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
  • asked a question related to Confocal Fluorescence Microscopy
Question
1 answer
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!
Relevant answer
Answer
It would be better for mTurquise2 and YFP pair.
  • asked a question related to Confocal Fluorescence Microscopy
Question
1 answer
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?
Relevant answer
Answer
This technique will suit you with some modifications.
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
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
Relevant answer
Answer
Try gram staining of microorganisms and you can also distinguish them buy once.
  • asked a question related to Confocal Fluorescence Microscopy
Question
1 answer
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!!
Relevant answer
Answer
Calcein AM is a non-fluorescent cell-permeable derivate of Calcein that is widely used in cell viability measurement. The carboxylic acid groups on Calcein are modified with AM (acetomethoxy) groups, which endows Calcein AM with high hydrophobicity, facilitating its penetration through cell membrane. Once inside the cell, AM groups are hydrolyzed by intracellular esterases. The fluorescent Calcein molecule is restored, which is trapped in the cell and emits strong green fluorescence.
Since dead cells lack esterase activity, only live cells are labeled and detected. The fluorescence intensity will be proportional to esterase activity. Calcein-AM has been proved to be both specific and sensitive for detection and tracking of apoptosis in living cells. The preservation of membrane integrity is one of the most significant features of apoptosis with respect to necrosis. In the presence of membrane defects, Calcein leaks out of the cell and the signal also vanishes in the presence of residual esterase activity.
On the other hand, Propidium iodide (PI) which is a red-fluorescent nuclear stain is not permeant to live cells or cells which are dead but still have an intact membrane (such as the primary apoptotic cells). In late apoptotic and necrotic cells, the integrity of the plasma and nuclear membranes decreases, allowing PI to pass through the membranes, intercalate into nucleic acids, and display red fluorescence.
Calcein generated from esterase in viable cells emits a strong green fluorescence with an excitation and emission maximum at 494nm and 517nm, respectively, while PI once bound to DNA has a maximum emission wavelength at 617nm when excited at 535nm.
There is something that must have gone wrong with your reagent or your process. You may have cells that are either alive or dead, but not both. Cells which are dead but still have an intact membrane (like the primary apoptotic cells), PI is not permeant to these cells.
You may repeat the experiment. Initially, observe the cells in bright field. Then observe the cells in the green fluorescence channel. The live cells will be stained by green, fluorescent Calcein. Follow it by observing the cells in the red fluorescence channel. The dead cells will be stained by the red fluorescent, PI. Then finally you merge the image of green and red channels.
Good Luck!
  • asked a question related to Confocal Fluorescence Microscopy
Question
7 answers
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.
Relevant answer
Answer
May consider using Permai fluorescence dye.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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!
Relevant answer
Answer
You might like to try nucleic acid blocking reagent sigma 11096176001. I suspect your aptamer is binding to the histones in the nucleus.
  • asked a question related to Confocal Fluorescence Microscopy
Question
1 answer
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?
Relevant answer
Answer
You can use (cellprofiler) software for identification and measurment of cells features. Then use the exported data for machine learning of cell diffrentiation with (cellprofiler analyst) software. Both are free.
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
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!
Relevant answer
Answer
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
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!
Relevant answer
Answer
I agree with Randall Scott Gieni, 300µM tissue section will not only be difficult to permeabilize but your antibody penetration will be difficult.
Good luck,
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
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!
Relevant answer
Answer
Hi, i think you can use a concentration of 2ug/mL.
  • asked a question related to Confocal Fluorescence Microscopy
Question
6 answers
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.
Relevant answer
Answer
Dear Fatemeh,
Maybe you look through this protocol:
Reagents
  1. Experimental animals (129S6 wild type mice- Taconic Laboratories) ! CAUTION Experiments involving live rodents must conform to local and national regulations.
  2. Paraformaldehyde (Sigma-Aldrich, USA) ! CAUTION Avoid skin and eye contact- Vapor is carcinogenic and toxic.
  3. Phosphate Buffered Saline- pH 7.4 (Sigma-Aldrich, USA)
  4. Cell TakTM cell and tissue adhesive (BD Biosciences)
  5. 0.1 M sodium bicarbonate, pH 8.0, filter sterile the buffer
  6. Triton X100 (Sigma-Aldrich, USA)
  7. MitoTracker Deep Red 633 for staining mitochondria (Molecular Probes, USA)
  8. Alexa Fluor 568 phalloidin for staining F-actin (Molecular Probes, USA)
  9. SYTO 11 Green-Fluorescent Nucleic Acid stain for staining the nucleus (Molecular Probes, USA)
  10. Bovine Serum Albumin (Sigma-Aldrich, USA)
REAGENT SETUP
  1. Tyrode’s solution- The modified Tyrode’s solution (pH 7.4) contained the following (mM): 126 mM NaCl, 4.4 mM KCl, 1.0 mM MgCl2, 18 mM NaHCO3, 11 mM glucose, 4 mM HEPES, 30 mM butanedione monoxime (BDM), and 0.13 U/ml insulin, and was gassed with 5% CO2/95% O2 (For a detailed description of the cardiomyocyte isolation procedure, please refer to Su et al. 200110
  2. Culture medium: composed of 5% fetal bovine serum, 47.5% MEM (Gibco Laboratories, Bethesda, MD), 47.5% modified Tyrode’s solution, 10 mM pyruvic acid, 4.0 mM HEPES, and 6.1 mM glucose and finally maintain the isolated cardiomyocytes in a 5% CO2 atmosphere at 30 °C until use.
  3. Fixative: Freshly prepare 100 ml of 4% PFA (wt/vol). Dissolve 4g PFA in 100 ml PBS. ▲CRITICAL STEP This solution must be made fresh. To dissolve the PFA efficiently, heat the solution to ~70 °C under constant stirring with a magnetic stirrer in a fume hood. Cool the PFA solution, filter it to avoid precipitates in the fixative.
  4. Coating chambered coverglass with Cell Tak cell adhesive: Coat the chambered coverglass with Cell Tak adhesive (1.7 µg/mm2). From the size and number of vessels to be coated, calculate total surface area. The best density of BD Cell-Tak depends on specific application, or cell type. A preliminary dose-response experiment is recommended to determine optimal density. High densities will not necessarily improve performance, so the “minimum effective density” should be determined empirically. Dilute the correct amount of BD Cell-Tak into the buffer, mix thoroughly, and dispense within 10 minutes. ▲CRITICAL STEP If the pH in the coating buffer is not between 6.5 – 8.0, Cell-Tak will not perform optimally. An aid to attaining this pH window is to use a volume of 1N NaOH equal to half the volume Cell-Tak solution used in combination with a neutral buffer. For example: Use 10 μl Cell-Tak, 285 μl Sodium Bicarbonate, pH 8.0 and 5 μl 1N NaOH (added immediately before coating) to make 300 μl Cell-Tak solution. A minimum incubation of 20 min is recommended, but longer times will not adversely affect adsorption, even if all the liquid evaporates. Pour off, or aspirate, the Cell-Tak and wash with sterile water to remove bicarbonate. If vessels are to be used later, they should be airdried and stored at 2-8 ºC up to two weeks or with dessicant up to 4 weeks.
Equipment
  1. Temperature controlled centrifuge
  2. Nutator
  3. Chambered coverglass (Lab-Tek, Nalgene Nunc, USA)
  4. FV300 confocal IX81 microscope (Olympus Microsystems, USA) and Leica TCS SPE confocal microscope with an oil immersion objective of 60x (NA 1.45) for image acquisition or similar
Procedure
  1. Isolation of adult ventricular cardiomyocytes: Adult mouse ventricular myocytes were obtained from the laboratory of Dr. William H. Barry (Cardiology Division, University of Utah, Health Sciences Center, Salt Lake City, USA) and were isolated from 129S6 wild type mice (Taconic Laboratories) according to previously reported methods (10) (Note: As this protocol deals with immunocytochemisry and confocal microscopic imaging, isolation of adult ventricular cardiomyocytes is not elaborated).
  2. Suspend the isolated cardiomyocytes in culture medium composed of 5% fetal bovine serum, 47.5% MEM, 47.5% modified Tyrode’s solution, 10 mM pyruvic acid, 4.0 mM HEPES, and 6.1 mM glucose. Maintain the cells in a 5% CO2 atmosphere at 30 ºC until use.
  3. Label the cells with MitoTracker Deep Red 633 for staining mitochondria (100 nm in the culture medium, M-22426, Molecular Probes) and incubate for 30 min while at the CO2 incubator ▲CRITICAL STEP staining for mitochondria should be done in live myocytes as the MitoTracker will stain mitochondria only when it is alive. The concentration and timing for staining should be standardized by the end user.
  4. Wash the cells with PBS twice and resuspend in fresh PBS ▲CRITICAL STEP All the steps after labeling with MitoTracker should be performed in the dark or by covering the tubes containing the cells with aluminum foil to avoid photo-bleaching of the dye.
  5. Fixing and processing of cardiomyocytes for immunocytochemistry: Pellet the isolated cardiomyocytes using low g force (300 g for 1 min, 30 ºC). Suspended the pelleted cells in 4% paraformaldehyde in PBS maintained at 30 ºC and fix for 30 mins with gentle mixing of the contents by inverting the tube using a nutating mixer. ▲CRITICAL STEP It is important to maintain the cells at 30 ºC when they are viable and centrifuged at low g force to pellet cardiomyocytes, since these would affect cell viability and morphology. ?Troubleshooting
  6. After fixing, pellet down the cells (300 g for 1 min) and resuspend in PBS.
  7. Layer the cells over chambered cover glass coated with Cell-Tak cell and tissue adhesive. ▲CRITICAL STEP Adhering the cardiomyocytes to the chambered coverglass is a difficult process due to its size, and it is important to coat the cover glass with a cell adhesive.
  8. Leave the fixed cells layered over the chambered cover glass undisturbed for 2 hours at room temperature. Once the fixed cells settle to the glass surface, wash the non-adherent cells using PBS.
  9. Permeabilization of cardiomyocyte using Triton X-100: Permeabilize the fixed cardiomyocytes which are adhered to the Cell Tak coated cover glass surface using 0.1% Triton X-100 in PBS (v/v) for 3 min at room temperature. Wash the cells with PBS (2×2 min) and process for immunostaining.
  10. Blocking: Treat the permeabilized cardiomyocytes with blocking solution containing 0.01% BSA in PBS (w/v) for 30 mins at room temperature. ▲CRITICAL STEP This step helps to prevent non-specific binding of the fluorophores and is important while using primary and secondary antibodies.
  11. Immunostaining of cardiomyocytes: Label the fixed cells with Alexa Fluor 568 phalloidin for staining F-actin (1:40, A-12380, Molecular Probes), and SYTO 11 Green-Fluorescent Nucleic Acid stain for staining the nucleus (1: 500, S-7573, Molecular Probes) for 30 mins at room temperature in dark ▲CRITICAL STEP The concentration and timing for staining should be standardized by the end-user. If the cells are stained with a primary antibody, then the user has to incubate a secondary antibody after washing the cells with PBS (2×2 mins). The timing and concentration of the primary and secondary antibodies should be standardized by the end user.
  12. After incubation with the fluorescent stains, wash the cells with PBS (3×3 mins) and maintain in PBS with antibiotics added to it. ■ PAUSE POINT The chambers containing the cells can be maintained in dark at 4 ºC until imaged to avoid photo bleaching of the fluorophores. It is highly recommended to image the immunostained cells as soon as possible.
  13. Confocal imaging: Images were obtained and processed using FV300 confocal IX81 microscope (Olympus Microsystems) and Leica TCS SPE confocal microscope with an oil immersion objective of 60x (NA 1.45) at the University of Utah School of Medicine, Cell Imaging Facility, Salt Lake City, UT, USA. The excitation lasers used were Argon 488 to image nucleus stained with Syto 11, HeNe laser 543 and 633 to image F-actin stained with Alexa Fluor 568 phalloidin and mitochondria stained with MitoTracker Deep Red 633 respectively. Three dimensional z-projection views were obtained by deconvolution and volume rendering of the z-stacks using Olympus FluoView software and Leica Confocal Software (Leica Microsystems, Version 2.5, LCS Lite, Mannheim, Germany). Volume visualization of the stacks and 3D movies were generated using Voxx (http://www.nephrology.iupui.edu/imaging/voxx/). (11)
Timing
  • Steps 1–2: 2-3 h
  • Step 3-4: 1 h
  • Step 5: 1 h
  • Steps 6-8: 2-3 h
  • Step 9: 10-15 min.
  • Step 10: 30-45 min.
  • Step 11-12: 1-2 h
  • Step 13: 2-3 h (depending on the number of channels, image quality, step size, kalman averaging, availability of equipment and expertise of the user, this timing varies)
Troubleshooting
Steps 1-4 It is important to handle the cardiomyocytes as gentle as possible while they are viable. The optimum temperature should be maintained and low-speed centrifugation in a temperature controlled centrifuge should be carried. The pipette tips should have a wide bore (by cutting the tip of the pipette tips) so that the cardiomyocytes are not strained while transferring.
Anticipated Results
With applying the above-described protocol, it should be possible to get images of isolated adult mouse cardiomyocytes which has been immunostained using confocal microscopy. The shape and morphology of the cardiomyocytes is retained even after fixation.
Figure 1 Confocal images of fixed cardiomyocytes with pseudo-colors for each staining.
Figure 1a Isolated mouse ventricular cardiac myocyte stained with MitoTracker Deep Red 633 for staining mitochondria shown as blue, Alexa Fluor 568 phalloidin for staining F-actin shown as red and SYTO 11 Green-Fluorescent Nucleic Acid stain for staining the nucleus shown as green. The images are deconvolved using Fluoview software from a series of image stacks obtained using Olympus Fluoview confocal microscope FV300.
Figure 1b Isolated cardiac myocyte stained as above shown as X-Y view. The orthogonal projection on the left of the image is the cross-section passing through the myocyte along the Y-Z view. The orthogonal projection on the bottom of the image is the cross-section passing through the myocyte along the X-Z view (images obtained using Leica TCS SPE confocal microscope).
Supplementary Figure 1a-d Isolated mouse cardiac myocyte pseudo-colored showing different staining in different panels and the fourth image on the bottom right is a merged image of all three panels (images obtained using Leica TCS SPE confocal microscope).
Supplementary Movie 1 The above described image stacks were used to construct a 3D image to visualize the distribution of cytoskeletal proteins and organelles within the cell.
References
  1. G. Bkaily, N. Sperelakis, and J. Doane, Am J Physiol 247 (6 Pt 2), H1018 (1984).
  2. N. Cambon and M. A. Sussman, Methods in Cell Science 19 (2), 83 (1997).
  3. M. A. Sussman, S. Welch, N. Cambon et al., Journal of Clinical Investigation 101 (1), 51 (1998).
  4. F. Appaix, A. V. Kuznetsov, Y. Usson et al., Exp Physiol 88 (1), 175 (2003).
  5. E. Ehler and J. C. Perriard, Heart Fail Rev 5 (3), 259 (2000).
  6. R. R. Kaprielian and N. J. Severs, Heart Fail Rev 5 (3), 221 (2000).
  7. A. V. Kuznetsov and R. Margreiter, Int J Mol Sci 10 (4), 1911 (2009).
  8. A. V. Kuznetsov, J. Troppmair, R. Sucher et al., Biochim Biophys Acta 1757 (5-6), 686 (2006).
  9. A. V. Kuznetsov, Y. Usson, X. Leverve et al., Mol Cell Biochem 256-257 (1-2), 359 (2004).
  10. Z. Su, K. Sugishita, M. Ritter et al., Biophys. J 80, 1230 (2001).
  11. J. L. Clendenon, C. L. Phillips, R. M. Sandoval et al., Am J Physiol Cell Physiol 282 (1), C213 (2002).
Sathya Srinivasan, Experimental Imaging Centre, University of Calgary
Correspondence to: Sathya Srinivasan (sathya_sr70@hotmail.com)
Source: Protocol Exchange (2011) doi:10.1038/protex.2011.235. Originally published online 11 May 2011.
Best regards,
Alexandr
  • asked a question related to Confocal Fluorescence Microscopy
Question
5 answers
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
Relevant answer
Answer
Thank you
Johanna Marie Dela Cruz
This looks like what I'm after. Does the setMinAndMax command run independent of the run("Enhance Contrast") or do you need to use one before the other?
Thanks again!
Sam
  • asked a question related to Confocal Fluorescence Microscopy
Question
12 answers
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
Relevant answer
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
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.
Relevant answer
I work mainly with mammalian cells and have no clue about the plant cells but I feel this could help you with your DAPI problem. DAPI is not great at staining a live nucleus. DAPI most often cannot enter cell membrane/wall and staining requires fixation and permeabilization. I suggest you try Hoechst 33342 which has similar excitation and emission wavelength so you don't have to change your protocol.
Please note that this works for mammalian cells and may or may not work for plant cells but something to think about.
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
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.
Relevant answer
Answer
Dear Alexandr,
Thank you for the answer. Unfortunately, I already know these works really well!
I can't use neural crest markers in IF as most of them are either cytoplasmic or downregulated before the enteric neurons become terminally differentiated.
Sox10 works quite well up to a certain point, but it's absent in mature neurons: its expression is maintained in enteric glia only.
  • asked a question related to Confocal Fluorescence Microscopy
Question
7 answers
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 :)
Relevant answer
Answer
Propidium iodide cannot be used as a viability dye in fixed cells. Propidium iodide is usually used to measure DNA content in fixed cells, but that requires methanol fixation and RNase treatment. And even then, I wouldnt rely on the sub-g1 population as a reliable for viability assays.
If you want to know the viability of fixed cells, look into amine reactive fixable dyes such as life technologies live/dead dyes, biolegend's zombie dyes, or the ghost dyes. You must first stain your cells with one of these amine reactive dyes and then you can fix the cells without losing your dead cell staining.
These dyes tend to have sharper excitation/emission spectra as well as numerous color options. You should be able to stain for both viability and phalloidin, just avoid fluorophores with spectral overlap.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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!
Relevant answer
Answer
It will depends of your FRET design. If a fluorochrome such as DAPI is not emitting or absorbing the light wavelength needed in your FRET system you should not have any problem. However, DAPI is a fluorochrome with a very broad spectrum of absorption and emission so it is maybe not be recommended. You could use phalloidin coated with an Alexa fluorochrome that is not having interference with your FRET system.
You can check the fluorochrome spectra of individual dyes and their overlap with other (if any) in:
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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()
Relevant answer
Answer
I Just checked your macro and it looks okay to me. Maybe you should try and include in the last step
run("Flatten");
I sometimes experience errors with merged images in TIFF format and this seems to help
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
I have isolated mitochondria from tissue. I would like to check them by fluorescence imaging. Can I use Mitored dye for these isolated mitochondria?
Relevant answer
Answer
Debaprasad Koner if you are seeing fluorescence with mitoRed then you are on the right track. Mitotracker red is membrane potential specific so what you see are mitos with reserved MP, and the stain is retained post fixation. However if you want to do a mito count then mitotracker green is the best since it is not mp dependent. Some mitos will lose MP based on treatment, mito fitness, lysis etc. MitoGreen also retains well post fixation.
  • asked a question related to Confocal Fluorescence Microscopy
Question
1 answer
I want to detect intracellular bacteria in urine by confocal fluorescence microscopy
Relevant answer
Answer
Hello,
You may coat your coverslips with Poly-L-Lysin for example and let your cells to sediment and attach on the glass.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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?
Relevant answer
Answer
You can also click on > Analyze tab > Click Set Scale and reset the scale (this is to convert your pixels to microns where you should have a scale bar of known size).
Sathya Srinivasan
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
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?
Relevant answer
Answer
Hi Huijing, the goal of confocal is to reduce the intensity from out-of-focus planes, but the cutoff is never a sharp one, so strong signals from planes not too far from the focus can be observed in the image. Make sure that your pinhole is not too large. If larger than 1AU, you will have increased contribution from out of focus. Other factor to consider is that DAPI signals are often very strong, compared to your FP, so you may see some faint out-of-focus signal from DAPI but not from the FP.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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.
Relevant answer
Answer
Dear Cy,
First, be sure that your cells are not contaminated by mycoplasma because the cytoplasm is not stained by HOECHST. Second, when you observe your stained cells by a conventional fluorescence microscopy, the light dispersion causes the nucleus seems as integrated round blue units. Whereas by using confocal fluorescence microscopy, the unfavorable light dispersion will be eliminated, hence you'll observe the stained nucleus with more details. Since the HOECHST is intercalated into the DNA, and regarding the fact that chromosomes don't occupy all the space of nucleoplasm, I think that your microscopic image is absolutely perfect.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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
Relevant answer
Answer
My pleasure, good luck with your work!
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
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
Relevant answer
Hi Saulo,
Thank you for the response. I was concerning if the GFP fluorescence can be damage after the fixative method. Do you have any ideia about it.
Thanks
  • asked a question related to Confocal Fluorescence Microscopy
Question
1 answer
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?
Relevant answer
Answer
If you are using Imaris to open these images (which you hashtagged in your question so I am assuming), then after opening the images, imaris gives you the option to click on the image using the cursor and reposition it the way you want it.
Uncheck the "frame" option to get rid of the frame. Take a snap and open it using imageJ, and in there you could re construct the xy axis (make sure you know the pixel size before you uncheck the 'frame' option, and draw the xy axis accordingly) hope this helps.
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
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.
Relevant answer
Answer
In addition to the previous answer also try to increase the speed of the spinning disk if you can find an option to do that.
  • asked a question related to Confocal Fluorescence Microscopy
Question
6 answers
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).
  1. Incubate mouse anti-antigen primary antibody at 4 degree overnight (1:100; used at same concentration as in indirect method staining)
  2. Incubate biotinylated goat anti-mouse secondary at RT for 90 mins (1:150)
  3. 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!
Relevant answer
Answer
I was actually going to ask you if you had double checked your filter/Cy3 combination, but since you were already using Cy3 successfully I figured you were good to go. You might throw some of that SA-Cy3 dilution between a coverslip/slide and just make sure you can see it with your 514 set up.
I wish I was being more helpful!
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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!
Relevant answer
Answer
Primary vs secondary antibodies and direct vs indirect staining
Primary antibodies provide specific recognition of the target antigen. Secondary antibodies bind to primary antibodies based on the species they were raised in. Primary antibodies that have been raised in rabbit or mouse are amenable to detection with anti-rabbit or anti-mouse secondaries, respectively.
The pairing of a primary antibody and secondary antibody (specific for the host species of the primary) that is conjugated to a fluorophore is known as indirect staining. The use of primary antibodies conjugated to fluorophores, without secondary antibodies, is known as direct staining. Direct staining methods save time and allow multiplexing using antibodies raised in the same host species. Indirect staining traditionally offers higher sensitivity thanks to signal amplification that occurs when multiple secondary antibodies bind to a single primary antibody.
Other variations on these techniques are available, such as using biotinylated primary antibodies with avidin/streptavidin-conjugated fluorochrome. Fluorophore-conjugated F(ab’)2 secondary antibody fragments (lacking the Fc domain) can help reduce background. In mIHC, horseradish peroxidase (HRP)-conjugated secondary antibodies are employed to catalyze deposition of tyramide-fluorophore conjugates.
  • asked a question related to Confocal Fluorescence Microscopy
Question
5 answers
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!
Relevant answer
Answer
Brian,
I would not recommend to use MitoSox with fixed tissue!
1. MitoSox is a membrane permeable weak acid. In live cells it distributes into the mitochondria because , as long as the mitos maintain their membrane potential, the mitochondrial matrix is the most alkaline place in the cell. Once MitoSox arrives in the matrix it gives off a proton. Now electrically charged it is no longer membrane permeable which effectively traps it inside the mitos. Thus, for proper localization it depends on the presence of membrane potentials which are missing in fixed tissue.
2. MitoSox becomes fluorescent after reaction with superoxide. The stain is meant to report superoxide as it is produced by the mitochondria, typically by activity of the electron transport chain (normal or pathologic). Fixed mitochondria are not metabolically active any more and therefore should not be able to produce superoxide any longer. Superoxide is diffusible, reactive (ROS - duh!) and therefore has a very short half-life. Thus, the superoxide that has been present at time of fixation will be long gone at the time you stain your fixed tissue slices.
Anything you find with MitoSox in fixed tissue will probably be caused by non-specific effects, such as autoxidation, and will be more confusing than useful.
I recommend to follow Alexandr's hints and concentrate on immunodetection of permanent ROS-damage to the tissue (protein, nucleic acids).
Good luck with your project!
Bernhard
  • asked a question related to Confocal Fluorescence Microscopy
Question
1 answer
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!
Relevant answer
Answer
For confocal imaging, if you have two probes, red and green then emmission light of green will not excite red. It is designed in a way that you will see only what you want to see. If you want to see red along with green then you have to program the scope in a way that you excite both red and green separately to see them.
If you want to do FRET, your microscope should be equipped with a different protocol for FRET.
  • asked a question related to Confocal Fluorescence Microscopy
Question
6 answers
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
Relevant answer
Answer
Hi Fran,
A mild heat-induced antigen retrieval on PFA fixed frozen brain sections has been performed for some IHC assays. At 70-80 C for 30 min or at 40 C overnight in a water bath. Unlike FFPE sections, tissue detachment from the slide might be an issue with HIER on the frozen section Tried several adhesive slides for HIER, Truebond 380 slides found better tissue attachemt or use the floating section method. Good luck
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
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.
Relevant answer
Answer
Thank you all for your help :) I will read the articles that you have shared.
  • asked a question related to Confocal Fluorescence Microscopy
Question
11 answers
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.
Relevant answer
Answer
Other aspect to think about is the primary ab. What is the epitope recognized by this ab? Some ab are good for western blot but not for IF because of the epitope.
  • asked a question related to Confocal Fluorescence Microscopy
Question
6 answers
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
Relevant answer
Answer
Hello, I'm reviving this old post since I'm dealing with a similar situation. I have a recombinant protein with 10xHis tag on the N' terminal and a Myc tag on the C' terminal. The protein is molecular weight is ~42.4 kDa. I would like to use the protein for analyzing antigen-specific memory B cells. I have in mind staining the protein with a fluorophore-conjugated anti-Myc or anti-His, and then using the labeled protein to stain the B cells population, followed by Flow cytometry analysis. Does anybody have an experience with such a procedure aimed at B cells? I am aware of the alternative of using the Molecular probes kits, but the first methods seems cheaper and more straightforward. Thank you!
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
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?
Relevant answer
Answer
Hi there,
The answer above is all right and is more of a BatchProcessing. You have also another option which is to assign a shortcut to your Macro so you just have to press a key to perform the function. In the example attached here is "F9". Btw, it will be much faster if you decide a value for each one of the channels for B&C adjustment so you can write it in the macro, this will prevent you for adjusting it in each channel each time. If you want to use different values for different sets of images, just rewrite the macro. The macro here automatically does all what you were doing, but image by image, by pressing F9. It will save the final image in the same folder where your original image was.
To use it:
Download the *.ijm file attached here.
Go to Plugins>Macro>Install...
Then open the image and press F9 and it will be done.
It also creates a white scale bar of 10*1 um at x=2um y=2um of your image. If you want to change those dimensions or the position, just modify the values 10 and 1 in the run("Specify..." line.
One warning, this is assuming that your C1=Red, C2=Green and C3=Blue. If it is not the case you have to modify the run("Merge Channels..." line. c1, c2 and c3 are always red, green, blue. If you have more channels, you will have to add them following the same reasoning.
Hope it helps,
Cheers
macro "adjust BC [F9]"
{
title = getTitle();
filename_clean = File.nameWithoutExtension();
path_to_dir = File.directory;
path_to_save = path_to_dir+filename_clean;
run("Z Project...", "projection=[Max Intensity]");
close(title);
run("Split Channels");
selectWindow("C1-MAX_"+title);
run("Brightness/Contrast...");
waitForUser("Apply optimal Brightness and contrast settings, then press OK");
selectWindow("C2-MAX_"+title);
run("Brightness/Contrast...");
waitForUser("Apply optimal Brightness and contrast settings, then press OK");
selectWindow("C3-MAX_"+title);
run("Brightness/Contrast...");
waitForUser("Apply optimal Brightness and contrast settings, then press OK");
run("Merge Channels...", "c1=C1-MAX_"+title+" c2=C2-MAX_"+title+" c3=C3-MAX_"+title+ "");
run("Specify...", "width=10 height=1 x=2 y=2 scaled");
setForegroundColor(255, 255, 255);
run("Fill");
saveAs("tiff", path_to_save+".tiff")
run("Close All");
}
  • asked a question related to Confocal Fluorescence Microscopy
Question
6 answers
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?
Relevant answer
Answer
Might try a non-iterative method like Wiener if the PSF is known.
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
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
Relevant answer
Answer
Hi Karl, sorry to barge into the discussion, can I ask you which clearing technique have you used? I have spheroids with brain cell types that are almost 2 mm in diameter. I realize there are several kits available and I need to check which one suits for my purpose. I plan to do 4 color fluorescent staining on these spheroids then image using regular confocal microscope. Thanks!
  • asked a question related to Confocal Fluorescence Microscopy
Question
7 answers
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 !
Relevant answer
I Agree With Sayantan Mitra
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
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.
Relevant answer
Answer
I agree with Sönke Weinert . Probably the easiest is trypan blue. This is a common technique often employed with phagocytosis assays, and a technique we use with our Vybrant Phagocytosis assay kit (link below). Since the FITC is on the surface, the trypan blue works through absorbance quenching to quench the FITC signal. Since trypan blue is not cell permeant and not taken up via phagocytosis, the phagocytoses bacteria then becomes unquenched and fluoresces green upon entering the cells.
So, all you'd need to do is put a subset of the labeled bacteria into a solution of trypan blue, compared to a control with no trypan blue, and see if the signal is quenched. If it isn't quenched, then the dye is internalized in the bacteria. If it is quenched, then the dye is on the outside of the bacteria surface.
Be aware, though, that if the bacterial cell wall and/or plasma membrane are compromised, the trypan blue will also enter the cell and quench internal signal.
Trying to determine the localization of signal via high-resolution imaging is reliant upon having a very high-performance microscope or super-resolution system. Most standard widefield or even confocal microscopes are not reliably capable of determining it with enough precision:
Vybrant kit link:
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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!
Relevant answer
Answer
As suggested by the others, ImageJ/ Fiji is a very nice tool to analyse microscopy images. If you're new to bioimage analysis and would like to get a better understanding, I can really recommend you the following channel:
Concerning comparing you samples: it it not be the best idea to compare fluorescent intensities! Samples bleach due to storage, antibody performance varies and different laser intensities/ illumination times can bias your results. Try to find a more reliable way, like quantifying cell numbers or marker-positive area.
This paper could be interesting for you: DOI:10.1038/s41598-017-16797-1, however, the there used ImageJ plugin is currently broken. But it can give you some ideas about possible acquisition and evaluation steps and perhaps the issues get fixed soon.
If you are having troubles with certain analysis steps, you can find help here: https://forum.image.sc/
Happy imaging!
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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.
Relevant answer
Answer
If you have a single channel grayscale image there's probably nothing you can do to get the original color channels. Such image contains only the information about intensity averaged across all three channels per each pixel.
If you change the format of your grayscale image and treat it as an RGB, splitting will give you the same image for red, green and blue, all identical to the original grayscale image. This is because only a mix of these 3 colors in equal and specific intensities results in the same corresponding intensity of white = certain shade of gray in every pixel ("grayscale" image).
If you had an RGB image made by merging of original red and green images, each pixel would still contain information about the intensities of red, green and blue channels separately. You would get the original channels by splitting the RGB image with one difference: If the original data were e.g. 12- or 16-bit, splitting an RGB image will provide only 8-bit result (ImageJ doesn't work with higher quality RGB in default, there might be some plug-ins), which means that you are losing information and quality. You could discard the blue channel which would be all black = zero intensity values in your case. I just tried all of this with one of my images and it functions the way described. Note that this method can only be used if the original image was made of red, green and blue channels or their subset, nothing else!
Anyway, this should be the last emergency solution to be used only when
a) you work with an image from someone else (watch for copyright) and you don't have an access to the original data or
b) you've lost your own original data.
Of course, the best practice is to keep your original data before any processing (and possibly some intermediate processing steps) ideally with a back-up and use those.
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
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.
Relevant answer
Answer
A Ferreol mentions, DeconvolutionLab2 is mainly a learning tool. However if you are processing only 2D images it could be good enough for your needs. If you are processing many, large 3D images the speed could be an issue. It is possible to use GPU deconvolution in ImageJ, though right now that requires an advanced set up to get imagej to use the fast GPU math and FFT libraries (if interested ask about it on the ImageJ forum https://forum.image.sc/ ). It looks like you are having issues dealing with a multiple channel image. In this case each channel would have a slightly different PSF. To process them properly you need to split the image into separate channels, generate a PSF for each channel, then merge them back after deconvolution. In ImageJ you may need a specialized script to do the entire workflow.
  • asked a question related to Confocal Fluorescence Microscopy
Question
5 answers
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?
Relevant answer
Answer
DAPI stains RNA as well, unless you pre-treat the cells with RNAse. Maybe try Hoechst (33458?) because it does not bind to RNA, just to DNA (it binds to the "rungs" of the DNA ladder, which RNA doesn't have).
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
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,
Relevant answer
Answer
Use fluorescent conjugated secondary antibodies ex. Alexa fluor conjugated antibodies
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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!
Relevant answer
Answer
Are you looking for 3D information like volume and surface area? If so, you need to analyze them as a volume with 3D Objects Counter. If you are looking for 2D information, then you don't need the 3D Objects Counter plugin and you can do standard 2D measurements. If the projection isn't too filled with structures (which can happen in large z stacks filled with dense structures), then I prefer to measure in the sum intensity projection rather than the maximum intensity projection. A max projection is only selectively displaying some brightest pixels from the volume and skews the measurements. The Sum projection shows the integrated value of all pixels, so especially if you are quantifying intensity this is important. or, if the information you need is contained within individual planes then you can measure a representative plane or planes.
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
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?
Relevant answer
Answer
It seems that it would depend on the species of cyanobacteria.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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.
Relevant answer
Answer
Hi Fan,
Sectioning and staining of lymph node is not different from other tissues.
But you know that thickness of lymph node compare to other tissue. Therefore, you have to make very thin slice of tissue. If you already inject dye labeled nanoparticles then you can just use nuclear stain and visualize under confocal microscope.
  • asked a question related to Confocal Fluorescence Microscopy
Question
7 answers
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?
Relevant answer
Answer
Many thanks for your help.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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
Relevant answer
Answer
The best results can be achieved with high-pressure freezing, freeze substitution and flat embedding.
Good luck!
  • asked a question related to Confocal Fluorescence Microscopy
Question
5 answers
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.
Relevant answer
Answer
Hi. If you are planning to use primary cells, it is better to establish your own stable cell lines. I have 2 recommendations. 1) Choose actin-GFP, not GFP-actin because attaching GFP before actin seems to affect cell's behavior (https://www.jstage.jst.go.jp/article/csf/42/2/42_17016/_html/-char/en)
2) If you are trying to establish a stable cell line, try lentiviruses (better pre-packaged and ready-to-use, such as from here https://www.systembio.com/products/imaging-and-reporter-vectors/cyto-tracers/pct-actin-gfp-cmv/), or go with transposable elements (piggyBac plasmids).
  • asked a question related to Confocal Fluorescence Microscopy
Question
7 answers
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.
Relevant answer
Answer
So this is quite late, but you are all correct in that the problem was the camera simply not being set up to detect far red wavelengths. I have switched to a proper microscope (not confocal, but it can detect more than three colours) and not had a problem since. Thanks for all your replies!
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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?
Relevant answer
Answer
This answer is copied from earlier discussion on researchgate under similar question that might help you
By Daniel Lee Adams   If your going after a surface, extracellular, protein then there is no need to permeabilize. If the protein is intracellular or transmembrane then you need to permeabilize. Saponin, tween and triton are all good options use 0.1%-1%. Methanol or ethanol are also options if all else fails. However all proteins and cells are different, start with saponin or tween-20, then move on to the triton, then as a last resort ethanol, then methanol. This is also the order of stringency, so the saponin/tween will do the least damage to the proteins/lipids, while methanol does the most damage.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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?
Relevant answer
Answer
Hi Chem,
unfortunately, percentages do not mean much, since the actual power on the sample strongly depends on the light source you are using, the objective you are employing, and the other settings of the microscope, so you have to experiment a bit with your settings.
As a general guideline, photodetector gain should be kept quite high when you are using your confocal microscope in live mode to find the focal plane and image area you want to image. Just turn it up until you start seeing visible noise in dark areas of the image, this way you know you will not miss any fluorescent object just because the gain is not high enough. Once you find the are of your sample you are interested in, turn down the gain to try to limit noise as much as possible, in order to get a "good" image. Ideally you should lower the gain until dark areas are completely black, and slow down the scan speed and/or increas the excitation power until the bright areas are almost at the maximum possible signal (but be careful not to saturate the image).
As for the excitation power, it really depends on whether your sample is fixed or fresh and living. If it is live, the lower the power, the better, as exposure to high intensities of light can interfere with the biological processes you are trying to observe. If the sample is fixed, however, the limiting factor is photobleaching, meaning the more you acquire images, the more you "burn" the fluorescent molecules, and the less bright the next image will be. I would suggest, when you are just looking at your sample to find the area you want ot image, to keep the power as low as possible, as long as you see something, and only turn it up once you start acquiring the images you want to save.If you are collecting a 3d stack, keep the power lower, as photobleaching happens even in the planes above and below the one you are imaging. Finally, if you can spare the time,slowing down the acquisition speed or averaging more images will in general always lead to better results than simply turning the power up, so try to be patient and only increase the power as a last resort.
Good luck!
  • asked a question related to Confocal Fluorescence Microscopy
Question
1 answer
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?.
Relevant answer
Answer
Have you tried BTS?. It works in skeletal muscle inhibiting the ATPasa activity of Myosin heavy chain.
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
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
Relevant answer
Answer
If you want to stain live cells, formalin fixation is not possible. After 2 days in formalin everything will be dead.
I could imagine that it depends on the cellular position of the antigen you want to stain: Staining cell surface proteins will be much more likely than proteins sitting in the mitochondrial membrane for example as you need to get access to the epitope.
Good luck, it sounds like an interesting but difficult idea!
  • asked a question related to Confocal Fluorescence Microscopy
Question
11 answers
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.
Relevant answer
Answer
Some news :
It works really well for me with Hoechst under 405 excitation, with very few toxicity on one hour visualization. I used low laser power (can't remember how but less than 1%).
Note that I work on confocal zeiss LSM 780.
Thanks all for your answer, find attached a picture of my staining:
[url=http://www.noelshack.com/2018-46-3-1542190510-hoechst.png][img]http://image.noelshack.com/minis/2018/46/3/1542190510-hoechst.png[/img][/url]
(Hela Cells)
Have a good day
  • asked a question related to Confocal Fluorescence Microscopy
Question
5 answers
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.
Relevant answer
Answer
Hello, here the info page, you can find much more than you looking for. https://www.olympus-lifescience.com/en/microscope-resource/primer/techniques/confocal/fluorophoresintro/
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
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
Relevant answer
Answer
Gustavo:
I would suggest using the Photon Counting Histogram (PCH) ; see the seminal papers below.
PCH and FCS are determined on the same time series of events. To extract the distribution of the duration of the fluctuations we use a math based on calculation of the correlation function, or FCS. To extract the distribution of the amplitude of the fluctuations, we use a math based on the PCH distribution.
FCS provides the diffusion coefficient D and the number of particles <N> in the observation volume; PCH provides the brightness ε ( the number of photons emitted in the unit time per molecule) and the number of particles <N> in the observation volume. The brightness may change in your system depending upon the number of fluorophores you have.
1. The Photon Counting Histogram in Fluorescence Fluctuations Spectroscopy"; Chen Y1, Müller JD, So PT, Gratton E. ; Biophys J. 1999 Jul;77(1):553-67.
2. Characterization of Brightness and Stoichiometry of Bright Particles by Flow-fluorescence Fluctuation Spectroscopy. Johnson, J.1., Chen, Y., Mueller, J.D. ; Biophys J., 2010, 99(9), 3084-92.
  • asked a question related to Confocal Fluorescence Microscopy
Question
5 answers
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.
Relevant answer
Answer
Hi. Can you please expalin what you need help with? Are you trying to quantify the percentage of your cells which was affected with the drugs Or?
With more info I might be able to help.
  • asked a question related to Confocal Fluorescence Microscopy
Question
7 answers
Did anyone observe the suppression of low fluorescence signals from one organelle due to high signals from other organelles?
Relevant answer
Answer
The word you are looking for is "dynamic range". The dynamic range in your images seems to be too small to see the weak signal (peroxisome) without saturating your strong signal (plastid). If I understood you correctly.
There are several ways you can try to improve on this. First, what bit-depth are you using, usually you can set the readout from the PMT to 8 or 12 bit, 16 bit on some systems. While this not increase the dynamic range as such, it subdivides it into more steps (256 for 8 bit, 4096 steps for 12 bit). Higher bit will make the files larger and might affect acquisition speed, but might give you more details on your weak structures.
Another option is HDRi (high dynamic range imaging), which you might know from your phone. In principle this can also be applied to microscopy images. A series of exposures is taken with different settings and then combined into a single image by software. Some microscope software has this function integrated but it can be done externally (e.g. using imageJ). However, be aware that this is a non-linear operation and the resulting image will not be quantitative anymore.
  • asked a question related to Confocal Fluorescence Microscopy
Question
5 answers
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
Relevant answer
Answer
Hi Mona and Muneeb. Thank you so much for your suggestion.
I was wondering if the filaments with microfluids chambers are axon, I could differentiates axon. I took the thick filaments which are at proximity of soma (cell body) as dendrites. So I am confused in the selection appropriate axons. In the image I have attached, soma in not under focus as I scanned the image with prim focus on neurites (axon and dendrites). I have already taken and scanned images for only cell body(soma). That's why I need to distinguish axon in the image I have attached. Your suggestion would be highly appreciable. Thank you once again.
  • asked a question related to Confocal Fluorescence Microscopy
Question
8 answers
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.
Relevant answer
Hi,
It's depend on the nature of the sample (If was fixed or not)
I recommend the use of hoechst 33342 or Sybr GreenI.
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
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?
Relevant answer
Answer
Thanks Timothy for the inputs.
I'll see what works best for my samples as I need to be heating my samples at 80C for 30 min.
Regards
  • asked a question related to Confocal Fluorescence Microscopy
Question
7 answers
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 ...
Relevant answer
Answer
Hi. I have used Prolong Gold when imaging samples using STED and it has worked very well. There is minimal sample distortion and it is readily available. Make sure to follow the manufacturer's advice with regards to the curating period (>24h) and to use high quality coverslips.
  • asked a question related to Confocal Fluorescence Microscopy
Question
5 answers
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!
Relevant answer
Answer
Thanks for all the inputs!
I used ImageJ as suggested.
If anyone sees this question later, looking for help, here is my Excel sheet.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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
Relevant answer
Answer
If you just put a drop of antifade, and then gently put the coverslip upside down to allow to sink by the viscous antifade. It will be OK.
Regards
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
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
Relevant answer
Answer
When you used FITC to conjugate the graphene oxide, did you notice any change in the color or still its native color ??
for my knowldge that FITC is  high reactive with minor amounts of free amino groups. so to ensure the deficiency in conjugation , you must measure the content of the dye in your produced conjugate and should be following the recommended ration to get you the preferable fluorescence.
  • asked a question related to Confocal Fluorescence Microscopy
Question
7 answers
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.
Relevant answer
Answer
Dear Dr.Subir Das,
I have studied Efficiency of the second-harmonic generation with the zeroth-order bessel-profile beams in uniaxial crystalsand and I have also learned how to create the zero order Bessel beam. To create the zero order Bessel beam we can use annular slit as in the work of Prof.D. MCGLOIN * and Prof. K. DHOLAKIA.
However, the most convenient method is probably to use an axicon.
  • asked a question related to Confocal Fluorescence Microscopy
Question
1 answer
I am interested in understanding how the surface area changes when adherent cells are brought into suspensions. I am looking for data supported responses. 
Relevant answer
Answer
No one is interested in measuring the surface area in both attached and suspended cells. Due to tremendous change of cell niche in both conditions, stress, cytoskeletal system, metabolic events, the resulting signaling system received within the cell will be totally different. Therefore, it is pointless to discuss, so, no papers. Just imagine monomeric actin molecules and dimeric tubulin molecules either polymerize or depolymerize in a brink of time within the cells. This cell movement accompanies with different metabolic activities. The membranes are not a fixed boundary, and endomembane system is always flowing from the plasma membranes, cis-, medial-, and trans-Golgi membranes, and ER membranes to the nuclear membranes and ever-changing. Full appreciation of these basic biological systems can only tell that there is no surface area change in both states of cells, but changes occur in cytoplasm. Just like a completely air-leaked balloon and maximally inflated balloon!!! Still doubt?
You may be interested in the followings;
Cell 16 (4), 909-918 (1979)
Biotechnol Prog 9 (4), 362-365 (1993)
  • asked a question related to Confocal Fluorescence Microscopy
Question
20 answers
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
Relevant answer
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
Chromosome counting using fluorescent microscope....
Relevant answer
Answer
If you want a good imaging of chromosomes with both conventional fluorescence microscopes, semi-confocal technologies and/or confocal microscopes (even super-resolution and nanoscopy microscopes) then forget about DAPI mounting medium, specially Vectas..... this will cause an high background, diffused two-photons emission that will coock your samples and inefficiency in your overall imaging. I'd like to suggest you to use Hoechst 33342 or picogreen for DNA visualization (compatibly with other fluorocromes you need in your preps), stain, wash, rinse and mount with a DAPI-free mounting medium. Optical index is something unknown by most but really important for imaging. Your mounting medium OI should match those of your coverslip and immersol (immersion oil) if used. Glycerol is really good (90% glycerol with clean PBS is fine). Antifading and antiquencing is extremely important to me so bear in mind that. Other commercial mounting medium are extremely well made in that sense. Also Mowiol powder could be used to make an excellent mounting medium but as well as glycerol you must add an antifading at least. Always clean coverslips and never autoclave them, they're not pirex glass and they'll be coocked.
feel free to ask any other question.
  • asked a question related to Confocal Fluorescence Microscopy
Question
6 answers
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?
Relevant answer
Answer
Stable: DNA is integrated into host genome, cannot be lost. Both daughter cells receive a full genome copy, complete with transgene.
Transient: DNA is on a plasmid, and can be lost (if a cell divides and only one daughter cell gets plasmid copies, the other daughter lineage has now lost the plasmid forever).
You are using transient transfection, so your DNA will eventually be lost by your cells.
You don't specify whether your plasmid contains a eukaryotic resistance cassette or not: this is a common method to ensure plasmid retention. If, for instance, your plasmid confers neomycin resistance, then your cells can be grown in neomycin and any that lose the plasmid will die.
if you don't have a resistance cassette, then...yes, your DNA will eventually be lost by your cells.
It shouldn't happen that fast, though, and the CAG promotor is (in my hands) generally very high expression and pretty robust, so even if your plasmid is lost, your cells should remain detectably green for several divisions (the green signal will simply halve with each division).
I am unclear as to why you would WANT to isolate a clonal cell population if you suspect they might have kicked out your plasmid anyway, however.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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,
Relevant answer
Answer
If you use excitation wavelength of 594nm, for FCS calibration, Atto 655 derivatives (-NHS ester, carboxylic acid, maleimide) could be used. I think Alexa 647 and Alexa 633 can be considered to use as well. Their absorption at wavelength of 594nm is not so strong but I think it is acceptable and their diffussion coefficient are known.
You can find the diffussion coefficient value of those dyes in this file: https://www.picoquant.com/images/uploads/page/files/7353/appnote_diffusioncoefficients.pdf
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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!
Relevant answer
Answer
I doubt you could have a source of autofluorescence that you could just assume to be stable and unchanging. If you have not yet done the experiments, I would suggest using fluorescent beads with wavelengths that are close but not overlapping.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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.
Relevant answer
Answer
I agree with Alex. Cell profiler will enable you to count cells, assess colocalization of your antibody staining pattern, and measure intensity values across your images very easily. The best part is it will batch process hundreds of images while you go for coffee! And it's free!
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
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,
Relevant answer
Answer
Hi Daiki,
it really depends on the camera you are using. Have a look at the manual, it usually reports an accurate calibration of slider value vs. actual multiplication gain.
For example, on my old Cascade II 512B, the relationship is exponential (page 16 of the manual https://www.photometrics.com/support/pdfs/manuals/Cascade2Manual.pdf), but on newer cameras it is linear (for example in this case: https://www.photometrics.com/products/datasheets/evolve_128.pdf).
If you really dont trust the manual you can of course try and measure the relationship yourself, taking a sequence of images with increasing values of the gain, and comparing the intensities. Just make sure you are not photobleaching the sample during your measurements.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
Laser scanning confocal microscopy
Relevant answer
Answer
@Jinhhe Yuan. Please suggest me anything so I can proceed with analysis for making application.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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. 
Relevant answer
Answer
You are welcome. I wish you the best for your research work.
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
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?
Relevant answer
Answer
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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.
Relevant answer
Answer
Try the rolling ball background subtraction in ImageJ.  It does wonders for unevenly illuminated fields.
The better solution is to use a Chroma slide to check that your beam is centered.  TIRF uses single mode fiber with a Gaussian beam profile, so it will always be brighter in the center and fall of at the edges.  Put the cells in the center of the field. 
  • asked a question related to Confocal Fluorescence Microscopy
Question
6 answers
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 
Relevant answer
Answer
When doing fixation after DAPI and PI staining  (as recommended above) I would recommend to wash your cells multiple times (with large volume of buffer and high concentration of protein =( 10% serum), then buffer alone with minimum 1-2% of protein) to "neutralize" staining  before fixation as DAPI and PI tend to leak from dead cells into solution and may stain  "de novo dead" (after fixation) cells, if they are not washed well enough. But  from practice I would not recommend to do this kind of staining. For  dead/apoptotic stain is better to use Annexin V.
There  are also Fixable viability  amine dyes available  like Aqua dead, Zombie, etc. that   brightly stain cells that were  already "dead" before fixation, and much less stain cells that "died after fixation".   This dyes  bind amines, and are recommended for flow cytometry, but there are some that can be used for fluorescent  microscopy as well, depending on filters availability.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
I am using confocal microscope and using sequential scanning but still there is crosstalk between them
Relevant answer
Answer
Many thanks for your reply.
Actually the hardware setting was the problem. It is over now and I can use the laser 552 to excite cy3 without affecting FITC.
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
I am looking to buy a fluorescent phalloidin product to use with confocal microscope?
Relevant answer
Answer
Any labelled phalloidin works well for actin labelling on cultured cells fixed with PFA and or Glutaraldehyde
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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?
Relevant answer
Answer
Check if filtering the sample helps.
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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
Relevant answer
Answer
Objective with higher magnification does not give better signals. The numeric aperture of an objective however does affect the amount of light collected by the objective. Oil immersion objective already has a numeric aperture surpassing water immersion and air objective. Changing the objective will not improve the signals as long as the objective has no obvious defects. Optimizing the correction ring of the oil immersion objective to better match the refraction index of material in which the sample is embedded however can improve the signals.  If the contrast between signals and noises is high enough, one can decrease the exposure time or decrease the excitation intensity to minimize photo-bleaching.  
By the way. for most of research grade fluorescence microscopes, it is possible to save and reuse customized settings for every user, so one does not need to worry so much about compromising the performance of the microscope for other users. It can be a problem of course for less advanced microscopes as such an option may not exist.
  • asked a question related to Confocal Fluorescence Microscopy
Question
6 answers
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
Relevant answer
Answer
Record the absorption spectrum and the emission spectrum of the hydrogel without the cells and the stains and identify the regions where the hydrogel is absorbing and emitting. Then select the wavelength where the hydrogel does not absorb and use it for excitation of the stained cells in the hydrogel.
  • asked a question related to Confocal Fluorescence Microscopy
Question
2 answers
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.
Relevant answer
Answer
You could try fluorescence imaging. Check for fluorescence emission from the drugs loaded in nanoparticles if the nanoparticles are non-fluorescent. In the absence of fluorescence from nanoparticles, the fluorescence has to come from the drug present in the nanoparticles.
But there are other methods such as infrared spectroscopy. You could identify unique characteristic IR bands of the drug molecule and check their presence in the sample confirming the presence of drug inside the nanoparticle.
  • asked a question related to Confocal Fluorescence Microscopy
Question
9 answers
Does anyone know if the streptavidin will conserve the biotin-binding capacity after a fixation by the paraformaldehyde?
thanks in advance
Relevant answer
Answer
Very interesting question.
But , when i had fixed cells with 4% PFA , it didn't reduced much signal.For erythrocyte, 2% PFA also work fine.
Best regards, 
Babu
  • asked a question related to Confocal Fluorescence Microscopy
Question
3 answers
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
Relevant answer
Answer
Hi Magdolna, I had the same before, used alpha-acetylated tubulin antibody on human airway epithelial cells grown in liquid culture and I only saw cytoskeletal structure (with the confocal). Cilia would normally appear in air/liquid interphase cultured, therefore structually differenciated cells. So if you dont see cilia on the wide-field I think the chances are good that there are no cilia. Best, Anita
  • asked a question related to Confocal Fluorescence Microscopy
Question
5 answers
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?
Relevant answer
Answer
Dear Iwan
1) You should use confocal imaging, otherwise the membrane support will obscure your image. Suppose you have an upright microscope, right?
2) When using CLSM, the distance between the cells and the coverslip is of great importance, the closer the cells are to the coverslip, the better the image (since the light intensity is inversely proportional to the square of the distance from flourophore to objective, and since your objective has a certain working distance). We normally cut the insert in tiny bits (we typically divide an insert into 4-6 pieces), place a coverslip on the table, place a piece of the insert on the coverslip with the cells facing downwards (facing the coverslip), and then add a bit of mounting medium (or PBS as Amlan describes) on the "back" of the insert. Then we turn the coverslip around , place it on a glass slide, seal with polish and make CLSM imaging. The transwell inserts tend to bulge, but if you have small bits, there is a fair chance that a part of the insert will be in close contact with the cover slip. Search for these regions before yo do the actual imaging, the closer the cells are to the cover slip, the brighter a signal you will get.  The procedure can be tricky though, it is important to keep track of whats "up" and "down" regarding the insert bits..
Best wishes
  • asked a question related to Confocal Fluorescence Microscopy
Question
1 answer
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?
Relevant answer
Answer
Hi Sebastian,
it depends on your technical onset. There are plenty of instruments that you could use for your approach. However in the case you would like to use a "normal" fluorescence microscope I it's more difficult.
At least you would need an automated motorised table to change between the different wells. Than you have to do a first run and measure the time the microscope needs to get to all wells and acquire an image. Your max amount of wells you could analyse is then defined by by the measured wells within 5min.
Keep in mind that not all fluorochromes are that stable over time, the medium should be colorless and you would need a proper control to generate a baseline (you will loose signal over time due to bleaching) on which you adjust your data (like % of control at timepoint x).
If there wouldn't be the need for a single cell anlysis you could use a platereader.
  • asked a question related to Confocal Fluorescence Microscopy
Question
1 answer
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?
Relevant answer
Answer
Concentrations of 10^(-4) M is sufficient for your experiment. To verify whether the dye is dissolved whole, slightly heat the solution and using a magnetic stirrer, for instance 2 h. Then cool the solution to the initial temperature and repeat the measurement of the fluorescence spectrum under the same experimental conditions. Repeat use of heated mixers and increasing time, for example up to 4 hours. Again, repeat measurement of fluorescence under the same experimental conditions. Finally, just keep the solution for a day with a little heated. The next day, refrigerate the solution and repeat the measurement of the fluorescence spectrum under the same experimental conditions. Compare four spectra. If there are no changes, your dye is highly soluble in oil. Use the same type of calibrated cell .
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
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?
Relevant answer
Answer
Hi Joe, in my opinion this is the way it should be.
You can also do statistics within your groups, but this will only show your inter-experimental variation (technical n).
  • asked a question related to Confocal Fluorescence Microscopy
Question
6 answers
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.
Relevant answer
Answer
What is your positive control, and is it the same between both lots?  One thing that isn't well-known about this dye is that H2O2 is not a good positive control for it.  Also, PBS is probably not the best choice.  I suggest something more physiological, such as HBSS, phenol red-free media, or FluoroBrite DMEM.
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
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
Relevant answer
Answer
Hi Masoud.
I attach you a link of an spectra viewer that will let you compare among a bunch of fluorochromes. You also will be able to apply filters (excitation/emission wavelenghts, beamsplitters, and filters).
Also, the majority of the fluorochromes manufactoring companies have their own spectra viewers.
Hope this works for you.
  • asked a question related to Confocal Fluorescence Microscopy
Question
1 answer
.
Relevant answer
Answer
Hi Vipin,
the accurcy of your localization is strongly depending on signal to noise, pixel size and the size of the point spread function. You might want to take a look into this publication doi:10.1038/nmeth.1447
The experimental localization accuracy is often measured as the FWHM of the distribution of a big number of localizations from one emitter. For example: localize one bead a thousand times and analyze the distribution of the thousand localizations.
Best,
Thorge
  • asked a question related to Confocal Fluorescence Microscopy
Question
4 answers
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?  
Relevant answer
Answer
It depends on what kind of optical setup you want. In the upright microscope, the laser light comes from the upside so it is little difficult to align the setup. However, both the microscopes are the better choice for fluorescence imaging of fix cell. In my experience, I had faced many difficulties with the upright microscope during alignment. Because all the optical components need to use on the upper side of the optical table.
  • asked a question related to Confocal Fluorescence Microscopy
Question
5 answers
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.
Relevant answer
Answer
Hi Sonia,
I absolutely agree with the suggestions above.I had experienced something similar. After investigating, I've had to use a different promoter since, the used promoter was very weak. Which Promoter do you use in your construction? 
Here are some other suggestion to find the reason: 
-  sequenced the entire promoter region, your insert and the detection tag in order to insure the insert is in frame?
- The protein you try to express may be degraded immediately after it has been synthesised. 
Good Luck
Ladan