex formation.
  5. Transfection: a. During the incubation period, replace the cell culture medium in the multi-well plate with fresh, antibiotic-free medium. b. Add the transfection reagent-DNA complexes dropwise onto the cells, ensuring even distribution. c. Gently swirl the plate to ensure uniform distribution of the complexes.
  6. Incubation and analysis: a. Incubate the cells at 37°C in a humidified incubator with 5% CO2 for 24-72 hours, depending on the experiment’s objectives. b. Assess the transfection efficiency by observing the cells under a fluorescence microscope (if using a reporter gene, such as GFP) or by measuring gene expression or protein levels using techniques like qPCR, western blotting, or flow cytometry.

Note: Optimal transfection conditions (e.g., cell density, DNA amount, and transfection reagent volume) may vary depending on the cell type, plasmid size, and specific transfection reagent used. It is essential to optimize these parameters for your specific experimental setup. Additionally, some cell types may require the use of other transfection methods, such as electroporation, nucleofection, or viral vectors, for efficient transfection.

Human Polioviruses 1

Human poliovirus type 1 (PV1) is one of the three serotypes of poliovirus that can cause poliomyelitis, a highly contagious viral infection affecting humans. Poliovirus is an enterovirus, belonging to the Picornaviridae family. It is a non-enveloped, single-stranded positive-sense RNA virus.

Poliovirus is primarily transmitted through the fecal-oral route or, less frequently, by ingestion of contaminated water or food. The virus initially infects the cells lining the throat and intestines, where it replicates. In most cases, poliovirus infection is asymptomatic or causes mild, flu-like symptoms. However, in approximately 0.5% of cases, the virus can spread to the central nervous system, particularly the spinal cord and brainstem, leading to paralysis or even death.

PV1 is the most virulent and widespread of the three poliovirus serotypes. It is responsible for the majority of paralytic polio cases worldwide. The other two serotypes, PV2 and PV3, are less virulent and have been declared eradicated.

The development of effective vaccines has been instrumental in controlling and preventing polio. There are two types of polio vaccines:

  1. Inactivated poliovirus vaccine (IPV): Developed by Jonas Salk in the 1950s, IPV is based on inactivated (killed) poliovirus and is administered via intramuscular injection. IPV induces a strong antibody response, providing protection against all three serotypes of poliovirus without the risk of vaccine-associated paralytic poliomyelitis (VAPP).
  2. Oral poliovirus vaccine (OPV): Developed by Albert Sabin in the 1960s, OPV is a live, attenuated vaccine administered orally. OPV is highly effective in providing both individual and community immunity due to its ability to spread from vaccinated individuals to unvaccinated contacts. However, in rare cases, OPV can revert to a neurovirulent form, causing VAPP or circulating vaccine-derived polioviruses (cVDPV).

Ongoing global efforts, such as the Global Polio Eradication Initiative (GPEI), aim to eradicate all serotypes of wild poliovirus, including PV1. As a result of widespread vaccination campaigns, the number of polio cases has decreased dramatically worldwide, and the disease is now endemic in only two countries: Afghanistan and Pakistan. The complete eradication of polio would make it the second human disease, after smallpox, to be eradicated.

Encephalomyocarditis Virus


Encephalomyocarditis virus (EMCV) is a non-enveloped, single-stranded positive-sense RNA virus belonging to the Cardiovirus genus within the family Picornaviridae. EMCV primarily infects rodents but can also cause disease in a wide range of animals, including pigs, primates, elephants, and occasionally humans.

In rodents, EMCV infection can result in encephalomyelitis, myocarditis, and reproductive disorders. The virus is transmitted mainly through ingestion of contaminated food or water, as well as via respiratory secretions and feces. Rodents, particularly rats and mice, serve as the primary reservoirs for the virus, which can be shed into the environment.

In pigs, EMCV can cause a range of clinical signs, including sudden death, fever, weakness, and respiratory distress. Myocarditis and reproductive failure, such as abortions and stillbirths, are also observed in infected pigs. Outbreaks of EMCV infection can lead to significant economic losses in the swine industry.

In non-human primates, EMCV can cause myocarditis, encephalitis, and sudden death. The virus has been implicated in several outbreaks in primate research facilities and zoos.

Human infections with EMCV are rare, but some cases of encephalitis, myocarditis, and flu-like illnesses have been reported. The risk of zoonotic transmission is considered low, and the clinical significance of EMCV infection in humans remains uncertain.

There is no specific antiviral treatment for EMCV infection. Management is primarily focused on supportive care and addressing the symptoms of the disease. Prevention of EMCV transmission involves controlling rodent populations, maintaining good hygiene practices, and implementing biosecurity measures in animal facilities.

Human Adenovirus 3

Human adenovirus 3 (HAdV-3) is a non-enveloped, double-stranded DNA virus that belongs to the Adenoviridae family, specifically the Mastadenovirus genus. There are more than 50 distinct serotypes of human adenoviruses (HAdVs), which are classified into seven species (A to G) based on their genetic and biological properties.

HAdV-3 is a member of species B and is associated with various respiratory infections, predominantly in children. Infections caused by HAdV-3 can range from mild to severe and may include symptoms such as fever, sore throat, cough, and runny nose. In some cases, HAdV-3 infection can lead to more severe conditions like bronchitis, pneumonia, or acute respiratory distress syndrome (ARDS).

Transmission of HAdV-3 primarily occurs through respiratory droplets, contact with contaminated surfaces, or close personal contact with infected individuals. The virus can be shed in respiratory secretions, urine, and feces, and it is known for its ability to survive on surfaces for extended periods. Outbreaks of HAdV-3 infection can occur in settings where people live in close quarters, such as schools, military barracks, and hospitals.

There is currently no specific antiviral treatment for HAdV-3 infection, and management typically involves supportive care to alleviate symptoms and prevent complications. In severe cases, hospitalization and supplemental oxygen therapy may be required.

Prevention of HAdV-3 infection involves general hygienic measures, such as regular handwashing, avoiding close contact with infected individuals, and disinfecting contaminated surfaces. Although there are no commercially available vaccines for HAdV-3, researchers are investigating potential vaccine candidates to prevent adenovirus-associated respiratory infections in high-risk populations.

Eradicated Polio

Polio has not yet been fully eradicated, but significant progress has been made in reducing the number of polio cases worldwide due to widespread vaccination efforts. The Global Polio Eradication Initiative (GPEI), launched in 1988 by the World Health Organization (WHO) and its partners, has played a crucial role in the decline of polio cases.

In 1988, polio was endemic in 125 countries, with over 350,000 cases reported annually. As of 2021, wild poliovirus remains endemic in only two countries: Afghanistan and Pakistan. The number of reported polio cases has decreased by over 99% since the launch of the GPEI, with less than 200 cases reported in 2020.

One of the three serotypes of wild poliovirus, type 2 (WPV2), was declared eradicated in 2015, and type 3 (WPV3) was declared eradicated in 2019. Type 1 (WPV1) is the only remaining wild poliovirus serotype, and its eradication remains a top priority for the GPEI and global public health organizations.

The key strategies employed by the GPEI to eradicate polio include:

  1. Immunization: Widespread use of oral poliovirus vaccine (OPV) and inactivated poliovirus vaccine (IPV) to protect children from polio and interrupt the transmission of the virus.
  2. Surveillance: Monitoring the circulation of wild poliovirus and vaccine-derived polioviruses (VDPVs) through the testing of acute flaccid paralysis (AFP) cases and environmental samples.
  3. Outbreak response: Rapid identification and containment of polio outbreaks through vaccination campaigns and enhanced surveillance.
  4. Targeted interventions: Implementing tailored strategies to reach children in hard-to-reach or conflict-affected areas, where vaccination efforts are often more challenging.

The complete eradication of polio would make it the second human disease, after smallpox, to be eradicated. Achieving this goal requires sustained global commitment, resources, and efforts to ensure that every child is vaccinated and that the remaining reservoirs of wild poliovirus transmission are eliminated.