Science topic
Ribosomes - Science topic
Multicomponent ribonucleoprotein structures found in the CYTOPLASM of all cells, and in MITOCHONDRIA, and PLASTIDS. They function in PROTEIN BIOSYNTHESIS via GENETIC TRANSLATION.
Questions related to Ribosomes
Dear Colleagues:
I have multigene sequences of my target species(18S and 28S ribosomal genes) for a phylogenetic analysis with GenBank sequences of other taxa to see the relationship among related families. The result using each gene looks ok, and now I am trying to do additional analysis based on the sequence concatenation. The problem is that the GenBank sequences of different genes were generated separately using different individuals by different researchers, even though they are from same species. My question is, therefore, is it ok to make concatenate alignment of the sequences from different individuals for my research purpose?
Thank you.
Sincerely yours
Jeongho Kim.
Hello all,
I have been trying to grow E. coli cultures for the past few weeks but every time I extract total RNA, it does not show the 23S and 16S rRNA bands on the agarose gel. I have tried multiple times and reduced the incubation time from overnight to 6 hours. I can't tell if my E. coli is in lag phase or if I have contamination causing RNA degradation. Any help is appreciated.
In someone who gets a bivalent mRNA shot, what happens, exactly, when the mRNA for BOTH subvariants of the spike is active in the same cell? Such cells are NOT going to just produce two kinds of spikes (Wuhan and Omicron). They’re going to produce hybrid spikes using various combinations of the proteins that normally combine to form the spikes of the two subvariants. The spikes are each made of many subunit peptides and proteins that are generated by ribosomes from the mRNA and THEN self-assemble to form the subunits containing three ribosome-generated copies of each spike subunit protein that then have to assemble together to form each spike. So does this mean that such cell will “normally” turn out a large number of DIFFERENT spike proteins? It does.
I note that per https://www.nature.com/articles/s41401-020-0485-4 :
“The total length of SARS-CoV-2 S is 1273 aa and consists of a signal peptide (amino acids 1–13) located at the N-terminus, the S1 subunit (14–685 residues), and the S2 subunit (686–1273 residues); the last two regions are responsible for receptor binding and membrane fusion, respectively. In the S1 subunit, there is an N-terminal domain (14–305 residues) and a receptor-binding domain (RBD, 319–541 residues); the fusion peptide (FP) (788–806 residues), heptapeptide repeat sequence 1 (HR1) (912–984 residues), HR2 (1163–1213 residues), TM domain (1213–1237 residues), and cytoplasm domain (1237–1273 residues) comprise the S2 subunit (Fig. 2a) [13].”
We can confirm in Fig. 2a of this peer-reviewed article clear confirmation that the spike is NOT created in one go by a ribosome reading and connecting the 1274 aa (amino acids) in sequence. Rather, these proteins and peptides are made from S1 and S2 proteins, which in turn are made of NTD, RBD, FP, HR1, NR2, TM, and CT proteins.
There are mutations within (that is, differences between) the genetic sequences of at least each of these proteins: NTD, RBD, FP, and HR1 between tozinameran and riltozinameran or famtozinameran.
So with 2 options for each of 3x4=12 locations, we have 2^12 - over one thousand combinations. We don’t know how each of these thousand different spike proteins will act in the human body. We have no idea. It’s likely they would all be created, but we don’t know. (I speculate that maybe some combinations wouldn’t self assemble, or would assemble into something totally unexpected.) Yet we’re (indirectly) injecting them into billions of people. They have only existed since this bivalent vaccine started being used, and ~99.8% of them have not been studied at all.
What am I missing?
[edit: Edgar, your “answer” below demonstrates no understanding of the question and makes no attempt to answer it – the actual question I asked. I’m asking a scientific question. Please don’t use it as a soapbox for proselytizing. I'm a fan of the mRNA therapy platform - and this wrinkle is concerning. I’m looking for an answer that shows expertise in protein assembly.
- Also, technically, you’re spreading misinformation as, for example, it is inaccurate to claim that the bivalent vaccines have been through reported clinical trials. They have not. (If that changes in the future, do point to it at ClinicalTrials dot gov or point to the literature if there’s one I’m not aware of.) Also, “no evidence” you say? Heck, *according to Pfizer*: “Myocarditis (inflammation of the heart muscle) and pericarditis (inflammation of the lining outside the heart) have occurred in some people who have received … Pfizer-BioNTech COVID-19 Vaccine, Bivalent. The observed risk is higher among adolescent males and adult males under 40 years of age than among females and older males, and the observed risk is highest in males 12 through 17 years of age. In most of these people, symptoms began within a few days following receipt of the second dose of vaccine. The chance of having this occur is very low” <sic>
Hi everyone,
I am looking for a ribosome complex on an mRNA .PDB file to visualise the interaction in the PyMol. I was so far unsuccessful at finding it in PDB or Uniprot databases, but want to learn more of how mRNA interacts with the ribosome.
If you have this file, I would be grateful if you could please share it with me.
Thanks,
Maria
The popular IRES from HCV or EMCV was used in quite a few plasmid. I am wondering any one try them in fish or bird?
Hi ResearchGate friends, I have found some visible bands between 18s and 28s rRNA while running the formaldehyde-denaturing agarose gel on total RNA (please check the attached images). This is seen in my experiment group, not in the control group, though the total RNAs in the two groups are co-extracted using the same method.
Anyone can kindly give me a hint if they are precursors or fragments of rRNA? Thanks!
I was trying to recalculate the RIN value using the Agilent Bioanalyzer. because it couldn't calculate the RIN value due to 'unexpected ribosomal ratio' error flag. I checked the setpoint for the ribosomal ratio for RIN calculation and it was 0.7. I then edited the fragment size of the 28s and got a ratio of 1.1 but still the error says the same and no RIN value. So, does that mean it is impossible to recalculate the RIN value? Thanks in advance for your kind help.
Text book always said that most important job of RNA is protein synthesis - assembling amino acids into proteins. Messenger RNA carries instructions from nucleus to ribosomes. Transfer RNA transfers each amino acid to the ribosome as needed by the code of the mRNA molecule.
However, it seems that it never mentioned the metals in the protein.
May I miss some knowledge in this field?
Where are the metals?
How the RNA to decide the metal center in the protein?
Any hints?
Running a sucrose gradient and I am only interested in the disome peak, would also like to separate out the monosome peak. Not interested in polysomes past this.
We usually run a gradient 10-50% sucrose to get the full range of ribosomes - from subunits to the high polysomes.
Should I attenuate the glucose density gradient for this more narrow focus? Would 10-30% be reasonable?
Hi everyone,
I performed ChIP-seq analysis on E. coli cells using antibodies against one of a transcription factor. During data analysis, I validated ChIP samples against corresponding "input" samples (sonicated DNA, without immunoprecipitation). After sequencing, I got a lot of sequences encoding tRNAs and ribosomal subuints. It is doubtful that the analysed protein interacts with sequences coding tRNAs. What could be the problem? I have to add that during sample preparation I used an excess of RNAse for DNA sample purification. An external company performed library preparation and sequencing.
Does someone have a similar problem with ChIP-seq data analysis?
I am preparing to design primers to clone a Leu2 maker from a plasmid into Saccharomyces cerevisiae (BY4742) in place of a native gene via homologous recombination. However, I struggling to get my head around the genetic workings of the gene orientations of both the Leu2 gene (from plasmid PAG305GPD-ccdB) and Htz1 from (BY4742). From what I can see, both the Leu2 and Htz1 are coded for on their respective negative strands.
As I understand it, transcription always takes place 5'-3 'direction against the antisense strand, which in this case would create mRNA strands with this kind of sequence 5- XXXXXXXXGUA-3'.
My confusion is this - How is it then translated into a protein if the 'AUG' recognition codon is in reverse. Can a ribosome move from the 3'-5 'end of a strand or is it rather that in this case transcription takes place against the sense strand? Or am I misunderstood something fundamental along the way?
I need to find a Mammalian cell line that can translate mRNA of a bacterial gene
I would like to do an MD simulation of ribosomes. However, I do not want to simulate the entire ribosome because of limited computational power.
I want to simulate a spherical subsection of the ribosome and restrain all the atoms outside the sphere. Is there any way to do this?
I am using Gromacs for the simulations.
Thanks in advance.
Dear members,
I would like to ask for your opinions about the process of yeast cell aging.
Which theory is in your opinion more trustful for explaining yeast cell string?
Thanks for an interesting discussion.
Regards,
Marco
My goal is RNA extraction from yeast's ribosome; still now I made this with TRIsure but the result is terrible. Can someone help me?
Hi All,
I have a protein sequence cloned in pET21+ vector.
Below is the part of the sequence starting from T7 promoter to the ATG (of my protein):
TAATACGACTCACTATAGGGGAATTGTGAGCGGATAACAATTCCCCTCTAGGATCCTCTAGAAGGAGGACAACCATG
Protein is not expressing at al, I am using BL21 DE3 for the expression. I am only curious to know that does this sequence have Ribosome binding site/SD? Or is this sequence correct for the expression of my protein?
I am doing polysome fractionation for stress conditions for mammalian cell. it seems that peaks of 40S and 60S disappear during stress condition compared to control. I am wondering whether all ribosome are struggling with translation since our translation analysis show us there is an increase in translation.
- His-tagged protein immobilized a on Ni coated NTA chip.
- Both the reference channel and ligand bound channel shows binding with analyte (ribosome)
- negative RU in the resultant curve.
- Reference channel is Ni free.
- Running buffer contains hepes,NaCl,EDTA,Mg(OAc)2
- Both the ligand and analyte are prepared in the running buffer.
- Reference channel was tried to be coated with BSA and still it's showing non-specific binding.
- Tris buffer as running nuffer gave the same non-specific binding but the resultant curve gave positive RU. As suggested in some paper I am using Hepes Buffer now. If anyone has any idea please suggest me a way.
We know about that ribosome is composed of proteins and rRNA and rib is the precursor of proteins for ribosome composition the proteins is synthesis by which?
During Replication, helicase, primase, DNA polymerase and ligase proteins travel from wherever they are in the nucleus to a specific region of the whole DNA. Why and how do the proteins reach till the 5 prime end?
During transcription, RNA polymerase travels towards the promoter region. How does this movement happen?
During translation, ribosomes travel towards mRNA and tRNA travels towards ribosomes. How?
Is the movement of protein/ chains random? or is it precise?
If it is precise how does the polymease/tRNA know where the DNA/ribosome is and how does it move in that direction?
Prokaryotes take aid of Shine–Dalgarno sequence to detect actual translation initiation AUG among the many AUG codons in the m-RNA. Is there any such kind of sequence in eukaryotes that help ribosomal machinery to detect actual initiation AUG among the various AUG codons? Or is it the case that there are no any AUG codon before the actual translation initiation site in eukaryotes so that recognition sequences are not essential for translation initiation? Or there is different kind of initiation site recognition mechanism?
Hi,
When using the lambda red system, after removing the antibiotic cassette, scar sequences are left behind. Do these sequences vary from clone to clone? Do they contain stop codons? In the three phases?
This is importart when deleting a bacterial gene that is in an operon with translational coupling: it is essential that the deletion does not insert a premature stop codon, because this would cause the ribosome to fall, and the downstream gene would not be translated. In translational coupling, the first gene´s mRNA UGA must be juxtaposed to the second gene´s AUG (so that the translating ribosome can turn around from the TGA to the ATG).
And if anyone knows of simple systems for making seamless/scarless deletions in K. pneumoniae that would be great!
Thanks for your answers.
I shined uv to plants and got a kegg figure for ribosome pathway after RNA-seq. But what do these components circled in red and blue mean? thanks
I am using BSA as block. solution and I think it is not so good to block protein up to 20kDa by mixture of BSA with more than 70kDa. Can you recommend me some better, smaller solution please?
Thank you!
I constructed a synthetic DNA library (scFv, VH-VL orientation) with a 3' reporter and 8x histag. I cloned and expressed this gene following which I performed an ELISA. The ELISA results suggested binding activity of some of my scFvs against their target antigens. I sequenced the DNA of these 'binders' and the results were unexpected. I could detect the full and correct reading of the VL sequences but interestingly the upstream VH sequences had 2-3 stop codons interspersed and were not recognized as correct IgVHs by bioinformatics.
Can anyone please explain to me what may be going on here? A stop codon upstream should usually truncate the protein at that point but the ribosome seem to have read through the stop codons.
I guess what I am interested in knowing is whether the VH is aberrant and non-functional and whether the binding activity I observed with ELISA was contributed solely by functional light chain, VL.
Here is one of those sequences (VH, linker and lambda VL).
CCATG GCC GAGGTGCAGCTGTTGGAGTCCTGGCCGCAGTGAGGAAGGGAACTGACTGCCGAGGCCGGAGGGCAGCCGGCCTGCCGAGGGGAACAGCTGGATCTGGGTCCAGGTCATGGGTAGGTGGGCGCGCAGGCAGACTTTCGTGATGTTGTGCACTTGAGGTGTTGTGCTGGACTTCCACAGCAGACAGGAAGGGTGAACAGAGCAGCCCTTCAGAGAGAAGCACTGGGCAGTCACAGGGATTTGAGGGAGGGTGGAGGACACCCTGCAAGCCCTGCGTGGCCACCAACAGGCAGCGCCCTGCAGCAAGGGGGCCACCGTCTCCTCA
GCCTCCACCGGATCCGGTTCTGGTAGTGGTGCTACTTCTGGATCC
TCCTCTGAGCTGACTCAGGACCCTGCTGTGTCTGTGGCCTTGGGACAGACAGTCAGGATCACATGCCAAGGAGACAGCCTCAGAAGCTATTATGCAAGCTGGTACCAGCAGAAGCCAGGACAGGCCCCTGTACTTGTCATCTATGGTAAAAAAAACCGGCCCTCAGGGATCCCAGACCGATTCTCTGGCTCCAGGTCAGGAACTACAGCTTCCTTGACCATCACTGGGGCTCAGGCGGAAGATGAGGCTGACTATTACTGTAACTCCCGGGACAGCAGTGGTAAGAGTTGGGTGTTCGGCGGAGGGACCAAGCTGACCGTCCTA
PS: I sequenced 2x with different primers and in forward and reverse.
Thank you all in advance.
I am running a simple experiment with a co-culture of bacteria (P. aeruginosa and an E. coli strain) in LB to study their interactions with regard to biofilm formation. Is there a good or feasible way to separate these two bacterial species from this co-culture in order to separately analyze their respective ribosomal profiles?
Thanks!!
I actually have several questions:
1) Where do I go to learn about the physical chemistry of the interaction between ribosomes and RNA made with pseudouridine or 1-methylpseudourine?
2) Are there any concerns that a DNA vaccine will integrate into host genome in the complete absence of any sort of integrase?
3) Is an immune reaction against ordinary mRNA a problem? Are there any papers documenting instances of this?
4) My biggest question: how is an mRNA with modified nucleosides, a 5' cap, and poly(A) tail manufactured in the quantities necessary to inoculate an entire nation?
Hello everybody!
so I have a plasmid which has a synthetic intron (IVS) & an internal ribosome entry site (IRES), and I'm thinking to remove them as I'm cloning due to some matters related to preferable enzymes restriction sites, and my question is :
What will be the effect on the expression of my gene of insert after I remove them?
looking forward to answers and thank you already !
I did some ribosomally depleted RNA-seq in rat brain. Does anyone have recommendations on how to quantify/compare small RNA expression between samples? I have used RNA STAR to map reads back to the genome and determine reads per gene for mRNA, but I can't seem to repeat that for any kind of small RNA like lncRNA or miRNA and the pipeline keeps failing. Is there a specific GTF file or similar type that I should be using? Do I need a different pipeline? Help please!
I did the experiment following the 2M sucrose cushion method to isolate the ribosome from 293F cells. Theoretically, I should get 10-15 A260 units from 2x 10^7 cells. But I can only get 4 units at most, I tried many ways, like increase the centrifuge speed, time, or lysis buffer, but neither of them works. Anyone with experience has good suggestions? Thanks.
Hi all,
I try to figure out weather there is a change in my rRNA following treatment.
when I treat the cells (mouse cells) and isolated RNA, I see a major difference in RNA concentration, without any difference in cell number.
what is the best way (or easy for start) to see if there is any change in the ribosomal RNA amount, activity or biosensis?
Thanks,
Shani
Hello everyone,
I recently started several polysome profiling experiment and something bugs me out.
Every protocols I found use 12mL tube.
They load their lysate containing ribosomes attached to mRNA and then ultracentrifugate for 2h at 260000 g.
The more ribosomes attached to a given mRNA, the more it migrates. By analysing fraction of the gradient you can determine if your mRNA of interest is efficintly translated or not.
The tube contain a 10-50% sucrose gradient. This way, heavy complexes don't migrate to soon (and don'taccumulate at the bottom of the gradient) compared to light complexes.
I only have 4mL tubes instead of 12mL. So the lenght of migration is divided by 3. Hence, I also reduced time of centrifugation by 3.
But, is this "rule of thumb" accurate ?
I have tagged my 12kDa eIF1 protein with Fluorescein-maleimide (Ex: 485nm, Em: 530nm). The interaction with this particular protein with 40S ribosome should show anisotropy value change. 10nM of eIF1_FL and 40nM of 40S in proper binding buffer (25mM HEPES-KOH pH 7.5, 2.5mM Mg(OAc)2, 80mM KOAc, 2mM DTT) were incubated and vertical and horizontal polarization measured. Anisotropy values calculated (Taking G=1) from those polarization values shows negative digits:
Blank Buffer: 0.123
Only eIF1_FL (10nM): -0.295
eIF1_FL (10nM) + 40S (40nM): -0.302
eIF1_FL (10nM) + 40S (200nM): -0.301
Please suggest how to improve my reaction conditions to get a values in the range of expected one.
Hey fellow researchers,
one of the first things you learn as biologist is the structure of the cell. So basically, every biologist should be skilled in the basic terms. But I encounter very often (even in very high ranking journals, including Nature, Science and Cell) the mix-up of cytoplasm and cytosol.
The definitions, as far as I know, are very clear.
The cytoplasm is the complete content of the cell surrounded by the plasma membrane, which includes the cytosol and ALL membran-enclosed organelles (with excpetion of the nucleus), as well as solutes, the cytoskeleton with all components, ribosomes and so on.
The cytosol is the fluid phase of the cytoplasm without the membrane-enclosed organelles.
So the cytoplasm includes the cytosol but not vice versa. Therefore, sentences like "Protein X was detected in the cytoplasm, but not in ER and mitochondria" is wrong, since ER and mitochondria are part of the cytoplasm and one cannot exclude them when using the term cytoplasm as definition for a cellular location. And this is only one example of many.
Do I miss something or have the terms changed? Perhaps experts from the field of cell biology can contribute to this discussion and help to understand.
All the best and stay healthy,
Marc
Does anyone know if eukaryotic ribosome translation / integrity can be preserved in other buffers than HEPES and TRIS after purification through a sucrose cushion, by IP, or other methods?
Maybe a reference where Ribosome activity or integrity in various buffers (and not only Mg2+ and K+ concentrations) is tested? Could be in vitro translation, cryo-EM, or other methods.
All the best - with hopes for any helping thoughts,
Kasper
Hi,
I have stumbled upon an article about translocation of AKT into mitochondrial matrix upon IGF stimulation and as I tried to find some mechanism of this transport I found another articles about translocation of cytosolic proteins into mitochondria (GSK upon EPO treatment, HSP60 upon dehydratation).
I have always been taught in our cell biology lectures, that nuclear-encoded proteins are imported into mitochondrial matrix predominantly co-translationally and that only small portion of proteins are translated on free ribosomes but are kept unfolded by chaperones and then, still unfolded, transported into mitochondria, where they acquire native conformation.
Since this is not main focus of my work, but I find it interesting, I want to ask if somebody knows how well studied and how common this translocation of mature cytosolic proteins into mitochondria is and weather other such well-known proteins do this. And maybe suggest some review/article on this topic.
Thank you very much
Hello everyone, I have a question about the translation rate. I want to check the translation rate of isoforms belonging to one specific gene. For example, isoform A and isoform B of X gene, which one goes ribosome more and the translation rate is high. Can you give advice about technique and research paper? Thank you :)
translation process need amino acid add for elongation of polypeptide chain
Hello everyone, I recently came across something very peculiar called internal ribosome entry sites (IRES). These secondary structures are present at the 5' end of many viral as well as mamallian mRNAs and are required for cap-independent translation (ex- at the time of apoptosis). Can somebody explain how the mRNAs are folded into such complex secondary structures? Are there any factors involved that regulate this folding or the mRNA itself catalyses this reaction?
Experiment sample : Rhizobacteria.
After screening and isolation and the extracted of bacterial DNA by using 16s ribosomal technique to get sequencing result (in the form of nucleotide sequence).
Hi,
I want to separate 60S from 80S ribosomes on a polysome gradient. I usually run 10-50% sucrose (3h at 35k rpm), but then they bleed into one another and the resolution is not good enough to tell if my protein binds to 60S only or to both 60S and 80S. I have also tried 10-25% (4h at 35k rpm)and 10-30% (3h at 35k rpm), but they either completely get rid of the 80S or they still keep the 60S and 80S together.
Hi everyone,
I was wondering what antibodies or pcr primers I would have to use to control my fractions in a polysome profiling experiment ?
A quick search highligthed antibodies against small and large ribosomal subunit like rpl17a and rps3. And anti-Ssb1 for soluble fraction ?
Can you me give me advice on this topic please ?
Thanks,
Philippe.
Is it possible for ribosomes to recognize mRNA and initiate translation without the shine-dalgarno sequence and initiation codon? In the Nirenberg -Matthei experiment with poly U mRNA and E.coli cell free systems that helped in cracking the genetic code, there is no mention about these requirements. My doubt is that how was it possible to initiate translation in a poly U mRNA (or any other synthetic mRNAs) without the recognition and initiation signals?
I want to targeted MD simulation of macromolecule using AMBER software .As i am quite new to MD simulation field and of such a big complex .
I intended to perform polysome sequencing upon FAC-sorted C. elegans cells, but the required number of cells is not achievable for my application. Ribosome nascent-chain complex (RNC) sequencing seems like a viable alternative for which fewer cells are needed - but does anybody have an estimate of how much the minimum requirement is?
I use 4 concatenated molecular markers for phylogenetic inference. One of them, ribosomal 16s, shows a large amount of indels in my group of study, and I would like to use that information which is otherwise lost.
Could you point me to a thorough description of the best procedure for coding gaps as an additional partition and incorporating them in RAXml/MrBayes (substitution model, etc.)?
Ribosomes are described as ribonucleoprotein structural complexes with the definite function to direct protein synthesis. If the structure that is supposed to make proteins already has proteins as part of its composition, where were those proteins made giving that the protein components of ribosomes pre-date the protein-synthesizing apparatus?
I am using Trizol to do RNA extraction from peritoneal tissues (rat). The samples were stored in RNA later. I followed the Trizol protocol exactly but i get two (18 and 28s) for some samples and only 1 band for others. The RNA extraction for the samples were done on the same day and at the same time. Can anyone please help.
Thank you.
The En3hancer spray is no longer sold. Does anyone have a protocol for performing 3H-pulse chase analysis of yeast rRNA where the nylon (no the gel) is subjected to other tritium enhancer ?
Hi everybody!
The whidely used method to quantify the various ribosome fractions (40S, 60S, 80S and polysomes) are by sucrose gradient and fractioning.
Unfortunately this techniques uses very high amount of cells (more than 10 million or even more).
I need to make a knockdown in a suspension cell line of a gene throth siRNA and than to evaluate the relative 80S/40S/60S ratio (not interested in polysomes) but It is difficult to knockdown such high amount of cells.
Does anybody know another technique to quantify the ribosome fractions starting with a lower cell quantity?
Enzymes are synthesised as zymogens and then make a transition to active form when required, is this true for every enzyme or are there any exceptions to that?
Protein biosynthesis in an RNA world could have had the following components: ribozymes instead of ribosomes, flexizymes instead of aminoacyl-tRNA-synthetases, mRNAs, tRNAs, other kinds of RNAs. Is there anything more needed for protein biosynthesis? What?
(Does the environment of early genetic code evolution have enough ATP? Are proteins needed for proper tRNA folding?)
Are proteins needed for protein biosynthesis or is RNA in the right environment enough to build a translation process?
Thanks in advance.
The EM image of E.coli mRNA being transcribed by polysomes by Miller et al. in 1970 became iconic The sample preparation, however, seems particularly involved.
I want to know what options I have today if I want to image transcription (from a plasmid) and/or translation in E. coli?
Thank you very much
Title explains itself. I am trying to find information about a question in my mind.
For example if we divide a cell by regions like west east noth south, a tRNA going out to look for its spesific aminoacid, it advence in cytosol(for example, to the east from the west) and find the a.a it interacts(at east region of the cell), take it to the ribosome(for example at north of the cell) and take off.
But, how?
How tRNA knows that a.a is at east and how it knows the way to ribosome after binding a.a?
is there something like electrostatic forces made it to advence in correct direction? and cytosol act as conductor?
Could you give me articles about this issue, I could not find a study other than interactome studies but interactome studies is not the answer i am looking for. Interactomes only answers if molecules are interaction close. But I want to know how distant molecules find each other?
Thanks and regards....
I read an article where they said, they used 20-25 OD A254 sample for a ribosome separation on sucrose gradient. I know this is an important parameter but I am not getting it how much does that mean? Can anyone explain it please?
Thanks
Hello folks,
I'm interested in looking at ribosomes by negative-staining, and I understand that a lot of people use the FEI Morgagni for this purpose. However, the only TEM that I have access to is a Jeol 2000FXII. Is there anyone here that has used this EM for looking at ribosomes (or other macromolecular complexes?)
Best wishes,
Pramod
Dear sir:
Regards
Last year, I've studied the Tet resistance among Aeromonas spp. and I found that all tetracycline-resistant isolates carried at least one of the tet genes examined. Among efflux genes, tet (A) was most commonly observed in isolates, followed by efflux tet gene (tet B and C) and ribosomal protection protein (tet O). However, efflux genes tet (D and G) were not detected in any of the isolates.
look at the link below:
http://www.bldeujournalhs.in/article.asp?issn=2468-838X; year= 2017; volume= 2;issue=1;spage=22;epage=28;aulast=Al-Charrakh
Regards
Prof. Alaa Al-Charrakh
Babylon Univ., Iraq.
I am in an academic research project, where we want to introduce a gene that codes for obtaining an enzyme that degrades phenols in the organism P. Infestans, so we want to introduce a construct together with the enzyme of interest in the organism, and this It leads me to ask myself the following question: We want to introduce the gene near the ribosomal site since it is a very conserved region and therefore this would prevent the gene from being maintained in the organism, so could this be possible? Or is it better? place the construct in another part of the body's genome?
Greetings.
What is pathophysiological Significance of mutations in the Ribosomal DNA (rDNA)?
Hence in human molecular genetics, we focus on mutations/variants in the genes encoding mRNA and then proteins to predict their possible impact in genetic disorders.As rbosomes are assemblies of proteins and rRNA molecules that bind to and translate mRNA molecules to produce proteins.
1. If there is a mutation/variation in these genes encoding rRNA what would be their possible impact?
2. If the sequence of the transcribed rRNA is disturbed, how can it disrupt the ribosomal assembly i.e. binding of rRNA, ribosomes and mRNA?
3. If this assembly is staggered, then the transnational process might be definitely affected?
4. Is there any genetic disease reported with mutations in the genes for rRNA?
Membrane proteins can be inserted into membranes via different mechanisms and in different orientations. I am specifically interested in mammalian cells (not bacteria or yeast). Does anyone know how high the percentage of type I, type II, and type III proteins is among mammalian integral plasma membrane proteins? Is there a review out there that has some numbers?
I am removing enzymes, amino acids and other small molecules from ribosome complexes through density gradient centrifugation with sucrose. I have very little ribosome sample so I would behave with volume as small as possible to recycle my samples. Could anybody tell me what's the minimum volume of an efficient density gradient centrifugation? 100ul worked for me, but I am not sure if I behave 50ul still works without losing my ribosome.
I am basically interested in understanding the graph obtained after ribosome profiling and how can you know from the graph in which translational state are the mRNAs?
Hi, dear colleagues,
Recently, I really confused about ORF in plasmid. I checked my target sequence in my plasmid, in fact, after the promoter there are two possible ORFs, 1st ORF is close to my promoter, as a 56aa peptide, 2nd ORF is my target protein 219aa [CmR].
In my opinion, if the 1st ORF is translated, the 2nd ORF will keep silent and no protein production. The fact is that the 2nd ORF is translated well, the 1st ORF is silent, even it is closer to promoter. The sequence file is attached.
My questions,
Whether there is a limit for ORF length?
>In SnapGene default parameter, the ORF should bigger than 75aa, but I’m sure the short peptide could be translated. Maybe in plasmid not ??
Always the first start codon [M] could be recognized during the translation, I wonder whether the small ribosomal could choose the start codon that is included in a longer ORF. That’s really incredible, if so.
Thanks for your any help~
Yunlong
I am currently using a lac-based vector for periplasmic expression of proteins in E. coli. I now want to re-design the vector and express the proteins under the control of the T7 promoter in a DE3 strain. I found a lot of different vector sequences and am a bit confused which sequence parts I really need in my vector for IPTG- inducible expression of my proteins.
T7 promoter only? T7 promoter and T7 terminator? T7 promoter plus lac operator? (If I got that right, the T7 RNAP is under the control of a lac promoter in the bactrial genome, so I shouldn't need a lac operator in my vector?) Which ribosome binding motif?
I am looking forward to your replies!
If I mutate a start codon, how far along a transcript will a ribosome scan for a downstream AUG before it gives up and falls off?
Hi all,i'm currently studying a brain sc-seq data by seurat package,and my cluster analysis seems to be profoundly influenced by genes that relate to ribosome(named Rpxxx).A few clusters is characterized by expression of these genes,and known markers of cell types seems to have little relation with these clusters.I have regressed cell cycle by ScaleData function,so i'm confused about what these ribosome-related dada means.Shall i delete these data?Or shall i study these ribosome-related clusters in other way?Thank all!
BTW,i have deleted blood cells from my data by hemoglobin-related genes
Hi, I am new to this. I have done sequence alignment of modified tRNA genome sequences from E. coli and Mycobacterium tuberculosis against the whole genome database of tRNA and have not found any hit showing the similarity in their sequences. I am trying to do Mycobacterial translational complex studies and would like to know how relevant it is to reconstitute Mtb ribosome with E. coli tRNAs. My query protein binds to tRNA as well as mycobacterial ribosome in vivo. I am performing biochemical and structural studies.
Can anyone point me to some work in this area? In kid terms, how would a mitochondrion, nucleus or vesicle feel to the touch? What methods are used to study this? What about smaller things like proteasomes and ribosomes?
I am unsure what the differences between IRES1 and IRES2 are at the sequence level, whether IRES2 is a development of IRES1, and what the consequences are of any modifications to the original sequence.
I'd be very grateful for any pointers or literature references.
Thanks a lot,
Jonathan
Depending on the answers of my first question, I would also like to know how to get rid off these ribosomes from the mRNA after doing IP of a particular protein. In precise, I want the condition where I can only get the particular protein bound to translating mRNA.
Why all the amino acids that are ribosomically incorporated into proteins exhibits L-configuration not D-configuration?
All enzymes undergoes post-translational modification (PTM) to become a functional mature enzyme. Is there any enzyme which doesn't undergo PTM? any exceptional enzyme?
Just wonder is there any evidence exist between ribosome copies and number (or diversity ) of the proteins as end product in a cell?