Science topics: ChemistryBiochemistry
Science topic

Biochemistry - Science topic

This group is designed for the exchange of thoughts on Biochemistry
Questions related to Biochemistry
  • asked a question related to Biochemistry
Question
1 answer
Hello,
My lab recently acquired a combination ORP electrode, with the refrence being Ag/AgCl and the working electrode being platinum.
We are hoping to use this electrode to measure the reduction potential of buffers prepared using biologically active compounds, such as GSH and GSSG and probe redox active systems by artificially setting the potential.
Recently, I was trying to validate the electrode by preparing 1 mM total concentration solutions of varying ratios of GSH and GSSG. This was done as, to my understanding regardless of concentration the ratio of Ox vs Red determines the potential value of the solution via the Nernst Equation. However the readings I got were all positive, and nowhere close to the expected potential, even when correcting for the electrode difference between Ag/AgCl and SHE.
Secondly, in 1x PBS pH 7.4, I added increasing amounts of BME up to 1 M and got an exponential decay like curve asymptotically approaching ~-120 (SHE) mV.
I am having trouble making sense of these results, namely the GSH vs GSSG ratio, and why the readings would not follow the nersnt equation.
Can anyone explain how to use these ORP electrodes, and where I may be going wrong in these experiments? All the information I can find online are referring to waste water treatment.
Relevant answer
Answer
Dear Darius Chernitsky,
I see two serious mistakes in your reasoning concerning the Nernst equation as applied to the measurement of the redox potential, specifically, in the glutathione (GSH/GSSG) system.
First. The statement that “regardless of concentration the ratio of Ox vs Red determines the potential value…” is wrong, since the Nernst equation is defined in terms of activity, not concentration. (In simple terms, intermolecular interactions reduce the effective concentration of the particles in solution so that from the "viewpoint of an electrode", the perceived concentration is smaller than the actual concentration.) This means that activity coefficients both of the components of the redox couple should be taken into account. The data on activity coefficients in solutions of GSH/GSSG may not be available, especially in buffer systems in which you are going to measure the potentials.
Second. The Nernst equation is applicable for equilibrium conditions, both “inside” the redox system and between the electrode and the redox components in solution. From the fact that the equilibrium between GSH and GSSG in biochemical systems is established rapidly (due to the involvement of enzymes) does not follow that this will be so in model solutions. It is natural to expect the appearance of kinetic limitations. Thus, the simple solution to the problem that you hoped for is hardly possible.
Regards,
Rouvim Kadis
  • asked a question related to Biochemistry
Question
5 answers
I am attempting to synthesize a schiff base ligand by reacting 5-hydroxytryptamine (HCl salt) and pyridoxal (HCl salt) in methanol, but the final product is not crystallizing in the methanol to be recrystallized. The product is originally a sticky brown oil that solidifies as a whole. What should I try differently? Sulfuric acid catalyst and maybe glacial acetic acid? Anything else?
Relevant answer
Answer
I would throw in 2 equivalents of an organic base such as TEA (triethylamine) to trap the HCl. This would result in deprotection of the primary amino group (protected by HCl) of hydroxytryptamine and free pyridoxal from its aminium salt. Triethylammonium chloride would remain in solution while the Schiff product may precipitate out. Filter off, wash twice with methanol and dried in oven.
In case of inconclusive purity by TLC, redissolve precipitate in DCM, wash twice with water, dry over MgSO4 and column if necessary.
An alternative route would be to deprotect both reagents separately before the Schiff reaction. To this end, dissolve a quantity of reagent in water, adjust pH to 8-9, the precipitate that form is filter off and dried.
  • asked a question related to Biochemistry
Question
6 answers
Background Biochemistry
Relevant answer
Answer
I am interested in biochemistry research
  • asked a question related to Biochemistry
Question
4 answers
I tried glycine buffer but the yeast cells acidify the buffer so the pH goes down to 7-6 overnight. I need something that will stay around 9 and that isn't toxic to the cells.
Relevant answer
Answer
To maintain a high pH (pH 9) for yeast cell suspensions, you can use a buffer that is effective in this pH range. Tris buffer is commonly used for maintaining alkaline pH and may be suitable for your needs. Here's how you can prepare Tris buffer at pH 9:
  1. Tris Base (Tris(hydroxymethyl)aminomethane): Dissolve Tris base in distilled water to make a 1 M stock solution. The molecular weight of Tris base is 121.14 g/mol, so to make a 1 M solution, dissolve 121.14 g of Tris base in 1 liter of water.
  2. Adjust pH: Adjust the pH of the Tris solution to 9 using concentrated hydrochloric acid (HCl) or concentrated sodium hydroxide (NaOH). Use a pH meter or pH strips to monitor and adjust the pH as necessary.
  3. Final Dilution: Once the desired pH is achieved, dilute the Tris solution to the desired concentration for your experiment. Common working concentrations for Tris buffer range from 10 mM to 100 mM.
  4. Sterilization: Filter-sterilize the Tris buffer using a 0.22 μm membrane filter to remove any particulate matter and microorganisms.
  5. Storage: Store the Tris buffer at room temperature (if using within a few weeks) or at 4°C for longer-term storage. Avoid repeated freeze-thaw cycles.
Tris buffer is widely used in biological research and is generally compatible with yeast cell suspensions.
  • asked a question related to Biochemistry
Question
7 answers
Most of the researchers use to teach at university. In some careers, professionals who exert their profession without doing research share teaching spaces. When I was a chemistry student, 100% of my teachers were researchers ranging from PhD candidates to experts in their respective fields. While it may seem logical for researchers to be the best candidates to teach in fields such as chemistry or biology, what about healthcare-related fields like medicine, pharmacy, or biochemistry? Who is better suited to lead a class, a researcher or a professional, or both, each one in different subjects? We can distinguish between basic and clinical subjects. I am interested in hearing your thoughts on this matter.
Relevant answer
Answer
Being a proficient researcher doesn't necessarily equate to being a better professor. While research expertise can enhance teaching by bringing current knowledge and real-world applications into the classroom, effective teaching requires distinct skills such as communication, empathy, and the ability to engage students.
A good professor balances both research and teaching responsibilities, tailoring their approach to meet the needs of their students while contributing to their field through research. However, being a successful researcher doesn't guarantee effective teaching, as teaching requires its own set of abilities and dedication.
  • asked a question related to Biochemistry
Question
5 answers
In all academic sources, sucrose is identified as α−glucose (1-->2) β−fructose. However, I cannot find any explanation anywhere as to why the fructose monomer has to be in the β configuration. Maltose has both α and β anomers, same for lactose. Even trehalose, another non-reducing disaccharide with glycosidic linkage between two anomeric carbons, has α-α, α-β, and even β-β anomers. Why is sucrose special? And is there a disaccharide out there that has α−glucose (1-->2) α−fructose configuration?
Relevant answer
Answer
It's because in sucrose the anomeric C atoms of glucose and fructose are both involved in the glycosidic bond. That's why they can't change their configuration without breaking the glycosidic bond. In reducing disaccharides like maltose the anomeric C of the "second" glucose is "free" (hemiacetal only). Therefore, the open-chain form and all possible configurations (alpha, beta, furanose, pyranose) of the second sugar are available without breaking the glycosidic bond.
  • asked a question related to Biochemistry
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 Biochemistry
Question
1 answer
I need some suggestions for articles recently focused on the computational design of proteins, as well as the evaluation of the designed proteins by assessing various properties or values. If anyone is interested in this field and has read articles on this topic, please don't hesitate to share your suggestions below
Relevant answer
Answer
Hi, you can read some papers:
1. Computational Protein Design: Advances in Algorithms and Biophysical Validation
This article discusses recent advances in computational protein design algorithms and methodologies, focusing on biophysical validation techniques to assess the stability, structure, and function of designed proteins.
2. Machine Learning Approaches for Protein Design: Recent Advances and Future Directions
This review article explores the application of machine learning techniques in protein design, including deep learning approaches, and discusses methods for evaluating designed proteins using molecular dynamics simulations and experimental validation.
3. Rational Protein Design: Computational Approaches and Experimental Validation
This paper presents computational strategies for rational protein design, highlighting methods such as Rosetta and FoldX, and provides insights into experimental techniques for validating designed proteins through biophysical assays and structural characterization.
4. Multi-Objective Optimization in Computational Protein Design: Balancing Stability, Function, and Specificity
This article focuses on multi-objective optimization strategies in computational protein design, aiming to balance various design objectives such as stability, function, and specificity. It discusses evaluation metrics and experimental validation approaches for assessing designed proteins.
5. Integration of Structural Bioinformatics and Machine Learning for Protein Engineering
This study presents an integrated approach combining structural bioinformatics and machine learning for protein engineering, emphasizing the importance of accurate protein structure prediction and molecular dynamics simulations in evaluating designed proteins.
6. High-Throughput Screening Platforms for Computational Protein Design
This review article surveys high-throughput screening platforms used in computational protein design, including methods based on protein-protein interactions, enzyme assays, and cell-based assays, and discusses their applications in evaluating large libraries of designed proteins.
7. Protein Design in the Age of Synthetic Biology: From Theory to Experiment
This paper explores the intersection of protein design and synthetic biology, discussing computational tools for designing novel protein functions and experimental techniques for characterizing designed proteins in living systems.
These articles provide insights into recent advancements in computational protein design and offer strategies for evaluating the properties and functions of designed proteins through computational and experimental approaches.
  • asked a question related to Biochemistry
Question
2 answers
Dear Researcher Gate,
I inquired about adding a reviewer to the submitted manuscript in Biochemistry can you help me with this?
Thanks
maha saad
Relevant answer
Answer
Dear Maha Maki In addition to what is indicated by Michael J. Benedik you can come up wit suggestions by:
-Use names you know in your field of expertise
-Use names of those who published relevant papers in your field
-Use names of the publications you cited
Nowadays all the relevant contact info can be found in their publications.
Best regards.
PS. You do not have to know these people personally, the journal just want to increase their network of potential peer-reviewers with people you estimate to be able to review your manuscript.
  • asked a question related to Biochemistry
Question
3 answers
I am currently learning about PyMol to utilize in my project. I used PyMol to visualize potential H-bond interactions in specific amino acid residues. However, I have discovered that Arg465 and Ser461 show a distinct interaction, as shown.
Please help identify this interaction.
Relevant answer
Answer
The broken yellow line with the distance indicator (6.2) looks like a simple distance monitor which you generate with a "measure" command, although I do not know how you generated the blue tubes around it. At 6.2Å, the Ca-Ca distance indicated by the broken line is far larger than the sum of the carbon Van der Waals radii (3.4Å). It is just about short enough that you might classify the contact as a solvent excluding contact (hydrophobic interaction)
  • asked a question related to Biochemistry
Question
5 answers
An unopened Sigma-Aldrich (P4557) phenol solution bottle was shaken (prior to the addition of the Equilibration Buffer) and a gel-like layer formed at the bottom of the bottle. The upper phase is still liquid. The bottle was shaken briefly after the phenol solution was taken out of +4 C. What should be done? Should it be heated in order for it to return to liquid?
Relevant answer
Answer
All you need to do is to heat it up slightly, but not with real flame, do that by gently stirring it until it returns to liquid. If this does not work, contact the supplier/company for assistance. Thank you.
  • asked a question related to Biochemistry
Question
8 answers
Hi,
I am looking for build up a consensus or a group in clinical biochemistry for mutual exchange in scientific idea, research protocols, cooperation in proposal preparation, sharing in books, Research articles and reviews.
Relevant answer
Answer
I am in fir such collaboration. Cant wait to get started
  • asked a question related to Biochemistry
Question
8 answers
It is known that in the early stages of desminopathy the muscles most often affected are: Semitendinosus, Gracilis and Sartorius. What is the reason for the damage to these particular muscles?
Relevant answer
Answer
Desminopathy, also known as desmin-related myopathy (DRM), is a rare genetic muscle disorder that affects the protein desmin. Desmin is an essential component of the intermediate filaments that provide structural support within muscle cells. Mutations in the DES gene, which codes for desmin, lead to disruptions in the normal structure and function of muscle fibers.
The muscles you mentioned - Semitendinosus, Gracilis, and Sartorius - are often affected at the onset of desminopathy due to their specific characteristics and biomechanical roles.
1. Semitendinosus: The semitendinosus is one of the three hamstring muscles located in the back of the thigh. It plays a key role in knee flexion and hip extension. The semitendinosus muscle is frequently involved in desminopathy due to its high proportion of slow-twitch muscle fibers, which are more vulnerable to desmin-related abnormalities.
2. Gracilis: The gracilis muscle is a long, thin muscle located in the inner thigh region. It is involved in hip adduction and knee flexion. Similar to the semitendinosus, the gracilis muscle also consists of a high proportion of slow-twitch muscle fibers, making it susceptible to desmin-related abnormalities.
3. Sartorius: The sartorius muscle is a long, strap-like muscle that runs diagonally across the front of the thigh. It plays a role in hip and knee flexion and also assists in thigh rotation. The sartorius muscle is affected in desminopathy due to its similar composition of slow-twitch muscle fibers.
The predilection for these specific muscles in desminopathy may be attributed to their fiber type composition and the mechanical stress they experience during certain movements. However, it is important to note that desminopathy can affect other muscles as well, and the degree and pattern of muscle involvement may vary among individuals with the same genetic mutation.
It is advised to consult with a medical professional or genetics specialist for a more accurate assessment of muscle involvement and management of desminopathy.
  • asked a question related to Biochemistry
Question
2 answers
Here and Now: Dependent Clause (Truth: Millions of Colors, Shades of Grey)
Here and Hereafter: Independent Reasoning (Independence: School of Taught)
Hereafter and Evermore: Organized Biologics (Chemistry x Biology + Physics) = Location Location Location = L3 (Heart-Person, Soul-Public, Interest-Private)
Key: Spelling-PEAR$
Note: Commentary
Comment: oh holy nigh
Relevant answer
Answer
Entanglement of electrons in quantum theory seems to be correct but other entanglements seems to be intuitionism as being maybe some kind of telepathy which yet incomprehensible !?
  • asked a question related to Biochemistry
Question
8 answers
In biochemistry, most work is done at room temperature. Yet, basic thermodynamics tells us that affinities often change with temperature, in positive or negative directions depending on the entropy/enthalpy contributions. Thermal transitions can occur which both quantitatively and qualitatively change the behavior of the molecules of study. In enzymatic reactions rate-limited by diffusion-mediated product release, the increased rate of diffusion could increase the rate of product release above the rate of the chemical step, such that the chemical step becomes rate-limiting. If one discovers a compound that potently inhibits this enzyme at 25C, it may have reduced effect at 37C, or none at all. Likewise, if an enzyme is predominantly dimerized at 25C based mostly on enthalpic contributions, this dimer may not even exist at 37C. Screening compounds against the dimer may be of little relevance to the situation in vivo. The converse could happen if dimerization is entropically driven. Temperature-dependent changes in solution properties can also obscure the relevance of 25C results to 37C, such as viscosity.
I welcome everyone's two cents.
Relevant answer
Answer
I don't think it's true that "Biochemistry experiments are done at room temperature, not 37C." They are often done at 37C. However, I do most of my enzyme assays at room temperature because I usually work with 384-well microtiter plates, and it is difficult to establish a consistent elevated temperature across the whole plate on a short time scale. It is important to report the temperature in any publications. Surprisingly, this detail is often forgotten in Methods sections. Also, "room temperature" should be defined as a specific temperature or range of temperatures.
If a detailed mechanistic understanding of the biochemical mechanism is being investigated, it is very important to be clear about the temperature. Moreover, varying the temperature can provide useful information about the chemical mechanism, such as the activation energy of the reaction.
  • asked a question related to Biochemistry
Question
2 answers
Here is an excerpt from Kollmann et al. (2020, J. Physiol.):
"This ring was placed in a recording chamber continuously perfused with 37°C aerated Hepes solution containing (in mM) 136 NaCl, 10 glucose, 5 KCl, 10 Hepes, 1.2 MgCl2, 2.5 CaCl2 (pH 7.40) at a rate of 11 ml min-1. Hypoosmolality of the Hepes solution was established by reduction of the NaCl content to 33 mM (94 mOsm kg-1 H2O), 58 mM (144 mOsm kg-1 H2O) or 83 mM (194 mOsm kg-1 H2O)."
For validation, I tried to calculate the osmolality by myself. For example, when the NaCl content is changed to 33 mM, the osmolality should be
33*2 (NaCl)+10*1 (Glucose)+5*2 (KCl)+10*1 (Hepes)+1.2*3(MgCl2)+2.5*3(CaCl2)
which gives 107.1 mOsm/L, i.e., 107.1 mOsm/kg. But 107.1 is largely different from what the author stated, namely 94 mOsm/kg.
When the NaCl content is changed to 58 mM or 83 mM, the osmolality should increase by (58-33)*2=50 mM, or (83-33)*2=100 mM. This is consistent with the author's calculation, i.e., 144-94=50 mM, or 194-94=100 mM. So, my calculation is correct at least in terms of NaCl. But how can I calculate the contributions of the remaining solutes correctly?
Reference: Kollmann, P. et al. Submucosal enteric neurons of the cavine distal colon are sensitive to hypoosmolar stimuli. J. Physiol. 598, 5317–5332 (2020).
Relevant answer
Answer
Your calculations are correct if and only if you assume an osmotic coefficient of 1.0 for each solute. The manuscript doesn't describe how the osmotic coefficients were determined or if tabulated values were used, but does describe the use of an osmometer in at least one instance, so it is possible that these were experimentally determined, but this isn't clear from the text.
In any case, details about how the solutions were prepared are described in enough detail to repeat them accurately. I suppose you would need to find tabulated values for osmotic coefficients to arrive at the same number as the authors, or alternatively, you could use an osmometer to determine the NaCl needed to adjust your own solutions to the same osmotic pressures/osmolality.
  • asked a question related to Biochemistry
Question
5 answers
Is it correct to filter the extracts we obtain with Whatman filter paper and use them directly in experiments to investigate the bioactivity of phytochemical substances? Or is it correct to first centrifuge the extracts we obtain, then filter the supernatant and use them in experiments?
Relevant answer
Answer
You should first carry out a rough clarification of the extract by simply decanting, followed by a filtration step by using a six to seven layered muslin cloth. Then you may include a centrifugation step which may be required if the powder is too fine to be filtered.
I have attached a book below which may be helpful.
Best.
  • asked a question related to Biochemistry
Question
1 answer
Some (but not all) DNA polymerases such as Klenow fragment, Taq, and Phi 29 DNA polymerase can catalyze strand displacement synthesis. This is evidenced by opening of molecular beacons and Loop Mediated Isothermal amplification (LAMP).
in strand displacement synthesis, the primer strand is extended using the template strand and each nucleotide incorporated displaces the 3rd strand that was originally annealed to the template?
where is the third DNA strand? the one being displaced? crystal structures and cryo-EM structures of linear primer template dsDNA structures with magnesium and dNTPs and DNA polymerases are not informative about where 3rd strand of DNA is. The displaced strand is not the template and is not the primer.
Where is the displaced 3rd strand of DNA?
Relevant answer
Answer
Yes, that’s correct! In strand displacement synthesis, the primer strand is extended using the template strand, and each nucleotide incorporated displaces the third strand that was annealed initially to the template.
The third strand is a short, single-stranded DNA annealed to the template strand and acts as a stabilizing agent for the displaced strand . The displaced strand is the second strand of the double-stranded DNA being synthesized.
More at:
- Mechanism of strand displacement DNA synthesis by the coordinated activities of human mitochondrial DNA polymerase and SSB | Nucleic Acids Research | Oxford Academic (oup.com)
-D-loop - Wikipedia
  • asked a question related to Biochemistry
Question
6 answers
I need to dissolve sulphate to purify the nanozyme synthesized with magnesium sulphate
Relevant answer
Answer
thanks a million for your answer.
  • asked a question related to Biochemistry
Question
4 answers
  • Which is your favourite field of Medicine and why?
  • In which Medical field do you work?
  • What is your field of interest?
Relevant answer
Answer
Minha paixão é a psicanálise. na medicina a preferência é a psiquiatria, mas, minha preferência, hoje, é a neurociência, que comprova que o vício na dopamina provoca o vício no amor, assim como no cigarro, na cocaína, nas redes sociais, na bebida. ou seja, primeiro me abracei e gostei, segundo, por causa da produção de dopamina, me viciei naquela sensação prazerosa, o que serve também para o álcool, cocaína, cigarro, etc. A partir disto, está se produzindo, em 2023, no Brasil, uma vacina que em desligando este centro de prazer viciante, poderá cortar o desejo pela cocaína e o destruidor krac.
  • asked a question related to Biochemistry
Question
7 answers
Dear all,
I have been trying to knockdown my target protein using siRNA. The protein has several isoforms the effect of this knockdown has been showing up in the functional assay. However, I don't see the knockdown in the wesptern blot of the same test sample. Please suggest how to do an effective knockdown so that I can visualize that in the western blot as well.
Thank you in advance for your kind suggestions
Sincerely,
Prem
Relevant answer
Answer
You can definitely use multiple siRNAs. We use a pool of 4 as default regardless of isoforms.
  • asked a question related to Biochemistry
Question
2 answers
Hello,
We are trying to purify proteins using a secretion system but do not have TFF cartridges for our device. Does anyone know where we can purchase these? Or is there a better replacement? We want to downscale the volume from 3 L to 100-200 mL.
I've attached a picture for reference.
Thanks,
Thomas Newton
Relevant answer
Answer
PALL (now Cytiva) I think, used to sell TFF devices.
Tangential flow filtration | Cytiva (cytivalifesciences.com)
Another alternative could be Sartorius
TFF Systems For Ultrafiltration and Diafiltration | Sartorius
Hope it helps.
  • asked a question related to Biochemistry
Question
4 answers
I'm on the lookout for remote bioinformatics and computational biology opportunities where I can actively contribute to research projects. Compensation is not a priority for me; my main focus is to gain hands-on experience in these fields.
#biopython
#computational_biology
#bioinformatics
#biology
#R
Relevant answer
Answer
Avenues you can explore to find such opportunities:
1. Academic research institutions: Many universities and research institutions offer remote research positions or internships in bioinformatics and computational biology. Check their websites, job boards, and reach out to individual researchers or research groups who align with your interests.
2. Online job portals and platforms: Websites and platforms dedicated to remote work, such as LinkedIn, Indeed, and Upwork, often have listings for bioinformatics and computational biology projects. You can search for specific keywords like "remote bioinformatics," "computational biology," or "bioinformatics internships" to find relevant opportunities.
3. Open-source projects: Contributing to open-source bioinformatics projects can provide valuable hands-on experience. Explore bioinformatics software and libraries like Biopython, Bioconductor (for R), or other popular tools on platforms like GitHub. Contribute to their development, report issues, or collaborate with the community.
4. Online communities and forums: Engage with online communities and forums focused on bioinformatics and computational biology. These platforms, such as Bioinformatics Stack Exchange, BioStars, or community forums associated with specific software packages, often have job boards or project collaboration opportunities shared by researchers or organizations.
5. Networking: Attend virtual conferences, webinars, and workshops related to bioinformatics and computational biology. Connect with researchers, presenters, and fellow attendees to express your interest in remote research opportunities. Networking can often lead to potential collaborations or recommendations for available positions.
When searching for opportunities, it's important to tailor your search keywords to include relevant terms like "remote," "internship," "volunteer," or "project-based." Additionally, clearly communicate your enthusiasm, willingness to contribute, and desire for hands-on experience in your application materials or when reaching out to potential mentors or supervisors.
Hope it helps:credit AI.
  • asked a question related to Biochemistry
Question
7 answers
I intend to explore the integration of computational biology and artificial intelligence (AI) with laboratory and experimental work, encompassing animal models, cell culture, clinical trials, and molecular studies. As a clinical biochemistry student with a keen interest in AI, I believe this interdisciplinary approach holds immense potential for advancement and innovation.
However, I face the challenge of identifying relevant literature in this emerging field. I would greatly appreciate guidance on effective keywords and search strategies to navigate this landscape of research and achieve my research goals.
Relevant answer
Answer
I think there are several ways that they could be integrated. Each of those ways will have different ways to best approach this and a different literature review. For example what is the computational biology you are interested in - an DNA analysis is very different to molecular simulations or simulating a artificial hip. Biological laboratories can produce very different data a spead sheet is different to a image and the types of AI are different. It is common in computational science to want to compare simulation/computational results to experimental/observational results. I imagine this would be a good starting point for what you are interested in. In this case I think what you would want to do is use AI to segment or analyse images so that you have something to compare back to the simulation results. Can you give some examples of what you want to compare or research questions you want to be able to answer.
  • asked a question related to Biochemistry
Question
5 answers
My target protein is a membrane protein and I want to check its expression level in the test sample using western blot. Can anyone suggest which protein should be used for loading control, like we use beta-actin in cytosolic proteins. And is there antibody available for that ?
Relevant answer
Answer
When working with membrane proteins and performing Western blot analysis, it's essential to choose an appropriate loading control that is not affected by changes in membrane protein expression. Commonly used cytosolic loading controls, like beta-actin or GAPDH, may not be suitable in this case, as their expression levels can vary in response to changes in membrane protein expression.
Here are some options for loading controls when studying membrane proteins:
  1. Membrane Proteins as Loading Controls:In some cases, it may be possible to use another membrane protein as a loading control, especially if it is known to have stable expression across different conditions. Examples include various membrane transporters or receptors. However, finding a suitable membrane protein loading control depends on the specific context and characteristics of your experiment.
  2. Total Protein Staining:Use total protein staining methods, such as Ponceau S or Coomassie Blue, to visualize total protein on the membrane before antibody probing. This can serve as a general loading control, indicating the total amount of protein in each lane.
  3. Housekeeping Membrane Proteins:Identify housekeeping membrane proteins that are known to have relatively constant expression levels in the tissues or cells you are studying. Examples include Na^+/K^+ ATPase or V-type H^+-ATPase. However, it's essential to validate the stability of their expression in your experimental system.
  4. Use of Multiple Loading Controls:Consider using multiple loading controls to ensure the reliability of your results. For example, you could combine a membrane protein loading control with a total protein stain.
  5. Normalization to Total Protein Content:Normalize the intensity of your membrane protein of interest to the total protein content in each lane. This involves quantifying the intensity of your protein of interest and dividing it by the total protein intensity in the same lane.
As for the availability of antibodies, it depends on the specific protein you choose as the loading control. Antibodies against some common membrane proteins, such as Na^+/K^+ ATPase or V-type H^+-ATPase, are commercially available from reputable antibody suppliers. Ensure that the selected antibody recognizes the appropriate epitope and has been validated for Western blotting.
Before finalizing your loading control strategy, it's crucial to conduct preliminary experiments to validate the stability of expression of the chosen loading control(s) under your experimental conditions. Additionally, consider consulting the literature or seeking advice from researchers with expertise in the specific membrane protein and experimental system you are working with.
  • asked a question related to Biochemistry
Question
1 answer
I intend to explore the integration of computational biology and artificial intelligence (AI) with laboratory and experimental work, encompassing animal models, cell culture, clinical trials, and molecular studies. As a clinical biochemistry student with a keen interest in AI, I believe this interdisciplinary approach holds immense potential for advancement and innovation.
However, I face the challenge of identifying relevant literature in this emerging field. I would greatly appreciate guidance on effective keywords and search strategies to navigate this landscape of research and achieve my research goals.
Relevant answer
Answer
I think that Ross King (currently at Chalmers University of Technology) has a good number of publications that have made significant contributions to the subject of AI in science. You can look at some of his publications at one of the following links.
  • asked a question related to Biochemistry
Question
4 answers
I need to convert from micromoles per second per liter (µmol s⁻¹ L⁻¹) to millimoles per gram of dry cell weight per hour (mmol gDCW-1h-1)
Thanks!
Relevant answer
Answer
Wat bull. An "empirical formula" for a microorganism most of whose molecules are of profoundly greater molecular weight. But there's the "However..."
  • asked a question related to Biochemistry
Question
2 answers
Hello everyone! I am new to this platform, I am looking for this book: "Bioquimica del estres Oxidativo" (Diego Camps, 2010)
I would like to know if anyone has it in digital format to share it with me. Waiting for a reply. Thank you very much!
Relevant answer
  • asked a question related to Biochemistry
Question
2 answers
I need to extract the liver from frozen Emerald rockcod and sequence its RNA. To keep the RNA stable and prevent degradation, I would like to avoid thawing the fish for the dissection. However, I haven't been able to find any methods that keep the fish frozen. Does anyone have any tips on how to best achieve this?
Relevant answer
Answer
Probably would need to work in a walk in freezer that is just below 0.
  • asked a question related to Biochemistry
Question
4 answers
If an active site mutant knocks out product formation at all excessive concentrations of substrate and at all excessive concentrations enzyme, but substrate binding affinity via anisotropy shows no difference in binding affinity for wildtype versus active site mutant enzyme, is kcat = 0? But if kcat = 0, then by the relationship of Michaelis constant (KM) to kcat then KM = KD.
How do you show to reviewers that the active site mutant is dead? If the wildtype mutant starts producing product in seconds, are you supposed to measure the reaction for the mutant for an hour at zero concentration of substrate and at 100fold excess substrate concentration of the K_M for the wildtype enzyme for the active site mutant, and show the time course?
Are there any publications that show a mutant enzyme is not just slow to produce product but is rather incapable of producing product but can still bind substrate? Examples would be greatly appreciated!
Relevant answer
Answer
Short answer - I would say yes. If an enzyme can't turn over products on any time scale, its turnover number would be 0.
I agree that if kcat = 0, then effectively your KM is just the ratio of kon and koff, that is, it is equal to the KD. If one was simply measuring binding of the substrate to the enzyme, I can definitely see that replacement of a residue critical to the mechanism, but not the binding and orientation of the substrate, would generate a variant that does not turn over product but still has a similar or identical KD for the substrate. This says nothing about the turnover number (or lack thereof).
How would I show a reviewer that a binding-competent enzyme is catalytically inactive? I think what you have described is going above and beyond. Extending a reaction for 60 min at 100 x KM to prove no activity exists feels a bit extreme. Even if the measured kcat is very tiny but not zero, it would be a moot point as this has absolutely no biological relevance.
If your wild-type enzyme has a kcat > 1 s-1 like you mention, I think it would be reasonable to measure the initial rates of i) the wild-type enzyme, ii) the catalytic mutant, and iii) a matched enzyme-free reaction using the same reaction conditions for all on whatever time scale is appropriate for your enzyme and its relative rate.
With enough replicates, you can use appropriate statistics to compare the grouped samples. Showing that there is no statistical difference in the rate of reaction between an experiment with no enzyme at all versus a catalytic mutant would be a strong argument that no activity exists, i.e. kcat must equal 0.
You will see in this paper we use an LC-MS assay to detect the presence/absence of product in active site replacements as justification for a dead enzyme, in addition to a colorimetric assay where we did the above to justify activity as "n.d." We did not measure affinity to the substrate but the binding site is > 15 angstroms away in this case.
Hope this helps!
ACA
  • asked a question related to Biochemistry
Question
15 answers
Hello all,
I hope everyone is doing well.
We are preparing buttermilk from standardized milk. In this regard we need to increase the pH of the buttermilk. We used sodium citrate (0.4% & 4%) and sorbitol (0.4%) to check any change in the pH. Unfortunately, there were no significant changes observed.
Please suggest any recommendation to increase pH using any natural or food-grade chemical compounds and any other alternative options.
Thank you in advance.
Keep smiling and Stay healthy.
Relevant answer
Answer
If this is for cooking, a quick recipe swap is to add vinegar (the acidity here is acetic acid) to regular milk to mimic the tangy flavor of buttermilk. It's a spoonful or two (about 15-30 ml) vinegar per 240 ml milk.
  • asked a question related to Biochemistry
Question
4 answers
I am reaching out to #researchers in the field of #Biochemistry, #Biophysics and #Bioinformatics, for collaborative partnership in scientific research. The researcher should be academic staff at the tertiary institutions in following listed countries:
#Afghanistan
#Angola
#Bangladesh
#Belarus
#Belize
#Benin
#Bhutan
#Burkina Faso
#Burma
#Burundi
#CaboVerde
#Cambodia
#Cameroon
#CentralAfricanRepublic
#Chad
#Comoros
#Congo
#CookIslands
#Cuba
#Democratic People's Republic of Korea
#Democratic Republic of the Congo
#Djibouti
#Dominica
#EquatorialGuinea
#Eritrea
#Eswatini
#Ethiopia
#Gambia
#Ghana
#Grenada
#Guinea
#Guinea-Bissau
#Guyana
#Haiti
#Iran
#IvoryCoast
#Kenya
#Kiribati
#Kyrgyzstan
#Lao People's Democratic Republic
#Lebanon
#Lesotho
#Liberia
#Madagascar
#Malawi
#Maldives
#Mali
#Marshall Islands
#Mauritania
#Micronesia (Federated States of)
#Mozambique
#Myanmar
#Nauru
#Nepal
#Nicaragua
#Niger
#Niue
#Palau
#PapuaNewGuinea
#Moldova (Republic of)
#Rwanda
#SaintHelena
#SaintLucia
#SaintVincent and the #Grenadines
#Samoa
#SaoTome and #Principe
#Senegal
#Sierra Leone
#SolomonIslands
#Somalia
#SouthSudan
#Sudan
#Suriname
#Syrian Arab Republic
#Tajikistan
#Timor-Leste
#Togo
#Tokelau
#Tonga
#Tuvalu
#Uganda
#Ukraine
#Tanzania (United Republic of)
#Vanuatu
#Yemen
#Zambia
#Zimbabwe
Interested researcher should kindly email to [email protected] with the subject: Research Collaboration from "your country".
Thanks.
Toluwase H. Fatoki
Visionary @ Heze-Sapience International, Nigeria.
Lecturer @ Department of Biochemistry, Federal University Oye-Ekiti, Nigeria.
Relevant answer
Answer
And why don’t you want any collaboration from Nigeria?
  • asked a question related to Biochemistry
Question
3 answers
Is there any system with small molecule binders and a short protein tag that is higher affinity than 6xHIS-NTA?
Relevant answer
Answer
V5 and Flag tag are the only ones I've seen other than His.
Einhauer A, Jungbauer A. The FLAG peptide, a versatile fusion tag for the purification of recombinant proteins. J Biochem Biophys Methods. 2001 Oct 30;49(1-3):455-65. doi: 10.1016/s0165-022x(01)00213-5. PMID: 11694294.
Also consider the MBP that is engineered with a linker with a protease cut site. MBP sticks to the affinity column, and you use the protease to elute your protein of interest. Then of course the protease has to be purified out, so it should have an affinity tag or biotin.
Wikipedia has a long list.
Peptide tags
  • ALFA-tag, a de novo designed helical peptide tag (SRLEEELRRRLTE) for biochemical and microscopy applications. The tag is recognized by a repertoire of single-domain antibodies [5]
  • AviTag, a peptide allowing biotinylation by the enzyme BirA and so the protein can be isolated by streptavidin (GLNDIFEAQKIEWHE)
  • C-tag, a peptide that binds to a single-domain camelid antibody developed through phage display (EPEA)[6][7]
  • Calmodulin-tag, a peptide bound by the protein calmodulin (KRRWKKNFIAVSAANRFKKISSSGAL)
  • iCapTag™ (intein Capture Tag), peptide-based a self-removing tag controlled by pH change (MIKIATRKYLGKQNVYGIGVERDHNFALKNGFIAHN). Its patented component derived from Nostoc punctiforme (Npu) intein. This tag is used for protein purification of recombinant proteins and its fragments. It can be used in research labs and it is intended for large-scale purification during downstream manufacturing process as well. The iCapTag™-target protein complex can be expressed in a wide range of expression hosts (e.g. CHO and E.coli cells). It is not intended for fully expressed mAbs or membrane proteins[8][9][10]
  • polyglutamate tag, a peptide binding efficiently to anion-exchange resin such as Mono-Q (EEEEEE) [11]
  • polyarginine tag, a peptide binding efficiently to cation-exchange resin (from 5 to 9 consecutive R)
  • E-tag, a peptide recognized by an antibody (GAPVPYPDPLEPR)
  • FLAG-tag, a peptide recognized by an antibody (DYKDDDDK)[12]
  • HA-tag, a peptide from hemagglutinin recognized by an antibody (YPYDVPDYA)[13]
  • His-tag, 5-10 histidines bound by a nickel or cobalt chelate (HHHHHH)Gly-His-tags are N-terminal His-Tag variants (e.g. GHHHH, or GHHHHHH, or GSSHHHHHH) that still bind to immobilised metal cations but can also be activated via azidogluconoylation to enable click-chemistry applications[14]
  • Myc-tag, a peptide derived from c-myc recognized by an antibody (EQKLISEEDL)
  • NE-tag, an 18-amino-acid synthetic peptide (TKENPRSNQEESYDDNES) recognized by a monoclonal IgG1 antibody, which is useful in a wide spectrum of applications including Western blotting, ELISA, flow cytometry, immunocytochemistry, immunoprecipitation, and affinity purification of recombinant proteins [15]
  • Rho1D4-tag, refers to the last 9 amino acids of the intracellular C-terminus of bovine rhodopsin (TETSQVAPA). It is a very specific tag that can be used for purification of membrane proteins.
  • S-tag, a peptide derived from Ribonuclease A (KETAAAKFERQHMDS)
  • SBP-tag, a peptide which binds to streptavidin (MDEKTTGWRGGHVVEGLAGELEQLRARLEHHPQGQREP)[16][17][self-published source?]
  • Softag 1, for mammalian expression (SLAELLNAGLGGS)
  • Softag 3, for prokaryotic expression (TQDPSRVG)
  • Spot-tag, a peptide recognized by a nanobody (PDRVRAVSHWSS) for immunoprecipitation, affinity purification, immunofluorescence and super resolution microscopy
  • Strep-tag, a peptide which binds to streptavidin or the modified streptavidin called streptactin (Strep-tag II: WSHPQFEK)[2]
  • T7-tag, an epitope tag derived from the T7 major capsid protein of the T7 gene (MASMTGGQQMG). Used in different immunoassays as well as affinity purification Mainly used [18]
  • TC tag, a tetracysteine tag that is recognized by FlAsH and ReAsH biarsenical compounds (CCPGCC)
  • Ty tag (EVHTNQDPLD)
  • V5 tag, a peptide recognized by an antibody (GKPIPNPLLGLDST)[19]
  • VSV-tag, a peptide recognized by an antibody (YTDIEMNRLGK)
  • Xpress tag (DLYDDDDK), a peptide recognized by an antibody
  • asked a question related to Biochemistry
Question
1 answer
Dear all, please suggest or provide a link for the provider or company who can design or synthesize shRNA plasmid (Single vector system) that will directly express into the mammalian cells to knockdown the protein expression for long term. Any suggestions will be highly appreciated.
Thank you
with kind regards
Prem
Relevant answer
Answer
Hi Prakash,
The Santa Cruz Biotechnology, Inc. (https://www.scbt.com/home) company offers almost all ready-made shRNA or siRNA for known protein expressed in mammalian cells. You can search for your interested protein in HOME page. Some of their shRNA and siRNA used in our current and previous study is of good characteristics. I think this may be useful to your work.
Regards.
  • asked a question related to Biochemistry
Question
3 answers
Fields:
Cell and Molecular Biology
Biochemistry
Physiology
Microbiology
Relevant answer
Answer
If someone knows German and certifies with DAF exam, he/she may have incredible positive opportunities. Other than Germany there seems to be unfortunately no hope !
  • asked a question related to Biochemistry
Question
3 answers
I am trying to express several proteins at the same time, but I want to use a different promoter and terminator for each one to avoid the possibility of recombination.
The promoters that are available to me are: TEF2, PGK1, CCW2, TDH3 and HHF2. The available terminators are: ENO1, SSA1, ADH1, PGK1 and ENO2.
Has anyone ever used these combinations of promoters and terminators? In your experience, which combinations work the best?
Relevant answer
Answer
Hi there,
These are strong constitutive promoters and terminators. Any combination should be OK... I have successfully expressed simultaneously 6 human proteins from genomic insertions using the following combinations: ProPGK1+terTDH1; ProTDH3+TerADH1; ProHHF2+TerSSA1; ProCCW12+TerENO1; ProTEF1+TerENO2; ProTEF2+TerPGK1. The most crucial point being to optimize sequences for expression in yeast.
  • asked a question related to Biochemistry
Question
5 answers
Hi
I'm purifying some mutants of the protein I study. The wild type protein exists as a monomer and is 28kDa.
I have two mutants (same protein, same number of amino acids but with 8 amino acid substitutions at defined positions), one of the mutants (mutant 1) analysed using size exclusion chromatography with multi-angle static light scattering (SEC-MALS) and its MW was shown to be 33kDa and has an oligomerization state of 1.2. The other mutant, mutant 2, also measured by SEC-MALS was 59kDa with an oligomerization state of 2.2.
For the wild type to measure the concentration I've just been using the MW (28kDa) and extinction coefficient (calculated by entering the sequence into online software ProtParam) and using a NanoDrop measuring absorbance at 280. This gives the concentration in mg/ml which I then convert to molar concentration.
For the mutants I want to measure their concentration the same way - measuring A280 on the NanoDrop using the mutants MW and extinction coefficient and calculating molarity from mg/ml. I'm not sure if this is an obvious/stupid question but what MW weight and extinction coefficient would you use for the mutants on the NanoDrop? E.g. For example mutant 2 molecular weight (MW) of the protein based on its amino acids (AA) composition is predicted to be 28kDa, but SEC-MALS shows it is 59kDa as the protein forms an oligomer.
My instant is to use 59kDa and the computed extinction coefficient predicted from the AA composition - is this correct?
Thanks in advance!
Relevant answer
Answer
Thanks for your reply very helpful - so if I’ve understood you properly - it is incorrect to use the SEC-MALS molecular weight when measuring UV 280 absorbance to determine protein concentration of the mutants as the extinction coefficient is based on the protein being denatured. Rather I should use the predicted Mw just based on the amino acid composition.
I agree the deviation from whole numbers points to an oligomer mixture. I want to add these recombinantly purified mutants to permeabilised cells to observe their localisation (they are fluorescent proteins) so knowing concentration accurately is important for these experiments.
So for example for mutant 2 whose Mw just based on amino acid composition is 28kDa - if A280 on the nanodrop gives a concentration of 10 mg/ml which would be 0.35mM, the concentration protein is 0.17mM? (as the oligomerizaiton state is 2 taken the closest integer value). I should say the ability of these mutants to form oligomers is because the mutations increase their aggregation propensities.
Am I correct in this? Is the above way accurate or would something like the Bradford assay be better?
  • asked a question related to Biochemistry
Question
3 answers
Dear All,
Ph.D. full-time position in Bangalore with fellowship:
Eligibility: M.Sc. Chemistry/Biochemistry/Biotechnology/Microbiology/Bioinformatics with first class of 60%.
GATE or UGC-NET or UGC-CSIR or SLET or JRF should be qualified.
RS 25,000 per month for full three years will be given.
For further details, contact me on: +919182864256. Call or what's app me for further details.
Relevant answer
I apologize and excuse the owner of the post. I would like to invite you to read my ebook and discover why microorganisms are so fantastic. https://www.amazon.com.br/dp/B0CF1VKKK8
  • asked a question related to Biochemistry
Question
4 answers
PhD biochemistry candidate is required for the post of guest faculty. Interested candidates can contact me asap.
Relevant answer
Answer
PhD biochemistry from Iraq. From faculty of sciences /University of Kufa.
  • asked a question related to Biochemistry
Question
2 answers
I learned DNS assay to find out the quantity of maltose in the sample.
I realized the importance about the boiling water in this assay. But I got a several question after that.
I don't know the reason why do I have to cool-down the sample after heating up?
And also is ice a tool used to reduce cooling time, or is it used to cool quickly?
Please leave me a response about this problem.
Thank you for your help.
Joonseo_Cha The Student of Hallym University
Relevant answer
Answer
On heating the alkaline mixture of reducing sugar with DNS at elevated temperature (100°C) for 10 minutes, the reddish-orange color will develop, and at this point it will show a direct relationship with the concentration of the reducing sugar in the sample. The color developed is maximum at this time point (10 mins) which is standardized for this assay.
So, to stop the reaction immediately, the tube is dived in cold water. When the tube reaches room temperature, 300ul of the resulting reaction mixture is transferred to the well of a microtiter plate and the absorbance read at 540nm using microtiter plate reader.
So, when you use ice, it will cool down the sample quickly so as to stop the reaction in the tube.
Best.
  • asked a question related to Biochemistry
Question
19 answers
How many grams of K2Cr2O7 to dissolve it in 1 liter Distilled water to obtain 50 ppm of Chromium? to become aqueous solution, Is there a specific equation to apply? Thanks
Ali
Relevant answer
Answer
Why are so many methods and no answers the same? Other papers also mention the different results. What is actually the correct answer?
  • asked a question related to Biochemistry
Question
2 answers
The size differ little bit from one species to another, yet they have one size range. Also, the size of them in their native form so they don't lose their colour while isolation.
Relevant answer
Answer
Uniprot is a good website for researching specific proteins. For example, you can type phycoerythrin and it will pull up all proteins with that name. Each organism will have a separate entry. Then you can look at data that has been generated by other researchers on that specific protein, including size, function, sequence, etc.
  • asked a question related to Biochemistry
Question
3 answers
I saw these on a same step of conversion of Malonyl CoA to Malonyl-ACP. Could anyone clarify this? I add that reference link below.
KEGG PATHWAY: Fatty acid biosynthesis - Chlorella variabilis (genome.jp)
Relevant answer
Answer
In FAS i.e., fatty acid synthesis the chemical reaction is, Malonyl CoA+ Acyl-carrier protein↔ CoA+Malonyl-[acyl carrier protein]. In fact, there are two substrates for the enzyme concerned. Concerned enzyme is a transferase. The enzyme very often called as S-malonyltransferase may be termed as FabD, i.e., Acyl-carrier-protein- S- malonyltransferase.
  • asked a question related to Biochemistry
Question
3 answers
I'd like to perform a SEC step prior to on-column refolding of a protein I am expressing/purifying but am worried about the extended time the protein will be in the presence of urea (and subsequent protein carbamylation) in the current refolding workflow I have.
Is there any concern of protein modification with a 4-6 hour denatured protein purification workflow using 6M GndHCl?
Relevant answer
Answer
I haven't heard of this being a problem.
  • asked a question related to Biochemistry
Question
1 answer
I am a second year PhD student at Wayne State University looking to screen a library of photosensitizers (transition metal complexes) for activity with ultrasound irradiation in cell cultures (likely MCF-7A cells). I would ideally like to set this up in a 96-well plate to screen multiple compounds but I am unsure of the set up as previous literature has not been as helpful as I would hope. The instrument I have is 3 MHz with a 3.5 cm diameter probe (Model-Sonic 103, Preamonex). I am unfamiliar with soundwaves so I will briefly discuss what I know so far from previous literature.
Overall, 0.3 W cm–2, 3.0 MHz seems to be a fairly common treatment condition when combining compounds with cell cultures.
This source clusters samples in a 2x2 square in the well plate and places the probe underneath the plate per cluster (see SI). Does a coupling agent need to be used and can this be done while keeping the plate sterile for continued incubation? Do the ultrasound waves pass through the plate to other clusters of wells (in a way that would significantly affect the other clusters)? Is there a way to evenly apply ultrasound stimulation evenly across the whole plate?
This source works with nanoparticles and a 96-well plate, but details of the set up are not given. Unless I'm missing something?
This source appears to place a sponge in degassed water on the head of the 35 mm probe. Could this be a valid option for the cluster of wells (2x2 square) mentioned above?
For the purpose of transfection, a 6-well plate was put into an ultrasound water bath.
Ideally, I would like to use the probe (as opposed to a water bath) to keep the instrumentation consistent between in vitro and in vivo studies. As I mentioned, I'm very unfamiliar with sonication/ultrasound and its physics. Any assistance/advice on how to make an accurate and precise high throughput set up is appreciated.
Thank you in advance!
Relevant answer
Answer
Setting up a high throughput system for ultrasound irradiation/sonication, especially for your specific application of screening photosensitizers in cell cultures, requires careful consideration of various factors. Given your current understanding and equipment, here are some steps and considerations to help you set up an accurate and precise high throughput system:
Coupling Agent and Sterility:
  • A coupling agent is often used to facilitate the transmission of ultrasound waves from the probe to the sample. It helps minimize air gaps that can impede wave propagation. Sterile coupling gels or fluids can be used to maintain aseptic conditions for continued incubation.
  • Plate Positioning:
  • Based on the literature you mentioned, positioning the probe underneath the plate in a 2x2 square cluster seems to be a common approach. This can help focus the ultrasound energy on specific wells. The use of a coupling agent between the probe and the plate is crucial for effective transmission.
  • Plate Material and Ultrasound Transmission:
  • The choice of plate material can affect the transmission of ultrasound waves. Opt for plates that are compatible with ultrasound transmission, such as plastic or materials with low acoustic impedance. Glass plates might attenuate the ultrasound energy.
  • Even Ultrasound Stimulation:
  • Achieving even ultrasound stimulation across the entire plate can be challenging. The setup you mentioned with a probe underneath each cluster can help localize the energy, but it might not provide uniformity across the entire plate. Experimentation and optimization are key to finding the best setup for your specific needs.
  • Impact on Adjacent Wells:
  • Ultrasound waves can interact with neighboring wells. While some energy might pass through to adjacent clusters, the effect would likely be attenuated. However, it's important to monitor this potential interaction, especially if you're aiming for precise and controlled results.
  • Probe Configuration:
  • Since you're using a probe (as opposed to a water bath), consider whether modifying the probe configuration, such as using an array of smaller probes or optimizing the positioning, could help achieve more even energy distribution.
  • Calibration and Dosimetry:
  • Accurate dosimetry is essential for consistent results. Calibrate your ultrasound system to ensure that the intended energy levels are being delivered to the samples. This calibration can involve measuring the intensity, power, and frequency of the ultrasound waves at different points within the plate.
  • Temperature Monitoring and Control:
  • Ultrasound irradiation can lead to localized heating. Monitor and control the temperature during experiments to ensure that any effects observed are due to sonication and not temperature-induced changes.
  • Safety Considerations:
  • Ultrasound can induce cavitation, which may affect cells and compounds differently. Carefully consider safety protocols and ensure that the chosen parameters do not cause harm to the cells or compromise the integrity of your experimental setup.
  • Collaboration and Consultation:
  • Collaborate with experts in the field of ultrasound and sonication. Consult with colleagues, mentors, or specialists who have experience in similar setups to gain insights and advice.
Remember that achieving an accurate and precise high throughput system requires iterative experimentation and optimization. As you become more familiar with ultrasound physics and its interaction with your samples, you'll be better equipped to fine-tune your setup for optimal results.
  • asked a question related to Biochemistry
Question
4 answers
How often do you get new salts? What salts do you get "fresh" most frequently? All help is greatly appreciated. Thank you to everyone who reads or responds, hope you all have a fantastic day.
I am a Graduate Student at the University of Wisconsin Milwaukee. Our most recent electrophysiology experiment was going fantastic for about 3 weeks. Suddenly, every slice was very poor quality, and every cell we patched onto died within about two minutes. Even if you don't have an Electrophysiology background, all advice and input is appreciated. It's been two weeks since this problem first arose. We didn't alter any procedures, everything has been held constant. We don't know what could be causing the sudden change in slice quality.
So far we have three theories: DDH20 filter needs replacing, our glassware has somehow become contaminated, or our salts have gone bad.
The DDH2O water resistance reads about 14.8MOhms. We changed the filter about a year ago. From what I have read, DDH2O should be between 14 and 18 MOhms to ensure slice quality, so we think our water is fine.
Our glassware washing procedure is 3 rinses of tap water, 3 rinses of deionized water, and 1 rinse of DDH2O. We don't know how else we can safely clean the glassware for slice preparation. But we don't think this is the issue.
Our main theory is the salts have gone bad. Our lab is on the 4th floor, and it is usually quite warm and humid on our floor during the summer. Most of opened salt bottles we use were first opened between 2 and 7 years ago. (for example, our bottle of sodium phosphate monobasic monohydrate and our potassium chloride were both opened 6 years ago. Our magnesium chloride and sodium chloride were both opened 2 years ago)
We think with repeated opening of the salt bottles causes debris/water from the atmosphere to leach into the salts, resulting in a change in our solutions and causing the cell death we have been observing. Is this a real possibility? How often do you get new salts? What salts do you get "fresh" most frequently?
We checked the pH, the pH of our solutions is within .1 of what it should be. All help is greatly appreciated. Thank you to everyone who reads or responds, hope you all have a fantastic day.
Relevant answer
Answer
These salts cost very little, and I would simply throw them out and buy new ones. If the issue goes away, great, If not, then you have narrowed down the options very quickly with little cost. You could also buy a bottle of sterile water to rule out some issue with your production system (I think you can have contaminants that do not affect the resistivity).
  • asked a question related to Biochemistry
Question
2 answers
I am currently making bacmid and consequent baculovirus using pDEST8, pFastBac and 438a vectors and recently switched to DH10EmBacY cells instead of DH10Bac due to the added YFP signal to monitor transfection, etc.
I was wondering if there is any reason for me to not use either of the two competent cells interchangeably to make bacmid DNA?
Relevant answer
Answer
DH10Bac and DH10EmBacY cells are both used for the generation of recombinant baculoviruses for protein expression in insect cells. However, they have different characteristics and uses.
DH10Bac cells are used in the Bac-to-Bac system developed by Thermo Fisher Scientific. They are used to generate recombinant bacmids containing your gene of interest in a baculovirus genome. These bacmids can then be transfected into insect cells to produce recombinant baculoviruses.
On the other hand, DH10EmBacY cells are part of the EMBacY system, an alternative system for generating recombinant baculoviruses. This system also involves creating recombinant bacmids, but the process and the specific cells used differ from the Bac-to-Bac system.
In general, you should follow the protocol and guidelines provided for each specific system and use the recommended cells for that system. Interchanging the cells may not produce the desired results, as each system has its own optimized conditions and requirements.
  • asked a question related to Biochemistry
Question
1 answer
I observed an interesting pattern that resembles a "flattening" of the near-UV region of the absorbance spectrum of the rat intestinal mucus in the streptozotocin-induced model of sporadic Alzheimer's disease. I'm studying qualitative and quantitative alterations in mucus as there seem to be some changes in the gut-brain axis, intestinal redox homeostasis, and cell turnover ( , ). I observed a quite dramatic change in the number and responsiveness of goblet cells so the observed spectral shift may be driven by a component of mucus. Another possibility may be a component of bile, a peptide or bilirubin/biliverdin, or even complexation between bilirubin/biliverdin and some other molecule (e.g. I found a similar pattern in a publication by Klinke et al. who reported flattening of the biliverdin near-UV spectrum upon binding to bacteriophytochrome? ). Any ideas about what I may be looking at?
Thanks!
Relevant answer
Answer
The report of UV-visible wavelength range in quantitative tissue spectroscopy has increased over last few years. The optical spectroscopy findings quantitatively in the UV-wavelength region might be a focus not only for study of malignancy but also other ailments too. I do suggest to go through the publication- Quincy Brown J, et al. Curr Opin Biotechnol 2009; 20:119-1311.
  • asked a question related to Biochemistry
Question
2 answers
Dear community,
I am planning on conducting experiments for which I need to obtain a plasma-free platelet suspension from an aliquote of a platelet concentrate. Do you now any methodology/protocol that allows for washing without extensive cell activation? I need the cells to be a "functional" as possible.
Yours sincerely,
Michael
Relevant answer
Answer
Platelet washing is very much needed not only for research purposes but also for preventing some transfusional reactions like anaphylaxis, fever etc. Human platelet-rich plasma(PRP) prepared from blood collected in tri-sodium citrate might face its aggregation owing to lower ionic calcium concentration. To get rid of this, isolation and washing of platelet is done with acid-citrate-dextrose solution and the washed platelet cells are suspended in a buffer solution containing 2mM calcium ion solution.
  • asked a question related to Biochemistry
Question
5 answers
I would like to add BG-azide to cells for SNAP tag pulldown however I don't know whether it is going to be cell permeable. My instinct is to day yes it will be but neither me nor the supplier know whether that is true. I thought I would ask if anyone has tried something similar, even though that is unlikely... I have attached the structures in case that is informative
Relevant answer
Answer
Thanks Wolfgang, I appreciate the detailed answer!
  • asked a question related to Biochemistry
Question
6 answers
In a patient with desminopathy (mutation Thr341Pro DES in the heterozygous state) with the progression of the disease, we note signs and symptoms that are also characteristic of botulism: bradycardia, arrhythmia, AV blockade, a significant decrease in the average duration of motor unit potentials according to electroneuromyography, paresis and paralysis of the striated muscles, decreased sweating, paresis of the gastrointestinal tract, dry eyes, dry mouth, symmetry of neurological symptoms, hoarseness, impaired visual acuity, doubling of objects occurs, progressive muscle weakness. These signs and symptoms are characteristic of botulism, only when a case of desminopathy is detected, they proceed slowly.
Relevant answer
Answer
Your analogy is very interesting, dear colleague.
Although the main cause of any form of myofibrillar myopathy is a violation of the structure of the protein components of sarcomeres caused by genetic mutations, why not assume that due to mutations, the sensitivity of the postsynaptic membrane of myofibrils in myofibrillar myopathy to acetylcholine may also be impaired.
  • asked a question related to Biochemistry
Question
10 answers
Hi everyone.
I have protein with concentration of 0.6 mg/mL
The total volume of my protein is 4 mL.
Protein size is ~18 kDa
How can I convert my total protein concentration to micro molar?
Thank you.
Relevant answer
Answer
Philip G Penketh is correct.
The Dalton symbol Da, is also sometimes used as a unit of molar mass, with the definition 1 Da = 1 g/mol.
Therefore,
1Da= 1g/mol
1 kDa = 1000 g/mol
So, 18kDa = 18000g/mol
Now
18000 g ---- 1M ---- 1L
18g----------1M -----1ml
18000mg --- 1M ---- 1ml
The protein concentration is 0.6mg/ml.
Therefore,
0.6mg x 1M / 18000mg = 0.0000333M = 0.0333mM= 33.3μM.
So the protein concentration (in μM) as per the size (18kDa) and concentration (0.6mg/ml) is 33.3μM.
Best.
  • asked a question related to Biochemistry
Question
1 answer
There seems to be some consensus on the reaction mix for lactate dehydrogenase (LDH) activity assays, but the composition of the homogenization buffer (HB) differs wildly among studies. Maybe somebody can help me with the following three questions:
  • Imidazole in HB: used in ~50% of studies in concentrations of 50-150 mM. Alternatively TRIS-HCl is used (50-75 mM). Almost all studies use imidazole in the reaction mix. Are there benefits to using imidazole in the HB instead of TRIS-HCl?
  • Protease inhibitors (e.g., PMSF) in HB: used in few studies. Is this just for the workgroups that have more money to spend on chemicals?
  • Some studies use KCN in the reaction mix without any explanation (earliest study that I could find was Sidell et al. 1987). Why?
Any recommendations and/or tips regarding LDH assays are greatly appreciated. Thanks!
Studies polled for this question:
Chippari-Gomes AR, Gomes LC, Lopes NP, Val AL, Almeida-Val VMF (2005) Comp Biochem Physiol B Biochem Mol Biol 141:347–355. https://doi.org/10.1016/j.cbpc.2005.04.006
Driedzic WR, Fonseca de Almeida-Val VM (1996) J. Exp. Zool. 274:327–333. https://doi.org/10.1002/(SICI)1097-010X(19960415)274:6<327:AID-JEZ1>3.0.CO;2-Q
Gerringer ME, Yancey PH, Tikhonova OV, Vavilov NE, Zgoda VG, Davydov DR (2020) FEBS J 287:5394–5410. https://doi.org/10.1111/febs.15317
Heinrichs-Caldas W, Almeida-Val VMF de (2021) Fish Physiol Biochem 47:1759–1775. https://doi.org/10.1007/s10695-021-01000-0
Martinez ML, Chapman LJ, Grady JM, Rees BB (2004) J Fish Biol 65:1056–1069
Moon TW, Mommsen TP (1987) J. Exp. Zool. 244:9–15. https://doi.org/10.1002/jez.1402440103
Sidell BD, Driedzic WR, Stowe DB, Johnston IA (1987) Physiol Zool 60:221–232. https://doi.org/10.1086/physzool.60.2.30158646
Singer TD, Ballantyne JS (1989) J. Exp. Zool. 251:355–360. https://doi.org/10.1002/jez.1402510312
Relevant answer
Answer
If the assay is for serum LDH activity one has to be careful during collection of blood sample so that there is no hemolysis of RBCs as they have higher activity of LDH. In that case there might be false higher activity of serum LDH. This might be taken as recipe while assaying serum LDH activity.
  • asked a question related to Biochemistry
Question
2 answers
Hello everyone!
I recently received invitations to contribute an article to the current issue of the Journal of Brilliant Engineering (BEN) and Chemistry and Biochemistry. Can anyone with experience or knowledge about these journals confirm their originality?
Thank you.
Relevant answer
Answer
Rahim,
They're certainly original.
But I've read the 'mission statement' of BEN, and can find no compelling reason to submit work there.
Indeed, here's one reason to not submit to the journals run by Prof. Aydin:
I quote from:
"An artificial intelligence based English proof system checks the language of the paper."
Mmm.
Call me old-fashioned, but it shouldn't be too hard to find someone to read a submission who is a native english speaker.
  • asked a question related to Biochemistry
Question
3 answers
Hey all, I'm very interested in understanding more about the biochemistry behind all of the wash buffers that go into the streptavidin affinity purification protocol seen here (https://static1.squarespace.com/static/5617d7d8e4b09f2fdf34baa6/t/5fe2631ba775596e136098b6/1608672031257/Cho+NP+2020.pdf pg 3990, step #38).
I understand this is to help reduce non-specific interactors from being pulled down with the biotinylated proteins; however, I'm more curious to understand the necessity of the 2M Urea vs sodium carbonate vs KCl, etc....looks like previous iterations of this protocol have not used sodium carbonate and KCl buffers but I can't find substantiation for why they were added.
Thanks in advance!
Relevant answer
Answer
Just note that sodium carbonate Na2CO3 is NOT a buffer. To work as buffer, you'd need some acid to complement it (e.g. H2CO3 or NaHCO3). This is "just" alkaline solution used to dissociate some interactions.
For example, I have used sodium carbonate when I have fractionated proteins associated with cell membrane few years back.
  • asked a question related to Biochemistry
Question
11 answers
biofilm and quorum sensing genes of E.coli, not used drug but chemical material (Thiophene)
Relevant answer
Answer
Lack of aseptic techniques
  • asked a question related to Biochemistry
Question
2 answers
I basically have normalized fluorescence data generated from a series of protein dilutions. I need to calculate K, K should be the same in all dilutions, thus the need for a global fitting approach. I have been using XLfit, but would like to move into a script format. Thanks in advance! Thus far, I found the package renz, but to my understanding, it does not accomodate for global fitting. Thanks in advance!
Relevant answer
Answer
COPASI: Biochemical System Simulator.
free download from copasi.org
  • asked a question related to Biochemistry
Question
4 answers
Specifically interested in purine derivative as a cation of ionic liquid.
Any relevant reading suggestions are highly appreciated. Thanks.
Relevant answer
Answer
Yes, there are several purine-based ionic liquids that have been described in the scientific literature.
Examples of purine-based ionic liquids include 1-butyl-3-methylimidazolium adenosine monophosphate ([BMIM][AMP]), which was first synthesized and characterized in 2011 (reference: "Synthesis and characterization of a new purine-based ionic liquid: 1-butyl-3-methylimidazolium adenosine monophosphate" by L. Zhao et al., in Tetrahedron Letters, vol. 52, p. 1130-1133, 2011).
Other examples of purine-based ionic liquids include 1-butyl-3-methylimidazolium guanosine ([BMIM][GMP]) (reference: "Synthesis and characterization of a novel purine-based ionic liquid: 1-butyl-3 -methylimidazolium guanosine" by L. Zhao et al., in Journal of Molecular Liquids, vol. 170, p. 63-66, 2012) and 1-allyl-3-methylimidazolium hypoxanthine ([AMIM][Hpx]) (reference : "Synthesis and Characterization of a New Purine-Based Ionic Liquid: 1-Allyl-3-methylimidazolium Hypoxanthine" by L. Zhao et al., in Chemical Research in Chinese Universities, vol. 32, p. 157-161, 2016) .
  • asked a question related to Biochemistry
Question
5 answers
1-The conditions of the relationship between the pigment (Pyocyanin) with Gold (Au Nanoparticles)?
2-Dose the pigment (Pyocyanin) work as Reducing agent or not?
3-Dose the pigment (Pyocyanin) endures high temperatures or not?
Relevant answer
Answer
1.Pyocyanin can be adsorbed on the surface of gold nanoparticles. It does not oxidize or reduce gold. Gold is oxidized with potassium cyanide or aqua regia.
2. When interacting with gold ions, Pyocyanin (quinone) acts as an oxidizing agent.
3. Pyocyanin melting point 133 0С.
  • asked a question related to Biochemistry
Question
4 answers
One example would be MBP and MBP-74, an interaction which can be disrupted by maltose when it binds MBP.
Relevant answer
Answer
Figure 1 shows Imatinib disrupts the protein-protein interactions of Bcr-Abl with some, but not all interaction partners.
  • asked a question related to Biochemistry
Question
2 answers
Hansen Solubility Parameters full sets (dD, dP, dH and Ro) for
1) Cholesterol CAS#[57-88-5], and/or
2)Cholesterol Monohydrate with CAS#[5808-12-8] are needed.
Any or both experimental or calculated in HSPiP values are interesting.
Old published values (20.4, 2.8, 9.4 and 12.6) are off interest.
Relevant answer
Answer
HSPiP software is not free. I am interested in how dose it work.
  • asked a question related to Biochemistry
Question
4 answers
I am currently working on Co-IP of NRIP and MyoD by overexpressing both proteins in HEK293T cell. The question is I cannot get a specific band of MyoD. For my input, I think the protein level is too low to be detected so I will increase the protein amount to run a western blot. However, several bands appear when detected using MyoD antibody after co-IP. The band most likely MyoD is pointed with a blue arrow. Some of my colleagues say it may be endogenous MyoD or protein with similar conformation. Does anyone face a similar situation when working with this protein? or any reference paper suggested for studying this protein. Thank you.
Relevant answer
Answer
Hello Hui Yee you could also try to use PICO to detect your MyoD. They claim to be much more sensitive compared to Co-IP/WesternBlot workflows. Here is an explanation of the technology:
  • asked a question related to Biochemistry
Question
7 answers
Can alpha glucosidase and alpha amylase work at room temperature and at 37*C? If so why? The specifications for these enzymes are that they work at 37*C. But number of papers have modified their protocols 25*C. How can it work at both room temperature and at 37*C?
Relevant answer
Answer
Indumati Sharma Did you figure out the working temperature for this? I am going to perform this and am quite confused which temperature should I prefer? And can you please also specify the blank you used?
  • asked a question related to Biochemistry
Question
16 answers
Hello
I'm sure this question has been asked a lot but the protein I am purifying is not as clean as I would like and all the potential solutions I have read about have not worked for me.
I am purifying a protein that forms inclusion bodies. The construct is 14x His tag - Tev cleavage - protein of interest. I use BL21 Star (DE3) cells and TB media for growing the bacteria and induce at OD 0.6 for 4 hours at 30 degrees.The purification protocol is based on a previously published paper. Following sonication, lysis buffer and washing the pellet the protein is extracted from the inclusion bodies using 6M Guanidine Hydrochloride, applied o/n at room temperature to a Ni NTA agarose column. It is then eluted in 4M Guanidine. I then pass it through a RP-HPLC. As you can see by the attached image although I am getting a high amount of protein it has a large smear which I am not sure if it is contaminants or degredation. I overloading with sample when trying to judge purity but I think it's better to get an accurate representation of what’s going on rather than kid myself I am working with a pure sample.
The HPLC makes no difference so I think optimisation on the Ni-NTA purification is needed but nothing thus far has worked, I have tested different induction temperatures, leaving it on the Ni column at both 4 degrees and for a few hours (rather than O/N at RT like the protocol says), protease inhibitors and washes/step-wise elutions with different concentrations of imidazole (as in attached file) - yet none of this has worked. I've even thought about making the His tag smaller (14x seems quite large, but I cant see how this would impact purity other than needing more imidazole for elution).
If anyone has comments on the purity and has any tips that I have not tried it would be greatly appreciated!
Thanks!
Relevant answer
Answer
Thought I would update this as it might be useful for others in the future, we managed to get the protein quite pure and remove most the contaminants by testing loads of different things (compare image attached to original image when I asked the question).
Things we tested:
Protease inhibitors - we tried the protease inhibitor cocktail, PMSF, EDTA and different combinations of these and the protein always looked the same
IMAC resin - We tried Ni-NTA resin from a few different companies including suggested here and this also made no difference. Co-NTA was no good as we purify with 10mM DTT, and resins marketed as highly DTT also did not make a difference. Plain old Ni-NTA agarose from Qiagen worked just fine.
Cell line - This is what made a huge difference, when we used BL21 LysS - this massively reduced the impure smear we got compared to the BL21 Star DE3 ceolls. Other cells we tested were BL21 AI and Rosetta cell lines, these helped but the BL21 LysS was good. BL21 LysE was also good and removed nearly all contaminants but at a cost we got hardly any protein, so the LysS line was a good balance.
We could then wash off lots of the remaining contaminants with step wise imidazole gradient washes before final elution. We then did SEC under denaturing conditions (6M Guanidine) as a final clean up step.
Hope this helps anyone else struggling to get their protein pure.
  • asked a question related to Biochemistry
Question
2 answers
In speculating about plant migration from the oceans to land, I wondered if any plant species went back into the ocean. Since that is unlikely, this question arose to explain why.
In a study of plant genetics, this area of plant diversification is likely amazing, because of how the gene sequences are accessed and how complexity developed as new genes and biological systems.
Relevant answer
Answer
Tristan Beckwith my dear friend, there is evolution relationship between plants and algae. primitive plants are evolved from the these algae. the non vascular plants that are dominate in early earth are evolved from green algae. after, vascular plants are dominate on earth. that is brief history about plants evolution. in simple we can say freshwater green algae are ancestors of plant. After, these plants able to adapted to the various environment system due to their well develop vascular systems. some of them are adapted to the saline water. the some plants species which is able see in ocean belongs to the angiosperms.
Please find about much details, If i am wrong please correct me because knowledge of science is update in day by day. therefore thing I know can be bit change within time. therefore find about deeply about your matter.
Best wish Poornima Nadun
  • asked a question related to Biochemistry
Question
4 answers
Hello every body I want to assay lipase but I cant dissolve PNPP in isopropanol could you please help me in this way?
Relevant answer
Answer
Ramesh K Jha Can you guide me how to prepare p-NPA solution for extracellular esterase activity assay? I read different papers, they are ether using methanol, isopropanol, or a 1:1 mixture of isopropanol and acetonitrile. Which could be the be solvent to perform this assay?
  • asked a question related to Biochemistry
Question
5 answers
We are doing a research on Biofilm formation of Bacteria, for knowing each isolate strong, moderate or weak, by calculations tables of Standard deviation, Variance and Cutoff (Ct) etc… and with the help of Microsoft Excel but the results in the program differ from the hand written and don’t know the best way to calculate and compare the results, any help will be very appreciated, Thank you
Ali
Relevant answer
Answer
Dear Ali,
There are probably many ways you can differentiate bacterial biofilm formation and I'd suggest either following the procedure described in a research paper working on the species or strains you are interested in, or by using your own criteria. We characterise our air-liquid interface biofilms using a combined biofilm assay measuring biofilm strength, attachment levels and total growth - which means we can describe 'biofilms' using one or all three quantitative measurements. We tend not to use 'no biofilm' control strains, as many biofilm-formers produce such weak and poorly attached structures it is not clear when a 'no biofilm' becomes a measurable one (though we can use a sterile microcosm/culture as the negative control if appropriate).
I can't comment on your problems with Excel, but one simple way is to graph the mean results from your experimental replicates, and divide these into quartiles about the median (i.e., from the minimum values to Q1, from Q1 to the median, from the median to Q3, and from Q3 to the maximum value). You could then simply state that no or poor biofilm formers are min - Q1, biofilm formers are Q1 ≤ Q3, and good biofilm-formers are Q3 - max (if you liked, you could divide your bacteria into no biofilm, poor biofilms, good biofilms and very good biofilms – the number of categories is up to you, but you can't have lots when you have relatively few bacteria to distribute across these categories).
There are statistical tests you could use, and assuming that data (or residuals) are Normally distributed, you could say that no or poor biofilms are not significantly different to a no-biofilm control, etc., but this gets messy because it is hard to know when a very poor biofilm is effectively no biofilm, etc.
Andrew
  • asked a question related to Biochemistry
Question
10 answers
In the Column treatment of aqueous solution of Chromium Removal by Adsorption
Relevant answer
Answer
Treating bark with acid (HCl) first and then with base (NaOH) second is a common method for isolating plant compounds for analysis or extraction, such as tannins, lignin, and cellulose. This method is known as acid-base treatment or A/B extraction.
The reason for this order of treatment is that acid treatment hydrolyzes and breaks down the plant cell walls, making the cell contents more accessible for extraction. Acid treatment can also help to remove impurities, such as pigments and waxes, from the sample.
After acid treatment, the sample is then neutralized with a base, typically sodium hydroxide (NaOH), to restore the pH to a neutral or slightly alkaline state. The neutralization step is important to prevent the acid from interfering with subsequent analytical techniques or reactions.
The use of acid and base treatments in this order allows for selective extraction of different plant compounds based on their solubility and chemical properties. For example, tannins are more soluble in acidic solutions, while lignin and cellulose are more soluble in basic solutions.
Treating the bark with base first would result in a saponification reaction, where the ester linkages in the plant compounds would be hydrolyzed by the base, resulting in a loss of their original structure and properties. Therefore, treating the bark with acid first and then with base is the preferred order for A/B extraction of plant compounds from bark.
  • asked a question related to Biochemistry
Question
3 answers
Bio-sensor development demands a variety of fields in cooperation. However, this breakthrough would not result in the case of doing research with a single attitude. I am calling researchers from biology or biochemistry, genetics, or related science to come into the discussion. Let me know if somebody has any knowledge in this field.
#Bio-sensors #Biochemistry #Genetics
Relevant answer
Answer
More information of how to use receptor for biosensor application!
  • asked a question related to Biochemistry
Question
2 answers
My aim is to detect nascent protein synthesis by click-chemistry of L-homopropargyl-glycine-labelled proteins with biotin azide and detection by western blotting with fluorescently-conjugated streptavidin. So far, I have failed to detect labelling above what looks like background biotinylation. I'm using biotin azide from Biotium (Cat# 92167), but I've noticed there different biotin azide molecules with different molecular weights. I was wondering if there were differences in their applicability for copper-catalysed click-chemistry. Any help with troubleshooting this would be appreciated. Cheers!
Relevant answer
Answer
The spacer length should not affect reactivity. These molecules rather differ in the distance between biotin and the reactive end. However, for a good interaction between biotin and streptavidin, you need a certain spacer length, as the biotin needs to fit into one of the pockets of the streptavidin tetramer. To this end, I'd recommend at least an added C6 chain, as it is the case in biotin-X-Osu, or 2 or more PEG molecules. So better make sure the biotin-azide you are referring to has this spacer.
If your reaction is taking place in a biochemically active system, I'd strongly recommend to use copper free click chemistry instead of the azide/alkyne system, like DABCO (as Satyendra Singh already suggested), and another suitable diene like e.g., a tetrazine.
This brochure by Iris Biotech might give you some ideas on biocompatible click chemistry: https://media.iris-biotech.de/flyers/IF4_1_Click_Chemistry.pdf, theer are some DBCO and Tz labeled molecules which might help you succeed. A nice chart with some more exotic click structure is in this paper:
TMTHSI, a superior 7-membered ring alkyne containing reagent for strain-promoted azide-alkyne cycloaddition reactions; J. Weterings, C. J. F. Rijcken, H. Veldhuis, T. Meulemans, D. Hadavi, M. Timmers, M. Honing, H. Ippel, R. M. J. Liskamp; Chem. Sci. 2020; 11: 9011-9016. https://doi.org/10.1039/d0sc03477k
HTH,
Wolfgang
  • asked a question related to Biochemistry
Question
6 answers
Aside from inhibiting protease activity (not effectively working), I read some papers about doing modifications to the antibodies:
using unnatural amino Acids
using mPEG
But, are there better options, more commercially applicable?
Relevant answer
Answer
If you are talking about using the antibodies as the affinity matrix (such as in immunoaffinity chromatography), I would suggest that the way to avoid damage to the antibodies from proteases in a crude extract is to save the immunoaffinity chromatography for the last step in the purification, at which point the protease activity from the crude extract should be greatly reduced. Begin the purification with ion exchange, hydrophobic interaction, and/or gel filtration chromatographies. Use a protease inhibitor cocktail when making the crude extract.
  • asked a question related to Biochemistry
Question
5 answers
Please spread the word: Folding at Home (https://foldingathome.org/) is an extremely powerful supercomputer composed of thousands of home computers around the world. It tries to simulate protein folding to Fight diseases. We can increase its power even further by simply running its small program on our computers and donating the spare (already unused and wasted) capacity of our computers to their supercomputation.
After all, a great part of our work (which is surfing the web, writing texts and stuff, communicating, etc.) never needs more than a tiny percent of the huge capacity of our modern CPUs and GPUs. So it would be very helpful if we could donate the rest of their capacity [that is currently going to waste] to such "distributed supercomputer" projects and help find cures for diseases.
The program runs at a very low priority in the background and uses some of the capacity of our computers. By default, it is set to use the least amount of EXCESS (already wasted) computational power. It is very easy to use. But if someone is interested in tweaking it, it can be configured too via both simple and advanced modes. For example, the program can be set to run only when the computer is idle (as the default mode) or even while working. It can be configured to work intensively or very mildly (as the default mode). The CPU or GPU can each be disabled or set to work only when the operating system is idle, independent of the other.
Please spread the word; for example, start by sharing this very post with your contacts.
Also give them feedback and suggestions to improve their software. Or directly contribute to their project.
Folding at Home's Forum: https://foldingforum.org/index.php
Folding at Home's GitHub: https://github.com/FoldingAtHome
Additionally, see other distributed supercomputers used for fighting disease:
Relevant answer
Answer
Vahid Rakhshan I will definitely spread the word about this amazing initiative. It's great to know that we can contribute to such a noble cause by simply utilizing our excess computer power. Thank you for bringing this opportunity to my attention. Let's join hands in making a difference in the fight against diseases.
  • asked a question related to Biochemistry
Question
4 answers
When staining with hematoxylin and eosin of a muscle biopsy from a patient with T341P desminopathy, we observe accumulations of inclusions similar to nuclei (arrows in figures 1 and 2, x280). And outside of these accumulations - adipose tissue, which used to be muscle tissue. There are no such massive accumulations of inclusions in adjacent muscle fibers. We assume that clusters of inclusions are not nuclei? Figure 2 is the inverted figure 1.
Relevant answer
Answer
Dear Geir Bjorklund, Duc M. Hoang, John Hildyard, thank you very much for your answers and recommendations!
  • asked a question related to Biochemistry
Question
5 answers
For the protein and ligand preparation process what kind of structure should ı choose and what is the reason of that
Relevant answer
Answer
For receptors, crystal structures are in a higher position but if there wasn't a crystal structure a good structural model could be the next option. For ligands, the PubChem ligands could be good choices.
  • asked a question related to Biochemistry
Question
18 answers
We have a Flask that contains broth, and we want to inoculate it with Bacteria inoculum, Can we simply take a touch by the loop or by micropipette?
Relevant answer
Answer
Kseniya Kondrasheva Nikhita Madhav Chambhare Thank you very much, The Goal is Pyocyanin production, we tried 1:100 and yes by micropipette because the loop carry small volume
  • asked a question related to Biochemistry
Question
8 answers
I made several reactions with 3,5-Dinitrobenzoyl chloride. When I looked at similar reactions for this substance, no heat was ever given in the reaction (even though heat increases the yield).
The interesting thing is that this substance turns black as soon as the heat is given to the reaction environment. Due to the fact that, theoretically, the polymerization of this material is impossible. Do you know the reason why they do not give heat to this material?
  • asked a question related to Biochemistry
Question
5 answers
I need to find a mutant allele of my protein that is defective for homo-oligomerization, but can still bind a separate protein (heterodimer formation).
I think I will do alanine scanning mutagenesis of my protein.
However, my assays are very low throughput and I must assay each mutant protein individually. I simply cannot assay every single mutant allele in the time that I have.
I do not have any structural information. And Alphafold cannot solve the structure.
I am considering:
1. Mutate 3 residues at once. Is this too much? Will I simply destabilize my protein?
or
2. Only mutate charged residues, as oligomerization fequently relies on charged residues from my research. Does this seem incorrect?
Or is there better mutagenesis approach than alanine scanning?
Relevant answer
Answer
It is not clear what assay do you have to test whether the homodimer interaction is impaired while retaining the heterodimer interaction. If you have an yeast-based assay (2 hybrid), you can mutagenize (use error-prone PCR conditions) your template, and then co-transform the linear PCR product and a wild-type plasmid (yeast powerful homologous recombination will recombine and you do not have to clone all your PCR products). After the first round of identifying mutants that disrupt the homodimer, you can test them against binding to the heterodimer. You can pick the colonies and identify the mutants by sequencing (attached reference with conditions).
Hope it helps,
a
  • asked a question related to Biochemistry
Question
5 answers
I am doing my thesis for the last season of college and ı need to dock to ligand and protein for better binding affinity ı decided to combine the two ligand for moluecular docking so how can I combine the 2 ligand into 1 receptor molecular docking.
Relevant answer
Although theoretically, it may be best to combine both ligands within one docking simulation but is not really possible to do it directly with most of the free software.
However, all hope is not lost. The other way around it is to do it independently. Meaning, dock the first ligand first, then the second one.
You just need to make sure the parameters (#runs, grid box position, algorithm, etc.) remain consistent for both docking simulations.
Finally, compare both of the results based on the binding energy, population cluster, and/or formed interactions. The best one wins! (sorta)
Hope this help. All the best.
  • asked a question related to Biochemistry
Question
5 answers
Usually, plant phenols are best extracted with solvents like methanol and ethanol. But I would like to know if the extraction is carried out with water, how much usually is the efficiency of that extraction??
Relevant answer
Answer
alcohols like methanol and ethanol are the best solvents to extract phenolic compounds. but these can also be extracted with water but it depends on the part of the plant drug used (leaves, flowers, roots ........) you must also do a bibliographic research on the optimization of the various extracts used on your plant in order to obtain the best yield in polyphenols. good luck
  • asked a question related to Biochemistry
Question
8 answers
Hi everyone!
I project to measure levels of total phenolic compounds in peppers following the method of the Folin-Ciocalteu's reagent, and I am going to calculate them by reading absorbance using gallic acid as a standard.
Nevertheless, I am confused about what wavelenght should be setted: while some authors read absorbance at 765 nm (Dogan et al., 2018; Kupina et al., 2018); others read it at 760 nm (Ghasemnezhad et al., 2011); 750 nm (Toledo-Martín et al., 2015; Lwin et al., 2022) or even 725 nm (Vega-Gálvez et al., 2009).
I would be grateful if someone could give me some light about this.
Thanks to everyone in advance.
Pablo
Literature cited:
Dogan, A.; et al. (2016). Comparison of pesticide-free and conventional production systems on postharvest quality and nutritional parameters of peppers in different storage conditions. Scientia Horticulturae 207: 104-116.
Ghasemnezhad, M.; et al. (2011). Variation in phenolic compounds, ascorbic acid and antioxidant activity of five coloured bell pepper (Capsicum annum) fruits at two different harvest times. Journal of Functional Foods 3: 44-49.
Kupina, S.; et al. (2018). Determination of total phenolic content using the Foling-C assay: Single-Laboratory validation, First Action 2017.13. Journal of AOAC International 101 (5): 1466-1472.
Lwin, H.P.; et al. (2022). Perforated modified atmosphere packaging differentially affects the fruit quality attributes and targeted major metabolites in bell pepper cultivars stored at ambient temperature. Scientia Horticulturae 301: 111131.
Toledo-Martín, E.M; et al. (2015). Application of visible/near-infrared reflectance spectroscopy for predicting internal and external quality in pepper. Journal of the Science of Food and Agriculture 96: 3114-3125.
Vega-Gálvez, A.; et al. (2009). Effect of air-drying temperature on physico-chemical properties, antioxidant capacity, colour and total phenolic content of red pepper (Capsicum annuum, L. var. Hungarian). Food Chemistry 117: 647-653.
Relevant answer
Answer
In short it is basically as said here by the Sigma-Aldrich site (https://www.sigmaaldrich.com/NL/en/product/sial/f9252) “Addition of the phenol reagent generates chromogens that give increasing absorbance between 550-750nm. Normally, absorbance at the peak (750nm) or shoulder (660nm) are used to quantitate protein concentrations between 1-100 mg/ml while absorbance at 550nm is used to quantitate higher protein concentrations.”.
Or as said in one of the most cited papers when it comes to Folin–Ciocâlteu reagent (FCR) “Because of the breadth of these peaks and the fact that other components in biological samples do not absorb in this region, analysis can be carried out at a wide range of wavelengths, 760 nm generally being chosen for FC.” (Analysis of total phenols and other oxidation substrates and antioxidants by means of folin-ciocalteu reagent).
Although the overall message is that every wavelength between 750-765 nm is fine, personally, I would go for 765 nm. This is based on the official JAOAC method: https://academic.oup.com/jaoac/article/102/1/320/5658204 but as said every wavelength is fine as long as you clearly indicate in your method which wavelength you choose (and on which paper you based that choice).
Best regards.
  • asked a question related to Biochemistry
Question
2 answers
In the following article's ESI, it seems that in situ decomposition of 2,5-Dihydroxy-1,4-benzoquinone (DHBQ) lead to the formation of oxalate under hydrothermal conditions.
Can someone explain the mechanism/pathway behind it?
Ref.: Science 374, 1464 (2021)
Please correct me if I am wrong by any chance.
Thanks in advance.
Relevant answer
Answer
You may also see the retrosynthesis pathway from scifinder
  • asked a question related to Biochemistry
Question
7 answers
Hi All,
I am trying to use a technique used by previous graduate students that involves growing yeast in SC media -Uracil buffered to pH 6.75. All the literature published from our lab states that the growth media was buffered to pH 6.75 with 100 mM HEPES.
My problem is that I have tried multiple times to buffer the solution and have not gotten a stable pH through the growth cycle.
Initially, I prepared 1M HEPES buffer pH 6.75 and diluted it 10X to reach 1X media concentration, however upon dilution to 1x, the pH drops significantly. I then prepared the 1X media with the 100 mM HEPES, and adjusted the pH with NaOH and KOH (two separate attempts), however the pH was not stable through during growth and acidified significantly. I then buffered the 1X media with a pH 6.5 and pH 7.5 100 mM HEPES buffer, and the same issues persisted.
Does anyone have any insight into what I an missing here?
Relevant answer
Answer
Apologies,
I am on mobile as I am out of the lab, so the spell check is lacking.
We utilize a drug which is toxic to S. c. and is transported by our protein. It is expressed in the plasma membrane and its topology is such that it transports the drug into the cell, leading to a growth delay in inducing media that is proportional to the isoforms ability to transport the drug. (We occasionally pair this with tritiated drug uptake and the correlation is actually quite striking. I can send the references to you privately if researchgate has the functionality.)
The transport is highly dependent on the membrane potential, so when working at a low concentration of the drug, no isoform distinctions are seen without an increase in membrane potential.
  • asked a question related to Biochemistry
Question
18 answers
Hello everyone. I need a little help here.
can we use essential oils to reduce chlorinous odor in a hypochlorous acid solution without decreasing free chlorine or pH value? any toughts and propositions are much appreciated
Relevant answer
Answer
The reduction in odor is likely due to chlorination of essential oil constituents. Why are you using hypochlorite? As biocide pH should be ~6.5. Are you measuring av Cl ?
  • asked a question related to Biochemistry
Question
3 answers
i am a medical student, however i would like to learn molecular biology to the extent that makes me comfortable with putting a hypothesis, a theory or possible treatment and testing this with the help of a specialized research team and as such, i want to reach to the point where i fully understand modern molecular biology and specially the LAB TECHNIQUES. What is the process that i should follow in order to achieve this.
keeping in mind that i have near full access to springerlink, RSC and online wiley libraries
Relevant answer
Answer
I suggest you to start with the books "Molecular Biology of the Cell" by Bruce Alberts and "Molecular cloning: A laboratory manual" by Tom Maniatis. The first is an easy read which will help you to gain overall understanding of molecular biological processes and the second will provide a detailed knowledge regarding lab techniques used in this field.
  • asked a question related to Biochemistry
Question
3 answers
I am trying to express and purify nanobodies from a nanobody library in E. coli. I am using a pBAD vector to which I added the pelB sequence for periplasmic localization. The nanobody is fused to YFP with a His tag.
I induce the expression with arabinose and I can see on an SDS-PAGE gel that the nanobody is being expressed (although the expression seems to be lower than for some of the other proteins that I am expressing using the same vector), but I am having trouble with the extraction and purification steps.
I have tried to extract the nanobodies using lysozyme with PMSF following a protocol that usually works for me and I have also tried the osmotic shock protocol ( ) but the nanobodies seem to be stuck in the cell pellet even after lysis.
I do not have any experience with nanobodies so maybe there is an important step in the protocol that I am missing or not doing properly. I would appreciate tips or good protocols for expressing and extracting nanobodies.
Relevant answer
Answer
Maybe I was not clear enough in my original post, but I always analyse the supernatants and pellets by SDS-PAGE after osmotic shock/lysozyme extraction and the protein is present in the pellet, I am just not sure how fix that.
  • asked a question related to Biochemistry
Question
4 answers
Dear all,
I got results after performing an enzyme kinetics with 4 different substrate the kcat of B and D is respectively two times lower than A and C. However, I am seeing the higher Km of A i.e 174 nM as compared to B i.e 54 nM. Can anyone please suggest me how to rationalize this result ? The Kd value however, is always doubled for A and C when compared to B and D respectively. Below are the data attached for your reference. How could it be explained the research paper ?
Thank you
Relevant answer
Answer
The different kinetic constants for the different substrates reflect the chemical differences between the substrates, including the strengths and types of their interactions with the enzyme active site and differences in the activation energy of the reaction. The differences in the kinetic constants between the substrates in this case are actually quite minor, and could easily be within the range expected for experimental variation.
  • asked a question related to Biochemistry
Question
1 answer
Does the stability change with stock concentration?
Relevant answer
  • asked a question related to Biochemistry
Question
5 answers
I have preformed a dehyrogenase assay on an arid Negev soil. the calibration curve highest values were about 0.5 but the values for some of my samples were even 2 or 3 times that based on the protocol Alef, K. "Dehydrogenase activity." Methods in applied soil microbiology and biochemistry. London: Academic Press, 1995. 228-231.‏
also the results were much higher on the FDA protocol based on:
Adam, G. and H. Duncan. 2001. Development of a sensitive and rapid method for the
measurement of total microbial activity using fluorescein diacetate (FDA) in a range of
soils. Soil Biol. Biochem. 33:943-951.
the curve values were 0.05 to 0.1 and the values were 0.2 -0.7. is this normal for soils? some of them were poluted 5 years ago
Relevant answer
Answer
i will send you results it thats ok
  • asked a question related to Biochemistry
Question
5 answers
I want to visualise secondary structures of multiple proteins aligned (something similar to this figure). Any recommendations?
Thanks in advance
Relevant answer
Answer
try this one:
all the best
fred
  • asked a question related to Biochemistry
Question
2 answers
It requires about 5.3 kcal/mol (or 8 kBT) of energy to break one phoshodiester bond of DNA. How do these enzymes cut the DNA only by using thermal energy and not ATP? I am only considering the ATP-independent restriction enzymes (Type II). How do these enzymes manage to generate the necessary energy? I couldn't find the exact mechanism with energetics of restriction enzymes cleaving DNA. Please provide me any relevant references.
Relevant answer
Answer
No, the standard free energy of hydrolysis of the phosphodiester bond in DNA is -5.3 kcal/mol. It requires energy to forge a phosphodiester bond, while to break one requires only enough energy to overcome the activation energy barrier, which is lowered by enzymatic- , acid- or base catalysis. Under physiological conditions, hydrolysis is further facilitated by the high water concentration.
  • asked a question related to Biochemistry
Question
2 answers
Hi all,
A little question about PyMOL: Assuming that I know a coordinate, and now I want to see which atom this coordinate belongs to in the structure, what select command should I use? Namely, "select selection_name," + what?
I didn't find any materials on this.
Also, attached is the selection algebra of PyMOL (https://pymolwiki.org/index.php/Selection_Algebra).
Does anyone know why they say x<12.3, etc.?
Thank you very much!
Relevant answer
Answer
In command: select selection_name," + what. + what shows selection-expression (select selection-name, selection-expression)
For selection-expression use options of first column under coordinates operator. The number of selected atoms gets adjusted after varying the number upto the provided limit (12.3) for the selection.
For e.g.: select selection_name, X < 12.3
  • asked a question related to Biochemistry
Question
1 answer
3 dpf larvae of zebrafish will be used to evaluate the body burden concentration of a compound at different period of time.
Relevant answer
Answer
Whether spectrophotometry is a suitable method depends on whether the substance you are looking for has an "optical signature". That is, whether it has characteristic absorption peaks. Some substances can be detected indirectly by color reactions. Whether such methods can be used on live/whole fish, I do not know.
  • asked a question related to Biochemistry
Question
4 answers
Hi
The question is: Determining hexokinase activity in rat brain where 10 µl of a 10% (100 g/L) brain homogenate (containing hexokinase) is mixed with 1990 µL of reaction medium (containing the substrate glucose and the coenzymes ATP and NADP+ and the auxiliary enzyme glucose-6-phosphate dehydrogenase). The reaction mixture is placed in a cuvette (light path = 1 cm) in a spectrophotometer at 37°C, where the hexokinase reaction can be seen as an increase in absorbance at 340 nm. The reaction proceeds at a constant rate during the measurement period, and during 10 min a total increase in the light absorption at 340 nm of 0.10 is recorded. The absorption coefficient of NADPH = 6300 x M-1 x cm-1
I need to find the enzyme activity of hexokinase in µmol/min/g. I don't understand how I need to interpret the first line (10 µL of a 10 % (100 g/L) brain homogenate) - and how should I use these numbers?
Thanks in advance!
Relevant answer
Answer
10 µL of a 100 g/L (= 100 µg/µL) homogenate contains 1000 µg = 1 mg = 0.001 g.
That will be the number in the denominator when you divide by the number of g to calculate µmol/min/g.
The change in absorbance of 0.1 unit is converted to the concentration of NADPH formed using the extinction coefficient of 6300 M-1cm-1 and the Beer-Lambert Law: absorbance = extinction coefficient x pathlength x concentration.
An absorbance change of 0.1 for NADPH with an extinction coefficient of 6300 M-1cm-1 in a 1-cm pathlength cuvette corresponds to a NADPH concentration of 0.1/(6300 x 1) = 1.587 x 10-5 M = 15.87 µM.
This amount of NADPH was formed during 10 minutes, so the rate of NADPH formation was 1.587 µM/min. This occurred on a reaction volume of 2 mL, so it can also be expressed as 1.587 µmoles/(L-min) x 0.002 L = 0.00317 µmoles/min.
This happened when you used 0.001 g of material, so the specific activity was (0.00317 µmoles/min)/(0.001 g) = 3.17 µmoles/min/g.
  • asked a question related to Biochemistry
Question
7 answers
A patient with desminopathy survived Covid-19 six months ago without pneumonia, but with a temporary loss of smell and taste. After Covid-19, we note an accelerated progression of desminopathy, penetration accelerates, new muscles are quickly involved in the pathological process, muscle mass decreases, and heart function worsens. Perhaps the infection or its consequences are somehow connected with the mechanism of progression of desminopathy?
  • asked a question related to Biochemistry
Question
4 answers
Hello!
I was wondering if anyone could point me to any references that show that with increasing oligo length, there is a significant reduction in ssDNA generation efficiency. Im specifically curious at what point heating to separate dsDNA becomes ineffective and some sort of legitimate physical experiment to quantify this reduction with increasing DNA length.
Relevant answer
Answer
found an assay here, thank you all!
  • asked a question related to Biochemistry
Question
3 answers
Hello,
I'm going to be labeling different size thiol-activated polyethylene (PEG) molecules with Atto488-Maleimide. I'm wondering what is the best way to accurately measure the final concentration of the PEGs following labeling and removal of free dye.
Colourmetric assays like BCA and Bradford would not work right for PEGs, neither would measuring A280 on a Nanodrop - so the only thing I can think of is either:
a) measuring the UV absorbance of the atto488 dye and using dye concentration as a proxy for the amount of labeled PEG (assuming I have high labeling efficiency).
b) Lyophilize the final product and just go by dry weight
Has anyone got any advice as to what the best way would be/any other suggestions?
Thanks!
Relevant answer
Answer
Separating the unlabeled low-molecular-weight PEG from the unincorporated label may not work well using gel filtration. You may have to use something like hydrophobic interaction chromatography, or possibly an organic extraction.
If the labeling reaction works well (because the sulfhydryl is fully reduced), and the label is in excess, you should get a 1:1 labeling stoichiometry. Including TCEP in the reaction will keep the sulfhydryl reduced without interfering with the maleimide reaction.
If you want to check that the PEG is fully labeled, you need an analytical method that can distinguish labeled from unlabeled PEG. There is probably an HPLC method in the literature somewhere. Fluorescent dyes are quite hydrophobic, so it should be possible to separate labeled from unlabeled dye by reverse phase chromatography.
Another method that occurs to me is to test for the presence of free thiol in the product using Ellman's reagent. There will be none if the reaction has gone to completion.
The molecular weight of PEG is an average value of a distribution, so the concentration measurement is also an average.
  • asked a question related to Biochemistry
Question
9 answers
I need to characterize two extra cellular enzyme produced by bacteria . For that process the purification of enzyme step is necessary before the characterization process? otherwise can i use the crude enzyme extract (cell free Supernatant) for the characterization process?
Relevant answer
Answer
If you have a specific assay for your enzyme's activity, you can perform enzyme kinetics on crude preparations. Also helpful is a specific inhibitor of your enzyme (like ouabain for Na/K-ATPase), the activity you measure in its presence is due to other enzymes and hence subtracted.
Many other measurements can also be performed with crude enzymes, if the method is adapted. For example, size determination by analytical ultracentrifugation requires pure proteins, but methods for crude preparations are available (DOI:10.1016/0003-2697(89)90297-2, DOI:10.1016/S0021-9258(18)64180-8).
Indeed, some purification methods (especially solubilisation of transmembrane enzymes) are so harsh, that part of the purified enzyme may be non-functional or (worse) partially functional. This would make measurements of turnover number impossible.
  • asked a question related to Biochemistry
Question
7 answers
We would like to dephosphorylate purified proteins (in vitro). How do we decide what type of alkaline phosphatase to purchase?
Relevant answer
Answer
Maybe this Recombinant Human Alkaline phosphatase, placental-like (ALPPL2) works for this situation. You can view the details on this page:
  • asked a question related to Biochemistry
Question
8 answers
Kindly discuss your ideas and viewpoints on the origin of life and the RNA world hypothesis.
What are the contradictory views on why researchers are still unsure about the origin of life through RNA or such analogous molecular intermediate pre-cursors preceding its existence?
"The general notion of an “RNA World” is that, in the early development of life on the Earth, genetic continuity was assured by the replication of RNA and genetically encoded proteins were not involved as catalysts. There is now strong evidence indicating that an RNA World did indeed exist before DNA- and protein-based life. However, arguments regarding whether life on Earth began with RNA are more tenuous. It might be imagined that all of the components of RNA were available in some prebiotic pool and that these components assembled into replicating, evolving polynucleotides without the prior existence of any evolved macromolecules. A thorough consideration of this “RNA-first” view of the origin of life must reconcile concerns regarding the intractable mixtures that are obtained in experiments designed to simulate the chemistry of the primitive Earth. Perhaps these concerns will eventually be resolved, and recent experimental findings provide some reason for optimism. However, the problem of the origin of the RNA World is far from being solved, and it is fruitful to consider the alternative possibility that RNA was preceded by some other replicating, evolving molecule, just as DNA and proteins were preceded by RNA." - Robertson and Joyce
[This is as per the explanation by Michael P Robertson and Gerald F Joyce in the article: "The origins of the RNA world." published in the Cold Spring Harb. Perspect. Biol. 4, a003608 (2012).]
The scientific community must resolve this contradicting conjecture through rational discussion and debate backed by strong experimental evidence on what must be the pre-cursor molecule to the Origin of Life if it is not RNA!
Relevant answer
Answer
One of the issues that is holding the concept of an RNA world back from being more scientifically useful - irrespective of whether there ever was such a thing - is that we don't use the idea in the scientific way it was intended. Just like any other prebiotic scenario, it is not (nor has it ever been) a scientific hypothesis. In fact, scenarios are usually not intended as such. Scenario authors from all niches (including RNA world) have pointed out that scenarios themselves are untestable. However, they guide thinking and allow to conceive of hypotheses that are testable. If we go through the old literature we find very explicit passages to support this fact.
For the specific authors advanced in the question, G.F. Joyce and L.E. Orgel, we have a passage from 1999 in "prospects for understanding the origins of the RNA world". (The RNA World 2nd ed. 49-77).
"The presumed RNA World should be viewed as a milestone, a plateau in the early history of life on earth. So too, the concept of an RNA World has been a milestone in the scientific study of life's origins. Although this concept does not explain how life originated, it has helped to guide scientific thinking and has served to focus experimental efforts."
You can find this point of view expressed in foundational work for all niches related to the popular scenarios today. But you can also find it for scenarios most people in origins have never heard of. E.g. the idea that celllular life started with terpenoids found in G. Ourisson and Y. Nakatomi's "the terpenoid theory of the origin of cellular life: the evolution of terpenoids to cholesterol. (1994) Chem & Biol. 1 11-23".
"The hypothesis provides an attractive way of ordering the terpenoids: like all evolutionary theories, it cannot be tested directly. The ideas summarized here do, however, suggest a multitude of experiments having some bearing on the fundamental and fascinating question: how did the first cells appear? We hope to carry out some of them."
A related line of thought - but highly influential - is the exposition by Harold J. Morowitz from 1992 in his book "Beginnings of Cellular Life: Metabolism Recapitulates Biogenesis". If we go to the conclusion, we find this explicit clarification on the distinction between a genuine scientific theory and a scenario:
"at this stage of the thought process, it is important to focus on the hypothesis that intermediary metabolism recapitulates prebiotic chemical evolution. This hypothesis is not a strictly vulnerable theory in the Popperian sense, but it does provide us with a valuable heuristic method for using modern knowledge of biochemistry to search for events that have left their trace. If the intermediary metabolism of autotrophs does not recapitulate biogenesis, then the discontinuities will have to be explained."
More than 2 decades back, many authors made a clear distinction regarding this nuance. Scenarios are here to help: they guide thinking and design experiments. They only guide thinking in a scientifically meaningful direction as long as we can easily abondon scenarios and enthusiastically continue replacing them with new, more informed scenarios. A situation where a scenario gets entrenched and where researchers treat it as a scientific hypothesis is - by construction - hard to escape.
In fact, this is exactly the situation that many researchers have described around the 80s, when criticism mounted against the prebiotic broth scenario. The passage from Wächtershäuser's 1988 "Theory of a Surface Metabolism" is telling:
"The prebiotic broth theory has received devastating criticism for being logically paradoxical (11, 135), incompatible with thermodynamics (11, 144, 160), chemically and geochemically implausible (134, 136, 144), discontinuous with biology and biochemistry (160), and experimentally refuted (135, 160). The reason for the tenacity with which it is retained as accepted dogma has been forcefully and clearly stated by Scherer (126): "If this rejection is substantiated, there will remain no scientifically valid model of the selforganization of the first living cells on earth."
Clearly, the broth scenario had overstayed its welcome. One reason for this is that its 'claims' (which for a scenario can only be speculations) were too much in contradiction with claims from fields of science that do not suffer the same restrictions when it comes to testing and refuting their theories. One example of a very controversial idea that can be found in Haldane's formulation of a broth scenario, is the purported necessity of a long, highly functional protein randomly emerging from a soup, as an extremely rare event: we expect this to be prohibitively unlikely and hence a far from parsimonious explanation.
Quite a few of the critiques voiced against the prebiotic broth scenario are equally valid critiques of some scenarios we have today, including RNA world.
The RNA world is an old and multifaceted concept. There are contrasting formulations that make different claims (to be interpreted as speculations) about history. As with the prebiotic broth scenario (and any scenario), it has raised genuine scientific objections. These have remained largely unadressed, in spite of its long dominance.
It is instructive to bear in mind that scenarios don't come from nowhere. They're fairly detailed speculations about purported historical events. To make them, each author makes assumptions. Some of these concern speculations that later became testable, e.g. about chemistry and physics. You will find different scenario authors make different assumptions and different arguments (and flaws therein). There's an inevitable bias here with respect to the fields an author is trained in. Some of the foundational assumptions in popular scenarios like RNA world are at least 50 years old, but some unchallenged assumptions date back to a literature that is more than a century old. A time before IUPAC, modern quantum mechanics, genetics, and so forth.
That has been enough time to forget that scenarios like RNA world are by construction not testable hypotheses and that they were not intended as such. Scenarios are here to guide thinking, to inspire experiments. The best thing a scenario can do for us, is generate insights that spur us to change the way we think and thereby necessitate replacing our old scenarios with new ones, and repeat the cycle. The science coming out of the community today is a lot more conducive to doing that than previously.
The same cannot be said for the rather myopic RNA-centric framing of a question in the cited passage, which attempts to elevate RNA world to more than a scenario. Rather than forcing ourselves to think about the rather narrow and outdated proposal by Joyce and Robertson, ("consider the alternative possibility that RNA was preceded by some other replicating, evolving molecule"), it is more productive to critically revisit all the things that have been assumed and argued when the concept of an RNA world was conceived and how which of these premises are considered valid or plausible today, and which ones back then. Is there a formulation of RNA world for abiogenesis that is logically sufficient? And if so is it logically necessary that abiogenesis proceeded this way?
It is also instructive to check how much of the logic was sound. e.g. the rhetorical tricks employed in RNA world introduce all sorts of hidden assumptionsm.
As an example of the latter: some still justify an RNA world by the party trick 'chicken-and-egg' question 'protein or RNA, which came first?', only to conclude with 'RNA, it encodes proteins' and hastily conclude with an even stronger 'RNA-first' for abiogenesis. 'chicken-and-egg' fallacies are nothing new in origins. In fact, they were already identified as such long ago. E.g. in chapter 8 of "Seven Clues to the Origin of Life (1985)" by Cairns-Smith, there's an illustrated passage detailing that these types of paradoxes in origins frame the question in a manner that prevent us from considering scaffolds.
"
The fact is that even the so-called simple organisms such as E. coli are very complex enterprises with all sorts of things going on together. There is plenty of scope for accidental discoveries of effective new combinations of subsystems. It seems inevitable that every so often an older way of doing things will be displaced by a newer way that depends on a new set of subsystems. It is then that seemingly paradoxical collaborations may come about.
To see how, consider this very simplified model - an arch of stones: This might seem to be a paradoxical structure if you had been told that it arose from a succession of small modifications, that it had been built one stone at a time.
scaffolds that starts like this:
This might seem to be a paradoxical structure if you had been told that it arose from a succession of small modifications, that it had been built one stone at a time. How can you build any kind of arch gradually? The answer is with a supporting scaffolding. In this case you might have used a scaffolding of stones. First you would build a wall, one stone at a time:
Then you would remove stones to leave the 'paradoxical' structure.
"
It should be noted that in 2022, even in RNA-world, very few scholars remain that find RNA-first a convincing idea. As a scenario, however, it is not useless: it is instructive to consider what the underlying ideas are that at some point in time made such a highly specific idea compelling to so many of us.
A fixed motif in scenario papers is to start explicitly and implicitly assuming a few things about what chemistry can and cannot do and some properties of abiogenesis. These sort of assumptions used to be spelled out routinely, also outside scenario papers. Let me give two examples.
The original 1953 paper for the "Frank Model" "on spontaneous asymmetric synthesis", has the passages
".. the defining property of a living entity the ability to reproduce its own kind ...
confining attention to chemical molecules, the complexity of any having this essential property of life is likely to be great enough to make it highly improbable that it has a centre of symmetry."
(*I should point out that Frank makes an important error here: the capacity for molecular reproduction is not a molecular property but a property of a reaction network. If we add an additional thermodynamic criterion this property is autocatalysis and we can then check this claim from the IUPAC definition: https://goldbook.iupac.org/terms/view/C00876. It turns out there are trivial ways to make small networks that have this property https://chemrxiv.org/engage/api-gateway/chemrxiv/assets/orp/resource/item/60c74d67469df42226f44295/original/emergent-autocat-animation.gif.)
The point to retain here is that Frank considers it to be generally accepted that one can assume this property to be prohibitively rare in chemistry. This belief was wideheld, and we can e.g. read in "the units of selection" (1970) by Lewontin a summary on scientific views on abiogenesis
"The present view ... Since there was no autocatalysis, there was no reproduction or heredity and so no possibility of natural selection."
The coacervates in Oparins scenario were notably invoked to adress this issue.
When it comes to assumptions in scenarios, this systematically involved conjecturing that chemistry 'in the wild' intrinsically and deterministically becomes a 'mess', undergoing no meaningful complexification, and for which no reproduction and evolution can reasonably be expected. From there, it appears that no process of abiogenesis should conceivably occur naturally, and thereafter some specific sequence of exceptional events is proposed as plausible, because it appears to be the sole contender.
Let us make more explicit why this is not an innocent procedure:
We still find our understanding of 'basic chemistry' to be plagued with limitations and long-lived misinterpretations (e.g. 2 days ago we learned that methyl substitution destabilizes radicals instead of the textbook knowledge that it stabilizes them ).
Moving beyond the basics, we by and large lack a lot of formal theory, experiment, or even a simple reference frame for the things that happen then. Joyce and Robertson honor the tradition of purporting from the outset that 'chemistry in the wild' becomes an intractable mess. The issue is that we don't know at all if that's the case. We cannot assume this from the outset, we need to extensively study it. We require extensive experiments and theory and a reference frame for all the phenomenology associatied with complex systems (e.g. multiple components, compartments, multiple forms of nonequilibrium driving, length scales, time scales).
In making the routine assumption of 'messy, intractable chemistry that can neither complexify nor multiply', we have decided in advance that, once we finally understand 'chemistry in the wild' with its 'so-called intractible mixtures', it cannot have any bearing on abiogenesis. Let alone explain it.
That is a disproportionately bold conjecture about fundamental science, and a very consequential one: all historical scenarios - RNA world being one out of many - have been justified by formulating conjectures of this sort (many authors also insist on other properties, e.g. chemistry being deterministic). Clearly, it should be the first priority of everyone in the field to test this conjecture, by extensively and rigorously studying complex chemical systems as an end in itself. If the conjecture is correct, it provides an important validation for historical scenario approaches. If the conjecture turn out wrong, we are in a much better position to conceive of more scientifically informed scenarios, but potentially the approach will change entirely.
In presenting it as such, I am making it appear as if it could be an open question whether the chemical conjectures underpinning our scenarios in origins may be true or not. In fact, we have learned quite a few things in the meantime. And some clumsy mistakes were made elsewhere.
- Determinism:
When it comes to chemistry being deterministic (a key tennet of e.g. Sutherlands scenario and Wächtershäusers surface metabolism): upon critical evaluation of what is known of basic chemistry this idea becomes unacceptable, especially when considering the chemical processes on the surface of a planet, as opposed to a quick reaction in pyrex.
1) insofar as it is reproducible, modern chemistry owes much of it to big strides of standardization in glassware, methodology, synthesis protocols (e.g. usage of stirring bars).
2) lab chemistry exhibits many forms of contingency. This is particularly the case when it comes to phase behavior, e.g. habit modification, polymorphism. Aspirin purportedly has 8 reported polymorphs, phenobarbitone 13.
3) glassware is cleaned between reactions, thereby making successive reactions in the same glassware independent. In nature, this property of independence is absent. In fact, effort to make an evolutionary classification of minerals are rooted in the opposite: that certain minerals start to form conditional on the presence of certain others. (https://pubs.geoscienceworld.org/msa/ammin/article/104/6/810/570840/An-evolutionary-system-of-mineralogy-Proposal-for)
- Autocatalysis:
A first issue to get out of the way is the misconception that autocatalysis is prohibitively rare. A prominent PI in origins (RNA world, not a chemist) told me that chemists throughout history have found exactly one example. Claims about the contents of a literature one cannot realistically have read in a lifetime is a common error we can find in the origins literature. Below are some reviews.
I should stress that these reviews discuss examples from a few niches in chemistry. These reviews do at least allow to have 100s of counterexamples to dubious claims about no autocatalysis in chemistry, but it's only a small fraction. Virtually all branches of chemistry have regular reports of autocatalysis, but very few focus on autocatalysis in its own right. And hence most branches do not review their reported examples.
By critically examining the IUPAC definitions, one can show that autocatalysis is dramatially more widespread than long thought. In part, this is because the definition applies to a wealth of situations where the term is not routinely employed. By examinging the requirements of autocatalysis as an emergent network property, one can demonstrate that this property emerges particularly readily in a heterogeneous / multicompartment context. With the disclaimer that I'm an author I refer to the following:
- Messy chemistry:
Refreshing counterexamples are afforded by the literature on systems chemistry and dynamic combinatorial libraries.
In the context of origins, a recent work that is greatly aiding in fixing our misconceptions is : https://www.nature.com/articles/s41557-022-00956-7
Where a reaction of purported immense complexity is found to exhibit highly reproducible and ordered behavior as function of environmental inputs. How chemistry exactly works on this level is still poorly understood. I think I do, but it'll have to await peer review. But we cannot in good scientific conscience take for granted anymore that chemistry becomes messy and intractable. When we do the experiments, we see something very different.
in conclusion, I want to come back to the final point of the question
"The scientific community must resolve this contradicting conjecture through rational discussion and debate backed by strong experimental evidence on what must be the pre-cursor molecule to the Origin of Life if it is not RNA!"
No. The sientific community should strive to do what it can justify scientifically. Those that find it fruitful to relegate the RNA world - which is not a hypothesis - are justified in doing so. Notably because it is is founded on scientifically refuted premises and logical errors.
Those that find ways to make it fruitful to keep it are justified in doing so: it's a scenario, one can draw inspiration from it. Perhaps a thoroughly altered version can be developed that fixes previous issues.
Above all else, RNA is an amazing molecule that has been used for fundamental research that concerns everyone in origins, and will continue to do so irrespective of how serious the RNA world scenario is still taken.
What the origins of life community needs, first and foremost, however, is concern itself with more important matters.
Complex chemistry needs to be studied thoroughly on an experimental and theoretical level.
New scenarios are needed. And these scenarios should no longer require chemistry to have properties it doesn't have, and vice versa. These scenarios should also explicitly be appraciated for what they are, an for what they're not. They're here to help, to guide thinking, inspire experiments, produce testable predictions, update our beliefs. They are not scientific hypotheses in and of themselves.
  • asked a question related to Biochemistry
Question
2 answers
I have the assay CHEMBL1962204 (ChEMBL assay ID), which contains some compounds with negative % Inhibition value. Below are the bottom 20:
chembl477979 -2.82 % Inhibition
chembl495902 -2.51 % Inhibition
chembl160876 -1.56 % Inhibition
chembl1789962 -1.52 % Inhibition
chembl234838 -1.47 % Inhibition
chembl1909407 -1.38 % Inhibition
chembl1909408 -0.91 % Inhibition
chembl207410 -0.86 % Inhibition
chembl1909409 -0.42 % Inhibition
chembl205765 -0.3 % Inhibition
chembl1794068 -0.3 % Inhibition
chembl1794067 -0.19 % Inhibition
chembl1909402 -0.02 % Inhibition
chembl517171 -0.01 % Inhibition
chembl1909364 0.19 % Inhibition
chembl1785003 0.21 % Inhibition
chembl2062290 0.23 % Inhibition
chembl1909393 0.26 % Inhibition
chembl1909388 0.34 % Inhibition
chembl261849 0.34 % Inhibition
How should I treat these compounds in QSAR? Completely remove them or set their value to 0 %?
Relevant answer
Answer
Set them to zero. They are all close to zero and well within a reasonable range of experimental error from zero.
Large negative % inhibition could be due to activation of the target or an experimental artifact, but that is not the case here.