Science topic

Virus - Science topic

Explore the latest questions and answers in Virus, and find Virus experts.
Questions related to Virus
  • asked a question related to Virus
Question
2 answers
In 2018, I, Reginald B. Little (RBL), wrote a book wherein I proposed multiple stable isotopes of nonzero nuclear magnetic moments (NMMs) play essential roles in living organisms. Prior scientists had reasoned nuclei with spin angular momentum could in some instances be involved in life and affect biomolecules and organisms thereby, but RBL first in 2002 proposed an nuclear orbital angular momentum with such nuclear spin angular momentum as expressed by NMMs and the import of parity of NMMs by positive and negative NMMs by bare protons and bare neutrons. On the basis of such, RBL later in 2013 proposed unusual isotopic distributions in molecules can causes diseases. In 2018, in his book RBL proposed that HIV and some other viruses fractionate stable isotopes for the origin of the virus and infectivity and advancement of the HIV and some other viruses. In 2023 a group of scientists led by Dr. Vincent Balters in France proved RBL theory of HIV fractionating stable isotopes correct as they measured HIV infecting cells fractionates zinc isotopes with HIV infected cells enriching in 64Zn and the HIV virus in the infected cell expressing similar 64Zn enrichment. These scientists measured the surrounding media and cells enriching in heavier zinc isotope 66Zn, 67Zn and 68Zn. In 2024, I developed more my theory from March 2020 of using static magnetic fields, electric fields and electromagnetic waves to stimulate the virus in this case HIV in presence of feeding particular stable isotope to induce mutating the virus, in this case HIV virus. I thereby proposed in 2024 using gamma rays to irradiate HIV host for selective inducing resonance in the 64Zn to selectively stimulate 64Zn enriched HIV infected cells and 64Zn enriched zinc fingers in the HIV virus and capsid in such infected cells for altering the biochemistry by the gamma stimulating of 64Zn in the HIV infected cells and HIV therein for killing the infected cells and mutating the HIV in the cells and even in hard to reach resevoirs. But I have not done much research on nuclear gamma spectroscopy. I read that there is a history of the uniqueness of 64Zn and a few other metals (four or five other metals) for giant dipole resonance (GDR) and such research on GDR dates back to the 1950s. I open this discussion to inquire about the feasibility of selectively using gamma dipole resonance on 64Zn in living organisms to kill HIV infected cells and inactivate HIV. Can the gamma rays be tuned only to 64Zn? Is there any general affect of the gamma rays on other biomolecules, cells and tissues and organs? Thanks Sincerely Reginald B. Little (Stillman College, Tuscaloosa, Alabama)
Relevant answer
Answer
High levels of gamma rays of course can lead to cancer. But can low levels of gamma rays of specific wavelength only affect specific nuclei like 64Zn? Does the 64Zn have a specific frequency of gamma photon that only it will absorb and other atoms are not affected? As the 64Zn seems to have a natural giant dipole resonance of its nucleus can such resonance give sensitivity to low intensity gamma ray of such characteristic frequency so negligible effects on other nuclei of other elements and isotopes occur? Based on the answers to these questions and more it gives feasibility of a cure and the risk of side effects or creating other problems like cancer. This is a theory for a cure and the possibilities are interrogated . - Reginald B. Little (Stillman College ; Tuscaloosa Alabama)
  • asked a question related to Virus
Question
8 answers
I have developed an AFM, and I want to capture an impressive image with it. So far, I have only scanned calibration samples. The resolution power and sharpness of the images are quite good. I thought I could image a virus with the AFM. However, I am a physicist. Where can I obtain a non-hazardous virus sample that I can find locally and prepare with my limited knowledge? How can I prepare it?
Relevant answer
Answer
Isolating DNA is pretty simple but I'm not sure it will be sufficiently interesting for you. It certainly is small but is only going to look like a piece of string.
You can find viruses out in nature nearly everywhere. The density of viruses in seawater for example (or lake water) is about 10e6 / ml. however there is so much other crud that it may be hard to figure out what you are looking at. Possibly if you were to filter sea water through a small pore filter (such as 0.22micron filter) then you might have water highly enriched for virus particles. But you won't really know what you are looking at unless you are lucky enough to see something that looks like a typical tailed virus.
  • asked a question related to Virus
Question
2 answers
I focus on animal taxonomy, where everyone uses ancestral range estimation method to reconstruct species migration in the speciation process. I've operated BEAST many times, and have helped some researchers do their Bayesian stochastic search variable selection (BSSVS) analysis for virus spread. I think this method is wonderful that it can even estimate a rough migration trace. Though, I haven't seen any of my realm's studies used it instead of ancestral range eatimation and I want to know the reason. Doesn't this model have a good accuracy across species, or on a MYA-scaled timeline?
Relevant answer
Answer
I think many interesting tools developed by people in phylodynamics are largely underutilized in other branches of evolutionary biology. Indeed, many models might be useful in analyzing processes at macroevolutionary scales. Still, these tools were designed to study viral evolution: viral phylodynamic data typically contain high temporal and spatial information (well, we can track the process itself in a real-time process in many cases), enabling the use of parameter-rich models like complex diffusion models. Unfortunately for the long-term macroevolutionary processes, we often lack such informativeness, and thus the data might not have enough power for parameter inference, leading to nonsensical results.
However, I do think that these approaches might be effective, at least for some types of clades, if their dispersal dynamics are not so complicated and species are well sampled, although further simulation study and empirical validation might be needed to completely justify their uses.
I am not an expert on this topic and my suggestions might not be optimal but I think these studies might be useful to start your investigation on applying diffusion-type models in biogeography at a macroevolutionary scale:
I hope this helps.
  • asked a question related to Virus
Question
4 answers
Dear experts it may concren,
We want to use stereotaxic injunction technique give AAV for up-regulating a protein in whole cerebella in P0 mice. Firstly, we have to try different injection locations and volumes for whole cerebella being infected. Normally, we should wait 4w to see the virus expression. Do you know someways to visualize AAV spreading area in 12h or 24h for saving time and reducing unnecessary harm to more mice ?
Thank you!
Relevant answer
Answer
Thank you, again. Winda Ariyani
  • asked a question related to Virus
Question
2 answers
Hello,
I just want to know your thoughts of using the terms "capsid particles" and viral particles. I feel most people use these two terms interchangeably.
They are not the same thing right? since the viral particles can contain genome while also having capsid.
Thoughts?
Relevant answer
Answer
It will depend if you deal with an enveloped or naked virus. for naked virus (no lipid no membrane) the capsid is the protein structure that forms the virus. it can be filled with the genome or empty (in that case it is sometime called CLP or VLP (capsid/Viral like particle; mainly if it has been produced with recombinant proteins). For envelopped viruses the capsid is the core of the virus (made of proteins that surround the genome) without the membrane part.
  • asked a question related to Virus
Question
1 answer
1. Is there any significant correlation between Chinese university students’ Corona Virus Anxiety and generalized anxiety disorder?
2. Is there any significant correlation between Chinese university students’ Corona Virus Anxiety and depression, anxiety, and stress?
3. Is there any significant correlation between Chinese university students’ Corona Virus and Grade Point Average (GPA)?
4. What are the negative educational and psychological consequences from the Chinese students’ perspectives?
Relevant answer
Frontiers in Psychology reports in 2020 examined the impact of COVID-19 on anxiety in Chinese university students. The study found that the mean anxiety score was significantly higher than the national norm, and there were notable differences in anxiety levels between male and female students2.
These studies suggest that COVID-19 anxiety can have a substantial impact on the mental health and academic performance of Chinese university students, potentially contributing to generalized anxiety disorder.
  • asked a question related to Virus
Question
3 answers
the genome of the virus is around 7 kb. what are the things I should pay attention to when taking measurements in the spectrophotometer and is there a calculation after the measurement like bacteria ?
Relevant answer
Answer
Yasemin Aş You can use 600 nm measurement.
see: Qing Zhou, Yi-Peng Qi, Feng Yang, Application of spectrophotometry to evaluate the concentration of purified White Spot Syndrome Virus, Journal of Virological Methods, Volume 146, Issues 1–2, 2007, Pages 288-292,
  • asked a question related to Virus
Question
1 answer
Hello,
I recently did a plaque assay using MHV A59. L929 cells were exposed to the virus for 1 hour, with slight plate tilting every 15 minutes. I added 1% agarose directly to the diluted virus and swirled the plates to ensure that the virus and agarose mixed well. 48 Hours later, my plaques look white. I am wondering if this would be considered contamination. Any tips and assistance are greatly appreciated.
Relevant answer
Answer
Did you include a control of cells where you added agar without the virus? That would be the simple way to know if you have contamination or if the plaques happen to look white.
  • asked a question related to Virus
Question
3 answers
Hi,
I'm planning to start standardization of absolute quantification of Influenza virus in our lab.
For standard curve, I'm planning to use the Influenza virus stock and do a 10-fold serial dilutions, extract and run qPCR in duplicates/triplicates.
My question is: we usually express the qPCR results as viral RNA copies/mL. Since I'm using virus stock which is in TCID/mL, how can I do the conversion or calculation to get the results of unknown samples in copies/mL? Or what can be done to do the conversion.
Any lead would be really helpful.
Thank you in advance.
Relevant answer
Answer
Not preferable because of the impurities of the media which give false results
  • asked a question related to Virus
Question
1 answer
Hi everyone,
I'm part of a newly established virology lab working with Cytomegalovirus (HCMV). We're currently facing issues recovering HCMV from the BJ-5ta cell line. We're confident the virus infects the cells, as they become green due to the GFP-tagged virus, but when it comes to recovering and titering the virus, we consistently get negative results.
Has anyone else experienced a similar problem or have any suggestions?
Relevant answer
Answer
Yes, you have bacterial infection
  • asked a question related to Virus
Question
1 answer
Hello, I am currently establishing a virus infection model in my lab. The cell I use is a vero cell and the virus is porcine epidemic diarrhea virus (PDEV). I plan to use the virus stock without concentration titration to make the new virus stocks, and I will conduct TCID50 analysis of the virus stocks in the future.
First, 0.5 ug/ml to 2 ug/ml TPCK-trypsin test was conducted in 96 well for the condition of virus infection culture (including 0.3% BSA). The virus infection culture was treated after washing twice with plain DMEM using 80-90% vero cells (Figure 1). Then, the appropriate concentrations (0.5 ug/ml, 0.75 ug/ml, 1 ug/ml) were selected and PEDV infection was performed.
Similar to the above process, after washing twice with plain DMEM, the virus stock of unknown concentration was diluted by virus infection culture (by TPCK-trpysin concentration) at 1:10. Finally 5 ml was dispensed into 100 dishes for 1 hour incubation. Next, I washed it twice with plain DMEM, added 10ml of the virus-infected culture medium, and incubated it for 3 days (Figure 2).
What I am curious about is the concentration (0.5 ug/ml or 0.75 ug/ml?) of TPCK-trypsin to establish a virus infection model and the timepoint of harvesting cell supernatant for virus stock manufacturing. What state should the vero cell be in to manufacture the virus stock? Please advise us to establish a virus infection model. Thank you!
Relevant answer
Answer
Vero cells either primary or continuous cell line prefered for porcine virus diarrhea because minimize mixed infection transmission and protective titer
  • asked a question related to Virus
Question
1 answer
resistant to pvy
Relevant answer
Answer
Yolanda Mutoti Many commercial potato varieties have a high tolerance to PVY, and despite some accumulation of the virus, they don't show any significant yield reduction. PVY can adapt to potato varieties with high (monogenic) resistance.
  • asked a question related to Virus
Question
4 answers
Dear all, I am studying A GENE influence viral replication and translation OR NOT. After knocking out THAT GENE by CRISPR, I infected the cells (both WT and KO) with virus at MOI of 0.02 and 0.2 . 3 days later, I collected the viral supernatant for TCID50 and extracted total RNA of supernatant and cell pellet (Mixture) by TRIzol. TCID50 showed that virus titer increased in KO groups, however, qPCR showed that virus copies decreased in KO groups. I am so confused which result was convincing, thank you all for kindly help.
Relevant answer
Answer
Didier Poncet Thanks again for your kindly help, and I have learned a lot from it! So i would take TCDI50 as consideration, for it represents infectious particles and shows the viral infectivity.
  • asked a question related to Virus
Question
3 answers
Hello everyone.
Various incidence rates are used in mathematical epidemiological models. Representing the rate of spread of the virus. Which incidence rate would be more meaningful to choose for which virus?
Or do you pay attention to choosing the incidence rate according to the virus when creating your model?
M.G.
Relevant answer
Answer
For deterministic model especially in epidemiology, firstly start with transmission mechanism and categories/groups in biology context. If is a physical phenomenon deal with the specific matter e.g ecology before presenting in variables for mathematical equations
  • asked a question related to Virus
Question
4 answers
Recombination within potyvirus species is well documented (OLIVEIRA, Alexandre Moisés Ericsson. Distinct recombination patterns in genomes of potyviruses. 2020, 115 f. Tese (Doutorado em Agronomia) – Universidade Federal de Uberlândia, Uberlândia, 2020. http://doi.org/10.14393/ufu.te.2020.253.). Is there any importance of interspecies recombination for Potato Virus Y evolution?
Relevant answer
Answer
Yes ....please see recent book about plant virology
  • asked a question related to Virus
Question
2 answers
I've been having difficulties inducing a proper amount of lung nodules in a KRAS-driven (KrasLSL-G12D) conditional mouse lung cancer model following this protocol:
Conditional mouse lung cancer models using adenoviral or lentiviral delivery of Cre recombinase
This is the Nature Protocol paper from Tyler Jacks lab that I have been using as a reference
Reagents
  • MEM (Sigma catalog #M-0268)
  • 2 M CaCl2
  • Adenovirus - University of Iowa (VVC-U of Iowa-5 Ad5CMVCre)*
Add 2.5 uL of Ad5CMVCre to 121.9 uL of MEM and mix well.
Add 0.6 uL of CaCl2 and mix well.
Let this mixture sit for ~20 minutes before use.
I have been using 2.5 x10E7 pfu for my experiments. Here are my questions:
1. When making the virus prep, is it a homogeneous solution after calcium phosphate precipitate formation? I wonder if one needs to flick the tube or pipette to mix it well after sitting on ice for 20 minutes and before giving it to the mice.
2. Can I make a "master mix" virus prep for all mice dosed on the same day? Or should I prepare one tube per mouse?
3. Is there a specific reason one must use 2M CaCl2 when making virus prep? Because sometimes 0.6 uL could be hard to pipette.
Relevant answer
Answer
Homogenization of the lesions with buffer solution and culturation on suspension growth medium the freezing and thawing ....several passages
  • asked a question related to Virus
Question
3 answers
Does bacteria, a virus, both or neither, cause aging? How? Why? Which?
Mendoza-Núñez, Víctor Manuel, and Ana Belén Mendoza-Soto. “Is Aging a Disease? A Critical Review Within the Framework of Ageism.” Cureus vol. 16,2 e54834. 24 Feb. 2024, doi:10.7759/cureus.54834. “However, although the proposal is qualified with said change, the codes XT9T (Ageing-related) and MG2A (Ageing-associated decline in intrinsic capacity) are maintained in the recently published ICD-11 [10], for which reason the WHO currently considers aging as a disease.”
Relevant answer
Answer
Interesting. Prof Stephen I. Ternyik gave a very good response. It seems to me when looking for references, the many did not really answer your question.
I would tend to feel that they do.
This article might bt relevant:
Harmful viruses and even friendly bacteria may cause premature ageing
  • asked a question related to Virus
Question
1 answer
I need to perform DNA extraction from amniotic fluid, but I do not have a specific kit for this purpose. I would like to know if anyone has used the QIAamp MinElute Virus Spin kit for extracting human DNA.
Relevant answer
Answer
Qiagen kits are suitable for almost all body fluids. it won't disappoint you..
  • asked a question related to Virus
Question
4 answers
I have infected Thp1 cells with lentivirus harboring GFP and my desired exogenous gene with the same promoter. Considering that everything is ok with the design of construct I expect to get 100 percent cells expressing both GFP and my desired gene expression. But, I have a population of cells with GFP expression but no expression of my desired gene. However, there is around 50 percent poulation expressing both GFP and the exogenous gene. By considering that my colleauge was susccessfull in getting the whole population with expressing both genes, there shoud be sth wrong in my hand. I just made one time virus packaging and infected my cells twice with the same batch of virus. Do you have any idea what is the problem.
I have always kept my cells in optimum density.
Relevant answer
Answer
Thanks for your response. I know that the promoter is the same for both GFP and GOI. we don't have the T2A sequence in the construct and mRNAs are probably seperated (I didn't design the construct, I have to check it). Currently, I am just checking the expression of GOI as external receptors by HA staining, and based on the FACS data, I don't have cells just expressing the GOI without GFP. So, one issue could be the problem in transferring of the receptors to the cell surface. But overall, since my colleague could get almost 98 pecent of cells expressing both GFP and GOI with the same plasmid, I am expected to get the same result. However, another previous lab member with the same plasmid had reported the same result as mine.
I appreciate again your time and your guidance in this matter.
  • asked a question related to Virus
Question
7 answers
I sequenced two isolates of a virus and constructed a phylogenetic tree base on their partial sequence. Although both sequences are 100% identical, they are separated from each other by another NCBI sequence that has 99% identity to my sequences.
However, the number of sequences submitted in GenBank is limited (about four sequences) and when I constructed the tree based on a shorter sequence (but more sequences), this problem will be solved.
Is it possible the low number of sequences cause this issue? and which tree is more reliable? a tree with more sequences but shorter length or a tree with low number of isolates but longer sequence?
Relevant answer
Answer
This question refers to something from a long time ago, so I don't recall exactly what I did. However, I suppose the sequences were incorrect. I replaced them with the correct sequences, so you may want to check the accuracy of your sequences.
Another issue might be related to selecting an inappropriate outgroup. Make sure to choose one that is significantly distant from the other sequences.
  • asked a question related to Virus
Question
2 answers
I know that depends of the person, but i don't understand why.
I would thank you answers
Relevant answer
Answer
Thank you :)
  • asked a question related to Virus
Question
1 answer
I have virus (viral hemorrhagic septicemia virus) in suspension and the experiment will not involve cells. What level of TCID50 is preferred?
Relevant answer
Answer
It depends upon the level of details that you want. If you want to study the surface morphology then SEM can be good option.
If you are interested to know the internal structures TEM can be better.
But microgrid sample and operation for TEM can be quite challenging. If you are using for 1st time.
  • asked a question related to Virus
Question
5 answers
I have done 3 blind passages to concentrate the viral stock in the supernatant without adding TPCK Trypsin. After passage 1 my Ct value for INF A was 36.96. After P2 the value decreased to 33.36. However, I was expecting a much lower value in the mid-20s. I am freeze-thawing the flasks at -20 degrees to lyse the cells and release the virus into the medium. Is it the correct approach? and can I get a much higher titre without adding TPCK Trypsin?
Relevant answer
Answer
Dear Colleague,
I hope this message finds you well. Culturing Influenza A virus in MDCK cells typically involves the addition of TPCK trypsin to facilitate viral replication and ensure high viral titers. TPCK trypsin cleaves the hemagglutinin (HA) protein, enabling the virus to become infectious. However, you are inquiring about the possibility of obtaining high viral titers without the addition of TPCK trypsin. Below is a detailed and logical discussion on this topic.
Culturing Influenza A Virus Without TPCK Trypsin
Role of TPCK Trypsin
  1. Cleavage of Hemagglutinin (HA):TPCK trypsin cleaves the HA protein of the Influenza A virus, which is essential for viral entry into host cells and subsequent replication. Without this cleavage, the virus remains non-infectious, and the replication cycle is interrupted, leading to lower viral titers.
Alternative Approaches
  1. Trypsin-Free Culture Media:Using trypsin-free culture media generally results in significantly lower viral titers because the HA protein remains uncleaved. Some labs have experimented with alternative proteases or conditions that might support HA cleavage, but TPCK trypsin remains the most reliable and widely used method.
  2. Genetically Modified Virus:One potential approach is to use a genetically modified strain of Influenza A that can replicate without the need for HA cleavage by trypsin. However, such strains are not typically used for standard virology studies.
  3. Endogenous Proteases:Some cell lines express endogenous proteases that can partially cleave HA. However, MDCK cells typically require exogenous TPCK trypsin for optimal viral replication.
Experimental Considerations
  1. Optimization of Conditions:pH and Temperature: Ensure that the culture conditions (pH, temperature) are optimal for both MDCK cell growth and viral replication. Infection Dose: Use a higher multiplicity of infection (MOI) to compensate for the lower efficiency of viral spread without trypsin.
  2. Protease Activity Monitoring:Monitor the culture for signs of cytopathic effect (CPE) and viral replication using assays such as plaque assays, hemagglutination assays, or qPCR. Assess the presence of cleaved HA using Western blotting or similar protein analysis techniques.
  3. Alternative Proteases:Experiment with other proteases that might be able to substitute for TPCK trypsin. However, this requires careful validation to ensure they effectively cleave HA and do not adversely affect cell viability or viral integrity.
Practical Recommendations
While it is theoretically possible to culture Influenza A in MDCK cells without TPCK trypsin, achieving high viral titers under these conditions is challenging and often impractical. TPCK trypsin remains the gold standard for this purpose due to its reliability and effectiveness. If you decide to experiment with trypsin-free conditions, I recommend conducting parallel cultures with and without TPCK trypsin to directly compare the effects on viral titers.
Example Protocol with TPCK Trypsin
  1. Cell Preparation:Seed MDCK cells in culture flasks or plates and grow to 80-90% confluency.
  2. Virus Infection:Infect cells with Influenza A virus at the desired MOI. Allow the virus to adsorb for 1 hour at 37°C in a CO2 incubator.
  3. Media Change:Replace the inoculum with serum-free DMEM containing 1-2 µg/mL TPCK trypsin. Incubate the cells at 37°C in a CO2 incubator.
  4. Monitoring and Harvest:Monitor cells daily for CPE. Harvest the supernatant when CPE is observed (typically 2-4 days post-infection) and determine viral titers using a plaque assay or other appropriate method.
Conclusion
While culturing Influenza A virus in MDCK cells without TPCK trypsin is not impossible, it poses significant challenges and typically results in lower viral titers. TPCK trypsin facilitates efficient viral replication by cleaving the HA protein, making it a critical component for high-yield virus production. If you proceed with trypsin-free cultures, careful optimization and parallel comparison with TPCK trypsin-containing cultures are recommended.
With this protocol list, we might find more ways to solve this problem.
  • asked a question related to Virus
Question
4 answers
We do this for SARS-COV-2 detection from patients' swap samples. The virus is showing fluctuations in copy number in progress and there is no standard sampling, so we have trouble in deciding about the results of these samples with high Ct values.
Your suggestions would be of great help for us.
Relevant answer
Answer
Dear Colleague,
I hope this message finds you well. Distinguishing between primer-dimer formation and true amplification of low-copy number templates in qPCR with TaqMan probes is critical for accurate quantification, especially at high cycle threshold (Ct) values. Here are detailed and logical steps to help you achieve this distinction:
Understanding the Issue
  1. Primer-Dimers:Primer-dimers are non-specific products formed by the annealing of primer pairs to each other rather than the target sequence. They typically appear at late cycles and can contribute to false-positive signals, especially in low-template scenarios.
  2. True Amplification:True amplification reflects the specific amplification of the target sequence. High Ct values indicate low starting template amounts but should still show exponential amplification.
Strategies to Distinguish Primer-Dimers from True Amplification
  1. Melting Curve Analysis:Although TaqMan probes do not usually require a melting curve, running a melt curve analysis can help identify primer-dimers, which typically melt at lower temperatures than the specific product. Protocol: After the qPCR run, slowly increase the temperature and record fluorescence. Analyze the melting peaks to distinguish specific products from primer-dimers.
  2. Gel Electrophoresis:Run the qPCR products on an agarose gel to separate and visualize the amplified products. Protocol: After qPCR, load a portion of the reaction mix onto an agarose gel (2-3% agarose) and run electrophoresis. Specific products will appear at expected sizes, while primer-dimers usually appear as smaller, non-specific bands.
  3. Design Optimization:Primer Design: Ensure primers are designed to minimize the formation of primer-dimers. Use software tools to check for secondary structures and dimerization potential. Concentration: Optimize primer concentrations. Too high concentrations can increase the likelihood of dimer formation.
  4. Reaction Optimization:Annealing Temperature: Optimize the annealing temperature. Higher temperatures can reduce non-specific binding. MgCl2 Concentration: Adjust the MgCl2 concentration, as it affects primer binding and specificity. Enzyme Choice: Use a hot-start polymerase to reduce non-specific amplification during reaction setup.
  5. Ct Value Analysis:High Ct Values: True amplification of low-copy templates should show a clear exponential phase, albeit at high Ct values. Primer-dimers typically show a more linear amplification phase. Replicates: Run technical replicates. True amplification should be reproducible across replicates, while primer-dimer formation may not be consistent.
  6. Fluorescence Monitoring:Probe-Based Specificity: TaqMan probes increase specificity since the probe only fluoresces upon binding to the target sequence. Non-specific products like primer-dimers usually do not lead to probe cleavage and fluorescence. Baseline and Threshold Setting: Set the baseline and threshold levels correctly in your qPCR analysis software to distinguish true signals from background noise.
Example Protocol for Optimization
  1. Primer Design and Validation:Use primer design software to ensure specificity and minimize dimer formation. Validate primer efficiency using a standard curve with serial dilutions of known template concentrations.
  2. qPCR Setup: Reaction Mix:
  3. 代码- Template RNA: variable (as low as needed for sensitivity) - Forward Primer: 0.3 µM - Reverse Primer: 0.3 µM - TaqMan Probe: 0.2 µM - 2x qPCR Master Mix: as per manufacturer’s instructions - Nuclease-free water: to final volume (e.g., 20 µL) Cycling Conditions: 代码- Initial denaturation: 95°C for 10 minutes - Denaturation: 95°C for 15 seconds - Annealing/Extension: 60°C for 1 minute (adjust based on optimization) - Number of cycles: 40-45
  4. Post-PCR Analysis:Melting Curve: If using SYBR Green or equivalent, perform a melting curve analysis. Gel Electrophoresis: Run products on a gel to verify the size and specificity of the amplified products.
  5. Replicate Analysis:Perform qPCR in triplicates or more to ensure reproducibility of the results.
By carefully following these strategies and optimizing your qPCR conditions, you can effectively distinguish between primer-dimer artifacts and true low-copy target amplification, ensuring accurate and reliable results.
Should you have any further questions or require additional assistance, please feel free to reach out.
With this protocol list, we might find more ways to solve this problem.
  • asked a question related to Virus
Question
7 answers
Hi,
I'm transducing Ly1 and L363 cell lines using our standard protocol for retroviral transduction. The cells are successfully transduced as evidenced by GFP expression. However, after 4-5 days they start dying off and look really stressed. I'm suspecting polybrene since we've got a new batch. The cells look really weird, irregular and start forming clumps which they don't normally do in standard cell culture. I've tried using the same polybrene concentration (8ug/ml) in standard culture medium without the virus to check toxicity and it appears that it is decreased. Which concentrations do you normally use? Should I make a polybrene concentration curve to find the minimal nontoxic condition?
Relevant answer
Answer
Dear Colleague,
I hope this message finds you well. Polybrene (hexadimethrine bromide) is commonly used to enhance the efficiency of viral transduction, particularly with retroviruses and lentiviruses, by reducing the charge repulsion between the virus and the cell surface. However, polybrene toxicity can sometimes be an issue, manifesting as reduced cell viability or altered cell behavior. Here are some insights and troubleshooting tips to address polybrene toxicity during viral transduction:
Understanding Polybrene Toxicity
  1. Mechanism of Toxicity:Cell Membrane Interaction: Polybrene interacts with the cell membrane to facilitate viral entry. At higher concentrations, this interaction can disrupt membrane integrity, leading to cell toxicity. Cell Type Sensitivity: Different cell types have varying sensitivities to polybrene. Some cells tolerate higher concentrations, while others are more susceptible to its toxic effects.
Troubleshooting and Mitigating Polybrene Toxicity
  1. Optimizing Polybrene Concentration:Concentration Range: Polybrene is typically used at concentrations ranging from 2 to 10 µg/mL. Start with a lower concentration (e.g., 2 µg/mL) and gradually increase it while monitoring cell viability. Cell Type Specificity: Determine the optimal concentration for your specific cell type. Perform a titration assay to find the highest concentration that enhances transduction efficiency without causing significant toxicity.
  2. Shortening Exposure Time:Reduced Exposure: Limit the exposure time of cells to polybrene. Shorter incubation periods (e.g., 2-4 hours) can reduce toxicity while still enhancing transduction efficiency. Wash Steps: After the desired exposure period, wash the cells thoroughly to remove any residual polybrene, replacing it with fresh culture medium.
  3. Using Alternatives or Supplements:Poloxamer 407 (Pluronic F-68): This non-ionic surfactant can be used as an alternative to polybrene to enhance viral transduction with reduced toxicity. Serum Supplementation: Ensure that your culture medium contains serum (e.g., 10% fetal bovine serum), which can help mitigate the toxic effects of polybrene by providing protective factors.
  4. Monitoring and Validating:Cell Viability Assays: Use viability assays (e.g., MTT, CellTiter-Glo) to quantitatively assess the impact of polybrene on cell health. This helps in optimizing the concentration and exposure time. Transduction Efficiency: Evaluate the efficiency of viral transduction by measuring the expression of the reporter gene or transgene (e.g., GFP, luciferase) to ensure that reduced polybrene toxicity does not compromise transduction efficacy.
Example Protocol for Polybrene Optimization
  1. Cell Seeding: Seed your cells in a 24-well plate at a density that allows them to reach 70-80% confluency the next day.
  2. Polybrene Titration: Prepare a series of polybrene concentrations (e.g., 0, 2, 4, 6, 8, 10 µg/mL) in your viral supernatant or culture medium.
  3. Viral Transduction:Add the viral supernatant containing the different concentrations of polybrene to the cells. Incubate the cells for 2-4 hours at 37°C. After incubation, replace the medium with fresh culture medium without polybrene.
  4. Assessment:Cell Viability: Assess cell viability 24-48 hours post-transduction using a viability assay. Transduction Efficiency: Measure the expression of the transgene 48-72 hours post-transduction to determine the optimal polybrene concentration.
By carefully optimizing the polybrene concentration and exposure time, you can enhance viral transduction efficiency while minimizing toxicity to your cells.
Should you have any further questions or require additional assistance, please feel free to reach out.
Reviewing the protocols listed here may offer further guidance in addressing this issue
  • asked a question related to Virus
Question
2 answers
I have some Covid-19 virus data and aI want a perform a CAI analysis using human codon usage data at CODON-W software.
My question is can I do that? and is there any chance that the presence of an internal stop codon can affect my results? as it is the Whole viral genome.
Thank You in Advance.
Relevant answer
Answer
Epstein-Barr Virus (EBV), a member of the Herpesviridae family, is known for causing various conditions, including infectious mononucleosis and certain malignancies. EBV remains latent in the host after the initial infection and can be reactivated under specific conditions. Reactivation of EBV can lead to symptoms or contribute to disease, particularly in immunocompromised individuals.
Here are conditions and factors known to reactivate EBV:
**1. Immunosuppression
**1.1 Immunosuppressive Medications:
Steroids: Corticosteroids used for conditions like asthma, autoimmune diseases, or organ transplants can lead to EBV reactivation.
Chemotherapy: Agents used in cancer treatment can suppress the immune system, facilitating EBV reactivation.
Immunosuppressive Drugs: Medications used to prevent organ rejection (e.g., calcineurin inhibitors, mTOR inhibitors) can also trigger reactivation.
**1.2 HIV/AIDS:
Immunodeficiency: Advanced HIV/AIDS can weaken the immune system, leading to opportunistic infections and reactivation of latent EBV.
**2. Stress
**2.1 Physical Stress:
Illness or Injury: Severe physical stress, including surgery or chronic illness, can contribute to EBV reactivation.
Extreme Fatigue: Prolonged physical exhaustion or stress can impact immune function and potentially lead to reactivation.
**2.2 Psychological Stress:
Emotional Stress: High levels of psychological or emotional stress have been associated with reactivation of latent viruses, including EBV.
**3. Co-Infections
**3.1 Other Viral Infections:
Cytomegalovirus (CMV): Co-infection with CMV or other viruses can influence EBV reactivation.
Influenza or Other Respiratory Viruses: Acute viral infections can sometimes trigger EBV reactivation.
**4. Immunological Factors
**4.1 Autoimmune Diseases:
Systemic Lupus Erythematosus (SLE) and Rheumatoid Arthritis: Autoimmune conditions can affect immune regulation and lead to EBV reactivation.
**4.2 Immune System Modulation:
Immunotherapy: Treatments aimed at modulating the immune response, such as checkpoint inhibitors, may affect EBV latency and reactivation.
**5. Hormonal Changes
**5.1 Pregnancy:
Hormonal Fluctuations: Changes in hormone levels during pregnancy can influence immune function and potentially trigger EBV reactivation.
**6. Other Factors
**6.1 Aging:
Immune Senescence: As people age, changes in immune function can impact the control of latent EBV and lead to reactivation.
**6.2 Chronic Conditions:
Diabetes or Chronic Kidney Disease: Chronic conditions that impact overall health and immune function may contribute to EBV reactivation.
Summary
EBV reactivation can be triggered by a variety of factors, including immunosuppression, stress, co-infections, immune system disorders, hormonal changes, and other health conditions. Monitoring and managing these factors, particularly in individuals who are at higher risk, can help mitigate the risk of EBV reactivation and associated complications.
l With this protocol list, we might find more ways to solve this problem.
  • asked a question related to Virus
Question
8 answers
I would like to monitor cell viability during an experiment (in a quantitative way if possible) using the very same cells I'm using for the experiment. I'd be using poly IC and viruses, and would like to make sure that the potential effects are not due to cells dying, but they are actual responses by living -but stressed- cells. One option is to use a Caspase assay on a parallel set of cells, but the ideal would be to use the same cells. What are the best, most used methods to test this?
Relevant answer
Answer
For in vivo cell viability assays in virus work, several methods are commonly used to assess the health and viability of infected or treated cells. The choice of assay depends on the specifics of the experiment, such as the type of virus, the cell type, and the objectives of the study. Here are some of the best in vivo cell viability assays for virus research:
**1. MTS Assay (CellTiter 96 AQueous One Solution Cell Proliferation Assay)
**1.1 Description:
Colorimetric Assay: Measures cell viability by converting a tetrazolium compound (MTS) into a formazan product, which is soluble in the cell culture medium.
**1.2 Applications:
Cell Proliferation: Useful for assessing cell proliferation and viability in response to viral infection or treatment.
Advantages: Sensitive, easy to perform, and provides quantitative data.
**1.3 Considerations:
Solubility: The formazan product is soluble, so it’s suitable for assays where cells are not lysed.
**2. MTT Assay
**2.1 Description:
Colorimetric Assay: Measures cell viability based on the reduction of MTT dye to purple formazan crystals by viable cells.
**2.2 Applications:
Viability and Proliferation: Commonly used for evaluating cell viability and proliferation in various experimental conditions.
**2.3 Considerations:
Cell Lysis: Requires solubilization of the formazan crystals, which involves additional steps.
**3. Trypan Blue Exclusion Assay
**3.1 Description:
Dye Exclusion: Viable cells exclude the dye, while non-viable cells take up the dye and appear blue.
**3.2 Applications:
Cell Counting: Useful for assessing cell viability and counting live and dead cells.
**3.3 Considerations:
Manual Counting: Requires manual counting under a microscope or using an automated cell counter.
**4. Annexin V/PI Staining
**4.1 Description:
Flow Cytometry: Uses Annexin V to detect early apoptotic cells and propidium iodide (PI) to identify late apoptotic or necrotic cells.
**4.2 Applications:
Apoptosis and Viability: Provides detailed information on cell death pathways and stages.
**4.3 Considerations:
Flow Cytometry: Requires flow cytometry for analysis, which may not be available in all labs.
**5. Lactate Dehydrogenase (LDH) Release Assay
**5.1 Description:
Cytotoxicity Assay: Measures the release of LDH, an enzyme released from damaged cells, into the culture medium.
**5.2 Applications:
Cytotoxicity: Useful for assessing cell membrane integrity and cytotoxic effects.
**5.3 Considerations:
Non-Specific: High LDH levels can indicate cell damage or death, but other factors can also influence LDH levels.
**6. XTT Assay
**6.1 Description:
Colorimetric Assay: Measures the conversion of XTT to a colored formazan product, similar to the MTS assay.
**6.2 Applications:
Cell Viability and Proliferation: Used for assessing cell viability in various experimental conditions.
**6.3 Considerations:
Solubility: The formazan product is soluble, so no cell lysis is needed.
**7. Live/Dead Cell Assays
**7.1 Description:
Fluorescence-Based: Uses fluorescent dyes to differentiate live and dead cells based on membrane permeability.
**7.2 Applications:
Real-Time Analysis: Allows for live imaging and assessment of cell viability in real-time.
**7.3 Considerations:
Fluorescence Microscopy: Requires fluorescence microscopy or flow cytometry for analysis.
**8. Cell Titer-Glo Assay
**8.1 Description:
Luminescence Assay: Measures ATP levels, which correlate with viable cell numbers.
**8.2 Applications:
Cell Viability: Provides a quantitative measure of cell viability based on ATP levels.
**8.3 Considerations:
Luminescence Detection: Requires a luminometer for measuring luminescence.
Summary
Each assay has its strengths and limitations, so the best choice will depend on your experimental needs. For general cell viability and proliferation studies, MTS, MTT, and XTT assays are widely used. For more specific apoptosis studies, Annexin V/PI staining is recommended. LDH assays are useful for assessing cytotoxicity, while live/dead cell assays are suitable for real-time analysis. Consider the specific requirements of your virus work and choose the assay that best meets your needs for sensitivity, accuracy, and ease of use.
Check out this protocol list; it might provide additional insights for resolving the issue.
  • asked a question related to Virus
Question
5 answers
I synthesised from Viral RNA to cDNA.
And next, I performed PCR. But in the gel, no band and only smearing.
I changed primer concentration, template concentration, annealing temperature, template.. but same results were obtained.
So, I want to check my cDNA condition.
Can I check if cDNA exists?
I'm so sorry about my english skills.
Relevant answer
Answer
it is generally not possible to see the cDNA (except if you use radioactive nucleotide precursors) ... did you do random priming? oligo dT priming ? or with specific primers ? do you have the PCR conditions OK from your cloned gene of interrest? (ie not to have to workout RT AND PCR conditions) for me RT with random primers is the more convenient as you can then add the primers you want after from the same RT reaction. To control your RT use an already validated primers couple with a small insert to amplify. then if it is OK with these primers work the PCR condition with your GOI primers..
  • asked a question related to Virus
Question
2 answers
Hi,
I have made multiple attempt to cultivate DENV2 and DENV3 virus from frozen stock using vero cells. ATCC recommended using LLC-MK2 cells, but since I don't have the cell line, I figured I could use vero cells since it is known to be infected by the virus. I had to get a new vial of the virus from ATCC after my first attempt didn't work.
Does anyone know how to go about this and how long DENV incubate for upon the first cultivation from frozen stock
Relevant answer
Answer
Dengue is a slow-growing virus in cell culture. It usually takes 3 days to get resealable titers, so small culture volumes are needed to achieve a higher virus concentration. We grow DENV on Vero and Huh-7 cells, where we can get high viral titers. However, after 2 days of infection, the titers are really low.
Best,
Jochen
  • asked a question related to Virus
Question
1 answer
I get two different bands from BCMV after PCR
Relevant answer
Answer
The PCR reaction may need to be optimized. Additionally, contamination may be occurred. I suggest you have it (both two bands) sequenced and checked.
  • asked a question related to Virus
Question
2 answers
Hey everyone,
I've been working with GRIN lenses (Inscopix) for almost 1 year and still had no luck seeing strong fluorescence on them. When I look at them, I can see good blood flow and, sometimes, some fluorescence, but never enough to get significant conclusions.
I've tried different virus concentrations (currently we're using 1:6), intervals between virus injection and lens implant (2-4 weeks), checking the calcium signal after 4-6 weeks, and still, I can't see a nice fluorescence.
It's important to highlight that when I perfuse the animals to check the virus expression, it is not as high as other regions/mice strains (since we depend on the D1 expression in the mPFC) but I'm still able to mark and visualize the GFP neurons in the confocal.
I've also injected in the striatum since it's another region that is involved in the type of behavior that I study, but I haven't done this immuno yet (probably in the following days).
In light of everything mentioned, does anyone have any suggestions to improve the fluorescence? Maybe some tips for the surgeries, intervals, virus, regions, concentrations etc. Every suggestion is more than welcome.
I'm attaching an example of one animal that I checked the fluorescence.
Let me know if you need further information and thank you very much for the help.
P.S. Relevant information:
- Target region: medial prefrontal cortex (mPFC) - DV -2.3mm
- Lens: Proview Integrated lens 0.5x4.0mm Inscopix
- Mice strain: D1-cre
- Virus: pAAV.Syn.GCaMP6f.WPRE.SV40 (Plasmid #100837 - Addgene)
- System: nVista Inscopix.
Relevant answer
Hey Anat, thanks for the quick reply. Please find below my comments:
1. Yes, I always clean the surface with cold saline during the procedure and avoid blood. I only start the lens implant when there is no blood around.
2. Thanks for the reminder, I'll try harder to improve the placement to avoid the boundaries.
3. I'm not familiar with this strain. I'll look for more information (although I don't think it's feasible at the moment).
4. We contacted the Inscopix support before choosing the best dimension and the 0.5mm was their suggestion (since we also had the 1.0mm), but I'll consider the 0.6mm as well.
5. Yeah, here we use a needle to track the path before the implant and we do it very slowly and gently.
Once again, thank you for the help.
Regards.
  • asked a question related to Virus
Question
2 answers
I am working on two viruses, when I do PCR for them separately they both showed on gel with their respective band sizes but when I multiplex them only one virus show different sizes and the other did not show at all.
Relevant answer
Answer
You have more template dna and primer in the multiplex compared with the single pcr. It may be that there is a pcr inhibitor in the dna which affects one pcr more than the other. Inhibitors often work by removing Mg from the pcr mix. I would try increasing the Mg concentration to 2mM or even 2.5mM and run dilutions of the mixed templates . 2 or 3 dilutions maybe 1:2, 1:4 and 1:8 of the dna sample. I am assuming that there is not a strong primer dimer band which may be removing one or both primers from the failing pcr set
  • asked a question related to Virus
Question
3 answers
We have a couple of primers but when we make a blast for those primers we see two results: When I use Blast from NCBI, I found many lineages which anniling with many lineages. But when I want to see in which part of the genome has an anniling, it was not possible to find the specific sequence.
Those are the primers:
F: TCAAGGAACTCCACACATGAGATGTACT
R: TGTATGCTGATGACACAGCAGGATGGGACAC
Thanks for your help.
Relevant answer
Answer
Exact results depends on original spcimens how ist pure you got exact results
  • asked a question related to Virus
Question
3 answers
Infected potato plant was negative for PVY, PVX, PVM, PVS. What can it be?
Relevant answer
Answer
Some more pics of these infected potato plants
  • asked a question related to Virus
Question
2 answers
Hello everyone,
I am extremely confused about analysis of qPCR data after virus infection or stimulations, and I need help. I hope I can explain properly.
Let's say we have two different cell lines.
1. HFF WT and
2. HFF protein A knockout.
We infect (or stimulate) these cells with virus and then do IFNB1 and GAPDH qPCR.
So, we will have these conditions:
  • Mock HFF WT
  • Virus-infected HFF WT
  • Mock HFF protein A KO
  • Virus-infected protein A KO
Let me try to explain what I understand how people analyse this kind of data:
First, you get the Cq values of IFNB1 and GAPDH.
Then, you do (IFNB1 - GAPDH)
Using (IFNB1-GAPDH) values, then you do (Virus infected HFF WT- Mock HFF WT) and (Virus infected HFF protein A KO - Mock HFF protein A KO).
Then you are doing POWER(2; -(Virus infected HFF WT- Mock HFF WT))
or POWER(2; -(Virus infected HFF protein A KO - Mock HFF protein A KO).
My problem is:
Since the mock cells are not stimulated, they usually give either negative or very high Cq value of IFNB1. (for example: 36, 38, 39). And this value is totally randomly changes experiment to experiment. Sometimes Mock HFF WT is 36, and the Mock HFF protein A KO negative. Sometimes vice versa. These also changes between biological replicates all the time. (I usually write 40 when it is negative, which is my cycle number).
In this case, can I just consider all the mock cells will have negative value and consider their Cq is 40 (eventhough I get 35-39). Of course they can have some IFNB1 even they are not stimulated/infected and this can change between these two cell lines, but having different mock results changes the whole outcome.
Note:
My IFNB1 Cq values are usually 20-26 in infected cells.
My GAPDH Cq values are usually 20-25.
I use GoTaq.
I convert total of 400 ng of RNA to cDNA (in 20 uL).
Then I take 1 uL for qPCR, so 20 ng. Can increasing the cDNA amount help me to get positive values from Mocks and equalize them?
Thank you very much.
Relevant answer
Answer
Can Justin Kiessling Thank you very much!
  • asked a question related to Virus
Question
1 answer
Hello everyone,
I'm currently working on a project where I need to isolate viruses in culture from incubation with PCR-positive serum samples, but I'm facing the challenge of dissociating antibodies bound to the virus. Does anyone have a reliable protocol or references that could share for pre-treatment of infectious sample for isolation purposes?
Any guidance or suggestions would be greatly appreciated! Thank you in advance for your help.
Relevant answer
Answer
Dear Aldo
You can probably do this by lowering the pH using an appropriate buffer over a short period of time, or by a competitive approach via adding recombinant proteins similar to the surface proteins of the virus.
  • asked a question related to Virus
Question
1 answer
we plan to detect a cell receptor (protein) that a certain type of virus with a certain serotype binds to it.
we will use Chicken embryo liver cells (CEL) from SPF chickens and make three replicates (three flasks) and one control (one flask). Next, we will infect the treatment group (3 flasks) with the virus and leave the control group uninfected once all flasks are confluent. then, we will store the infected flasks and proceed with the identification of the protein by other downstream tests (IP, SDS-PAGE and mass spectrometry). I wanted to know if:
1. using 3 replicates and one control would be enough
2. is the use of the below hypothesis correct or we do not need to use the hypothesis?
3. what statistical test should be used (in case if hypothesis is necessary) to reject/accept the null hypothesis?
thank you for your assistance,
our null hypothesis is as follows:
Membrane proteins of hepatocytes in epithelial islands of CEL cells are not the target site for virus X fiber knob protein attachment.
Relevant answer
Answer
You may check here www.Stats4Edu.com
  • asked a question related to Virus
Question
3 answers
Hello all. I grew some cells for virus infection. I infected the cells with CMV (Cytomegalovirus) 8 days ago. The cells have not started lysis yet, but the plate is about to get dry. I wonder if I can add 5 mL of growth medium (DMEM) to the plate containing CMV-infected cells to prevent dryness at this moment (8 days after infection). Is there any risk to do so? Any advice is appreciated.
Relevant answer
Answer
Yes
  • asked a question related to Virus
Question
3 answers
Hello all. I grew ARPE-19 cells in cell culture and infected them with a virus (Varicella-zoster virus). After the virus reached a high infection rate, I harvested everything in the plate and use freeze & thaw technique to release viruses into the supernatant. Now I want to store my viruses in -80 freezer. What is the composition of freezing medium for VZV? Are DMSO and FBS enough? Or do I need to add sucrose or something else? Any advice is appreciated.
Relevant answer
Answer
Use 0.2M sucrose phosphate as a cryoprotectant for varicella-zoster virus.
You may want to refer to the article attached below.
However, virus infected cells may be frozen in 70% culture media + 20% FBS + 10% DMSO.
Best.
  • asked a question related to Virus
Question
5 answers
I want to estimate the half-life value for the virus as a function of strain and concentration, and as a continuous function of temperature.
Could anybody tell me, how to calculate the half-life value in R programming?
I have attached a CSV file of the data
Relevant answer
Answer
Estimating the half-life of a virus involves understanding its stability and decay rate under specific environmental or biological conditions. This is a crucial parameter in virology, impacting everything from the design of disinfection protocols to the assessment of viral persistence in the environment or within a host. Here's a structured approach to estimating the half-life values for a virus:
  1. Defining Conditions:Environment: Specify the environmental conditions such as temperature, humidity, UV exposure, and presence of disinfectants, as these factors significantly affect viral stability. Biological: In biological systems, consider the impact of host factors such as immune response, tissue type, and presence of antiviral agents.
  2. Experimental Setup:Sampling: Begin by preparing a known concentration of the virus under controlled conditions. Time Points: Collect samples at predetermined time points that are appropriate based on preliminary data or literature values suggesting the expected rate of decay.
  3. Quantitative Assays:Plaque Assay: One of the most accurate methods for quantifying infectious virus particles. It measures the number of plaque-forming units (PFU) which reflect viable virus particles. PCR-Based Assays: These can measure viral RNA or DNA but do not distinguish between infectious and non-infectious particles. Adjustments or complementary assays might be required to correlate these results with infectivity. TCID50 (Tissue Culture Infective Dose): This assay determines the dilution of virus required to infect 50% of cultured cells, providing another measure of infectious virus titer.
  4. Data Analysis:Plot Decay Curves: Use logarithmic plots of the viral titer (e.g., PFU/mL or TCID50/mL) against time. The decay of viral concentration should ideally follow first-order kinetics in the absence of complicating factors. Calculate Half-Life: The half-life (t1/2) can be calculated using the equation derived from the slope (k) of the linear portion of the decay curve on a logarithmic scale:�1/2=ln⁡(2)�t1/2​=kln(2)​Statistical Analysis: Ensure statistical methods are used to analyze the data, providing estimates of variance and confidence intervals for the half-life.
  5. Validation and Replication:Replicate Studies: Conduct multiple independent experiments to validate the half-life estimation. Variability in viral preparations and experimental conditions can affect the reproducibility of results. Peer Review: Consider external validation or peer review of the methodology and findings to ensure robustness and accuracy.
  6. Interpretation and Application:Contextual Interpretation: Understand that the estimated half-life is context-specific. Results obtained under laboratory conditions may differ significantly from those in natural or clinical settings. Application in Risk Assessment: Use the half-life data to inform risk assessments, disinfection strategies, or predictive modeling of viral spread and persistence.
By meticulously following these steps and ensuring the precision of each phase of the process, one can accurately estimate the half-life of a virus under specific conditions. This information is essential for developing effective control strategies and understanding the dynamics of viral infections.
Perhaps this protocol list can give us more information to help solve the problem.
  • asked a question related to Virus
Question
7 answers
A number of definitions are found in publications ranging from 6 days to 14 days. Most frequent definitions mention 7 and 14 days. Some use onset of symptoms as staring point. Others use first detection of virus material as starting point. The end of shedding is usually defined by the last detection of viral material followed by negative sampling results. Usual duration of shedding (about 6 days) should probably be considered when the definition is chosen. Any suggestions?
Relevant answer
Answer
Prolonged viral shedding refers to the continued presence of influenza virus in respiratory secretions beyond the typical duration observed in immunocompetent adults. In immunocompetent individuals, influenza viral shedding usually lasts for about 5 to 7 days after symptom onset but may persist for longer periods in some cases. Prolonged shedding in immunocompetent adults may be defined as shedding persisting beyond 10 days after symptom onset.
In immunosuppressed individuals, such as those with HIV/AIDS or undergoing immunosuppressive therapy, viral shedding can be prolonged further due to compromised immune responses. In these cases, prolonged shedding may be defined as viral presence beyond 14 days after symptom onset. However, the exact definition of prolonged viral shedding can vary depending on the specific context, such as the severity of illness, the presence of complications, and individual patient factors.
  • asked a question related to Virus
Question
24 answers
  • asked a question related to Virus
Question
1 answer
This question was extracted from the information from the journal “Tenants of Specimen Management in Diagnostic Microbiology” written by Rajeshwar Reddy Kasarla and Laxmi Pathak. The journal has relayed that most samples in Diagnostic Microbiology used for bacteriological examination or virus isolation were incubated, refrigerated, or incubated. Once the sample is received to be processed, what should be done to bring the sample back to its optimal temperature?
Relevant answer
When transporting samples for bacteriological examination or virus isolation, it's crucial to maintain their integrity by preventing temperature fluctuations that could affect their viability and accuracy of analysis. Gradual warming at room temperature is a recommended method because it allows the sample to adjust slowly without experiencing sudden thermal shocks. This approach is especially important for delicate samples, as rapid changes in temperature can lead to cell lysis or denaturation of proteins, compromising the results of the examination. Using a water bath further enhances the control and gentleness of the warming process. Water baths are designed to provide a stable and uniform temperature environment, which is ideal for bringing samples back to their optimal temperature. By setting the water bath to the desired temperature and placing the sample container within it, you ensure that the sample warms evenly and without the risk of overheating. Continuous monitoring of the sample's temperature during the warming process is essential. It helps to prevent any unintended temperature fluctuations and ensures that the sample reaches the optimal temperature required for accurate bacteriological examination or virus isolation.
References:
Khandpur, R.S., (2019). Compendium of Biomedical Instrumentation, 2099–2101. https://doi.org/10.1002/9781119288190.ch398
  • asked a question related to Virus
Question
4 answers
Does anyone knows the pH acceptable range for virus transport medium (VTM) for Sars cov 2 samples? I supose that it depends if you are only testing by PCR or if you need viability for culture but does anyone has experience in this subject?
Found a studie that defends that in normal individuals with no history of reflux or eustachian tube dysfunction, the pH values range from 6.10 to 7.92 with an average pH of 7.03 (SD, 0.67) so i believe that VTM should be buffered around pH 7 (with a variation of plus or minus 1) but need to confirm that.
Thank you and be safe.
Relevant answer
Answer
For the effective transport of SARS-CoV-2 samples, the virus transport medium (VTM) plays a crucial role in preserving the viability and integrity of the virus until it can be processed in the laboratory. The pH of the VTM is a critical factor that must be carefully controlled to ensure the stability of SARS-CoV-2, as well as the safety and accuracy of subsequent diagnostic tests.
Optimal pH Range for VTM:
The acceptable pH range for virus transport mediums used for SARS-CoV-2 samples generally falls between 7.2 and 7.4. This slightly alkaline pH range is conducive to maintaining the structural integrity and infectivity of the virus particles during transport and storage, thereby ensuring that the samples remain representative of the in vivo state.
Rationale Behind the pH Range:
  1. Virus Stability: SARS-CoV-2, like many other enveloped viruses, has a lipid membrane that is sensitive to pH changes. A pH that is too acidic or too alkaline can destabilize this membrane, leading to the loss of viral infectivity and compromising the sample.
  2. Cell Preservation: Some VTMs are designed to preserve not only the virus but also the host cells present in the sample. Maintaining a physiological pH is crucial for preventing cellular degradation over the transport period.
  3. Enzymatic Activity: The preservation of enzymatic activity, which may be necessary for certain types of diagnostic tests, requires a pH close to physiological conditions. Deviations from this range can denature enzymes and affect the sample's suitability for analysis.
Monitoring and Adjusting pH:
  • Quality Control: Regular monitoring of the VTM pH is necessary, especially in large-scale production or when using newly prepared batches. pH indicators or strips can be used for quick checks, while precise measurements may require a pH meter.
  • Adjustment: If the pH of the VTM is found to be outside the acceptable range, it can be adjusted using dilute hydrochloric acid (HCl) to lower the pH or dilute sodium hydroxide (NaOH) to raise the pH. After adjustment, thorough mixing and re-measurement of the pH are essential to ensure uniformity throughout the medium.
Conclusion:
Maintaining an optimal pH range of 7.2 to 7.4 in the virus transport medium is essential for preserving the integrity and infectivity of SARS-CoV-2 samples during transport to the laboratory. This careful control of the pH ensures that the samples remain viable for diagnostic testing, thereby contributing to the accuracy and reliability of COVID-19 detection and research. Regular monitoring and adjustment of the pH, as part of the VTM quality control process, are critical practices for all handling and diagnostic facilities.
Perhaps this protocol list can give us more information to help solve the problem.
  • asked a question related to Virus
Question
5 answers
Waiting for your suggestions.
Thanks
Uchurappa M
Relevant answer
Answer
I hope this message finds you in the midst of exciting and fruitful research endeavors. Your question regarding the prevention of contamination during lentivirus infection processes is of paramount importance, as contamination can significantly impact the validity and reproducibility of experimental results. Below, I outline strategic measures and best practices designed to minimize the risk of contamination when working with lentiviruses.
Establishing a Sterile Workflow:
  1. Preparation and Planning:Prior to beginning your work, ensure that all materials, including culture media, reagents, and pipettes, are prepared and sterilized. Plan your workflow to minimize the time cultures are exposed to non-sterile environments.
  2. Use of Personal Protective Equipment (PPE):Always wear appropriate PPE, such as gloves, lab coats, and face masks, to protect both yourself and your cultures from cross-contamination. Change gloves frequently, especially after handling potentially contaminated items.
  3. Sterile Technique Practices:Employ aseptic techniques at all times when working in the biosafety cabinet (BSC). This includes flame sterilizing tools, such as forceps and scissors, and using sterile pipette tips and tubes. Avoid direct contact between the container openings or the pipette and non-sterile surfaces.
  4. Biosafety Cabinet (BSC) Use:Perform all manipulations involving open lentiviral containers within a certified BSC. The BSC should be properly maintained and certified annually to ensure it provides a sterile environment.
Managing Lentivirus Preparations:
  1. Quality of Lentivirus:Use high-quality, purified lentivirus preparations. Contaminants in viral stocks, including bacterial endotoxins, can impact cell health and experimental outcomes.
  2. Thawing and Handling:Thaw lentiviral aliquots quickly at 37°C and minimize freeze-thaw cycles by aliquoting stocks. Keep the virus on ice once thawed and immediately prior to use to maintain its stability and reduce the risk of degradation.
Cell Culture Practices:
  1. Regular Monitoring:Monitor cell cultures regularly for signs of contamination, including changes in medium clarity, pH, and unexpected cell behavior. Use microscopy to check for microbial contamination.
  2. Dedicated Media and Reagents:If possible, dedicate specific media and reagents to your lentiviral experiments to avoid cross-contamination between different cell culture projects.
  3. Proper Disinfection:Disinfect all work surfaces before and after procedures with an appropriate disinfectant. Additionally, properly dispose of all waste materials, including pipette tips and culture vessels, in biohazard containers.
Environmental Controls:
  1. Minimize Traffic and Limit Access:Work in an area with controlled access to minimize traffic and reduce the introduction of contaminants. Limit the number of individuals who have access to the cell culture area.
  2. Equipment and Facility Maintenance:Ensure that cell culture incubators, BSCs, and other equipment are regularly cleaned and maintained. This reduces the risk of equipment being a source of contamination.
Adhering to these guidelines will significantly reduce the likelihood of contamination during your experiments with lentivirus. It is the meticulous attention to detail and adherence to strict aseptic techniques that ultimately safeguards the integrity of your research.
Should you have further questions or require additional guidance, please do not hesitate to contact me. I am here to assist you in navigating the challenges of your research endeavors.
Best regards,
This list of protocols might help us better address the issue.
  • asked a question related to Virus
Question
2 answers
osmosis process occurs in bacterial and other cells.
But is it possible to virus?
Relevant answer
Answer
I hope this message finds you well. Your question regarding the possibility of osmosis in viruses is intriguing and delves into fundamental principles of biology and virology. To address this query, it's essential to first understand the concept of osmosis and the basic structure of viruses.
Understanding Osmosis:
Osmosis is a process by which solvent molecules move across a semipermeable membrane from a region of lower solute concentration to a region of higher solute concentration, aiming to equalize solute concentrations on both sides of the membrane. This phenomenon is crucial for the maintenance of cellular homeostasis in living organisms.
Viral Structure and Osmosis:
Viruses are complex entities that straddle the line between living and non-living matter. A typical virus is composed of genetic material (either DNA or RNA) encapsulated within a protein coat known as a capsid. Some viruses also possess an outer lipid envelope. However, viruses lack the cellular structures and metabolic machinery that are characteristic of living cells, such as cytoplasm, organelles, and cell membranes capable of regulating osmosis.
Considerations Regarding Viruses and Osmosis:
  1. Lack of Semipermeable Membrane: Unlike cells, viruses do not possess semipermeable membranes required for osmosis. The capsid of a virus does not function like a cellular membrane in regulating the movement of solutes and water.
  2. Absence of Metabolic Processes: Viruses are obligate intracellular parasites that require a host cell to replicate. They do not engage in metabolic processes, including osmoregulation, independently of a host.
  3. Osmotic Pressure and Viral Stability: While viruses themselves do not undergo osmosis, the osmotic pressure of the surrounding environment can affect viral stability and infectivity. For example, extreme osmotic conditions can lead to the disruption of the viral envelope or capsid in enveloped viruses.
Conclusion:
In summary, the process of osmosis, as traditionally defined in the context of cellular and physiological processes, does not apply directly to viruses due to their lack of semipermeable membranes and metabolic activity. However, environmental osmotic conditions can influence viral stability and infectivity, highlighting the importance of osmotic balance in virology research and applications.
I trust this explanation clarifies the relationship between osmosis and viruses. Should you have any further questions or wish to explore related topics, please feel free to reach out.
Best regards,
Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to Virus
Question
2 answers
Dear Researchers:
Could you please share some simple cures or prevention for COVID-19, Cold, Flu or Influenza, and possibly Other Viruses, and Cancers?
Updates on Oct. 10, 2023: First, many thanks to all contributors to this discussion. Here are some Natural Approaches found from surveying literature in medicine to Boost our Immune Systems against viruses such as COVID-19, Cold, Flu or Influenza infections and to avoid/minimize developing further inflammations in the lungs and hearts caused by some of those viruses:
Give it a try, please! Especially if you increase your Vitamin D level to a required level and consume Vitamin C sources, e.g., oranges, on a daily basis, you can check how rarely you would catch the virus. Or, even after catching the virus, the virus will likely develop very mild symptoms in your body.
1- Daily uptake of Vitamin D pills up to 100 IU per 1 kg weight is safe and very important, recommended by Afshar et al. (2000) and Dr. Hamid Sajjadi in an interview, to RAISE the Vitamin D level in our body to the POINT which is REQUIRED to BOOST our IMMUNE SYSTEMS against Viruses and Diseases including Cancers.
Vitamin D daily use needs to be adjusted based on our body weight.
Please read the following article by Afshar et al. (2000) about the importance of vitamin D and the required daily dose of it (Up to 100 IU per 1 kg weight) to boost our Immune Systems.
Please also read the following Review article by Jordan et al. (2022) about the importance of Vitamin D on the level of infection & disease progression for COVID-19. You may find in the article the importance of our Forgotten SUN.
Vitamin D is rarely available in food sources, except in fatty fish which needs to be eaten high enough to get the required amount of Vitamin D for a body.
Another good natural source is daily sunbathing with naked skin; however, in cloudy regions such as Europe, sunbathing doesn't work well.
Vitamin D helps to absorb Calcium in our intestines and thus, in order to avoid excessive absorption of Calcium by our body, it would be better to use Vitamin D pills with Calcium sources such as warmed-up milk and Magnesium sources such as bananas on a daily basis. Because magnesium competes with calcium in our intestines to get absorbed.
Here is a text from A Review article by Kulie et al. (2009) about some of the importance of Vitamin D on our health:
"Vitamin D is a fat-soluble vitamin that plays an important role in Bone Metabolism and seems to have some Anti-Inflammatory and Immune-Modulating properties. In addition, recent epidemiologic studies have observed relationships between low vitamin D levels and multiple disease states.
Low vitamin D levels are associated with increased overall and Cardiovascular mortality, Cancer incidence and mortality, and Autoimmune Diseases such as Multiple Sclerosis. Although it is well known that the combination of vitamin D and calcium is necessary to maintain Bone Density as people age, vitamin D may also be an independent risk factor for falls among the Elderly."
2- Having Good Nutrients including Protein sources, Minerals, and Other Vitamins, e.g., C, A, and E, sources from fresh fruits, vegetables, and nuts. For example, the good sources of fruits and vegetables for these vitamins could be a daily use of 1-2 Oranges for Vitamin C, Carrots for Vitamin A, and Almonds or Sunflower Seeds for Vitamin E.
As Vitamin C is a water-soluble vitamin, the excess of it will be excreted from the body, it needs to be consumed every day to provide everyday vitamin C requirements for the body, as it is the 2nd most important vitamin after Vitamin D to boost our Immune Systems against viruses and diseases.
And, Vitamin B family from grains, poultry, and meat sources.
3- After the infection by those viruses, gargling salty water to disinfect the throat to avoid further movement of the virus into the lungs as the virus may stay in the throat for a few days
4- Inhaling Steamed Fresh Leaves, if not available, the Oil, of Eucalyptus 4-5 times a day for several continuous days to kill the virus in the lungs.
Here is A Review article by Mieres-Castro et al. (2021) about the "Antiviral Activities of Eucalyptus Essential Oils: Their Effectiveness as Therapeutic Targets against Human Viruses"
Australian Aboriginals are very much using Eucalyptus to Treat Infections.
5- Having plenty of Warm Drinks to wash out the virus from our body and dilute the blood to avoid blood clotting.
6- Having enough sleep and daily activities/exercises
7- Kids are proven to have High Immunity Against COVID-19, likely due to having a high amount of Melatonin, the Sleep Hormone, in their blood. So, that is why kids sleep very much as you know.
Melatonin production in our body usually decreases with increasing age. Thus, we may use daily melatonin pills after the infection based on what physicians may prescribe for us.
Here is A Review article by Carrillo-Vico et al. (2013) about the Importance of Melatonin on the Functionality of Our Immune Systems:
8- Avoid Fear/Panic as it Substantially Deteriorates the Functionality of Immune Systems against viruses and diseases.
Here is an interview by Dr. Lauren Deville about How Fear Affects Our Immune System:
Relevant answer
Many thanks, Rohan RANJAN Waliya, for contributing to this discussion!
  • asked a question related to Virus
Question
1 answer
Hi, I've done a plaque reduction assay to analyze my possible plant activity, I used a HSV 2 virus at 6,5x10^3 as control, and I count (added file) the PFU... do I need to calculate something or I can make a graph with PFU results?
Relevant answer
Answer
That's not a great graph, because it uses a thick bar to show means and a detonator to show standard errors. It does not give any actual information about how much data you had or how they were distributed. The two strange values for group D are not evident. And the asterisk is a bit of a mystery.
What is your hypothesis? Then maybe someone can help with a recommendation for testing. Since your groups differ by dosage, it would make more sense to see if the observed values are a function of dose, rather than treating each group as a distinct treatment. This gives you a directional hypothesis, and a lot more statistial power.
  • asked a question related to Virus
Question
2 answers
Akin to the oral "polio vaccine technology", is it possible for a healthy human to build antibodies against a said "bacterium, an example being a Steptococcal infection?".
In my opinion, yes we can. Our bodies have the required armoury "an adept immune system and the relevant enzymes", to tackle bacterial infections.
Please elucidate in detail, how you think our bodies could fight against a bacterial infection.
The previous discussion had been answered so well by "Rizzi". Looking forward to an answer like that of "Rizzi'.
Relevant answer
Answer
Creating antibodies against a specific bacteria typically involves vaccination or immunization strategies. However, the specific technique used depends on various factors such as the characteristics of the bacteria, its virulence factors, and the desired immune response The techniques exist for creating antibodies against bacteria, including traditional vaccines, conjugate vaccines, recombinant DNA technology, and toxoid vaccines, the oral poliovirus vaccination technique is not applicable for bacterial infections due to fundamental differences in pathogenesis and immune response mechanisms between bacteria and viruses.
  • asked a question related to Virus
Question
1 answer
I have been having an issue with my plaque assay for a few weeks now, I am working with a soil sample I inoculate a certain amount of virus PFU/mL into the soil then use some recovery method to recover the virus sample and run a plaque assay using methyl cellulose as an overly but the problem I keep having was mold growing on the sample which prevents seen any place on the sample. I would greatly appreciate any technical advice on how to get rid of the mold from the sample as we don't want to autoclave the soil or use UV radiation I would prefer using any mold inhibitory substance I have used Anti-Anti which it did not work as it only inhibits bacteria and fungi so is not efficient for my work. Thank you.
Relevant answer
Answer
Different fungicidal compounds act only on certain groups of fungi. When I was doing bacterial isolation from soil samples I used nystatin + cycloheximide. Even then I would get the odd fungus that was resistant to both. Nipagin is also good for stopping fungal growth but it also inhibits bacteria as well. I advise checking your plates 24h after setting them up and using a scalpel to remove any fungi that are persisting.
  • asked a question related to Virus
Question
5 answers
I am after the sequence for the murine leukemia virus-derived MND promoter (myeloproliferative sarcoma virus enhancer, negative control region deleted, dl587rev primer-binding site substituted), MNDU3. Can someone help me out?
Thanks Karin
Relevant answer
Answer
Dear Esteemed Colleague,
Greetings. I hope this message finds you well and deeply engaged in your genetic research endeavors. Your inquiry regarding the complete sequence of the MNDU3 promoter is both important and specific, highlighting your pursuit of precision in your scientific exploration. The MNDU3 promoter, recognized for its strong transcriptional activity in a broad range of cell types, is indeed a tool of significant interest for gene therapy and molecular biology applications.
Accessing the MNDU3 Promoter Sequence
  1. Literature and Databases:To obtain the complete sequence of the MNDU3 promoter, I recommend consulting reputable scientific literature and genomic databases. Databases such as GenBank, EMBL, and the NCBI Nucleotide database are invaluable resources for genetic sequences. Utilizing specific search terms related to the MNDU3 promoter can yield relevant entries or publications where its sequence might be detailed.
  2. Collaboration and Scientific Community:Engaging with the scientific community through forums, research networks, and conferences can also be a fruitful approach. Researchers working with similar promoters or in related fields might have insights or unpublished data regarding the MNDU3 promoter sequence.
  3. Contacting Authors:If the MNDU3 promoter sequence is referenced in specific research articles, reaching out directly to the authors for more detailed information or unpublished sequences could provide the necessary data. Authors are often willing to share such details for collaborative and scholarly purposes.
Considerations and Best Practices
  • Verification: Upon obtaining the sequence, verify it through multiple sources when possible to ensure its accuracy and completeness. Discrepancies in sequences can arise due to mutations, alternative splicing, or transcription start sites.
  • Legal and Ethical Use: Be mindful of any intellectual property rights or usage restrictions associated with proprietary sequences. Obtaining sequences from public databases or through academic sharing is generally permissible for research purposes, but commercial applications may require licensing agreements.
  • Functional Validation: After incorporating the MNDU3 promoter into your experimental vectors, empirical validation of its activity in your system or cell lines of interest is crucial. Reporter assays can quantify promoter strength and specificity across different conditions.
Conclusion
While I am unable to provide the exact sequence of the MNDU3 promoter directly, the pathways suggested above should facilitate your access to this critical piece of genetic information. The pursuit of such specific and foundational elements of gene expression underscores the meticulous nature of your research.
Should you require further guidance in navigating genomic databases, or if you have additional inquiries related to promoter sequences or their applications, please do not hesitate to reach out. I am here to support your scientific journey and assist in the advancement of your research projects.
Warm regards.
Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to Virus
Question
4 answers
I want to design tagman probe and primers to detect a virus.  I had searched the complete DNA in NCBI. 
What should I do next?
What is the standard procedure to design PCR probe and primers in general?
Relevant answer
Answer
Dear Esteemed Colleague,
Greetings. I trust this message finds you well and progressing steadily in your research endeavors. Your inquiry about determining the orientation (forward or reverse) of a PCR product in sequence data is a matter of great significance in molecular biology, particularly in the context of cloning, sequencing, and subsequent analyses. Below, I outline a methodical approach to ascertain the orientation of your PCR product using sequence data, ensuring precision and clarity in your experimental outcomes.
1. Reference Sequence Alignment
  • Obtain a Reference: Begin by securing a reference sequence for the gene or genomic region from which your PCR product was amplified. This reference should be from a reliable database, such as the National Center for Biotechnology Information (NCBI).
  • Alignment Software: Utilize sequence alignment software or online tools such as BLAST (Basic Local Alignment Search Tool) available on the NCBI website. These platforms allow you to compare your PCR product sequence against the reference.
2. Preparing Your Sequence Data
  • Quality Check: Prior to alignment, ensure your sequence data is of high quality. Trim any sequences of low quality or primer sequences to avoid misalignment.
  • Format: Ensure your sequence data is in a format compatible with your chosen alignment tool. Common formats include FASTA and plain text.
3. Performing the Alignment
  • Upload Your Sequence: Input your PCR product sequence into the alignment tool, along with the reference sequence.
  • Analyze the Results: Once the alignment is complete, the tool will display how your sequence aligns with the reference. A direct alignment (with no need for reverse complementing your sequence) indicates that your PCR product is in the forward orientation relative to the reference. Conversely, if the tool suggests reverse complementing for optimal alignment, your PCR product is in the reverse orientation.
4. Verification
  • Use Primers as Markers: If your sequence data includes regions amplified by your forward and reverse primers, their position in the alignment can serve as an additional verification of orientation. The presence of the forward primer sequence at the start of the aligned sequence (and the reverse primer at the end, in the correct orientation) confirms a forward orientation.
  • Consult the Electropherogram: Reviewing the original electropherogram (if available) for the sequencing reaction can provide clues about the orientation based on known sequences or primer binding sites.
5. Considerations and Troubleshooting
  • Multiple Alignments: If your PCR product can align in both orientations, further analysis may be required to resolve the ambiguity. This can include designing additional primers for sequencing or performing restriction enzyme mapping.
  • Sequence Variants: Be mindful of sequence variants or mutations that might affect alignment. In such cases, aligning to a closely related reference or using multiple references might be necessary.
Determining the orientation of your PCR product with respect to the reference sequence is crucial for accurate downstream applications, such as cloning, mutation analysis, and gene expression studies. The approach detailed above provides a robust framework for achieving this with a high degree of confidence.
Should you encounter any challenges or require further clarification on this process, please do not hesitate to reach out. I am here to support your scientific journey and contribute to the advancement of your research.
Warm regards.
Take a look at this protocol list; it could assist in understanding and solving the problem.
  • asked a question related to Virus
Question
11 answers
A patient with desminopathy (mutation Thr341Pro DES in a heterozygous state) with the progression of the disease has a decrease in taste and smell, immunosuppression, and an increase in IgA in the blood.
Oddly enough, but all this is characteristic of infections, including viral ones. For example, it is known that if the hepatitis C virus is not treated, then death will occur in 20 years.
In the identified case of late onset desminopathy, muscle weakness manifests itself at the age of 30, and death occurs 20 years after the onset of the disease.
Could the desmin mutation in myofibrillar myopathy be caused by an infection?
Perhaps the infection contributes to the progression of desminopathy?
Relevant answer
Answer
Dear Esteemed Colleague,
Greetings. I trust this message finds you deeply engaged in your research and seeking answers to complex questions within the realm of genetics and molecular pathology. Your inquiry regarding the potential role of infection in causing desmin mutations in myofibrillar myopathy is both intriguing and indicative of a keen scientific mind exploring the multifaceted nature of genetic disorders.
To address your question with the precision and clarity it deserves, it is crucial to first understand the nature of myofibrillar myopathies and the role of desmin within this context. Myofibrillar myopathies are a group of neuromuscular disorders characterized by the progressive weakening of muscles and the disintegration of muscle fibers at a cellular level. Desmin, a type of intermediate filament protein, plays a pivotal role in maintaining the structural integrity and function of muscle cells. Mutations in the DES gene, which encodes the desmin protein, are directly linked to certain forms of myofibrillar myopathy.
The genesis of these mutations, particularly those affecting the desmin protein, is primarily genetic, resulting from inherited or de novo mutations in the DES gene. These mutations lead to the production of an abnormal desmin protein, which disrupts the normal architecture of muscle cells, leading to the symptoms associated with myofibrillar myopathy.
Addressing the specific question of whether an infection could cause desmin mutations, it is essential to differentiate between the origins of genetic mutations and factors that may exacerbate the phenotype of a genetic disorder. Genetic mutations, including those affecting the desmin gene, arise from alterations in the DNA sequence. These alterations can be inherited from parents, occur spontaneously during DNA replication, or be induced by certain environmental factors, such as exposure to specific chemicals or radiation. Infections, while capable of causing a wide array of health issues, do not directly induce genetic mutations in the DNA sequence of the genes like DES. However, it is conceivable that certain infections could exacerbate the clinical manifestations of myofibrillar myopathy in individuals already predisposed or carrying a desmin mutation, by stressing the muscular system or triggering inflammatory responses that may further compromise muscle function.
In conclusion, while infections can have significant impacts on overall health and may interact in complex ways with genetic disorders, the mutations in the DES gene that cause myofibrillar myopathy are not directly caused by infections. The mutations are genetic in origin, and the relationship between infections and the severity or progression of myofibrillar myopathy would be more accurately viewed through the lens of infection exacerbating pre-existing conditions rather than causing the genetic mutation itself.
I hope this elucidation addresses your inquiry comprehensively. Should you have further questions or require additional clarification, please feel free to reach out.
Warm regards.
This protocol list might provide further insights to address this issue.
  • asked a question related to Virus
Question
8 answers
Dear colleagues,
I defended my Ph.D. thesis in October 2016 and now I am looking for a postdoctoral position in microscopy (AFM, TEM, SEM) and biophysics of microorganisms (especially, viruses, I like them :)).
My CV is attached. If there is an open position in your lab, please, write me.
Best regards,
Denis  
Relevant answer
Answer
That sounds like an exciting field! Here are some steps you can take to find a postdoctoral position in microscopy and physics of microorganisms:
  1. Identify Research Groups: Look for research groups or labs that specialize in microscopy and physics of microorganisms. Search university websites, scientific journals, and research databases for relevant publications and projects.
  2. Networking: Attend scientific conferences, workshops, and seminars related to microscopy, microbiology, and physics. Network with researchers in the field and express your interest in potential postdoctoral opportunities. You can also reach out to professors or researchers whose work you admire to inquire about available positions.
  3. Online Resources: Explore online platforms and job boards dedicated to academic and research positions. Websites like Nature Careers, Science Careers, and ResearchGate often list postdoctoral positions in various scientific disciplines.
  4. Collaborations: Consider collaborating with researchers who are conducting interdisciplinary work at the intersection of microscopy and microbiology. Collaborative projects can provide valuable insights and connections within the scientific community.
  5. Tailored Applications: Customize your application materials, including your CV, cover letter, and research statement, to highlight your expertise in microscopy and physics of microorganisms. Emphasize relevant skills, research experience, and achievements that align with the requirements of the position.
  6. Funding Opportunities: Look for postdoctoral fellowship programs or research grants that support projects in your area of interest. Many funding agencies offer fellowships specifically for early-career researchers pursuing research in microscopy, microbiology, or physics.
  7. Stay Informed: Stay updated on the latest developments and advancements in microscopy techniques, microbiology, and physics research. Familiarize yourself with emerging trends and technologies that could enhance your research interests and expertise.
  8. Persistence and Patience: Finding the right postdoctoral position can take time and persistence. Be proactive in your search, maintain a positive attitude, and keep refining your skills and qualifications to increase your competitiveness as a candidate.
By following these steps and leveraging your expertise in microscopy and physics, you can increase your chances of securing a rewarding postdoctoral position in this exciting field of research.
l Perhaps this protocol list can give us more information to help solve the problem.
  • asked a question related to Virus
Question
11 answers
Various pandemic diseases have taught us various lessons from time to time, lastly, the spread of corona virus spread has shown how fickle human condition or survival is in face of sudden outbreak of dangerous diseases!
What are the human security implications of 'corona virus spread' around the world?
Relevant answer
Answer
Dear Esteemed Reader,
The spread of the coronavirus, specifically referring to the COVID-19 pandemic caused by the SARS-CoV-2 virus, has posed unprecedented challenges to human security around the globe. The impacts of this pandemic extend far beyond the immediate health crisis, affecting various dimensions of human security, including economic stability, access to essential services, social cohesion, and the functioning of governance systems. Below, we delve into the multifaceted ways in which the spread of the coronavirus threatens human security worldwide.
1. Health Security:The most direct impact of the coronavirus is on health security. The rapid spread of the virus has overwhelmed healthcare systems in many countries, leading to a shortage of medical supplies, hospital beds, and healthcare personnel. This situation has not only jeopardized the care for COVID-19 patients but also disrupted routine healthcare services, affecting the management of other diseases and health conditions.
2. Economic and Livelihood Security:The pandemic has triggered a global economic downturn, affecting livelihoods, increasing unemployment rates, and exacerbating poverty. Lockdown measures, while necessary to contain the virus's spread, have led to the closure of businesses, disruptions in global supply chains, and significant losses in income for workers and families. The economic impact is particularly severe for vulnerable populations and those working in informal sectors.
3. Food Security:Disruptions in agricultural production and supply chains have raised concerns over food security. In some regions, the pandemic has affected food availability and accessibility, increasing the risk of hunger and malnutrition. The economic fallout from the pandemic further compounds food insecurity, as more individuals and families struggle to afford basic necessities.
4. Social Security:The pandemic has strained social fabrics, leading to increased isolation, mental health issues, and domestic violence. Social distancing measures, while crucial for public health, have disrupted traditional support systems and community networks. The situation is exacerbated by the stigma and discrimination associated with COVID-19, affecting certain groups more profoundly.
5. Political and Community Security:The coronavirus has tested the resilience of governance and political systems. In some cases, it has led to the postponement of elections, restricted civic freedoms, and heightened tensions among communities. The effectiveness of governmental responses to the pandemic has also become a source of public scrutiny, influencing trust in public institutions.
6. International Security:On a global scale, the pandemic has impacted international relations and cooperation. Issues such as vaccine nationalism, restrictions on travel, and competition for medical resources have highlighted the challenges of managing a global health crisis in a politically fragmented world. The pandemic underscores the need for strengthened international cooperation and solidarity to address shared threats.
In conclusion, the spread of the coronavirus represents a profound threat to human security, touching upon all aspects of life. Addressing these challenges requires a holistic and coordinated approach that goes beyond immediate health responses, encompassing economic support, social protection, and international collaboration. As we navigate through and beyond this pandemic, the lessons learned will be crucial for building more resilient societies capable of withstanding future crises.
Should you require further insights or wish to discuss specific aspects in more detail, please do not hesitate to reach out.
Best regards,
With this protocol list, we might find more ways to solve this problem.
  • asked a question related to Virus
Question
1 answer
I have knocked down my gene of interest by lentiviral transduction. I revived the vials and continue growing cells for 72 h without any selection. After confluency, I passaged the cells later 24 hours I have used puromycin 1ug/ml, 3ug/ml, 5ug/ml for selection of transduced cells. After 2 days, most of my virus transduced cells die at all the puro conc. The viral transduction itself doesn't seem to be toxic as cells were growing up until selection. Any suggestions would be greatly appreciated!
Relevant answer
Answer
What kind of cells? What is a vector design? How are You sure if the lentivirus entered the cells and integrated?
  • asked a question related to Virus
Question
7 answers
I would like to increase the titre of my virus before spin infection to increase my infection efficiency. 
Relevant answer
Answer
Concentrating retrovirus before spin infection (or centrifugal enhancement of infection) can significantly increase the efficiency of gene transfer, especially when infecting hard-to-transduce cells. There are several methods for concentrating retrovirus, each with its own advantages and considerations. Here are the most common techniques:
1. Ultracentrifugation
Ultracentrifugation is a widely used method for concentrating retrovirus. This process involves spinning the viral supernatant at a high speed to pellet the viral particles.
  • Procedure:Collect viral supernatant 48-72 hours after transfection of producer cells. Filter the supernatant through a 0.45 µm filter to remove cell debris. Transfer the filtered supernatant to ultracentrifuge tubes and spin at approximately 70,000-100,000 x g for 2 hours at 4°C. Carefully discard the supernatant and resuspend the viral pellet in a small volume of appropriate medium or buffer (e.g., PBS), overnight at 4°C for best results. The concentrated virus can be aliquoted and stored at -80°C for long-term use.
2. Precipitation Methods
Precipitation methods involve adding a compound to the viral supernatant that facilitates the precipitation of viral particles, allowing them to be collected by lower-speed centrifugation.
  • Polyethylene Glycol (PEG):Add PEG (e.g., PEG 8000) and NaCl to the filtered viral supernatant to final concentrations of 8% and 0.15 M, respectively. Incubate overnight at 4°C to allow the virus to precipitate. Centrifuge at 1,500-3,000 x g for 30 minutes at 4°C. Discard the supernatant and resuspend the viral pellet in a small volume of PBS or medium.
3. Concentration Using Filtration Devices
Commercially available filtration devices, such as centrifugal filter units with a molecular weight cutoff suitable for viral particles (typically around 100 kDa), can also be used for virus concentration.
  • Procedure:Filter the viral supernatant to remove cell debris. Load the filtered supernatant into the filtration device. Centrifuge according to the manufacturer's instructions until the volume is reduced to the desired concentration. The concentrated virus can be directly used or stored at -80°C for future use.
Considerations
  • Efficiency: Each method has its pros and cons regarding yield, purity, and practicality. Ultracentrifugation typically provides high purity and concentration but requires specialized equipment. Precipitation is simpler but may result in lower purity. Filtration is easy and effective but may require multiple filtration units for large volumes.
  • Application: The choice of concentration method might also depend on the specific application of the retrovirus. For instance, high purity might be more critical for in vivo applications.
  • Storage: Concentrated virus should be aliquoted to avoid freeze-thaw cycles, which can reduce viral titer.
Experimenting with different methods and optimizing conditions based on your specific requirements and available resources is advisable to achieve the best results.
l Check out this protocol list; it might provide additional insights for resolving the issue.
  • asked a question related to Virus
Question
4 answers
The patient is being kept alive by 100% O2 input.
Relevant answer
Answer
Reviving lungs that have been infected by the swine flu virus, or any influenza virus, largely depends on the severity of the infection and the damage incurred. Influenza A virus H1N1, commonly known as swine flu, can range from a mild to severe respiratory illness, and the body's ability to recover varies significantly among individuals. Here are several key points regarding recovery and treatment:
Mild to Moderate Infections
  • Immune Response: In cases of mild to moderate infection, the body's immune system can often clear the virus effectively on its own. Supportive care and symptomatic treatment may be all that's required.
  • Antiviral Medications: Drugs like oseltamivir (Tamiflu) or zanamivir (Relenza) can be prescribed to reduce the severity and duration of symptoms, especially if taken within the first 48 hours of symptom onset.
Severe Infections and Lung Damage
  • Hospital Care: Severe cases, particularly those involving significant lung damage or complications like pneumonia, may require hospitalization. Treatment can include antiviral therapy, antibiotics (to prevent or treat secondary bacterial infections), oxygen therapy, and sometimes mechanical ventilation support.
  • Recovery and Rehabilitation: Recovery from severe lung infection may involve a prolonged period of rehabilitation. The lungs can heal from the damage over time, but the recovery process can vary widely and may take weeks to months. Some individuals may experience long-term respiratory issues following a severe influenza infection.
Preventive Measures
  • Vaccination: Getting vaccinated against the influenza virus is a key preventive measure that can reduce the risk of severe infection and complications.
  • Hygiene Practices: Regular hand washing, wearing masks during outbreaks, and avoiding close contact with infected individuals can also help prevent the spread of the virus.
Experimental and Supportive Therapies
  • In cases of severe lung damage, experimental therapies such as ECMO (extracorporeal membrane oxygenation) may be used. ECMO is a life-support technique that oxygenates the blood outside the body, allowing the lungs to rest and heal.
  • Research into regenerative medicine and stem cell therapy offers potential future treatments for repairing lung tissue damaged by viral infections, though these are still in the experimental stages.
Conclusion
While the body can often recover from mild to moderate swine flu infections with appropriate care, severe infections requiring hospitalization can result in significant lung damage that necessitates more intensive treatment and a longer recovery period. The extent to which lungs can "revive" or heal depends on the severity of the damage, the overall health of the individual, and the treatments applied. Continuous advances in medical treatments and supportive care improve outcomes for those affected by severe influenza infections.
l Reviewing the protocols listed here may offer further guidance in addressing this issue
  • asked a question related to Virus
Question
6 answers
Q1
We have animal behavior scores of 4 group, Normal+ctrl virus, Normal+down-regulation virus, Model+ctrl virus and Model+down-regulation virus. It has two factors(Independent Variable): Model and virus. Editors suggested we use two way ANOVA to analyze, and now we obtained main effects of Model (F(1, 56)=201.18, P<0.0001) and virus (F(1, 56)=11.17, P=0.00427), as well as Model × virus interactions (F(1, 56)=16.13, P=0.0007).
If we should continue to calculate? For example, Model+ctrl virus vs. Model+down-regulation virus. We want to confirm the role of virus in Model animals.
Q2
Next, we used chemical drug to treat the Model animals and Normal animal. It has 4 drug concentration. Should we still use two way ANOVA to analyze the behavior scores? We want to know the role of different drug concentration in Model animals. And what do we do after two way ANOVA?
Thanks very very much!!!
Relevant answer
Answer
Dear Esteemed Colleague,
Following the completion of a two-way ANOVA, which serves to ascertain the effects of two independent variables on a dependent variable, as well as any interaction between these independent variables, your subsequent steps should be methodically oriented towards a comprehensive interpretation and validation of the results obtained. Here is a structured approach to guide your post-ANOVA analysis:
  1. Examine ANOVA Assumptions: Prior to delving into further analysis, it is crucial to ensure that the assumptions underlying the two-way ANOVA have been met. These include the assumptions of normality, homogeneity of variances (homoscedasticity), and independence of observations. Tools such as the Shapiro-Wilk test for normality and Levene's test for equality of variances can be employed to assess these assumptions. Should any assumptions not be satisfied, corrective measures such as data transformation or the use of non-parametric tests may be considered.
  2. Interpret Main Effects and Interaction Effects: The core of your analysis will involve interpreting the main effects of each independent variable and any interaction effects between them. A significant main effect indicates that different levels of an independent variable have significantly different impacts on the dependent variable. A significant interaction effect, on the other hand, suggests that the effect of one independent variable on the dependent variable varies depending on the level of the other independent variable. It is essential to carefully interpret these effects in the context of your research question.
  3. Conduct Post Hoc Tests for Multiple Comparisons: In the event that your ANOVA results indicate significant effects, post hoc tests are necessary to determine which specific groups differ from each other. Techniques such as Tukey's HSD (Honestly Significant Difference) test, Bonferroni correction, or Sidak adjustment are commonly employed for pairwise comparisons while controlling for the family-wise error rate. The choice of post hoc test depends on the specific characteristics of your data and the comparisons of interest.
  4. Evaluate the Magnitude of Effects: Beyond statistical significance, assessing the practical significance of your findings is vital. This can be achieved by calculating effect sizes, such as partial eta squared (η²) or Cohen's d, which provide insight into the magnitude of the differences or relationships observed. These measures help to contextualize the importance of your findings in real-world terms.
  5. Graphical Representation of the Results: Visualizing your data and the results of the ANOVA can greatly aid in their interpretation. Interaction plots, for example, are particularly useful for visualizing how the levels of one independent variable affect the outcome across the levels of another independent variable. Box plots and bar charts can also be effective in displaying the central tendencies and variabilities within and across the groups.
  6. Report Your Findings: The final step involves a detailed and coherent reporting of your methodology, analysis, results, and interpretations. This should include a summary of the ANOVA results, post hoc tests, effect sizes, and any graphical representations. It is crucial to discuss the implications of your findings in the context of existing literature and your research objectives, including any limitations and suggestions for future research.
By following these steps, you will ensure not only the rigorous analysis of your two-way ANOVA results but also the meaningful interpretation and reporting of these results within the broader context of your research field.
Should you require further assistance or clarification on any of these steps, please do not hesitate to reach out.
Warm regards.
This protocol list might provide further insights to address this issue.
  • asked a question related to Virus
Question
5 answers
Hi, here in my lab I have a kit for RT-PCR with probes (for the real time quantification). Can I run my reaction without adding the probes, in order to obtain en “end point” product? I was thinking about running two reactions, one without the probes and one with the probes, in order to obtain the product amplified but also its quantification.
I want to do this because I have to amplify a gene from a ssRNA+ virus and insert it into a plasmid.
Do you think it is possible? Does the absence of the probes impacts negatively on the Taqman polymerase? Thank you for your help
Relevant answer
Answer
@Yoav Lubelsky yes, I will use oligos for the RT. I also have designed primers to the CDS that I need
  • asked a question related to Virus
Question
1 answer
Physalis rugose mosaic virus (PhyRMV) is a Sobemovirus that causes severe damage to Physalis peruviana L., affecting vegetative parameters, fruit quantity and quality.
It was reported in Brasil and some other parts of South America. We are looking for photo of typical symptoms caused by this virus on host plants, especially on tomato.
Relevant answer
Answer