Science method
Confocal Fluorescence Microscopy - Science method
Confocal fluorescence microscopy is a microscopic technique that provides true three-dimensional (3D) optical resolution. This technique has gained popularity in the scientific and industrial communities and typical applications are in life sciences, semiconductor inspection and materials science.
Questions related to Confocal Fluorescence Microscopy
Thank you for reading my question.
Are newer RFPs like mRuby3 or mScarlet better complements for eGFP? Or should I stick to the eGFP/mCherry pair?
I am new to FRET and I am planning a FRET experiment to study interactions between a mutated soluble mammalian protein (which forms aggregation) and a wild type protein in cytosol.
My cells are HEK293T transferred with the two strains of plasmids. Confocal Laser Scanning Microscopy will be applied to conduct a short live-cell imaging. The microscope is Zeiss LSM880.
Feasible emitter wavelengths are: 405 nm, 488 nm, 543 nm, 594 nm, and 633 nm.
Both vectors carrying a fusion protein of object-eGFP have been already constructed and validated in previous experiments.
After conducting some literature review, it seems that CFP/YFP pairs and GFP/RFP pairs meet my needs. Considering that I have object-eGFP vectors and the blue emitter (488nm) is not working, I prefer a GFP/YFP pair.
Effective pairs like mNeonGreen/mScarlet-I and mClover/mRuby3 have been validated.
To the best of my knowledge, The pair eGFP/mCherry has been well-established and reliable. However, relatively low Quantum Yield and Extinction Coefficient of mCherry still raise my concern.
Thank you in advance for your reply!
I have prepared an oil in water nano emulsion and would like to visualise it for confocal microscopy, however i dont have any fluorescent compounds in my emulsion. I know that nile red is a lipophillic dye and would like to know how to stain (protocol with concentrations) it so that i can visualise the droplet morphology better using confocal/fluorescence microscopy?
I fabricated microbeads using hydrogel (20-50 micrometres) to mimic human cells' size and mechanical properties; I want to stain them to be able to recognize them under an optical microscope. Which staining should I use? I don't have confocal microscopy at the moment, so I think fluorescence inks will not be helpful here; I am using the microscopic camera to view those beads
Hi! I was staining my tissue with both PI and Calcein AM dyes, but I realize a lot of cells were stained by both PI (red) and Calcein AM (green), for example, the cell at the center of the attached image. I thought PI stained for dying cells while Calcein AM stained for live cells.. so I am curious to know if those cells are actually dying or alive (or both...??).
Also, some of the red signal was really blurry.. could it because those cells at the late stage of dying already (DNA was fragmented and released into interstitial space?)?
Thank you in advance!!
It doesn't matter the color of the fluorophore or the localization (nuclear, cytoplasm). I just need a good one, because the ones I tested so far didn't give great result. Any suggestions from people with experience in that. Thank you very much for helping.
Hello guys, I'm meeting a big problem that my cytoskeleton staining always colocalize exactly with nucleus. I used aptamer conjugated with Cy5 or Alexa647 (I tried them both) to target cytoskeleton, but all of them targeted to nucleus and overlapped with Hoechst (nucleus staining).
For my opinion, I assume that the problem might caused by different charges of aptamer and nucleus. But I don't know how to avoid this non-specific binding. Do you have any ideas to fix it? Thank you so much for your help!
We are seeking to count cell death stain from confocal image stacks; however, Neurolucida is quit expensive. Are there any alternatives that you recommend? Is ImageJ viable?
Does anybody know of a live membrane dye which works well with tissues and isn't internalised or pumped out of the cells in a short time frame? I'm looking to perform 1 or 2 hour imaging experiments on live vessels (ex vivo), however most of the live membrane dyes my lab has previous experience with are only really suitable for very short-term experiments. I should also add that due to the nature of the experiments I plan to carry out, using probenicid to retain the dyes for longer won't be an option.
Any advice would be greatly appreciated! Thanks!
I am trying to immunostain whole (micro)tissue samples, and I think there is an antibody penetration issue. Would longer permeabilization procedures or harsher permeabilization agents help with this?
Thanks for sharing your experiences!
Hello,
Could you please recommend DAPI concentration for nuclei staining in mesenchymal stem cells during chondrogenic differentiation?
I have used 1 μg/ml DAPI for 6 min, then as usual PBS washing. but I got fuzzy colour and lots of unspecific binding (but still fuzzy).
If that matters - the brief protocol:
- PBS washing 3x
- permeabilization 0.2% Triton X-100/PBS, 4 min, PBS washing 3x
- blocking 0,2% BSA/PBS 1h, PBS washing 3x
- staining of collagen I, II and X (each with a pair of antibodies, with 3x washing in PBS in between steps).
Thank you!
Hello everyone.
I am going to analyze differentiated cardiomyocytes (CM) via confocal microscopy. However, I struggle to attach CM properly to the glass slide/plate. Although I coated the glass slides with LM-E8, the CM could not attach firmly and started to detach after adding 4% PFA to cell staining.
Does any appropriate coating for CM culture over the glass?
I appreciate any recommendations and comments.
Best,
Fatemeh.
Dear all,
I'm just starting to teach myself how to make macros to process confocal images in ImageJ (FIJI). I've managed to make a macro that does most of my processing using the record macro function (it has already saved me an enormous amount of time!), however I'd like to add brightness and contrast adjustment to the processing.
My current process involves:
1. Splitting the multi-channel images
2. Applying grey LUT to all
3. Saving all in a temporary folder
4. Creating a merged image of the channels is various colours
5. Creating a panel with split and merged images
6. Adding a scale bar
7. Saving and closing all windows
However, when I look at the recorded code produced by the brightness and contrast adjustment dialogue, I get code which doesn't seem right to me:
//run("Brightness/Contrast...");
run("Apply LUT");
I've also tried the following code, however it automatically maximises the contrast, where I'd like to use the brightness and contrast adjustment to reduce background in some channels.
run("Enhance Contrast", "saturated=0.35");
Is there anyone else using macros for brightness and contrast adjustment in ImageJ?
Many thanks,
Sam
Hi everyone.
I am facing a very intriguing phenomenon. My immunofluorescence and confocal microscopy analyses showed a nucleus that is positive for MnSOD (which should be present only within mitochondria). Please, find attached representative images of cells stained for either nucleus or MnSOD positivity, along with the merged image.
Of note, it should be highlighted that the anti-SOD2 primary antiobody we used is extremely specific and that Western immunoblots showed only the specific band.
Did anyone else find MnSOD (SOD2) in the nucleus in some experimental or physiological condition?
Thanks a lot in advance.
Stefano Falone
Hello everyone!
Looking for some useful tips for fluorscence/confocal microscopy of plant root tissue.
I am mostly working with the root tissue of Arabidopsis thaliana where I take confocal/fluorscence images of plant root samples expressing proteins of interest with GFP/mcherry tags also often with Propidium Iodide/DAPI stains. Usually, I don't fix the tissue and take images as such and here are a few problems I face most of the time and I will be glad if you can advise me some suggestions to fix or minimize the following issues-
1.The amount of protein expression in various layers of root tissue is variable ranging from low to very high expression so in a Z-stack often I get highly staurated layers or if I adjust laser power/analog gain to minimize that oversaturation I end up losing signal where protein is less expressing. What should I do in such case?
2. how do you prepare live tissue for imaging, like if you use any specific buffer that does not change the physiological state of the plant tissue but helps in maintianing the GFP fluorescence for a longer duration?
3. Also, no matter how carefully I prepare the slide I never get the root tissue lying uniformly over the slide which means I get regions with good focus and in perfect plane wherease at some regions it gets blurry, out of the focus, how to avoid it?
4. I too have problem with DAPI staining of the root tissue as in most cases barely I see any nuclei but most often I end up with images showing DAPI at the cell borders just like Propidium Iodide.
Thanks a lot in advance for all the incoming beautiful suggestions.
Hello everyone,
I am looking for a nuclear marker, like a transcription factor, expressed in human enteric neurons.
I would like to characterize my human iPSC-derived culture with immunofluorescence stainings. The problem is that my neural progenitors give rise to enteric neurons and enteric glia (GFAP+), so I cannot use Sox10 throughout the whole maturation. So far I haven't found anything this specific.
Does any of you have any suggestions about a good antibody, or a marker that would be useful in this case?
Thanks a lot in advance.
Hello,
I am currently using Jasplakinolide to treat my Be2-C cells and I want to measure cell viability. I want to fix the cells in PFA and mount them with Vectasheild. However, I have seen a lot of different ways to use PI with PFA fixation, and I am a little unsure the best way to do it. I am thinking I will first treat the cells with PI by adding it directly to the media in the incubator for 1 hour and then fix with PFA, stain with DAPI, and mount? Any advice/suggestions?
Also, has anyone used this and also stained for phalloidin? I am worried there will be too much cross talk to use both.
Thank you in advanced :)
Hey!
I have a FRET system between the inner and outer nuclear membrane, and I was wondering if I stained with DAPI or any other fluorescent antibody if that would mess up my FRET readout? As of now I am using acceptor photo bleach and I am working with neuroblastoma cells (Be2-c). I can do either fixed or live cell imaging, depending on which would work best if this even possible.
Thank you!
I made a script to try to convert 4 colour nd2 files (not stacks) to run in the process> batch function of Fiji (see https://www.youtube.com/watch?v=fU104OU3Kk4 at 3:28) to create single tiffs, of all colours, merged. When I check the results in Fiji I get only the DAPI channel from the script below. However, if I run the macro in Fiji itself without the batch command, just on a single open nd2 file, I am left with the file that I want! So I am not sure why it is only saving the DAPI. Does the batch command save the earliest tiff that it sees and not let the macro finish? I am an inexperienced Fiji user so I am not sure what I am doing wrong.
Code is:
title = getTitle();
run("Split Channels");
one = "C1-" + title;
two = "C2-" + title;
three = "C3-" + title;
four = "C4-" + title;
run("Merge Channels...", "c1=["+one+"] c2=["+two+"] c3=["+three+"] c4=["+four+"] create");
run("RGB Color");
selectWindow(title)
close()
I have isolated mitochondria from tissue. I would like to check them by fluorescence imaging. Can I use Mitored dye for these isolated mitochondria?
I want to detect intracellular bacteria in urine by confocal fluorescence microscopy
I would like to build a lateral projection of a z-stack with the correct linear size ratio for the final illustration, how can I change the pixels size ratio in ImageJ?
I'm observing a small tumor with confocal. I imaged the expression of a fluorescent protein, which is supposed to express everywhere. However, I can see some vague DAPI signals, where I can't observe the fluorescent protein. Is this normal? If so, wouldn't the images that I obtained for a single layer be always interfered with vicinity layers?
I am wondering why my hepg2 cells nucleus looks fuzzy and unclear under the confocal?
Is this mycoplasma contamination?
I used Hoechst staining, 1:200 dilute with 3%BSA.

Hi everyone,
I am working with cells labelled with Mcherry (membrane) and EGFP (cytoplasm). Following my experiment, I want to quantify the green signal which relocated to the membrane. How can I extract and quantify in Fiji the amount of green signal on the membrane (e.g. where the red signal is)?
Thanks in advance for all your answers and suggestions!
Sissi

I was wondering if someone could share a protocol to fix my plant tissue that has some GFP-tagged endophytic bacteria. Can I fix it with aldehydes? If so, how long I can presente the GFP bioluminescence?
Best regards,
João
I have z-stack confocal images of whole-mount tissue IF staining, marking the tight junction of the cell. And I would like to do 3D reconstruction of the cells to measure cell circularity and area using Imaris.
However, since it's challenging to mount my tissue perfectly flat, my z-stack images are slanted.
This makes the cell reconstruction challenging since the cell membrane signal for a certain cell is spread across multiple z slices. I attached few screenshots to demonstrate.
I'm wondering if there is a way to rotate my z-stack image and reassign the xy reference angle to computationally make the image flatter?

I am using a BD pathway imager. When the spinning disk gets in the light path I get very blurry images( see attachment). Does anybody know how to fix this issue. First image is with regular fluorescence image. The second is with confocal at the same focal plane.


I am working on amplifying a signal for confocal imaging using the Labelled Streptavidin/Avidin Biotin (LSAB) technique and ran into a surprising outcome: No staining. This is odd as I've employed this stain previously using the traditional indirect method and see decent specific signaling so I thought by amplifying the signal I'd get better, not worse, staining. Below I'll leave the basic outline as to my design (barring washes in between and counterstains).
- Incubate mouse anti-antigen primary antibody at 4 degree overnight (1:100; used at same concentration as in indirect method staining)
- Incubate biotinylated goat anti-mouse secondary at RT for 90 mins (1:150)
- Incubate Cy3-conjugated streptavidin for 90 mins (1:1000)
Any tips as to how to optimize this protocol and get it to function would be greatly appreciated!
When performing an indirect immunofluorescence experiment, it is not favorable to use a primary antibody raised in the same host as the tissue you will be staining (ie. mouse anti-x on mouse tissue, rat anti-x on rat tissue) due to the increased background you will get from the secondary antibodies binding with endogenous IgGs. Logically, it follows that if I use a fluorophore conjugated primary antibody for direct detection, this issue will not arise despite the antibody being raised in the same host as it is reactive for because there are no secondary antibodies involved. Can anyone verify that my assumption is true? If not, please share with me why my rationale was incorrect. Thanks!
I am working with FFPE sections of mice ileum and am trying to image changes in the mitochondrial ROS production between a treatment and control group. I'm not sure if there are any stains or antibodies that can specifically target mtROS once the tissue is fixed and I have seen another researcher propose using MitoSox Red on tissue prior to fixation to get an assessment of mitochondrial ROS in the live animal. Has anyone else tried this successfully?
We also dabbled using a pimonidazole hypoxyprobe as a measure of total ROS, however the staining in the ileum and colon differ for physiological reasons. If anyone has some ideas as to how to better optimize this stain or an alternative method of detection that works well with ileal tissue, please let me know!
Hello,
If a sample has both GFP and RFP expression, and GFP is activated by a laser, then will its emission light be absorbed by RFP and cause RFP to shine? Another toy example would be, will the emission light of Alexa Fluor 555 become the excitation light of Alexa Fluor 647?
If not, why? If so, how could this be resolved during confocal imaging of dual-color? I don't think this is FRET, as FRET doesn't involve an actual photon being emitted..
Thank you so much!
Hi,
I'm having problems with the Recombinant Anti-MAP2 antibody from Abcam (Recombinant Anti-MAP2 antibody [EPR19691] (ab183830)) antibody on my frozen, 4%PFA perfused, mice brain tissue.
I was wondering if someone could elaborate better the note from the application part of the data sheet; Antigen retrieval: Heated citrate solution (10mM citrate PH 6.0 + 0.05% Tween-20).
To my knowledge AR is not used with frozen tissue, as it is to rough, and can damage the tissue, while also causing tissue detachment from the glass!?
Does someone have any experience with AR and frozen tissue?
I did try using 1/100 dilution, 0,2% triton X-100 and 4%PFA fixation as described in the images part of the datasheet: Immunohistochemical analysis of 4% paraformaldehyde-fixed, 0.2% Triton X-100 permeabilized frozen Mouse Cortex tissue labeling MAP2 with ab183830 at 1/100 dilution.
However I did not observed cytoplasmic staining of my neurons (image as uploaded for reference)!!
My protocol was the following:
4%PFA fixation, 15 min RT
3xPBS, 5min
10 min PBS+0.2% Triton x-100
Blocking solution (3%BSA, Without Triton x-100, 1h)
Anti-MAP2 1:100 dilution in 1%BSA (try both overnight and 2h RT)
3xPBS, 5min
AF-647 1:500, 30-45 min RT
3xPBS, 5 min
DAPI, 5 min
3x PBS, 5 min
dH2O, 5 min
Antifade and coverslip.
The antibody works excellent in IHC-P.
Thanks for the help,
Fran

Hi Everyone,
I am trying to characterize the microstructure (localization of carbohydrates, fats, and proteins) of coconut milk powder using confocal microscopy. From the review of the available literature, I have found that fats and proteins can be non covalently labeled using fluorescent dyes. However, I am having trouble deciding the labeling protocol for the carbohydrate. Two common methods that I have seen in the papers are as follows:
1. Use antibodies or lectins conjugated with fluorophores to label carbohydrates
2. Covalent labeling of carbohydrates with FITC
If I use the first method, the signal from antibodies or lectins may interfere with the signal from protein present in the sample.
In the second method, the FITC (probe used for covalent labeling) also shows an affinity for proteins. So, I am afraid that the use of such a probe may also interfere with the protein signal (from the food sample). Is there any other probe available that I can use?
What would be the best way to label carbohydrates such that I can simultaneously image three components in the food mixture?
Thanks.
I did a Western Blot for a protein that should only be present in hematopoetic cells. The WB shows a very clear picture, present in k562(hematopoetic) and not present in HeLa and 293T. However when using the exact same antibody for IF on HeLa cells we do see clear signal from our protein of interest (see picture). Can anyone provide insights to how this is happening? in controls without primary/secondary antibody we see no signal.

Hi! I am about to use AGR2 protein in my antibody display system. I want to check wherther protein can bind to cell surface displayed antibody or not. I need suggestions on how to fluorescently label (with red dye) AGR2 protein in order to view binding and also if someone could suggest efficient and cheap method to label protein that would be really great. please give me some insight and useful tips!
Thank you
Hello,
I have dozens of confocal images to process in Fiji. It is neither practical nor a productive use of my time to do them all out by hand, so I want to use a macro. However, I am fairly new to research; I only started a few months ago, so I don't have much experience writing macros in Fiji. What I'm doing is taking an image split into 3 channels, applying the grayscale LUT, and z-projecting to max intensity. Then I manually adjust brightness/contrast depending on the quality, merge channels, and stack to RGB. I also add a scale bar, but that I adjust manually i.e. 10 microns, 20 microns, etc.
Does anybody know if such a macro exists (i.e. in the Fiji user guide) or how to write such a macro? I have made one by using the record function, but that turned out to be inefficient because I had to change the file name every time I ran the macro. is there a way to get around this? Also, how would I incorporate into the macro the pauses when I have to make manual adjustments?
I use Richardson-Lucy deconvolution algorithm (flowdec package) with custom generated PSF.
And after several iterations, granularity is observed in the cytoplasm, but the protein must be distributed diffusely. Random noise fluctuations resulting from deconvolution become bright points in the cytoplasm.
How can I reduce noise influence and prevent occurrence of an artifacts? And/or could I implement regularization (Tikhonov regularization f. ex.) in flowdec package?
Looking for recommendations on what to use for imaging spheroids and organoids using a confocal scope. Would it be feasible to carefully transfer spheroids to glass bottom dish with coverslip, or better to use glass bottom microplate? Or another method altogether? I am experienced in the formation of spheroids but not so much the characterization. Thanks in advance
Basically I would like to visualize (either by confocal or electron microscopy) differences in transcriptional activity inside a tissue, to see if I have cells that are more active in gene transcription then others . I am using an entire organism, a marine invertebrate animal, not a cell culture.
For ex. a staining method that will show euchromatin (non-condensed DNA) activity in the nucleus?
Also, somebody mentioned to me using the bromouridine labeling (Bru) to asses changes in RNA synthesis. Did anyone try this method on an organism?
Thank you !
I have a protein (conjugated with FITC fluorophore) that may adsorb and cover the bacterial cell surface or be taken up into the cytoplasm.
Using confocal (fluorescence) microscopy, how can you tell the difference with any confidence? (LSM700)
The cells are rather small at 200 x 1200 nm.
I'm relatively new to microscopy imaging analysis so I'm seeking some help! I have z-stack images (.czi files) from zebrafish using a Zeiss LSM 880 confocal microscope at 40x water immersion objective. My advisor has suggested using the ZEN software to do a maximum intensity projection and then using orthogonal view. The images still look "messy" after conducting these steps in the ZEN Blue v3.1 software, so I'm wondering if you have any suggestions or protocols to analyze images. Ultimately, I would like to compare fluorescent intensities, myelin sheaths/olig, and/or internode length across my samples. (also- should I implement a deconvolution step?)
Thank you in advance!
I have a grayscale image of which the original RGB image was formed by combining Green and Red channels which represent different types of cells (live/dead). Splitting the channels in RGB mode is giving Red, Blue and Green to all the cells. I want to confine green to the live cells and red to the dead cells using ImageJ.
Dear fellow image enthusiasts,
I just recently started doing a lot of confocal fluorescence microscopy imaging and I am getting good images, but I still thought about increasing their quality. I immediately considered deconvolution methods. Only problem is, I have never used a deconvolution tool before.
Could anybody help me out with kind of a "quick start protocol"? I want to use imageJ and donloaded deconvolutionlab 2 (http://bigwww.epfl.ch/deconvolution/deconvolutionlab2/) as well as the PSF generator they recommend (http://bigwww.epfl.ch/algorithms/psfgenerator/).
First thing:
Which method should I chose to generate a PFS which would work for deconvoluting my 2D image?
Which parameters do I have to know (both while taking the image and for generating a proper PSF)?
- I do know the refractive index if my immersion oil, do I also need to consider the refractive index of the NPG used for covering and/or of the glass cover slide?
- I also know, the NA
Which method should I chose for decovoluting the 2D image?
I made a couple of test runs, but the output looked much worse than the input and particularly proper colour information was lost.





+1
I am working on Symbiodinium cells (symbiotic dinoflagellates) and am trying to calculate the mitotic index. All the papers I have referred to use DAPI. I want to know if there are other stains or techniques that an use to distinguish between the doublets and just single cells. Is the resolution of DAPI good enough for me to see the mitotic spindle?
I have localised a protein in Endoplasmic reticulum of Saccharomyces cerevisiae. I have tagged this protein with HA tag. Now i want to label this protein with anti-HA fluorescent antibody for FACS and microscopy purpose, If anyone can suggest me a protocol for this then it will be a great help.
Thanking you,
I have been imaging many z stacks of Drosophila mid/hindgut tumors using a leica confocal microscope. I'm wondering what the best way to quantify these tumors would be. They are GFP+.
I know both leica and image J have quantification programs. Any preference? Any recommendations would be greatly appreciated!
I need to image cyanobacteria using fluorescent microscopy, I have easily done this before with good results using the fluorescence of chlorophyl on a traditional fluorescent microscope. However, when I attempted to do this on a confocal system I had very poor results when imaging Cy5.5 along side the chlorophyl.
What excitation and emission wavelengths should I use for chlorophyl, specifically for cyanobacteria?
Should I use a different second dye like NBD or rhodamine instead of Cy5.5?
I'm running a study in which I will inject some nanoparticles into mice and then section the lymph node and visualize it with confocal. Does anyone have the protocal for the lymph node sectioning and staining? Thanks.
Hi,
I'm working with a Leica SP8 confocal microscope,
and I'm trying to analyze the sum intensity of z-stacks (16-bit).
For particle analysis I'm making a max intensity image first which later I redirect its ROIs to the SUM.
A) in certain images I'm using the "make binary" and the output image is kind of clean B&W, in other images (from the same project and settings) I just get a cloud of pixels (I guess it's over saturation) among my cells.
B) in order to maintain a proper workflow I'm trying to batch analyze my files (8-bit->8-bit->threshold->analyze particles etc. etc.).
Any recommendations?
I would like to analysis the S. aureus biofilms to know the effect of certain quorum sensing inhibitor on ultra structure. Could someone elaborate the method to prepare sample for TEM analysis (Fixation, dehydration, drying, staining, embedding, ultra sections etc)? Many thanks for your valuable time and help
Hi Everyone,
We are attempting to study actin/cytoskeletal function under different conditions in live cells. Does anyone know of an existing cell line that stably produces GFP-actin for purchase? I know that this can also be accomplished by transducing cells with viruses if you have any products in particular you recommend.
We are also ok with accomplishing this by labeling. I have tried BacMam and SiR-actin with fairly low efficacy in HeLa cells just as a trial (ideally something that works with easy to transfect and primary cells preferred). Considering trying LifeAct products but not sure if they are any better. Any recommendations on products and exactly how you applied them (number of cells, volume, inc time, concentration, etc) would be highly appreciated.
In a nutshell, I cannot get a decent signal for Alexa Fluor 647 on the epifluorescent microscope I am using.
Setup:
Nikon Eclipse Ti-S inverted microscope
Nikon Ti-FL epifluorescence illuminator
Lumenera Infinity 2-1RC CCD Camera
BFP-A-Basic filter cube
GFP-1828A filter cube
YFP-2427B filter cube
TRITC-B filter cube
Cy5-4040C filter cube
Samples:
Mouse leg muscle cryosections (6um thickness) mounted in ProLong Gold medium, stained for various muscle protein epitopes with AF350, AF488, AF555, and AF647.
The problem:
The AF647 should sit perfectly in the far-red channel (Cy5-4040C), but it is extremely dim. It cannot be seen down the eyepiece at all, and only with much imagination using the camera, with no ND filters, at 2000ms exposure and with contrast and brightness cranked all the way up later in ImageJ. Semrock, the makers of the filter cube, even claim that this filter set is ideal for use with AF647, and AF647 is supposed to be bright and relatively stable.
Things that I have tried:
- Replaced mercury lamp. Old lamp had <100 hours on it, but upon examination it was quite blackened. Replacement, alignment and refocusing did improve brightness slightly, but not enough.
- Tried a secondary antibody with a different fluorochrome. Worked really well, hence it is not the antibody, dilutions used or the epitope that is the problem. Using a different fluorochrome is not a viable solution since I need to use the far-red filter cube to get all the stains in.
- Imaged a droplet of pure antibody. This is visible, even with the naked eye, but not as blindingly bright as the other fluorochromes. I.e. it can be detected, but in a proper stain it will be less intense than pure antibody.
- Imaged using a confocal microscope. Fantastic staining and brightness, just as it should be, but I can’t use a confocal long-term for other reasons. Again, this shows that there shouldn’t be an issue with the antibody or the epitope.
I am at my wit’s end. AF488 and AF555 are perfectly fine, AF350 is dim too but I’m pretty sure it’s because it’s slightly out of range for the filter cube, I might replace it with AF405. Is there something really basic I am missing?
I would be very grateful for any ideas or advice.
Dear All,
Can we use Tween 20 instead of triton X in 0.1% PBT-T(I assume T stands for triton X only) for immunofluorescence ?
Also, can tween 20 be replaced by tryton in IP wash buffer?
I found that the signals for my dye are weak in tissue sample, what is the maximum limit to which I can increase the detector gain and laser power percent for my dye?
I am going to mesure the mitochondrial calcium uptake with Rhod-2AM in permeabilized neonatal rat cardiomyocites, but the fluorescence signal is altered due to cell contraction, generating an artifact signal.
I read about 2 pharmacological agents to disrupt the excitation-contraction coupling: -blebbistatin and 2,3-butanodione monoxime. It seems to be that both pharmacological agents are not frequently used, and have been descibed different work concentrations.
What is the best option to inhibits the excitation-contraction coupling? and what is the work concentration?.
Hello there,
The literature is plenty of examples of immunostaining in live cells, but is hard to find the same in live organotypic culture. Does anyone have experience with this? Is there (a priori) any caveat about this technique?
Thanks,
J
By looking excitation spectra I would say no (~5% excitation), but its maybe enough in practice ? Has anyone already tried ?
Thanks, have a good day.
I would like to observe microgel particles inside fibres using UV microscope, thus I came acrross some difuculties when choosing the appropriate UV dye. The microscop can emit light at 405 nm, 488 nm, 555 nm and 633 nm. The dye has to be soluble in water, as other solvents like methanol and DMSO damage the fibres.
Dear all,
I've measured the diffusion coefficient of a fluorescently labeled glycosidase interacting with its substrate (a polysaccharide) by using fluorescence correlation spectroscopy (FCS). An example can be seen in the attached figure. The substrate was in its solubilized form and in saturating concentration (5 mg/ml). After FCS curve fitting, I found that 75% of the species corresponded to the free enzyme (Rh = 2 nm), while 25% were a much bigger particle (Rh = 37 nm). This value matches very well to the Rg of the polysaccharide (31 nm) according to the manufacturer. The effect of viscosity is already taken into account. However, since many enzyme molecules can be simultaneously associated to the carbohydrate, these 25% may not correspond to the actual proportion of enzymes in the bound state.
If my reasoning is correct, my question is: How to determine the average number of enzymes associated to the carbohydrate? I suppose I should analyze the photon counts over time, but I'm not sure how to do that.
Thank you in advance!
Best regards,
Gustavo
I have a set of confocal microscopy images. I am finding little difficult to conclude from the data, so need help from experts on this. I will upload the images as someone is interested.
Did anyone observe the suppression of low fluorescence signals from one organelle due to high signals from other organelles?
Hi, I want to quantify total tau, phospho tau and tubulin in the axon and dendrite of hippocampal and cortical neurons by Confocal microscopy analysis. I'll appreciate if you could suggest me the method to distinguish axon from dendrites, and the method to quantify them using imageJ software.
I have attached the image file/picture I scanned using microscope (red channel: total tau, green channel: tubulin, yellow channel: phospho tau). The image is focused on neurites.
Thank you.
Sincerely,
Saroj
I want to microscopic studies of cultured unicellular algal cells. I want to know which dye is most widely used for nucleus staining for algae.
Hi everyone,
I'm studying the gelation process of certain bio-polymers. When I form the final gels and cut it into small pieces for observation under the Confocal microscope. This process seems to hamper my gel structure. Is there any other process wherein I could form the gels in accessories that could be directly used for observation under the microscope. Please bear in mind that I have to heat the gels to induce gelation, so the material needs to resistant to heat. I have read of quart glass dishes can anyone suggest anything else?
Leica and Abberior say: ¨Please do not use Vectashield, Vectashield Hard set or (other) embedding media containing p-phenylenediamine as antifading reagent." (I do not understand why, but I have no choice to trust them)
Prolong Gold transforms the cells (10 - 12 um of the thickness) in 2 um pancake
The list of allies of a microscopist becomes short ...
I am currently writing my first master project. I have several images of tissue stained with Alexa Fluor 488, and I need to determine difference in fluorescence intensity between my control and sample group.
The image files are .TIFF.
Huge thanks in advance!
I would like to image differentiated Caco2 cells grown on a transwell and stained with fluorophore (ie Phalloidin -Alexa 488) without perturping the 3D morphology of the cells.
I have read that you can cut with a scalpel the filter and place it on a glass slide. I gess I should than add antifadding, but then? Should I add a coverslip? Will the coverslip not scratch out the apical surface of the CaCo2 cells?
Do you have a protocol to recommand? any suggestions are welcome
I have tried to fluorescently label graphene oxide sheets once with Fluorescein sodium salt and another time with Fluorescein isothiocyanate (FITC) by mixing 500 uM of Fluorescein (any of them) with 0.5 mg/ml GO overnight then washing with DI water 3X and then re-suspending the GO in DI water. After washing I take a drop of the suspension and place it on a glass slide, leave it to dry then look under the optical microscope using blue light (of wavelength 450-495nm) to excite it but I can't see it fluorescing while the Fluorescein on its own provide green fluorescence at this wavelength. I will be grateful to obtain any advice regarding how to fluorescently label GO sheets.
Thanks
I want to create a zero-order Bessel beam. But I am a little bit confused how will I create zero-order?. As far I know the most simple way to create a Bassel beam is with an Axicon lens. So my queries are below-
1. Can I use axicon lens to create zero-order Bessel beam?
2. Does apex angle of the axicon lens play a critical role to create zero-order Bessel beam?
3. Is there any other way to create zero-order Bessel beam?
Please help me. Thank you in advance.

I am interested in understanding how the surface area changes when adherent cells are brought into suspensions. I am looking for data supported responses.
Hi all,
I need expert opinions on this matter as my plant samples are detached during washing step of FISH. I used agarose as well as transparent nail polish for this purpose and to me agarose didn't work at all but nail polish works slightly. It provides a base to sliced plant tissue, when I put my hybridization slide into the washing buffer (50 ml falcon) and then in water bath; after heating up to 48 degree Celsius, the nail polish is no more stick and my plant samples swim in freely in the media. Though I doubt that it may not affect the hybridization but still I am not sure. Can you please share your practical experiences?
Thanks in advance
Chromosome counting using fluorescent microscope....
I am trying to isolate a monoclonal cell population by limiting dilution in 96-well plate. I want to isolate transfected cells which they express GFP. Are the cells reduce fluorescent properties by cell division or it is permanent? Should I mark my green cells in first two days prior to fluorescent disappear?
I’m looking for fluorescent dye with known diffusion coefficient excited by laser line 594. I need it to calibrate confocal volume in Fluorescence Correlation Spectroscopy (FCS) measurement where I use mCherry dye. Unfortunately, I haven’t line 561 which is also good for mCherry
Atto 590, Atto 594 or Atto Rho 13 look nice, but I haven't found their diffusion coefficients in literature.
Any help or suggestions would be greatly appreciated!
Regards,
I would like to be able to compare fluorescence measurements taken at different times in different samples directly with one another.
From previous experience I know that microscope settings need to vary between samples in order to optimise the image being taken. I use live tissue and variations in dye loading, tissue thickness, etc. prevent keeping the microscope settings (detector gain, amplifier offset, amplifier gain) the same between measurements of different samples. Obviously settings for different measurements made within the same sample are kept the same, and laser power and exposure time are also kept constant for different samples.
As such, is there a way to normalise fluorescence measurements? Ideally I'd like to know If I can normalise my fluorescence measurement of interest (e.g. TMRM) to some cellular autofluorescent species (something that shouldn't vary in my experimental conditions of course). Something essentially akin to the loading control used in Western blotting. I'd measure both my probe of interest and the 'loading control' under the same microscope settings (just varying the excitation and emission wavelengths as necessary). Is this at all possible? Or does having to change excitation and emission wavelengths make this impossible.
Alternatively, could I normalise measurements of the fluorophore of interest from my region of interest (I currently take measurements in my time-course experiments by selecting the nephrons in kidney tissue) to the background fluorescence of the same fluorophore? And if so what counts as the background? Interstitial tissue? The perfusate fluid that surrounds the tissue on the stage?
Any other suggestions? Some kind of physical standard like a fluorescence ruler, or fluorescent beads?
I do not want to manipulate the data with ImageJ or some other software as I think this will somewhat invalidate it.
Many thanks for your time!
I have several slides of cells stained for transcription factors and cytoskeletal proteins. The staining worked quite well and appears very specific to targets. Now I'd like to have some semi-quantitative analysis of the slides. What is a typical method of analyzing stainings? For example, can one us an arbitrary flourophore intensity to bin cells into either a 'high' or 'low' expression level, and count the number of each type per field of view? Do people try to produce an average intensity for the FOV? What is common here? Please link to papers if possible, thank you!
Update: Attached is an example field of view. These are cancer cells which I have probed for vimentin (red) and e-cadherin (white). All cells in every experimental group express both proteins to a degree and I am attempting to quantify potentially subtle expression differences.




I research on cell responses especially intracellular Ca2+ increase after exposure to pulsed electrical fields using Flou-8.
The software to monitor intracellular Ca2+ was SlideBook.
Now I have been using MetaMorph and thinking about proper values of laser to excite Flou-8 such as its intensity and exposure time. Another value to amplify the fluorescence is EM gain.
When SlideBook, I used to set the values up below.
Exposure time 1000 ms, Intensification 3500.
I am not sure the relation between EM gain and intensification.
I would like to know how much EM gain corresponds to 3500 of intensification.
I would really appreciate it if someone would give me some advice.
Best regards,
Laser scanning confocal microscopy
Except some traditional methods such as EEM, SEM, AFM as well as FTIR. The fouling might be microalgae, protein, polysaccharide. We want to confirm the membrane fouling in a big area but not in a really small area. We check that Optical Coherence Tomography might be a good methods.
Does anyone have recommendations for breathable plate seals for 96 well plates that are compatible with fluorescent (high content) imaging, in other words are not autofluorescent/ increase light scatter in the well?
I have YFP tagged STIM1, transfected in HEK293 cells and imaged in TIRF plane for formation of punctae/clusters upon ER store depletion with thapsigargin. I have been trying to analyze the number of clusters and area, intensity of each cluster using ImageJ.
However, I have uneven illumination and background in the fluorescent images in TIRF plane. And, the clusters are of different sizes. So, I am facing difficulty to subtract background and set a common threshold for both small and big clusters. I tried FFT filter, background correction, local threshold but nothing helped so far.
Has anyone analyzed such clusters? Any help is much appreciated.
PS: I have attached a sample image with clusters.
Hi everyone
I am trying to perform live dead assay in a mammalian cell line. I am using DAPi and PI, i.e. ideally DAPI staind nuclear material in all cells and PI must stain nuclear material of cells whose membranes have been compromised i.e dead cells. Howeevr, I am getting complete staining from both the dyes in all the cells. What could be going wrong? Dye concentraation? no of washings? staining protocol?
Plz help
I am using confocal microscope and using sequential scanning but still there is crosstalk between them
I am looking to buy a fluorescent phalloidin product to use with confocal microscope?
I did the fluorescence microscopy for a solution of FITC dye in my control experiment. What I expect is the completely dark/green image depending on the exposure time. However, I saw some pale green weird shape standing out from the quite dark background. I guess it may be dust or somehow the dye aggregates together or air bubble. Is there any idea about what I saw and how to verify that? And how to avoid that thing to get a dark or green image?
I have a lot of tissue sections with various treatments and for my control sections, I've determined the optimal exposure time for each fluorophore. These are fine for the DAPI and 488 channels but I'm concerned about the 594 channel exp time, which is 2 seconds. The problem is that for a minority of sections, the signal is strong in the 594 channel that I get the odd bleaching occurring. I'm keeping the exposure time the same for every single image I take.
While I'm able to eliminate this bleaching in subsequent analysis in FIJI, I'm worried that the 2 second exposure time may be too long. So I was wondering if there is a general rule of thumb for exposure times.
Thanks in advance
I am doing a DAPI/Phalloidin staining of cell seeded on top of my hydrogel. However, it is almost impossible to see the staining because I have a lot of background noise, because my hydrogel matrix has autoflourescence (the hydrogel alone without fixation and staining emits fluorescence
I tried blocking with 1% BSA but I wasn’t able to eliminate the background noise. Since my samples aren’t thin (1mm) someone recommended try a confocal microscope, but I don’t have one in my lab. Is there another alternative to eliminate the background noise?
Thank you
Is there any way to visualize a fluorescent drug loaded inside the nanoparticles? I think confocal microscopy won't help due to the small size of the nanoparticles and TEM won't be useful too.
Does anyone know if the streptavidin will conserve the biotin-binding capacity after a fixation by the paraformaldehyde?
thanks in advance
I found a lot of fluorescence pictures of primary cilium stained with alpha-tubulin antibody in the literature, but I am not sure if they made by confocal or epifluorescence microscope? Will I see it in epifluorescence mic. or do I need confocal? thanks
hi
i have been doing immunofluorescence on transwell membrane inserts and have been getting suboptimal images. i fix and stain the cells in transwell insert, and when i mount them i cut the membrane off, place it on glass slide with cells facing up. I then drop a few mounting media on membrane and then a coverslip over it. the pore size is 0.4um and does not permit migration.
1) i have tried fluorescence micoscopes and confocal microscopes but both are giving suboptimal imagings. What kind of microscope is best for this?
2) I notice that after placing the membrane on glass slide, and coverslip over it. there are still some slight creases (uneven regions of membrane that kinda forms bulging upwards). This could explain why when im focusing on one cell, the other regions get out of focused. I was thinking of putting pressure on the coverslip to straighten out the membrane. But will this affect morphology of the fixed cells?
any tips for people who had used this technique before?
I am looking for an approach that will allow me to follow the emission of fluorescent proteins (in single cells) in vivo over time. The time frame is 2-3 hours with measurements every 5 minutes. I need a convenient way to run samples in parallel to maximize data output. Any suggestions?
How can I determine the amount of a fluorescent dye to be used to stain my oils without having excessive dye remained? And how to make sure that all the dye I used is attached to my oil with no unreacted dye that might cause reading errors and inconvenience? I read something about using GPC for the second part of my question but could not find good details. Any help please?
I understand that if you have 3 mice and are generating data from microscopy images, you might want to average the data in such a way that you enter in 3 numbers to Prism for example.
Should you handle in vitro microscopy data in the same way? For example let's say you are interested in the intensity of a fluorescently stained protein on cells. You do the experiment 3 independent times (n = 3). For each experiment you have cells seeded on 4 coverslips. For each coverslip you take images from 5 fields. Within each field you might have 10-15 cells.
How many numbers do you ultimately report (i.e. how many numbers go into Prism). Do you put in every cell you analyze? Every coverslip? Or 3 numbers for each experiment? If you average them, should you take an average of the coverslip and then average the coverslips? Or just average all the cells in the experiment?
I have been using CellROX Green to label my cells in a live cell fluorescent imaging format. My protocol is very simple:
-wash cells with PBS
-Add 5uM CellROX Green
-Immediately perform 45 minute time lapse imaging of cells (to include the 30 min incubation and 15 minutes post incubation in images and quantitative detection)
In comparing fluorescent ROS detection of my diseased and non diseased cells, I initially noticed stark and expected differences in my images. When I switched to a new tube in the same lot, and since then a completely different lot #, almost all my fluorescence is extremely low in every cell and I can't replicate the images I've previously obtained. The only time I see clear/vivid fluorescence is when I accidentally photo-oxidixed the dye with light, which isn't what I want.
I've seen other people post issues with CellROX Green and that it requires a lot of optimization. Has anyone else experienced difficulty with consistency and reproducability?
Thanks.
I am relatively new in IHC. My confocal microscope is Zeiss LSM800.
This microscope has a smart setup program with a database to select Fluorophores, and it shows the excitation and emission spectrum of each one and the amount of crosstalk between them.
Is there any application or software to make the same comparison between fluorophores to decide for the best combination for ICH?
Your help is highly appricated.
Masoud
Hi,
Could you please share your experience regarding upsides and downsides of these microscopes? Which micrsocope is better for fluorescent imaging in fixed cells at 100x?
Hello everyone, I am studying a protein from E. coli that is only expressed in acidic media. I have replaced the whole ORF of this protein (since GTG start codon) by sfGPP in order to monitor the gene expression. However I was not able to see fluorescence by microscopy both in neutral and acidic media.