Springer Nature Experiments

Laser Scanning Confocal Microscopy: History, Applications, and Related Optical Sectioning Techniques

Series: Methods In Molecular Biology > Book: Confocal Microscopy

Overview | DOI: 10.1007/978-1-60761-847-8_2

  • Howard Hughes Medical Institute, Department of Molecular Biology, University of Wisconsin, Madison, WI, USA
  • Laboratory for Optical and Computational Instrumentation and Laboratory of Molecular Biology, University of Wisconsin, Madison, WI, USA

Full Text Entitlement Icon

Confocal microscopy is an established light microscopical technique for imaging fluorescently labeled specimens with significant three-dimensional structure. Applications of confocal microscopy in the biomedical sciences include the imaging of the

Confocal microscopy is an established light microscopical technique for imaging fluorescently labeled specimens with significant three-dimensional structure. Applications of confocal microscopy in the biomedical sciences include the imaging of the spatial distribution of macromolecules in either fixed or living cells, the automated collection of 3D data, the imaging of multiple labeled specimens and the measurement of physiological events in living cells. The laser scanning confocal microscope continues to be chosen for most routine work although a number of instruments have been developed for more specific applications. Significant improvements have been made to all areas of the confocal approach, not only to the instruments themselves, but also to the protocols of specimen preparation, to the analysis, the display, the reproduction, sharing and management of confocal images using bioinformatics techniques.

Figures ( 0 ) & Videos ( 0 )

Experimental specifications, other keywords.

laser scanning confocal microscopy experiments

Citations (37)

Related articles, a paintbrush for delivery of nanoparticles and molecules to live cells with precise spatiotemporal control, imaging microtubule network in rodent giant glutamatergic presynaptic terminals.

Author Email

Large-scale deep tissue voltage imaging with targeted-illumination confocal microscopy

Long-term imaging of endoplasmic reticulum morphology in embryos during seed germination, make it shine: labelling the er for light and fluorescence microscopy, nano/microformulations for bacteriophage delivery, observation of dietary lipids using microscopy techniques.

  • Conchello JA, Lichtman JW (2005) Optical sectioning microscopy. Nat Methods 2:920–931
  • Paddock SW (1999) Protocols in Confocal Microscopy. Methods Mol Biol 122. Humana Press, Totowa, NJ
  • Chalfie M, Tu Y, Euskirchen G, Ward WW, Prasher DC (1994) Green fluorescent protein as a marker for gene expression. Science 263:802–805
  • Fraser SE (2003) Crystal gazing in optical microscopy. Nat Biotechnol 21:1272–1273
  • Walter T et al (2010) Visualization of image data from cells to organisms. Nat Methods 7:S26–S41
  • Hibbs AR (2004) Confocal microscopy for biologists. Springer, New York
  • Price R, Jerome WG (2011) Basic confocal microscopy. Springer, New York
  • Minsky M (1988) Memoir on inventing the confocal scanning microscope. Scanning 10:128–138
  • Minsky M (1957) Microscopy apparatus U.S. Patent no. 3013467
  • Brakenhoff GJ, van der Voort HTM, van Spronsen EA, Linnemans WAM, Nanninga N (1985) Three-dimensional chromatin distribution in neuroblastoma nuclei shown by confocal laser scanning laser microscopy. Nature 317:748–749
  • Pawley JB (2006) Handbook of Biological Confocal Microscopy, 3rd edn. Plenum Press, New York
  • Amos WB, White JG (2003) How the confocal laser scanning microscope entered biological research. Biol Cell 95:335–342
  • White JG, Amos WB (1987) Confocal microscopy comes of age. Nature 328:183–184
  • White JG, Amos WB, Durbin R, Fordham M (1990) Development of a confocal imaging system for biological epifluorescence application in "Optical Microscopy For Biology". Wiley-Liss Inc, New York, NY, pp 1–18
  • White JG, Amos WB, Fordham M (1987) An evaluation of confocal versus conventional imaging of biological structures by fluorescence light microscopy. J Cell Biol 1987(105):41–48
  • Wilson T (1995) The role of the pinhole in confocal imaging system. Plenum Press, New York, pp 167–182
  • Paddock SW (2002) Confocal imaging of Drosophila embryos. In: Matsumoto B (ed) Cell biological applications of confocal microscopy, 2nd edn. Methods Cell Biol 70:355–372
  • Kosman D, Mizutani CM, Lemons D, Cox WG, McGinnis W, Bier E (2004) Multiplex detection of RNA expression in Drosophila embryos. Science 305:846
  • Brelje TC, Wessendorf MW, Sorenson RL (1993) Multicolor laser scanning confocal immunofluorescence microscopy: practical applications and limitations. Methods Cell Biol 38:98–177
  • Livet J, Weissman TA, Kang H, Draft RW, Lu J, Bennis RA, Sanes JR, Lichtman JW (2007) Transgenic strategies for combinatorial expression of fluorescent proteins in the nervous system. Nature 450:56–62
  • Murray JM, Appleton PL, Swedlow JR, Waters JC (2007) Evaluating performance in three-dimensional fluorescence microscopy. J Microsc 228:390–405
  • Van Roessel P, Brand A (2002) Imaging into the future: visualizing gene expression and protein interactions with fluorescent proteins. Nat Cell Biol 4:E15–E20
  • Paddock SW (2001) A brief history of time-lapse. Biotechniques 30:283–289
  • Pawley JB (2000) The 39 steps: a cautionary tale of quantitative 3-D fluorescence microscopy. Biotechniques 28:884–887
  • Heim R, Tsien RY (1996) Engineering green fluorescent protein for improved brightness, longer wavelength and fluorescence energy transfer. Curr Biol 6:178–182
  • Shaner NC, Patterson GH, Davidson MW (2007) Advances in fluorescent protein technology. J Cell Sci 120:4247–4260
  • Tsien RY (1998) The green fluorescent protein. Annu Rev Biochem 67:509–544
  • Giepmans BNG, Adams SR, Ellisman MH, Tsien RY (2006) The fluorescent toolbox for assessing protein location and function. Science 312:217–224
  • Fiala A, Suska A, Schulter OM (2010) Optogenetic approaches in neuroscience. Curr Biol 20:R897–R903
  • Mohler WA, White JG (1998) Stereo-4-D reconstruction and animation from living fluorescent specimens. Biotechniques 24:1006–1012
  • Provenzano PP, Eliceiri KW, Keely PJ (2009) Shining new light on 3D cell motility and the metastatic process. Trends Cell Biol 19:638–48
  • Paddock SW (2002) Confocal reflection microscopy; the “other” confocal mode. Biotechniques 32:274–278
  • Paddock SW, Mahoney S, Minshall M, Smith LC, Duvic M, Lewis D (1991) Improved detection of in situ hybridisation by laser scanning confocal microscopy. Biotechniques 11:486–494
  • Gaietta G, Deerinck TJ, Adams SR, Bouwer J, Tour O, Laird DW, Sosinsky GE, Tsien RY, Ellisman MH (2002) Multicolor and electron microscopic imaging of connexin trafficking. Science 296:503–507
  • Deerinck TJ, Martone ME, Lev-Ram V, Green DPL, Tsien RY, Spector DL, Huang S, Ellisman MH (1994) Fluorescence photooxidation with eosin: a method for high-resolution immunolocalisation and in situ hybridisation detection for light and electron microscopy. J Cell Biol 126:901–910
  • Piston DW, Kremers GJ (2007) Fluorescent protein FRET: the good, the bad and the ugly. Trends Biochem Sci 32:407–414
  • Pietraszewska-Bogiel A, Gadella TWJ (2010) FRET microscopy: from principle to routine technology in cell biology. J Microsc 240:111–118
  • Bastiaens PI, Squire A (1999) Fluorescence lifetime imaging microscopy: spatial resolution of biochemical processes in the cell. Trends Cell Biol 9:48–52
  • Borst JW, Visser AJWG (2010) Fluorescence lifetime imaging microscopy in life sciences. Meas Sci Technol 21:1–21
  • Conklin MW, Provenzano PP, Eliceiri KW, Sullivan R, Keely PJ (2009) Fluorescence lifetime imaging of endogenous fluorophores in histopathology sections reveals differences between normal and tumor epithelium in carcinoma in situ of the breast. Cell Biochem Biophys 53:145–57
  • van Royen ME, Dinant C, Farla P, Trapman J, Houtsmuller AB (2009) FRAP and FRET methods to study nuclear receptors in living cells. Methods Mol Biol 505:69–96
  • Lippincott-Schwartz J, Altan-Bonnet N, Patterson GH (2003) Photobleaching and photoactivation: following protein dynamics in living cells. Nat Cell Biol 2003:S7–14
  • Weigel A, Schild D, Zeug A (2009) Resolution in the ApoTome and the confocal laser scanning microscope: comparison. J BioMed Opt 14. Online article #014023
  • Wallace W, Schaefer LH, Swedlow JR (2001) A workingperson's guide to deconvolution in light microscopy. Biotechniques 5:1076–1080
  • Svoboda K, Yasuda R (2006) Principles of two-photon excitation microscopy and its applications to neuroscience. Neuron 50:823–839
  • Zipfel WR, Williams RM, Webb W (2002) Nonlinear magic: multiphoton microscopy in the biosciences. Nat Biotechnol 21:1369–1377
  • Helmchen F, Denk W (2005) Deep tissue two-photon microscopy. Nat Methods 2:932–919
  • Campagnola PJ, Loew LM (2003) Second-harmonic imaging microscopy for visualizing biomolecular arrays in cells, tissues and organisms. Nat Biotechnol 21:1356–1360
  • Keller PJ, Schmidt AD, Wittbrodt J, Stelzer EHK (2008) Reconstruction of zebrafish early embryonic development by scanned light sheet microscopy. Science 322:1065–1069
  • Keller PJ, Schimdt AD, Santella A, Khairy K, Bao Z, Wittbrodt J, Stelzer EHK (2010) Fast, high-contrast imaging of animal development with scanned light sheet-based structured-illumination microscopy. Nat Methods 7:637–642
  • Axelrod D (2003) Total internal reflection fluorescence microscopy in cell biology. Methods Enzymol 361:1–33
  • Schermelleh L, Heintzmann R, Leonhardt H (2010) A guide to super-resolution fluorescence microscopy. J Cell Biol 190:165–175
  • Sharpe A, Ahlgren U, Perry P, Hill B, Ross a, Hecksher-Sorensen J, Baldock R, Davidson D (2002) Optical projection tomography as a tool for 3D microscopy and gene expression studies. Science 296:541–545
  • St. Croix C, Zipfel WR, Watkins SC (2007) Potential solutions for confocal imaging of living animals. Biotechniques 43:14–19
  • Paddock SW (2001) Channel surfing: creating different colour combinations from multi-label images. Biotechniques 30:756–761
  • Waters JC, Swedlow JR (2007) Interpreting fluorescence microscopy images and measurements. Cell Online 37–42
  • Swedlow JR, Lewis SE, Goldberg IG (2006) Modelling data across labs, genomes, space and time. Nat Cell Biol 8:1190–1194
  • Swedlow JR, Goldberg I, Brauner E, Sorger PK (2003) Informatics and quantitative analysis in biological imaging. Science 300:100–102
  • Peng H (2008) Bioimage informatics: a new era of engineering biology. Bioinformatics 24:1827–1836
  • Swedlow J.R., Goldberg I.G., Eliceiri K.W., and the OME Consortium (2009) Bioimage informatics for experimental biology. Annu Rev Biophys 38:327–346
  • Linkert M, Rueden CT, Allan C, Burel JM, Moore W, Patterson A, Loranger B, Moore J, Neves C, Macdonald D, Tarkowska A, Sticco C, Hill E, Rossner M, Eliceiri KW, Swedlow JR (2010) Metadata matters: access to image data in the real world. J Cell Biol 189:777–782
  • Conn PM (2010) Techniques in confocal microscopy (reliable Lab solutions). Academic, New York
  • Goldman RD, Spector DL, Swedlow JR (2009) Live cell imaging: a laboratory manual. Cold Spring Harbor Press, New York
  • Murphy DB, Davidson MW (2012) Fundamentals of light microscopy and electronic imaging. Wiley-Blackwell, Hoboken, NJ
  • Fischer RS, Wu Y, Kanchanawong P, Shroff H, Waterman CM (2011) Microscopy in 3D: a biologist’s toolbox. Trends Cell Biol 21:682–691
  • Swedlow JR (2012) Innovation in biological microscopy: current status and future directions. Bioessays 34:333–340

Advertisement

Laser Scanning Confocal Microscopy: History, Applications, and Related Optical Sectioning Techniques

  • First Online: 01 January 2013

Cite this protocol

laser scanning confocal microscopy experiments

  • Stephen W. Paddock 3 &
  • Kevin W. Eliceiri 4  

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1075))

10k Accesses

37 Citations

Confocal microscopy is an established light microscopical technique for imaging fluorescently labeled specimens with significant three-dimensional structure. Applications of confocal microscopy in the biomedical sciences include the imaging of the spatial distribution of macromolecules in either fixed or living cells, the automated collection of 3D data, the imaging of multiple labeled specimens and the measurement of physiological events in living cells. The laser scanning confocal microscope continues to be chosen for most routine work although a number of instruments have been developed for more specific applications. Significant improvements have been made to all areas of the confocal approach, not only to the instruments themselves, but also to the protocols of specimen preparation, to the analysis, the display, the reproduction, sharing and management of confocal images using bioinformatics techniques.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Subscribe and save.

  • Get 10 units per month
  • Download Article/Chapter or eBook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
  • Available as EPUB and PDF
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
  • Durable hardcover edition

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

laser scanning confocal microscopy experiments

Advances in Confocal Microscopy and Selected Applications

General considerations for acquiring a three-color image by laser scanning confocal microscopy.

laser scanning confocal microscopy experiments

Tutorial: guidance for quantitative confocal microscopy

Conchello JA, Lichtman JW (2005) Optical sectioning microscopy. Nat Methods 2:920–931

Article   CAS   PubMed   Google Scholar  

Paddock SW (1999) Protocols in Confocal Microscopy. Methods Mol Biol 122. Humana Press, Totowa, NJ

Google Scholar  

Chalfie M, Tu Y, Euskirchen G, Ward WW, Prasher DC (1994) Green fluorescent protein as a marker for gene expression. Science 263:802–805

Fraser SE (2003) Crystal gazing in optical microscopy. Nat Biotechnol 21:1272–1273

Walter T et al (2010) Visualization of image data from cells to organisms. Nat Methods 7:S26–S41

Article   CAS   PubMed Central   PubMed   Google Scholar  

Hibbs AR (2004) Confocal microscopy for biologists. Springer, New York

Book   Google Scholar  

Price R, Jerome WG (2011) Basic confocal microscopy. Springer, New York

Minsky M (1988) Memoir on inventing the confocal scanning microscope. Scanning 10:128–138

Article   Google Scholar  

Minsky M (1957) Microscopy apparatus U.S. Patent no. 3013467

Brakenhoff GJ, van der Voort HTM, van Spronsen EA, Linnemans WAM, Nanninga N (1985) Three-dimensional chromatin distribution in neuroblastoma nuclei shown by confocal laser scanning laser microscopy. Nature 317:748–749

Pawley JB (2006) Handbook of Biological Confocal Microscopy, 3rd edn. Plenum Press, New York

Amos WB, White JG (2003) How the confocal laser scanning microscope entered biological research. Biol Cell 95:335–342

White JG, Amos WB (1987) Confocal microscopy comes of age. Nature 328:183–184

White JG, Amos WB, Durbin R, Fordham M (1990) Development of a confocal imaging system for biological epifluorescence application in "Optical Microscopy For Biology". Wiley-Liss Inc, New York, NY, pp 1–18

White JG, Amos WB, Fordham M (1987) An evaluation of confocal versus conventional imaging of biological structures by fluorescence light microscopy. J Cell Biol 1987(105):41–48

Wilson T (1995) The role of the pinhole in confocal imaging system. Plenum Press, New York, pp 167–182

Paddock SW (2002) Confocal imaging of Drosophila embryos. In: Matsumoto B (ed) Cell biological applications of confocal microscopy, 2nd edn. Methods Cell Biol 70:355–372

Kosman D, Mizutani CM, Lemons D, Cox WG, McGinnis W, Bier E (2004) Multiplex detection of RNA expression in Drosophila embryos. Science 305:846

Brelje TC, Wessendorf MW, Sorenson RL (1993) Multicolor laser scanning confocal immunofluorescence microscopy: practical applications and limitations. Methods Cell Biol 38:98–177

Livet J, Weissman TA, Kang H, Draft RW, Lu J, Bennis RA, Sanes JR, Lichtman JW (2007) Transgenic strategies for combinatorial expression of fluorescent proteins in the nervous system. Nature 450:56–62

Murray JM, Appleton PL, Swedlow JR, Waters JC (2007) Evaluating performance in three-dimensional fluorescence microscopy. J Microsc 228:390–405

Article   PubMed Central   PubMed   Google Scholar  

Van Roessel P, Brand A (2002) Imaging into the future: visualizing gene expression and protein interactions with fluorescent proteins. Nat Cell Biol 4:E15–E20

Article   PubMed   Google Scholar  

Paddock SW (2001) A brief history of time-lapse. Biotechniques 30:283–289

CAS   PubMed   Google Scholar  

Pawley JB (2000) The 39 steps: a cautionary tale of quantitative 3-D fluorescence microscopy. Biotechniques 28:884–887

Heim R, Tsien RY (1996) Engineering green fluorescent protein for improved brightness, longer wavelength and fluorescence energy transfer. Curr Biol 6:178–182

Shaner NC, Patterson GH, Davidson MW (2007) Advances in fluorescent protein technology. J Cell Sci 120:4247–4260

Tsien RY (1998) The green fluorescent protein. Annu Rev Biochem 67:509–544

Giepmans BNG, Adams SR, Ellisman MH, Tsien RY (2006) The fluorescent toolbox for assessing protein location and function. Science 312:217–224

Fiala A, Suska A, Schulter OM (2010) Optogenetic approaches in neuroscience. Curr Biol 20:R897–R903

Mohler WA, White JG (1998) Stereo-4-D reconstruction and animation from living fluorescent specimens. Biotechniques 24:1006–1012

Provenzano PP, Eliceiri KW, Keely PJ (2009) Shining new light on 3D cell motility and the metastatic process. Trends Cell Biol 19:638–48

Paddock SW (2002) Confocal reflection microscopy; the “other” confocal mode. Biotechniques 32:274–278

Paddock SW, Mahoney S, Minshall M, Smith LC, Duvic M, Lewis D (1991) Improved detection of in situ hybridisation by laser scanning confocal microscopy. Biotechniques 11:486–494

Gaietta G, Deerinck TJ, Adams SR, Bouwer J, Tour O, Laird DW, Sosinsky GE, Tsien RY, Ellisman MH (2002) Multicolor and electron microscopic imaging of connexin trafficking. Science 296:503–507

Deerinck TJ, Martone ME, Lev-Ram V, Green DPL, Tsien RY, Spector DL, Huang S, Ellisman MH (1994) Fluorescence photooxidation with eosin: a method for high-resolution immunolocalisation and in situ hybridisation detection for light and electron microscopy. J Cell Biol 126:901–910

Piston DW, Kremers GJ (2007) Fluorescent protein FRET: the good, the bad and the ugly. Trends Biochem Sci 32:407–414

Pietraszewska-Bogiel A, Gadella TWJ (2010) FRET microscopy: from principle to routine technology in cell biology. J Microsc 240:111–118

Bastiaens PI, Squire A (1999) Fluorescence lifetime imaging microscopy: spatial resolution of biochemical processes in the cell. Trends Cell Biol 9:48–52

Borst JW, Visser AJWG (2010) Fluorescence lifetime imaging microscopy in life sciences. Meas Sci Technol 21:1–21

Conklin MW, Provenzano PP, Eliceiri KW, Sullivan R, Keely PJ (2009) Fluorescence lifetime imaging of endogenous fluorophores in histopathology sections reveals differences between normal and tumor epithelium in carcinoma in situ of the breast. Cell Biochem Biophys 53:145–57

van Royen ME, Dinant C, Farla P, Trapman J, Houtsmuller AB (2009) FRAP and FRET methods to study nuclear receptors in living cells. Methods Mol Biol 505:69–96

Lippincott-Schwartz J, Altan-Bonnet N, Patterson GH (2003) Photobleaching and photoactivation: following protein dynamics in living cells. Nat Cell Biol 2003:S7–14

Weigel A, Schild D, Zeug A (2009) Resolution in the ApoTome and the confocal laser scanning microscope: comparison. J BioMed Opt 14. Online article #014023

Wallace W, Schaefer LH, Swedlow JR (2001) A workingperson's guide to deconvolution in light microscopy. Biotechniques 5:1076–1080

Svoboda K, Yasuda R (2006) Principles of two-photon excitation microscopy and its applications to neuroscience. Neuron 50:823–839

Zipfel WR, Williams RM, Webb W (2002) Nonlinear magic: multiphoton microscopy in the biosciences. Nat Biotechnol 21:1369–1377

Helmchen F, Denk W (2005) Deep tissue two-photon microscopy. Nat Methods 2:932–919

Campagnola PJ, Loew LM (2003) Second-harmonic imaging microscopy for visualizing biomolecular arrays in cells, tissues and organisms. Nat Biotechnol 21:1356–1360

Keller PJ, Schmidt AD, Wittbrodt J, Stelzer EHK (2008) Reconstruction of zebrafish early embryonic development by scanned light sheet microscopy. Science 322:1065–1069

Keller PJ, Schimdt AD, Santella A, Khairy K, Bao Z, Wittbrodt J, Stelzer EHK (2010) Fast, high-contrast imaging of animal development with scanned light sheet-based structured-illumination microscopy. Nat Methods 7:637–642

Axelrod D (2003) Total internal reflection fluorescence microscopy in cell biology. Methods Enzymol 361:1–33

Schermelleh L, Heintzmann R, Leonhardt H (2010) A guide to super-resolution fluorescence microscopy. J Cell Biol 190:165–175

Sharpe A, Ahlgren U, Perry P, Hill B, Ross a, Hecksher-Sorensen J, Baldock R, Davidson D (2002) Optical projection tomography as a tool for 3D microscopy and gene expression studies. Science 296:541–545

St. Croix C, Zipfel WR, Watkins SC (2007) Potential solutions for confocal imaging of living animals. Biotechniques 43:14–19

Paddock SW (2001) Channel surfing: creating different colour combinations from multi-label images. Biotechniques 30:756–761

Waters JC, Swedlow JR (2007) Interpreting fluorescence microscopy images and measurements. Cell Online 37–42

Swedlow JR, Lewis SE, Goldberg IG (2006) Modelling data across labs, genomes, space and time. Nat Cell Biol 8:1190–1194

Swedlow JR, Goldberg I, Brauner E, Sorger PK (2003) Informatics and quantitative analysis in biological imaging. Science 300:100–102

Peng H (2008) Bioimage informatics: a new era of engineering biology. Bioinformatics 24:1827–1836

Swedlow J.R., Goldberg I.G., Eliceiri K.W., and the OME Consortium (2009) Bioimage informatics for experimental biology. Annu Rev Biophys 38:327–346

Linkert M, Rueden CT, Allan C, Burel JM, Moore W, Patterson A, Loranger B, Moore J, Neves C, Macdonald D, Tarkowska A, Sticco C, Hill E, Rossner M, Eliceiri KW, Swedlow JR (2010) Metadata matters: access to image data in the real world. J Cell Biol 189:777–782

Conn PM (2010) Techniques in confocal microscopy (reliable Lab solutions). Academic, New York

Goldman RD, Spector DL, Swedlow JR (2009) Live cell imaging: a laboratory manual. Cold Spring Harbor Press, New York

Murphy DB, Davidson MW (2012) Fundamentals of light microscopy and electronic imaging. Wiley-Blackwell, Hoboken, NJ

Fischer RS, Wu Y, Kanchanawong P, Shroff H, Waterman CM (2011) Microscopy in 3D: a biologist’s toolbox. Trends Cell Biol 21:682–691

Swedlow JR (2012) Innovation in biological microscopy: current status and future directions. Bioessays 34:333–340

Download references

Author information

Authors and affiliations.

Howard Hughes Medical Institute, Department of Molecular Biology, University of Wisconsin, Madison, WI, USA

Stephen W. Paddock

Laboratory for Optical and Computational Instrumentation and Laboratory of Molecular Biology, University of Wisconsin, Madison, WI, USA

Kevin W. Eliceiri

You can also search for this author in PubMed   Google Scholar

Editor information

Editors and affiliations.

University of Wisconsin Dept. of Molecular Biology, Madison, Wisconsin, USA

Rights and permissions

Reprints and permissions

Copyright information

© 2014 Springer Science+Business Media New York

About this protocol

Paddock, S.W., Eliceiri, K.W. (2014). Laser Scanning Confocal Microscopy: History, Applications, and Related Optical Sectioning Techniques. In: Paddock, S. (eds) Confocal Microscopy. Methods in Molecular Biology, vol 1075. Humana Press, New York, NY. https://doi.org/10.1007/978-1-60761-847-8_2

Download citation

DOI : https://doi.org/10.1007/978-1-60761-847-8_2

Published : 24 August 2013

Publisher Name : Humana Press, New York, NY

Print ISBN : 978-1-58829-351-0

Online ISBN : 978-1-60761-847-8

eBook Packages : Springer Protocols

  • Publish with us

Policies and ethics

  • Find a journal
  • Track your research

laser scanning confocal microscopy experiments

Nikon Instruments | Nikon Global | Nikon Small World

Specimen Preparation and Imaging

laser scanning confocal microscopy experiments

The procedures for preparing and imaging specimens in the confocal microscope are largely derived from those that have been developed over many years for use with the conventional wide field microscope. The best approach in developing a new protocol for a specimen to be imaged with the confocal microscope is to begin with one known to be appropriate for conventional microscopy, and to modify it as necessary.

Figure 1 - Merged 3-Channel Image

Regardless of the specimen preparation protocol employed, a primary benefit of the manner in which confocal microscopy is carried out is the flexibility in image display and analysis that results from the simultaneous collection of multiple images, in digital form, into a computer. This is discussed in more detail below, but one elegant example of the image display possibilities is presented in Figure 1 , a triple-labeled Drosophila embryo at the cellular blastoderm stage. The specimen was immunofluorescently labeled with antibodies to three different proteins. After three corresponding images were collected in the red, green, and blue channels of the confocal system, the images could be rearranged by copying them to different channels. By evaluating the image resulting from merging the three, the most effective color-to-channel assignment for illustrating the various protein domains was chosen. Figure 1 presents the merged three-channel image (combined red, green, and blue channels).

Most of the methods that have proven successful in preparing specimens for the conventional wide field optical microscope are specifically aimed at reducing the amount of out-of-focus fluorescence, since this produces flare in the image that greatly reduces the resolution of the features of interest. Due to the optical sectioning achieved by the confocal approach, the confocal microscope undersamples the fluorescence in a thick specimen as compared to the conventional epifluorescence microscope. The result is that samples may require increased staining times or stain concentrations for confocal analysis, and if evaluated in the conventional microscope, may appear to be over stained.

Although the illumination in the typical laser scanning confocal system appears to be extremely bright, the average illumination at any given point on the specimen is relatively moderate, due to the fact that many points are scanned per second. At a typical scan speed of one point per 1.6 microseconds, the actual illumination at any given point is generally less than in a conventional wide field epifluorescence microscope. It is usually advisable to use the lowest laser power that is practical for imaging in order to protect the fluorophore. Although many protocols include an antibleaching agent to prevent fading of the fluorescent species, such additives may not be required with many of the more modern confocal instruments, such as the Nikon A1 HD25/A1R HD25 and C2+ systems.

The primary advantage and application of the confocal microscope is in improved imaging ability of thicker specimens, although the success can be limited by the specific properties of the specimen. Certain minimum physical requirements for the specimen apply; it must fit on the microscope stage, and the area of interest must be able to be placed within the working distance range of the objective lens. In some cases resolution may have to be compromised in order to accommodate a specimen, and to avoid damage to it or the objective lens. For example, a high resolution lens such as a 60x having a numerical aperture of 1.4 may have a working distance of 170 micrometers, whereas a 20x (having a typical numerical aperture of 0.75) might offer a relatively large 660 micrometer working distance, with the ability to access more restricted areas of a specimen without physical interference.

Specimens that have three-dimensional structure that is to be studied with the confocal microscope, have to be mounted in such a way as to preserve the structure. Some sort of spacer, such as fishing line or a piece of coverslip, is commonly placed between the slide and the coverslip to avoid deforming the specimen. When living samples are to be studied, it is usually necessary to mount them in a chamber that provides all of the necessary requirements for life, and that will also allow sufficient access by the objective lens to image the desired area.

Table 1 - Objective Lens Parameters and Optical Section Thickness

ObjectivePinhole
MagnificationNAClosed (1 mm)Open (7 mm)
60x1.400.41.9
40x1.300.63.3
40x0.551.44.3
25x0.801.47.8
4x0.2020.0100.0

Specimen properties that affect light transmission, such as opacity and turbidity, can greatly influence the depth of penetration of the laser beam into the specimen, and consequently the structures that can be imaged. Unfixed and unstained corneal epithelium of the eye, for example, is relatively transparent and a laser beam will penetrate it to a depth of about 200 micrometers. In contrast, unfixed skin is relatively opaque and scatters more light, limiting the laser penetration to about 10 micrometers. Many fixation protocols include some form of clearing agent intended to increase the transparency of the tissue.

If sufficient laser penetration cannot be achieved with a whole mount specimen, thick sections can be cut using a microtome. Fixed tissue is usually used for sectioning, but tissue (such as living brain) has been cut by vibratome and successfully imaged. To gain access to deeper parts of a section mount, it is possible to remove the specimen from the slide, invert it, and remount it, but this is usually not very successful. Images from a somewhat deeper part of a specimen can be obtained by using dyes that are excited at longer wavelengths (such as cyanine 5), as opposed to those that require shorter wavelength excitation. The use of longer wavelength illumination will, however, slightly reduce the maximum resolution that can be achieved in comparison to images acquired at shorter wavelengths. For similar reasons, multiple-photon imaging techniques allow images to be collected from deeper levels within a specimen (due to the use of red light for excitation).

The Objective Lens

For confocal microscope studies, the choice of the objective lens used is extremely important, as the light-collecting ability of the lens, measured as its numerical aperture, is a determinant of both resolution and optical section thickness. Holding other microscope variables constant, the higher the objective numerical aperture is, the thinner the optical section will be. As examples for one particular instrument, the optical section thickness using a 60x (numerical aperture of 1.4) objective with the pinhole diameter set at 1mm is approximately 0.4 micrometer, and with a 16x (numerical aperture of 0.5) lens, with the same 1-millimeter pinhole, the section thickness is on the order of 1.8 millimeters. By opening the pinhole to a larger diameter, the optical section thickness can be increased. Table 1 gives optical section thickness values (in micrometers) for various objective lenses at two different pinhole diameters for one model of LSCM . The image resolution is always poorer vertically than it is horizontally. For example, using the 60x, 1.4 numerical aperture, objective lens the horizontal resolution is approximately 0.2 micrometer, and the vertical resolution about 0.5 micrometer. Flatness of field and chromatic aberration are additional lens characteristics to be considered when choosing an objective lens. The degree of chromatic aberration correction is particularly important when imaging multilabeled specimens at different wavelengths.

Objective lenses that are capable of the highest resolution generally are those with the highest magnification and the highest numerical aperture. They are also the most expensive, so a compromise is often made between the area of the specimen that is scanned and the maximum resolution that can be achieved for that area. If imaging insect embryos and imaginal disks, for example, a 4x lens might be used to locate the specimen on the slide, a 16x (numerical aperture of 0.5) lens for imaging whole embryos, and a 40x (numerical aperture of 1.2) or 60x (numerical aperture of 1.4) lens for resolving individual cell nuclei within embryos or imaginal disks. For imaging larger tissues, such as butterfly imaginal disks, the 4x lens would be useful for whole wing disks, and the 40x or 60x for resolution of individual cells. Figure 2(a) illustrates use of a 4x objective to obtain an overall view of an entire butterfly fifth instar wing imaginal disk, and the additional nuclear detail provided by a 16x lens (Figure 2(b)). One way in which high resolution and large image fields of view can be combined is to acquire many images from adjoining areas and to combine them digitally into montages. Some microscopes have automated x-y stages that can be set up to move around the specimen and collect multiple images into a large-area montage.

Figure 2 - Objective Lens Zooming

One of the more useful features that is characteristic of most LSCMs is the ability to zoom an image using the same objective lens, with no loss of resolution. This capability is achieved simply by decreasing the area of the specimen scanned by the laser, by control of the scanning mirrors, while maintaining the same image display size or memory storage array size, effectively increasing the magnification. In this way several magnifications are achieved with a single lens without disturbing the specimen or losing track of reference points in the field of view. Whenever possible, however, a lens of higher numerical aperture should be used to maximize resolution, rather than zooming using a lens of lower numerical aperture. The capability of zooming with a single lens (40x) is illustrated in Figure 2(c-f). Panel (c) of the figure shows the additional nuclear detail provided by the 40x objective (compared to the 16x view of panel (b)). Panels (d) through (f) are images obtained by zooming the same 40x lens by progressive increments, accomplished by reducing the area scanned on the specimen.

A number of confocal instrument designs have an adjustable pinhole that limits the out-of-focus light that reaches the detector. Opening the pinhole to a larger diameter produces a thicker optical section and reduced resolution, but is often necessary to include more specimen detail or to increase the light striking the detector. As the pinhole is closed (diameter reduced) the optical section thickness and brightness decrease. Resolution increases until a certain minimum pinhole diameter is reached, beyond which resolution does not increase but brightness continues to decrease. The pinhole diameter at which this condition is reached is different for each objective lens.

Probes for Confocal Imaging

The development of confocal instrumentation continues to both influence and be influenced by the synthesis of novel fluorescent probes that improve immunofluorescence localization. Fluorochromes are being introduced that have excitation and emission spectra more closely matched to the wavelengths produced by the lasers supplied with most commercial LSCMs. Improved probes that can be conjugated to antibodies of current research interest are continually developed. As one example group of dyes, the cyanines have developed as alternatives to other long-established dyes, with cyanine 3 as a brighter alternative to rhodamine, and cyanine 5 finding increased use in triple-label strategies.

Fluorescence in-situ hybridization ( FISH ) has advantages in resolution and sensitivity of probe detection that are further enhanced when coupled with the LSCM, and is a valuable approach for imaging the distribution of fluorescently labeled DNA and RNA sequences in cells. In addition, brighter fluorescent probes are currently available for LSCM imaging of total DNA in both nuclei and isolated chromosomes.

A large number of fluorescent probes are available that, when incorporated in relatively simple protocols, specifically stain certain cellular organelles and structures. Among the plethora of available probes are dyes that label nuclei, the Golgi apparatus, the endoplasmic reticulum, and mitochondria, and also dyes such as fluorescently labeled phalloidins that target polymerized actin in cells. Such dyes are very useful in multiple labeling approaches to locate antigens of interest having specific compartments in the cell. For example, Figure 3 presents the employment of a combination of phalloidin and a nuclear dye ( ToPro ) with the appropriate antigen in a triple labeling scheme applied to whole mounts of butterfly pupal wing imaginal disks. As illustrated in Figure 3(a) , phalloidin can be used to accentuate cell outlines in developing tissues, with the peripheral actin meshwork being labeled as bright fluorescent rings. Panel (b) of the figure illustrates the dramatic specificity of the nuclear dye in labeling just that one cellular component. In addition to this cellular compartment labeling strategy, antibodies to proteins of known distribution or function in cells (such as antitubulin) can be usefully included in multilabel studies.

Figure 3 - Cellular Feature Labeling

If living cells are being imaged, it is critical to be aware of the effects of adding fluorochromes to the system. These probes can be toxic to living cells, especially when they are excited with the laser. Toxic affects are reduced in some preparation protocols by the addition of ascorbic acid to the cell medium. The particular cellular component that is labeled can affect the viability of the cells during imaging. For example, stains for the cellular nucleus tend to have more deleterious effects than do cytoplasmic stains. Probes are available that distinguish between living and dead cells (among these are acridine orange), and that can be used in assays of cell viability during imaging. Most such assays are based upon the premise that the membranes of dead cells are permeable to many materials, such as the dyes, that cannot penetrate them in the living state.

Fluo-3 and rhod-2 are examples of dyes that have been synthesized to change their fluorescence characteristics in the presence of certain ions such as calcium. New probes have been developed for imaging gene expression, including for example, the jellyfish green fluorescent protein ( GFP ), which enables gene expression and protein localization to be observed in vivo . The use of GFP has enabled the monitoring of gene expression in a number of different cell types including living Drosophila oocytes, mammalian cells, and plants (using the 488nm line of the excitation laser of the LSCM). Mutants of GFP with spectral variations are available for use in multilabeling experiments, and these have also found use for avoiding interference from autofluorescence in living tissues.

Autofluorescence

Autofluorescence of tissues occurs naturally in many cell types, and can be a major source of background interference during imaging. For example, chlorophyll in yeast and plant cells fluoresces in the red part of the spectrum. Certain reagents, such as glutaraldehyde fixative, are sources of autofluorescence, which can be reduced by treatment with borohydride. Autofluorescence can sometimes be avoided by using fluorophores that can be excited at wavelengths that are out of the range of natural autofluorescence. Cyanine 5 is often chosen since it is excited at a longer wavelength that avoids the shorter-wavelength autofluorescence.

Although it is most often considered a problem, tissue autofluorescence can be utilized for imaging overall cell morphology as a part of multilabeling studies. The contribution from autofluorescence to the total fluorescence can be assessed by viewing an unstained specimen at different wavelengths and noting the laser power and PMT settings of black level and gain. Autofluorescence can often be bleached out by brief exposure to the laser at high power, or by flooding the specimen with light from a mercury lamp. More sophisticated approaches to dealing with autofluorescence include using time resolved imaging, or removal using digital image processing techniques such as image subtraction.

Collecting Images

Beginning users of confocal microscopes can gain experience in several ways. The microscope manual provided by the manufacturer usually includes a series of simple procedures necessary for getting started. In most multi-user facilities, the person primarily responsible for operating the instrument may provide orientation sessions, or the facility manager may require a short training session and demonstration of a certain competence level before solo use of the instrument is allowed. Particular attention should be paid to the house rules of the facility. Information and training can also be gotten from training courses conducted by the microscope companies, from workshops on microscopy, and from a variety of publications.

Before work is done with experimental specimens, it is essential to be familiar with the basic operation of the imaging system. It is usually beneficial for the novice to begin trial imaging with a relatively easy specimen rather than a more difficult experimental one. Some better test samples include paper soaked in one or more fluorescent dyes or a preparation of fluorescent beads. Both types of specimen are brightly fluorescent and relatively easy to image with a confocal system. Another excellent sample is a slide of mixed pollen grains that exhibit autofluorescence at many different wavelengths. These can be easily prepared from pollen collected from garden plants, or can be obtained from commercial suppliers of biological specimens. The pollen grain images in Figure 4 were collected simultaneously with the same PMT black level and gain, and pinhole diameter settings, but reveal three types of pollen that each fluoresce at different excitation wavelengths. These specimens are valuable as test subjects because they not only have some interesting surface details, but also maintain their properties relatively well when exposed to the laser beam. For trials with living tissue, specimens prepared from onion epithelium or the water plant Elodea sp. are reliable, using either autofluorescence or staining with DiOC6.

Before attempting imaging, the confocal instrument should be set up to give the best possible performance. This requires optimal alignment, especially when one of the older confocal microscopes is being used. The alignment routine used is highly specific to the particular instrument, and is usually best done by the person who has overall responsibility for maintaining it. In no case should alignment be attempted without proper training and permission from the owner of the microscope. Improper procedures used to attempt alignment can result in complete loss of the beam and can, in the case of some instruments, require a service visit to rectify.

Figure 4 - Autofluorescence of Pollen

The confocal system is based on a conventional optical instrument, and the fundamental procedures and practices of optical microscopy should be followed at all times. It is extremely important that all glass surfaces in the optical path be clean because dust, oil, or grease on slides, coverslips, and objective lenses is a primary cause of poor images. The refractive index between the objective lens and the specimen must be appropriate to the lens in use. For example, the correct immersion oil must be used for a given objective numerical aperture, and the specimen must be mounted to be within the working distance of the lens. Coverslip thickness must be correct for the lenses used, especially for the higher power objectives, which require a No. 1 or No. 1.5 coverslip instead of a No. 2. The coverslip must be sealed to the slide using an appropriate medium, and mounted flat. Nail polish can be used for fixed specimens if care is taken to ensure that it is dry before imaging. A mixture of petroleum jelly, beeswax, and lanolin, or some other nontoxic sealing material must be used with living specimens. Following strict basic cleanliness procedures at the specimen preparation stage can save much time and effort later.

In preparation for confocal mode imaging, a region of interest is located using either brightfield or conventional epifluorescence microscopy. It is preferable to do this survey using the microscope of the confocal system, but it can be extremely difficult for the novice to find the correct focal plane using the confocal imaging mode alone. If conventional imaging modes are not available on the confocal system, then structures of interest can be located using a separate fluorescence microscope, and their positions marked using a diamond marker on the microscope, a marking pen, or by recording the position coordinates from the microscope stage. It is especially useful to be able to preview specimens with the actual microscope of the confocal system when attempting to image a rare phenomenon such as a gene expressed at a particular stage of development in a specimen containing perhaps hundreds of embryos of different ages. A great deal of time can be saved in this way, over having to scan many specimens using the confocal mode. Confocal instruments commonly have a low-resolution rapid scanning mode that makes the preliminary scanning more efficient. The best approach when searching for rarely occurring events, however, is to scan the slides using a conventional microscopy mode and then to immediately switch to confocal mode on the same microscope to collect the images.

Successful confocal imaging relies on the "secret" of mastering the interplay between objective lens numerical aperture, pinhole size, and image brightness, and using the lowest laser power possible to achieve the best image. The novice user should experiment with varying these parameters using test specimens and several objective lenses of different magnifications and numerical apertures to gain a sense of the capabilities of the microscope before attempting imaging on experimental specimens. A comparison should be made of images acquired using the zoom function of the confocal system with those obtained using objectives having higher numerical aperture. The particular specimen and features being imaged will determine which lenses and methods are most appropriate. Two examples of the many objectives suitable for confocal microscopy are illustrated in Figure 5 . The figure includes a 60x plan apochromat oil-immersion lens, and a 20x plan fluorite . The latter objective has an adjustment collar that allows it to be utilized with oil, glycerin, or water as an immersion medium.

Specific microscope settings appropriate to the specimen should be set up away from the primary region of interest to avoid photobleaching of the fluorescent species in valuable regions of the specimen. Usually this requires setting the gain and black levels of the photomultiplier detectors together with the pinhole size to obtain the best balance between acceptable resolution and adequate contrast, using the lowest laser power possible to minimize photobleaching. Many instruments utilize color lookup tables designed to aid in setting the correct dynamic range for the image. Such tables are designed so that the darkest pixels, having brightness values around zero, are arbitrarily displayed as green (for example), and the brightest pixels, with brightness values near 255 in an 8-bit system, are displayed as red. The microscope parameters such as gain and black level, and the pinhole diameter, are adjusted so that there are only a few green and red pixels in the image, ensuring that the full dynamic range from 0 to 255 is utilized, but with little cutoff at either end of the brightness range. Although these adjustments can be made by eye, the use of pseudocolor at the extremes of the dynamic range of the imaging system makes the adjustment much less subjective. In some cases images must be collected at less than the full dynamic range of the system because less than optimum laser power must be used or the specimen has uneven fluorescence, causing a bright region to obscure a dimmer region that is of interest in the frame.

Figure 5 - Objectives for Laser Scanning Confocal Microscopy

During scanning of the specimen an image averaging routine is usually employed to reduce random noise from the detection system and to enhance the constant (nonrandom) features in the image. An image equalization algorithm can be applied after collection of images to scale them to the full dynamic range of the display. Care should be taken not to apply this type of routine if measurements of fluorescence intensity are to be made unless a control image is included in the same frame as the experimental images before the equalization routine is applied. In using any type of image processing routine, it is a good strategy to save raw unprocessed images in addition to any processed ones.

The usual strategy in image collection is to save images onto the hard disk of the confocal system's computer, and later to back them up onto another mass storage device. In general it is always advisable to collect as many images as possible during a microscope session, and to discard unneeded ones during later review. Many images that seem unnecessary at first consideration become highly valuable at a later date after further review (especially with one's peers). If it seems wasteful to take seemingly superfluous images, consider that it much harder to prepare another specimen, and harder still to reproduce the exact conditions of an experiment or even to exactly duplicate the specimen preparation protocol.

A strategy for labeling image files in an informative way should be developed before imaging is begun. During imaging many notes should be taken or added to the image file along with the image if this capability is available on the system used. Tests should be done to ensure that any saved information is accessible after saving the images, keeping in mind that text and other information related to an image may be lost when it is subsequently transferred to image editing programs such as NIH Image or Adobe Photoshop on other computers. A well-organized notebook or laptop computer file may be preferable over other means of recording imaging session details, and should include filenames, comments, and details of the objective and any zoom factor used to allow calculating scale bars at a later date. Most confocal systems do not automatically record the objective lens used, and this information is important for calculating field widths and scale bars for later publication. Many modern systems utilize an image database that organizes file names and locations of the files, and that usually will display thumbnail files of the images. Care must be taken to follow image naming restrictions imposed by the system, such as the number of characters allowed in a file name and whether characters such as periods or spaces might be misinterpreted by the software.

Troubleshooting

In any experimental discipline, a protocol that has produced good results will sometimes inexplicably cease to work. When this occurs with confocal imaging experiments, there may be an initial reflex to blame the instrument rather than the specimen, but tests should be conducted to confirm that the specimen is not at fault before any troubleshooting is begun on the instrument. A good first test is to view the specimen on a conventional epifluorescence microscope. If some fluorescence is visible by eye, the signal should be very bright on a properly functioning confocal system. Having confirmed that fluorescence is present in the specimen, some tests of the confocal system should then be done using a known test specimen rather than the experimental one. For reference purposes, the confocal system should have a digital file of an image of the test specimen accessible to users of the microscope, including all parameters of its collection, such as laser power, pinhole diameter, objective lens and zoom value, and gain and black level of the detector.

If initial tests do not provide a clear solution, it is advisable to seek help from an expert who may have experienced the problem before. As a rule, if a user is not sure of something, it is best that they step back and ask for help before attempting any remedies. All of the companies that supply confocal microscopes have telephone help lines and websites that may be accessed for additional help.

Problems that are traced to the preparation protocols are usually caused by degradation of reagents, and this should be checked by performing a series of diagnostic tests. It is usually advisable for the person doing the experiments to make up their own reagents, or at least to obtain them from trusted co-workers. Antibodies should be allocated from frozen stock in small batches, then stored under refrigeration, and should not be reused unless absolutely necessary. Sometimes this is unavoidable with rare or expensive reagents, and often does not present a problem.

In experiments with multilabeled specimens, bleed-through from one channel into another can occur as the result of properties of the specimen itself, or due to problems with the microscope. Published reviews in the literature should be consulted for the details of causes of bleed-through and possible remedies. A good test of the instrument itself involves imaging of a test specimen with known bleed-through properties, using both multiple-label and single-label settings. Images of the test specimen should be stored along with records of all pertinent microscope settings, so that when problems do occur the test specimen can be re-imaged with the same instrument conditions and the images compared with the stored ones recorded when the instrument was known to be operating optimally.

Other tests that may be done when problems arise include a visual inspection of the color of the laser illumination and a check of the anode voltage of the laser. If, for example, the beam from a krypton/argon laser appears blue instead of white when scanning on a multiple-label setting then this may indicate that the red line is weak. In this case, the anode voltage will probably be too high, and can usually be reduced to an acceptable level by adjusting the mirrors of the laser. Such adjustments should be done by, or supervised by, the person responsible for maintaining the confocal system. If the voltage cannot be brought into an acceptable range, a replacement for the laser may be required.

Another problem that may be encountered is that antibody probes may have degraded or need to be repurified or otherwise cleaned. Specimens that have been prepared for some time may develop increased background fluorescence and bleed-through caused by the fluorochrome separating from the secondary antibody and diffusing into the surrounding tissue. If at all possible, imaging should be carried out on freshly prepared specimens. Sometimes changing the concentration and/or the distribution of the fluorochromes will help alleviate problems. As one example, fluorescein may bleed into the rhodamine channel, and can be switched so that rhodamine is on the stronger channel. The rationale for this is that the fluorescein excitation spectrum has a tail that overlaps with and is excited in the rhodamine wavelength range. In subsequent experiments, the concentration of the secondaries can be reduced.

Image Processing and Publication

Images acquired with the confocal microscope are usually saved as digital computer files in a format that allows them to be easily manipulated using the proprietary software provided as part of the confocal system. One of the most dramatically improved capabilities of current LSCMs is their display of confocal images. This is of great importance because the improvements in imaging using the confocal microscope are of little value if there is no means to effectively display the images or reproduce them as hard copy.

As recently as 5 or so years ago most laboratories were still using traditional photographic darkrooms and chemical processing of film and paper for their final hard copy of images. There was particular difficulty in reproducing color images, because they were usually printed by independent printers who often had little idea of what constituted correct color balance in a micrograph, and the cost of quality prints was high. To obtain hard copy of images now, the image files can be exported to a slide printer, a color laser printer, or to a dye sublimation printer for publication quality prints, with direct control of the image characteristics being maintained by the person who acquired the images. Photographs for prints or slides can be taken directly from the video monitor screen, and movie sequences can be published on the Web.

Most journals are now able to accept digital image files for publication, and this has resulted in a dramatic improvement in the quality of published images. The image quality achieved by the confocal imaging system can now be more faithfully reproduced in published articles. In some cases journals also make their articles available on CD ROM, which means that readers can have access to published images exactly as they appeared when collected on the confocal systems of those doing the research. Not surprisingly the technological advances in image acquisition, display, and publication are especially beneficial in the case of color images in that journals can now accurately reproduce images with their original resolution and color balance, and theoretically, at much lower cost to the author.

Contributing Authors

Stephen W. Paddock - Laboratory of Molecular Biology, Howard Hughes Medical Institute, University of Wisconsin, Madison, Wisconsin 53706.

Thomas J. Fellers and Michael W. Davidson - National High Magnetic Field Laboratory, 1800 East Paul Dirac Dr., The Florida State University, Tallahassee, Florida, 32310.

Related Nikon Products

Confocal microscopes.

Nikon offers both point-scanning and spinning disk confocal systems to meet the widest range of research needs.

World-class Nikon objectives, including renowned CFI60 infinity optics, deliver brilliant images of breathtaking sharpness and clarity, from ultra-low to the highest magnifications.

Objective Selector

Filter, find, and compare microscope objective lenses with Nikon's Objective Selector tool.

Share this article:

Introduction

Get updates on our social media channels:

Nikon Instruments Inc.

  • Cookie settings
  • Do Not Sell or Share My Personal Information
  • Terms of Use

© 2024 Nikon Instruments Inc.

Microscopy U - The source for microscopy education

main menu

AIP Publishing Logo

  • Previous Article
  • Next Article

Laser-scanning confocal vibrometer microscope: Theory and experiments

Electronic mail: [email protected]

Electronic mail: [email protected]

  • Article contents
  • Figures & tables
  • Supplementary Data
  • Peer Review
  • Reprints and Permissions
  • Cite Icon Cite
  • Search Site

Christian Rembe , Alexander Dräbenstedt; Laser-scanning confocal vibrometer microscope: Theory and experiments. Rev. Sci. Instrum. 1 August 2006; 77 (8): 083702. https://doi.org/10.1063/1.2336103

Download citation file:

  • Ris (Zotero)
  • Reference Manager

In this article we present a new laser-scanning confocal microscope for vibration measurements in microscopic mechanical and biological structures. Our system can map out-of-plane vibrations with picometer amplitude resolution while the locus transverse and depth resolutions are in the submicrometer regime. We have achieved this performance by using the measurement beam of a heterodyne laser-Doppler vibrometer as the scanned laser beam of a confocal microscope. The power of the heterodyne carrier is a measurement of the detected light intensity. Therefore, the laser-scanning confocal laser-Doppler vibrometer microscope (CVM) can also be used as common confocal microscope to image and measure geometries of three-dimensional structures. The focus of this article is on the analysis of the transverse resolution, which cannot be adopted from other techniques. Our realized CVM allows vibration measurements up to 20 MHz ⁠ . The measured minimum 1 ∕ e 2 -power spot diameter of 730 nm enables vibration analysis of submicrometer structures. In this article we discuss the fundamental limits of this new microscope type and we demonstrate the results of our realized system.

Citing articles via

Submit your article.

laser scanning confocal microscopy experiments

Sign up for alerts

laser scanning confocal microscopy experiments

  • Online ISSN 1089-7623
  • Print ISSN 0034-6748
  • For Researchers
  • For Librarians
  • For Advertisers
  • Our Publishing Partners  
  • Physics Today
  • Conference Proceedings
  • Special Topics

pubs.aip.org

  • Privacy Policy
  • Terms of Use

Connect with AIP Publishing

This feature is available to subscribers only.

Sign In or Create an Account

Evident Logo

  • Shopping Cart

Confocal Laser Scanning Microscope

Not Available in Your Country

Sorry, this page is not available in your country.

Applied Technologies

Ai solutions for confocal microscopy, configurations, specifications.

Transform your images with the new FLUOVIEW™ FV4000 confocal laser scanning microscope. Advanced imaging technology enables higher precision images to empower researchers with more reliable data from their samples. With our breakthrough SilVIR™ detector at the core of the system, achieve much lower noise, higher sensitivity, and improved photon resolving capabilities. With the FV4000 confocal microscope, researchers can acquire higher-quality, quantitative image data in less time and with less effort.

The FV4000 confocal microscope uses our advanced, silicon-based SilVIR™ detector that makes it easier than ever to acquire precise, reproducible data.

The SilVIR detector combines two advanced technologies—a silicon photomultiplier (SiPM) and our patented* fast signal processing design.


 

The system’s updated TruSpectral technology combined with high sensitivity SilVIR detectors enable you to see more by making it possible to multiplex up to six channels simultaneously.

The macro-to-micro workflow enables you to easily observe the target sample from the macro level—whole body or tissue—down to the cell or subcellular level.

Time-lapse imaging is easier with smart features:

: resonant scanner can acquire high-resolution images over a wide area : the scanner’s short pixel dwell time reduces the time the focused laser beam rests on a single spot : the SilVIR detector’s high sensitivity produces higher quality images at higher speeds : rolling average processing maintains qualifications and time resolution

The SilVIR detector has less sensitivity loss over time compared to previous-generation detector technologies. With our laser power monitor (LPM) and , achieve reproducible images under consistent conditions for better reproducibility. Different users on different days can acquire the same precise images using the same settings. Even the images acquired by different FV4000 microscopes can be compared and discussed using the same photon number intensity scale.

We designed the FV4000 system to be easy to maintain:

We stand behind our products with a commitment to fast service and technical support. We offer various support plans to keep your microscope running at peak performance at a predictable cost as well as remote support options, so you don’t need to wait for an engineer or specialist to visit if you’re having an issue.

 

The system’s enhanced technologies provided expanded multiplexing to see more in one image.

NIR imaging offers greater multiplexing capabilities by extending the excitation (λ_Ex) and detection (λ_Em) spectral profile of the FV4000 system. This enables additional dyes to be used to help minimize emission signal overlapping.

HeLa cells labeled by 6 fluorochromes.

HeLa cells labeled by 6 fluorochromes. Cell nuclei (DAPI; blue), cell membrane (AF488; green), nuclear pore (AF561; yellow), microtubule (Qdot605; magenta), mitochondria (MitoTracker DeepRed; cyan), actin (AF750 phalloidin; gray).

The FV4000 system's optical elements have a high transmission from 400 nm to 1300 nm, including the galvanometer and resonant scanner, which are coated in silver rather than aluminum.

Our award-winning are corrected for chromatic aberrations between 400–1000 nm. They also have a higher numerical aperture, excellent flatness, and very high transmittance from UV to NIR, increasing the multiplexing capabilities.

For improved colocalization reliability, our (ne~1.40) significantly minimizes chromatic aberration for strict colocalization analysis.

. ,

A unique combination of advanced technologies delivers high-quality images faster than conventional laser scanning microscope systems.

Capture super resolution images using the FV4000 microscope with no dedicated hardware.

When imaging thicker samples, the FV4000 microscope enables you to capture high-resolution, 3D images.

Typically, using longer wavelengths for fluorescence excitation for shorter periods of time is better for overall sample health. Using less phototoxic light means you can image for longer periods, enabling you to obtain more consistent and reproducible data from live cell imaging experiments.

The FV4000 system not only provides gentle time-lapse imaging via the 685 nm, 730 nm, and 785 nm lasers, but it also features a dedicated TruFocus Red Z-drift compensator to maintain the focus position. This upgraded TruFocus Red unit supports a larger range of wavelengths and is compatible with a wide range of objectives, including our high-performing X Line™ and A Line™ series.

Use our with the FV4000 microscope and achieve clear images of features and structures deep within your sample. Silicone oil has a refractive index close to that of live cells or tissue, greatly reducing the spherical aberration as compared to air, water, or other oils. With less aberration, you can achieve clearer images of your sample at depth. And silicone immersion oil does not dry out at 37 ℃ (98.6 °F), making it effective for long-term time-lapse imaging.

Take your confocal imaging to the next level and save time during data analysis. While the microscope’s signal-to-noise ratio is already very good, denoise can further reduce the noise for stunning, data-rich resonant images.

To speed up image analysis, you can pretrain an AI model so that the system can automatically segment your image data, greatly reducing the workload of this often time-consuming manual process. Then, further streamlines the analysis so that you can get your data quickly.

Improve your resonant scanner image quality by incorporating TruAI noise reduction. Although resonant scanner images are effective in capturing cellular dynamics at high speeds with low damage, this usually causes a compromise in the S/N ratio. TruAI noise reduction can improve these images without sacrificing time resolution using pre-trained neural networks based on the noise pattern of the SilVIR™ detectors. These pre-trained TruAI noise reduction algorithms can be used for on-the-fly processing as well as post processing.

Image analysis requires data extraction using segmentation techniques based on intensity value thresholds. However, this can be time-consuming and is affected by the sample conditions.

TruAI image segmentation using deep learning helps streamline image processing and minimize sample variables for more accurate image analysis. It enables you to segment very weak fluorescence images or tissues that are usually difficult to extract using the simple thresholding method.

The FV4000 microscope is engineered to be modular, making it easy for you to configure the system based on your applications and budget. You can start with a standard FV4000 and easily upgrade to multiphoton imaging by adding the as your research changes.

 

Multiphoton and single-photon combination imaging in one sample is also possible. The is capable of second and third harmonic generation imaging, so different researchers or users can make the most out of the system. If your research requires a custom setup, the microscope’s modularity and optional ports enable you to customize the system to add extra lasers, cameras, detectors, and more.

Scanner Galvanometer scanner
(normal imaging)
64 × 64 to 4096 × 4096 pixels, 1 μs/pixel–1000 μs/pixel
Resonant scanner scanner
(high-speed imaging)
512 × 512 pixels, 1024 × 1024 pixels
Field number (FN) 20
Spectral confocal detector Detector SilVIR detector (cooled SiPM, broadband type/red-shifted type)
Maximum channels Six channels
Spectral method VPH, detectable wavelength range 400 nm–900 nm
Laser VIS laser 405 nm, 445 nm, 488 nm, 514 nm, 561 nm, 594 nm, 640 nm
NIR laser 685 nm, 730 nm, 785 nm
Laser power monitor Built in
Image High dynamic range photon counting (1G cps, 16-bit)

Application Notes

Drug Efficacy Evaluation of 3D Cancer Spheroids Using an Imaging-Based Workflow

White Papers

Reliable Quantitative Confocal Fluorescence Imaging  using the Microscope Performance Monitor

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

The PMC website is updating on October 15, 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • HHS Author Manuscripts

Logo of nihpa

Fluorescence Microscopy: A Concise Guide to Current Imaging Methods

Christian a. combs.

NHLBI Light Microscopy Facility, NIH, Building 10, Room 6N-309, 10 Center Dr. Bethesda, MD 20892, Phone: 301-496-3236, Fax: 301-480-1477, vog.hin.iblhn@csbmoc

Introduction

Fluorescence microscopy is a powerful tool for modern cell and molecular biologists and, in particular, neurobiologists. It provides a window into the physiology of living cells at sub-cellular levels of resolution. This allows direct visualization of the inner workings of physiological processes at a systems level context in a living cell or tissue. Fluorescence microscopy enables the study of diverse processes including protein location and associations, motility, and other phenomenon such as ion transport and metabolism. This versatility explains why thousands of papers a year are published using variants of fluorescent microscopy techniques. Many new techniques have been developed over the last decade which enable more comprehensive exploitation of light for biologic imaging. These advances include the wide-spread use of fluorescent proteins (for review see ( Shaner et al., 2005 ), the myriad number of new fluorophores available (for reviews see ( Eisenstein, 2006 ; Suzuki et al., 2007 ), the growth of the utility of the basic confocal microscope, the use of multi-photon microscopy to optically image far deeper into tissues, and the breaking of the diffraction limit for “super-resolution”. Many of the new advanced techniques are now being commercialized, opening their use to a large fraction of modern biologists. However, for the biologist inexperienced in light microscopy, matching the best technique to the biological experiment can prove to be difficult. Optimal use of fluorescence microscopy requires a basic understanding of the strengths and weaknesses of the various techniques as well an understanding of the fundamental trade-offs of the variables associated with fluorescent light collection.

In a very simple form, the ideal light microscopy experiment can be viewed as optimizing the competing properties of image resolution (in the XY or lateral direction as well as the Z or axial dimension), imaging speed (and/or acquisition time), and the amount of signal collected from the fluorescing sample ( Figure 1 ). This is bounded by the limits imposed by photo-bleaching and/or photo-toxicity. In many experiments, light levels at the diffraction limited spot (focused by the objective) can be very high. This can lead to destruction of the fluorophore and unwanted biological consequences leading to cell death or changes in the physiology of the cells or tissue being illuminated. Given these constraints, these variables are difficult to balance and require careful attention to detail and systematic empirical testing. On top of these basic variables other secondary variables also can become important such as the cost of the necessary equipment and the difficulty of the technique.

An external file that holds a picture, illustration, etc.
Object name is nihms174227f1.jpg

Diagram of some of the critical opposing factors in an imaging experiment. The best image is one that can balance these factors to obtain the necessary information while avoiding photobleaching or phototoxic effects. Table 1 outlines how these factors differ between the various commercialized microscopy techniques discussed in this work.

This review is intended to expand and build-upon the last review of fluorescence microscopy in this series ( Coling and Kachar, 1997 ) which provided a foundation for understanding fluorescence microscopy and the basics of immuno-labelling. In this review, knowledge of the basics of fluorescence microscopy (including wide-field microscopy) presented in that paper will be assumed. The object of this review is to provide non-expert microscopists a concise description and guide to select techniques that may have the widest appeal and that are, or will soon be, commonly used in most light microscopy core facilities or advanced biological research labs. The techniques to be reviewed encompass the most basic (such as wide-field fluorescence microscopy) to cutting edge techniques like Stimulated Emission Depletion (STED) microscopy. An emphasis will be placed on explaining the strengths and weaknesses of these techniques in terms of balancing the variables discussed in Figure 1 . A table at the end of this review will summarize this discussion and should serve as a quick guide for choosing the appropriate imaging modality from among the techniques discussed.

Wide-field Fluorescence Microscopy (WFFM) Techniques

In the most basic form (WFFM) involves exciting the fluorophore(s) in the sample of interest using a fluorescence light source, a microscope, excitation and emission filters, and an objective. The resulting emission light, of longer wavelength, is observed through the microscope eyepieces or by a camera followed by computer digitization (for reviews see ( Coling and Kachar, 1997 ; Inoue and Spring, 1997 ). Over the last decade developments in microscope and camera design, light filters, and in new techniques have greatly improved resolution and light collection for WFFM.

One of the most significant advancements has been the development of electron multiplied (EMCCD) and very low-noise, cooled CCD cameras. These cameras allow for fast detection of low-light fluorescence (EMCCD) or allow for the gradual accumulation of fluorescence signal to be integrated with little noise (cooled CCD) while still maintaining high resolution. These advancements allow for faster imaging and better contrast at low signal levels such as when the excitation light is purposefully minimized to prevent photo-bleaching or photo-toxicity.

In addition to cameras, wide-field microscopy has also been improved by better light filters, mirrors, and objectives. Commercially available filters, for instance from Chroma Inc (Rockinham, VT), (Omega Brattleboro, VT), or Semrock (Rochester, NY), have very high transmittance or reflection values enabled through new sputter-coating technologies. Also, these filters can have very sharp wavelength dependencies which enable multi-color discrimination. In the last decade, all of the major microscope companies (such as Leica, Nikon, Olympus, and Zeiss) have also improved microscope objectives. These new objectives have very flat fields (which decreases objective induced gradients in intensity across an image), long working distances with good resolving power, improved light transmission from the near UV to the infra-red, and are available in varieties that match the refractive index of the sample being imaged.

The main advantages of basic WFFM are that it is the least expensive technique, it provides good XY dimension resolution (the ability to distinguish fine detail in a specimen in the XY dimension), can provide very fast temporal resolution (particularly with the new EMCCD cameras), and in many cases requires the least amount of excitation light ( Table 1 ). XY resolution (R xy ) in wide-field microscopy is a function of the NA of the objective and the wavelength of the excitation light according to Ernst Abbe's diffraction limit expression:

Comparison of selected characteristics of commercially available microscope techniques discussed in this unit (Black boxes are best in category, grey are worst).

TechniqueResolution XYResolution ZResolution TemporalImaging DepthEase of UseCostPhotobleaching or Phototoxicity
Wide-fielddiffraction limited (≈200nm)WeakBest (ms/ Frame, signal limited)Worstsimple$Best (usually μWatts)
Structured LightVaries (can be super-resolution)Varies (can be super-resolution)Varies based on number of images neededBetterVaries with resolution needed$$Varies with number of images needed
Laser- Scanning Confocal (LSC)Diffraction limitedGood (>700nm)Typically slow (1s/frame)Better (less than 100μM)Complex but most versatile$$$$Can be bad (μWatts of power focused to spot)
Multi-Point/Slit ConfocalRange to diffraction limitGood same as LSCGood (signal limited)Better (same as LSC)Better$$$Better (usually lower flux density than LSM)
TIRF Diffraction limited but low backgroundBest but only first 200-300nmGood (signal limited)<300nmGood$$Better
Two-Photon (TPFM)Diffraction limitedGood (slightly less than LSC)Typically slow (1s/frame)Best (hundreds of μMs)Complex$$$$$Can be bad (mWatts power focused to spot but limited to 1 plane)
STED Super-resolution (<70nm)Same as TPFMSlowest???Most complexVery high costWorst (second beam with many mWatts of power)

Were λ is the wavelength of the emission light, NA is the numerical aperture of the objective. For a high NA objective (NA 1.4) lens this limit is around 200nm. All of the techniques listed in Table 1 are approximately limited to this type of XY resolution except where super-resolution is indicated. The main disadvantage of basic WFFM is that that all of the emission light is integrated through the sample in the Z dimension. Therefore, it is difficult to tell where the fluorescence from a point in the sample originated in the Z-dimension. For samples that are thin or where Z-discrimination is not critical this may not be a limiting factor. For thick samples (such as live cells or tissues) where optical sectioning is critical or where out of focus light obscures details even in the XY plane, other techniques such as confocal or multi-photon microscopy may be more appropriate (see the following sections), although fluorescence deconvolution microscopy and structured light microscopy (SLM) are WFFM techniques that are commercially available.

Structured Light Microscopy (SLM) is a form of WFFM that enables optical sectioning. SLM works by inserting a moveable grid pattern into the optical path of the excitation light in the wide-field microscope. This produces a pattern in the images produced. The pattern is moved in the XY and even Z-dimension and the way that the detected fluorescence from the sample interacts with the pattern is then analyzed using a simple mathematical formula to create the optical sections ( Figure 2 ). Many commercial systems are available for SLM and also moveable grating patterns are available for those wishing to modify existing WFFM microscopes. In the last few years SLM has been shown capable of producing super-resolution images (≈ half the diffraction limit) ( Gustafsson, 2000 ; Schermelleh et al., 2008 ).

An external file that holds a picture, illustration, etc.
Object name is nihms174227f2.jpg

The basic principles of structured light microscopy are shown in panels A , B , and C. If an unknown pattern (such as a biological sample) represented in (A) is multiplied by a known regular illumination pattern (B) then a beat pattern (moiré fringes) will appear (C). The pattern is the difference between the sample and the regular illumination pattern and is course enough to be seen through the microscope even if the original pattern in the sample was not resolvable. By moving the grid and the sample in space and computationally processing the resulting data an image can be generated that has resolution at least 2× better than a conventional wide-field image. D and F. Confocal and structured light images respectively of the edge of a Hela cell showing the actin cytoskeleton. E and G show enlargements of the images in D and F. The apparent fiber diameters are 110-120nm in the structured light images compared to 280 to 300nm in the confocal image. Figure A, B, and C are reproduced with permission from ( Gustafsson, 2005 ) . Figures D and E are reproduced with permission from ( Gustafsson, 2000 ). Panels A-E were originally published in color and have been altered here to black and white.

The main advantage of SLM is that one can optically section using WFFM without the cost of expensive confocal systems and in some cases produce super-resolution images without the cost or technical complexity associated with other super resolution techniques such as Stimulated Emission Depletion Microscope (STED, discussed in a section later in the paper). The main disadvantage of this technique is that multiple images must be taken to provide optical sections. This can lead to photo-bleaching. Also the optical sectioning ability of SLM is negated if the sample moves while the different images are being captured (as would be the case in live cells).

Deconvolution fluorescence microscopy (DFM) is a form of WFFM. DFM requires prior knowledge of the point spread function (PSF, for more information on the PSF see ( Coling and Kachar, 1997 )) to allow optical sectioning. In this technique, multiple XY sections are imaged through the sample in the Z-dimension. The resulting stack of images, still lacking Z-dimension discrimination, are then analyzed using an empirical or idealized mathematical model of the PSF created by the microscope optics. This analysis results in a volumetric recreation of the sample in 3D space. DFM can also be used to enhance other techniques that are able to optically section such as confocal and two-photon microscopy (discussed below). Software for DFM is available commercially or through plug-ins for the free image analysis program ImageJ ( http://rsbweb.nih.gov/ij/ , NIH, Bethesda, MD). Although DFM is a powerful technique when used in capable hands (reviews include ( Boccacci and Bertero, 2002 ; Wallace et al., 2001 )), the novice user should be warned that optical sectioning is done indirectly using a mathematical model. Knowledge of the limitations of the model used for DFM is critical to understanding the images produced and interpretation of the data. Given these constraints it is beyond the scope of this paper and is not listed in Table 1 .

Modern Confocal Microscopy

The laser scanning confocal microscope (LSCM) remains a key piece of equipment in most imaging laboratories. Most modern LSCM systems offer a variety of advantages and are equipped with software to perform complex 3D (z-stack), 4D (z-stack over time), or even 5D (z-stack over time including spectral imaging) experiments. These microscopes often include software to facilitate data acquisition for complex methodologies such as spectral deconvolution, Fluorescence Recovery After Photobleaching (FRAP), and Fluorescence Resonance Energy Transfer (FRET). There have been many reviews written about confocal microscopy, but readers are encouraged to see the following texts for comprehensive information regarding all forms of confocal microscopy (as well as other microscopy techniques)( Hibbs, 2004 ; Pawley, 2006 ).

In the last few years, many changes have been made to improve these microscopes, but the fundamental design for optical sectioning remains largely unchanged. Figure 3 shows a simplified diagram of the light path of an LSCM. This figure shows that laser light is directed to the sample through collimating and beam steering optics, scanning mirrors (which sweep the laser beam over the field of view) and an objective that focuses the light to a diffraction limited spot in the sample. Emission light from the sample is directed to light sensing detector(s) (typically photomultiplier tubes, also known as PMT's) through a pinhole that is in the conjugate image plane to the point of focus in the sample. After spatial filtering by the pinhole, the light is sensed by the detectors, and a proportionate voltage is produced and amplified and converted into digital levels for image display and storage.

An external file that holds a picture, illustration, etc.
Object name is nihms174227f3.jpg

Basic architecture of a modern confocal microscope. Excitation light from laser is passed through the various collimating optics in a scan-head to either a variable dichroic mirror (Nikon, Zeiss, or Olympus) or an AOBS (Acousto-Optical Beam Splitter) (Leica) where it is reflected through the objective and focused to a point on the sample. Moveable mirrors in the scan-head before the objective scan the excitation beam over the sample, a point at a time, to build the image. Fluorescence emission light passes back through the objective, through the dichroic or AOBS to the light sensing PMT(s) (photo-multiplier tube). An aperture (pinhole) placed in the conjugate image plane to the point of focus in the sample allows only light from the focal plane to impinge on the sample and out-of-focus light is blocked. The pinhole can be made larger to allow for larger optical sectioning capability allowing more out of focus light to impinge on the PMT(s). In some models a diffraction grating or prism placed in the beam-path of the emission light can act as a variable band-pass filter or as a spectral detector if the polychromatic light is spatially spread on a number of PMTs.

At the heart of the confocal microscope is the pinhole. When placed in the conjugate image plane to the point of focus on the sample it enables optical sectioning ( Figure 3 ). The pinhole optically sections by acting as a barrier to light originating from other focal planes in the sample. Although the pinhole facilitates optical sectioning it must be understood that the axial resolution is still worse than the XY resolution (which is similar to WFFM). Axial resolution (R z ) in the confocal microscope is set by the expression:

were λ is the wavelength of the emission light, η is the refractive index of the mounting medium, and NA is the numerical aperture of the objective. At an intermediate emission wavelength when coupled with a pinhole and a high numerical aperture lens this would enable an ideal axial resolution of approximately 0.6 μm. In practical terms, axial resolution is likely to be between 0.6 and 1.0 μm. The difference between the XY and Z dimensions leads to a resolution limit that is ellipsoidal in shape in 3D space.

Most LSCM manufacturers also offer a spectral imaging option that will allow for either variable band-pass emission filtering or spectral detection on a per pixel basis. This works by placing either a diffraction grating or a prism in the light path before the PMT detector(s). In many cases, polychromatic (spectral) light is passed to a PMT array to detect a range of wavelengths either sequentially or simultaneously depending on the range of wavelengths desired. Although this option allows for more versatility and direct selection of the emission range it can come at the cost of less sensitive detection, due to the light loss in the additional optics required and in the spreading out of the light over a series of detectors to enable the spectral detection.

Table 1 shows the main advantages and disadvantages of LSCM. The main advantage of LSCM is that one may optically section while still doing complex experiments. Another advantage is the versatility of imaging capabilities and types of experiments one can perform. Most of these systems have multiple channels for multi-color, variable pinhole sizing for selecting the desired optical section thickness (usually sacrificing z-resolution for signal intensity), and software for variable ROI (region of interest) selection. Another example would be the ability to separate spectrally overlapping fluorescent proteins with spectral detection and spectral deconvolution methods. In addition, these systems, particularly in the inverted microscope configuration, can accommodate both live cells or fixed cells or tissue. Many manufacturers also provide options for small stage incubation systems. These systems allow long term experiments, particularly when coupled to automated acquisition software that enables auto-focusing algorithms in tandem with precise XYZ stage movement. Disadvantages of a modern LSCM system include the relatively low scan speed (as the beam must be swept through each pixel in the field of view), the relative price, and the amount of light impinging on the sample. The flexibility of the LSCM does mitigate many of the disadvantages, and in many instances, one can balance the imaging conditions among the variables listed in figure 1 to get the most out of a given experiment. For instance, if full-frame imaging speed is too slow to capture a physiological event in a live cell experiment one might use a small ROI to increase temporal resolution. Despite this flexibility, one concern always remains and should be considered when conducting LSCM and that is keeping the light levels low enough to avoid killing or bleaching the sample.

Another type of confocal microscopy is multipoint confocal microscopy, which includes Nipkow spinning disk, swept-field, and slit line scanning microscope systems. Each of these types of microscope systems shares the characteristic that multiple parts of the sample are imaged at once, thus increasing imaging speed. In the case of the Nipkow spinning disk and swept field systems, a sensitive camera (typically an EMCCD) is also employed. This allows for fast (usually tens to hundreds of milliseconds vs. the seconds timeframe of the LSCM), relatively low-light confocal imaging. Nipkow scanning systems have a drawback in that confocal sectioning can only occur with relatively high NA objective lenses and the pinhole size is fixed. In the case of the slit-scanning confocal microscopes, there is also a modest decrease in resolution for the X or Y dimension. All of these systems are usually less expensive than a LSCM system but can become relatively expensive if a very sensitive camera is also included.

Total Internal Reflection Fluorescence (TIRF) Microscopy

TIRF microscopy provides very good axial resolution (Z-direction, along the axis of illumination) to levels of approximately 200nm (for review see ( Toomre and Manstein, 2001 ). Not only is the axial resolution better than most other techniques but this also can greatly reduce background light (thus increasing the signal to noise ratio) that can obscure fine details. The setup for TIRF microscopy is very simple and is similar to wide-field microscopy except that it employs an oblique angle for the excitation light impinging on the sample. When the incidence angle is set to a critical angle relative to the coverslip, the excitation light is totally internally reflected ( Figure 5A ). This generates an electromagnetic field at the interface, called an evanescent wave, which excites fluorophores in nearly the same manner as conventional fluorescence excitation light. The key here is that the evanescent wave propagates only a short distance above the coverslip ( Figure 5B ). Therefore, only fluorescent molecules in close proximity to the coverslip are excited. Figure 5C shows a wide-field and a TIRF image of the fluorescence from EGFP labeled myosin in drosophila embryo hemocytes. As can be seen in the overlay, only myosin molecules in portions of the cell near the coverslip are excited, showing where the cell is closest to or touching the coverslip.

An external file that holds a picture, illustration, etc.
Object name is nihms174227f5.jpg

TIRF microscopy excites a shallow region above the coverslip using oblique laser excitation which is totally internally reflected and produces an evanescent wave for fluorophore excitation. A. Internal reflection. Light propagating through the periphery of a high numerical aperture objective (>1.38) is totally internally reflected by the coverslip and sent down the opposing side of the objective. B. Evanescent wave is formed w hen the critical angle θ C is reached and the light is the totally reflected. The reflection at the coverslip is due to the oblique angle of illumination and the mismatch of refraction index (n) between the oil and coverslip. Note that the evanescent wave only excites fluorophores where the cell attaches or is touching the coverslip. C and D (no D indicated) show a wide-field and TIRF image, respectively, of GFP tagged myosin V from Drosophila embryo hemocytes. Comparing the two images it is evident where the Myosin 5 is closest to the coverslip particularly in the bottom cell. Hemocytes courtesy of Amy Hong, NHLBI, NIH. Figure B was reproduced with permission from Mike Davidson (Florida State University and the National High Magnetic Field Laboratory) and the Molecular Expressions website.

As mentioned above, the decay of the evanescent wave is exponential to the distance above the coverslip. This relationship can be expressed as:

Where I(Z) represents the intensity at a given distance ( z ) from the coverslip, I(o) is the intensity at the coverslip, and d is the penetration depth. The penetration depth d decreases as the reflection angle (θ C , shown in Figure 5 ) of the incident beam grows larger. This value is also dependent on the illumination wavelength and on the refractive index of the media present at the interface. In a typical commercially available objective-based TIRF system, the reflection angle of the excitation light can be changed mechanically on a special illumination module attached to the epi-fluorescence port of a wide-field microscope. Turning of the micrometer changes the position of the beam traveling in the periphery of the objective back aperture, resulting in a change in the angle of the beam exiting the front element. Another requirement for the typical objective based TIRF system is that high numerical oil objectives (>1.4 NA) are required to generate the necessary reflection angles to establish the evanescent wave.

As is shown in Table 1 , the main advantage of TIRF is enhanced z-resolution. XY resolution benefits from a reduction in background fluorescence. Also, relative to other techniques such as confocal and two-photon, a commercial turn-key objective based TIRF microscope system is inexpensive. These systems only require a microscope, special illuminator(s), lasers, camera, and a high numerical aperture objective lens. The main disadvantage of TIRF is related to its main advantage in that only fluorophores in the first 200-300nm can be excited. This obviously limits imaging to near the coverslip but enable a z-resolution to the same depth as the penetration of the evanescent wave. Also, because the intensity of the evanescent wave decreases according to this relationship, fluorescence intensity will be a function of distance from the coverslip as well as the concentration of the fluorescent molecules. This makes quantification of depth from the coverslip or comparisons of molecular concentration difficult from TIRF images.

Two-Photon Fluorescence Microscopy (TPFM)

TPFM is a type of laser scanning microscopy that is particularly useful for imaging thick samples both in vitro and in vivo . It has been used to image hundred's of microns into tissues (for reviews see ( Diaspro et al., 2006 ; Svoboda and Yasuda, 2006 )). An example of this type of imaging is shown in figure 6C . Deep imaging is achieved by using pulsed near-infrared excitation light. Infrared light penetrates much deeper into tissue do to decreased scattering and absorption than the visible wavelengths used in standard confocal and wide-field microscopy. This technique is also good for limiting the excitation (and often photo-bleaching and possible photo-toxicity) to just one focal plane. This also has the added benefit of eliminating the need for a pinhole aperture for optical section as is used in confocal microscopy. In confocal microscopy the pinhole is used to reject out-of-focus emission light from reaching the photo-sensor (photo-multiplier tube or camera). In effect, the pinhole selects only a small portion of the emission light to achieve optical sectioning with much of the emission light “thrown away”. In TPFM it is the excitation pulse that provides the optical sectioning: therefore all of the light can be collected from the excited focal spot and none of the scattered or ballistic emission light photons need be wasted during collection. TPFM is a form of multi-photon imaging. Multi-photon imaging refers to techniques where more than one photon at a time is used to excite a fluorophore (other examples are Second Harmonic Generation (SHG) imaging and Coherent Anti-stokes Raman (CARS) microscopy. CARS and SHG are not fluorescence techniques and are outside the scope of this review).

An external file that holds a picture, illustration, etc.
Object name is nihms174227f6.jpg

Principals of two-photon fluorescence microscopy (TPFM). A shows a regular one-photon (e.g. confocal) and TPFM energy transitions in a Jablonski diagram. In TPFM two photons are absorbed nearly simultaneously to produce twice the energy. In this example GFP is excited with 960 nm light for TPFM and 488nm higher energy light for a confocal experiment. The emission is the same for both cases. It should be noted that TPFM absorption spectra for most fluorophores, including GFP, are very broad (in some cases hundreds of nanometers), and that the maximum is roughly a little less than twice the one-photon absorption maxima. B Two-photon fluorescence is generated in only one plane when a laser pulse train propagating through an objective is focused to a spot. Fluorescence is generated only at the point where the maximal photon crowding occurs and falls off from this plane at a rate of the fourth power from the center of the focal spot. C I n vivo TPFM image of a mouse neocortex genetically labelled with a chloride indicator. This image shows the remarkable depth to which TPFM imaging is possible. Figure C is reproduced with permission from ( Helmchen and Denk, 2005 ).

TPFM excitation is generated when a fluorophore absorbs two photons essentially simultaneously. This roughly doubles the amount of energy absorbed by the fluorescent molecules which drives their excited electrons to the same energy level as would the absorption of one photon at one-half the two-photon excitation wavelength ( Fig. 6A ). An example would be the excitation of GFP (typically excited around 488nm in a confocal experiment) at around 960nm using a pulsed laser. This is an oversimplification, as the actual TPFM absorption spectra for many fluorophores are over 100nm broad, and “selection rules” that govern the relative strengths of absorption bands vary between one-photon and two-photon excitation, but in most cases that is a good starting point for guessing where the maximal TPFM excitation occurs. The broad spectral absorption range of the typical two-photon fluorophore allows for multiple fluorophores in a sample to be excited at one wavelength simultaneously. Corresponding emission wavelengths for each fluorophores are then separated in different channels with the appropriate set of dichroic and emission filters or with spectral detection. The inherent optical sectioning ability of two-photon excitation occurs due to increased probability of two-photon absorption that occurs at the diffraction limited spot due to spatial energy crowding ( Figure 6B ). This can be seen in the equation for time averaged two-photon fluorescence intensity (I f ):

Where δ 2 is the two photon cross section for the fluorophore, η is the quantum yield of the fluorophore, P is the intensity (power) of the excitation light, τ P is the pulse width of the excitation pulses, f P is the repetition rate of the laser, NA is the numerical aperture of the objective, h and c are Plank's constant and the speed of light respectively, and λ exc is the wavelength of the excitation light ( Diaspro et al., 2006 ). In fact, the probability of two-photon absorption decreases as the fourth power of distance away from this focal region along the z-axis (NA dependence) and increases as the square of the intensity (mW of power are typically required). Another variable is the temporal pulse width, τ P , of the excitation light pulse as it reaches the sample. In general, short pulse widths (on the order of 100fs) are optimal for two-photon excitation.

Commercially available turn-key confocal systems usually consist of a modified point-scanning confocal microscope which includes a Ti:Sapphire pulsed laser (often automatically tunable over a broad range of wavelengths) and non-descanned detector channel(s). The non-descanned detector is a PMT mounted closer to the sample where the emission does not travel back through the scan-head. Since no pinhole is necessary, this configuration can be employed to reduce light losses that would occur if the emission light passed back through the scan-head. Typically, commercially available pulsed lasers produce approximately 100fs pulses at rate of 80mHz. Dispersion in the optics of the microscope and objective will lengthen these pulse widths by at least a factor of two. Recently, commercial lasers optionally include an additional unit for pre-compensation of this dispersion which can reduce the pulse length at the sample (which restores the two-photon fluorescence efficiency).

In summary, as is shown in table 1 , the main advantage of TPFM is the depth of imaging (hundreds of microns) into the sample. Another important advantage is that the bleaching and phototoxicity are limited to the focal plane, however in the focal plane the damage can be greater due to the higher light intensities (mW compared to μW in confocal) needed for TPFM. TPFM typically requires the same time frame for acquisition as traditional CLSM (on the order of 1s/frame). One big disadvantage is cost due to the need for a point-scanning microscope and a tunable pulsed Ti:Sapphire laser. This cost increases substantially if one also adds a pre-compensation unit to correct dispersion in excitation pulse lengths or selects higher power lasers whose gain enables lasing at the hard to tune to regions approaching 700nm or somewhat above 1000nm.

Stimulated Emission Depletion (STED) Fluorescence Microscopy

STED microscopy, developed by Stefan Hell and colleagues, is a relatively new super-resolution technique that has been shown to improve fluorescence microscopy resolution by approximately an order of magnitude over traditional diffraction limited techniques such as LSCM. STED can produce optical resolution to levels that were previously thought possible only with electron microscopy, and it has been used to examine key biological processes that no other technique could have examined ( Kellner et al., 2007 ; Willig et al., 2006b ). STED improves resolution by a direct reduction in the emission spot size by using a second laser beam (the STED beam) ( Figure 7 and explained below). It is important to note that the improvement in resolution is done directly without the need for post processing and mathematical redistribution of the light, as is done in deconvolution microscopy or by combining multiple images that has taken with respect to a moveable grid pattern, as is done in structured light microscopy. STED is so straight-forward that to the user this seems like a normal point-scanning technique such as LSCM or TPFM. In fact, Leica Microsystems (Wetzlar, Germany) has now commercialized this system on their current point-scanning microscope stand.

An external file that holds a picture, illustration, etc.
Object name is nihms174227f7.jpg

Technical principals of Stimulated Emission Depletion (STED) microscopy. A . The combination of the normal excitation beam with the phase modulated STED beam produces a sub-diffraction emission spot. The images on the right in ( A) show the doughnut pattern produced by the phase modulation of the STED beam. This beam when overlapped with the diffraction-limited excitation spot quenches emission where the beams overlap leaving the middle, sub-diffraction sized, spot for spontaneous fluorescence. B . Comparison of confocal (left) and STED (right) images reveals a marked increase in resolution by STED since more labeled particles are visualized. Scale bar, 500 nm. Figure reproduced with permission from ( Willig et al., 2006b ).

Figure 7A shows the setup for the STED technique. In the commercially available system (Leica Microsystems), two pulsed (picosecond time-domain) lasers are included. One excites the fluorophore as would normally occur in a LSCM experiment. The second, longer wavelength laser is used for the patterned quenching of the focal spot (STED beam). Specialized optics in the scanhead spatially shapes the phase of the STED beam wave-front to form a doughnut pattern (with a sharply decreased laser intensity at the central portion of the doughnut) at the focal spot. The STED wavelength must be red shifted (longer wavelength) such that it does not overlap the absorption spectrum of the fluorophore but does overlap with its emission spectrum. In this way, it quenches (forces the excited electrons into a lower energy state without giving off fluorescent light) the emission of the fluorophore in the area of the spot where the STED beam overlaps the excitation beam. This reduces the ultimate emitting region to that of the middle of the doughnut. The size of this region is related to the power of the STED beam according to the following equation:

where R is the lateral (XY) resolution, λ is the wavelength of the excitation light, NA is the numerical aperture of the objective, and ζ describes the intensity of the STED beam. Stefan Hell and colleagues have reported limiting the excitation size to around 30nm, or almost the size of a few fluorescent molecules ( Willig et al., 2007 ).

The main disadvantages of the STED approach are the cost of the system and the amount of power that impinges on the sample ( Table 1 ). The cost of the system is relatively high because two pulsed lasers are needed in addition to the already expensive laser scanning microscope system and very sensitive emission detectors (avalanche photo-diodes) required ( fast electronics and are sensitive in the far-red portion of the emission spectrum). Another disadvantage is the amount of power that is used for a STED system is high (tens of mW for the second beam). Since there is the potential for destruction of the probe or sample, only very photo-stable probes can be used. The list of probes that have been used to date are LDS721, certain ATTO dyes (AttoTech), and fluorescent proteins ( Willig et al., 2006a ). While the current commercial system can only improve lateral resolution, axial resolution can also be improved ( Hell, 2007 ). It remains unclear how deep into tissue this technique remains effective. It is likely that that the loss of coherence of the shape of the STED beam with depth is a limiting factor and will be tissue dependant. In general, STED is most effective for fixed tissue as movement or diffusion of the fluorescent marker during scanned imaging will negate any gains in resolution.

Final Considerations

Many of the microscope systems available from manufacturers have become very easy to run. While this provides easy image acquisition for many of the techniques listed in Table 1 , the danger remains that an incomplete understanding of the fundamental physics and limitations of the techniques can result in wrong, incomplete, or biased data (as recently noted in ( Pearson, 2007 ) ). It should be noted that Table 1 is only a rough guide to the commercially available fluorescence microscopy techniques and the reader should consult other sources to provide more complete understanding. There are many reviews available that are not listed here and many good web based resources such as Molecular Expressions: Exploring the World of Optics and Microscopy ( http://www.microscopy.fsu.edu/ ), The Molecular Probes Handbook ( http://probes.invitrogen.com/handbook/ ) and the Confocal Listserv ( http://listserv.acsu.buffalo.edu/cgi-bin/wa?A0=CONFOCAL&D=0&F=P&T=0 ). One final note is that one must also be able to analyze the data one has collected. There are many good commercially available image processing software packages available as well as the comprehensive free program ImageJ ( http://rsbweb.nih.gov/ij/ ). Last, the mention of any company producing microscopy related products in this work is in no way intended as an endorsement of them by the National Institutes of Health or the author.

An external file that holds a picture, illustration, etc.
Object name is nihms174227f4a.jpg

Maximum projection reconstruction from confocal images obtained through a 65 μm stack of mouse cerebellum labeled with a combination of fluorescent proteins. In the online color version of this image one can see the unique colors produced and spectrally detected by the genetic combinations of individual fluorescent proteins which the authors label as XFP's. These colors were used to trace and map the various synaptic circuits. This figure was reproduced with permission from ( Livet et al., 2007 ). This figure was originally published in color, and can be seen online in color, but has been altered for the print version of this article in black and white.

Acknowledgments

The author wishes to thank Dr. Jay Knutson, Dr. Paul Jobsis, and Dr. Aleksandr Smirnov for useful discussions and critical reading of this manuscript. The author also thanks Ethan Taylor and Alan Hoofring for help with many of the figures in this work. The author also thanks those who permitted reprinting of some of the figures in this article. Those authors are cited in the figure legends and permission was granted by the publishers. This work was supported by the intramural research program of the National Institutes of Health and the National Heart Lung and Blood Institute.

  • Boccacci P, Bertero M. Image-restoration methods: basics and algorithms. In: Diaspro A, editor. Confocal and Two-Photon Microscopy: Foundations, Applications, and Advances. Wiley-Liss; New York: 2002. pp. 253–269. [ Google Scholar ]
  • Coling D, Kachar B. Theory and application of fluorescence microscopy. Curr Protoc Neurosci. 1997; Chapter 2 :1. Unit 2. [ PubMed ] [ Google Scholar ]
  • Diaspro A, et al. Multi-photon excitation microscopy. Biomed Eng Online. 2006; 5 :36. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Eisenstein M. Helping cells to tell a colorful tale. Nat Methods. 2006; 3 :647–655. [ Google Scholar ]
  • Gustafsson MG. Surpassing the lateral resolution limit by a factor of two using structured illumination microscopy. J Microsc. 2000; 198 :82–7. [ PubMed ] [ Google Scholar ]
  • Gustafsson MG. Nonlinear structured-illumination microscopy: wide-field fluorescence imaging with theoretically unlimited resolution. Proc Natl Acad Sci U S A. 2005; 102 :13081–6. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Hell SW. Far-field optical nanoscopy. Science. 2007; 316 :1153–8. [ PubMed ] [ Google Scholar ]
  • Helmchen F, Denk W. Deep tissue two-photon microscopy. Nat Methods. 2005; 2 :932–40. [ PubMed ] [ Google Scholar ]
  • Hibbs AR. Confocal Microscopy for Biologists. Springer; 2004. [ Google Scholar ]
  • Inoue S, Spring KR. Video Microscopy: The Fundamentals. Springer; 1997. [ Google Scholar ]
  • Kellner RR, et al. Nanoscale organization of nicotinic acetylcholine receptors revealed by stimulated emission depletion microscopy. Neuroscience. 2007; 144 :135–43. [ PubMed ] [ Google Scholar ]
  • Livet J, et al. Transgenic strategies for combinatorial expression of fluorescent proteins in the nervous system. Nature. 2007; 450 :56–62. [ PubMed ] [ Google Scholar ]
  • Pawley JB, editor. Handbook of Biological Confocal Microscopy. Springer; 2006. [ Google Scholar ]
  • Pearson H. The good, the bad and the ugly. Nature. 2007; 447 :138–40. [ PubMed ] [ Google Scholar ]
  • Schermelleh L, et al. Subdiffraction multicolor imaging of the nuclear periphery with 3D structured illumination microscopy. Science. 2008; 320 :1332–6. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Shaner NC, et al. A guide to choosing fluorescent proteins. Nat Methods. 2005; 2 :905–9. [ PubMed ] [ Google Scholar ]
  • Suzuki T, et al. Recent advances in fluorescent labeling techniques for fluorescence microscopy. Acta Histochem Cytochem. 2007; 40 :131–7. [ PMC free article ] [ PubMed ] [ Google Scholar ]
  • Svoboda K, Yasuda R. Principles of two-photon excitation microscopy and its applications to neuroscience. Neuron. 2006; 50 :823–39. [ PubMed ] [ Google Scholar ]
  • Toomre D, Manstein DJ. Lighting up the cell surface with evanescent wave microscopy. Trends Cell Biol. 2001; 11 :298–303. [ PubMed ] [ Google Scholar ]
  • Wallace W, et al. A workingperson's guide to deconvolution in light microscopy. Biotechniques. 2001; 31 :1076–8. 1080, 1082 passim. [ PubMed ] [ Google Scholar ]
  • Willig KI, et al. STED microscopy with continuous wave beams. Nat Methods. 2007; 4 :915–8. [ PubMed ] [ Google Scholar ]
  • Willig KI, et al. Nanoscale resolution in GFP-based microscopy. Nat Methods. 2006a; 3 :721–3. [ PubMed ] [ Google Scholar ]
  • Willig KI, et al. STED microscopy reveals that synaptotagmin remains clustered after synaptic vesicle exocytosis. Nature. 2006b; 440 :935–9. [ PubMed ] [ Google Scholar ]

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Published: 19 September 2024

A cuproptosis nanocapsule for cancer radiotherapy

  • You Liao   ORCID: orcid.org/0000-0003-3765-4529 1 , 2   na1 ,
  • Dongmei Wang   ORCID: orcid.org/0000-0002-5995-631X 1 , 2   na1 ,
  • Chenglu Gu 1 , 2 ,
  • Xue Wang 1 , 2 ,
  • Shuang Zhu   ORCID: orcid.org/0000-0002-3962-0587 1 , 2 ,
  • Ziye Zheng 3 ,
  • Fuquan Zhang 3 ,
  • Junfang Yan   ORCID: orcid.org/0000-0002-0101-5947 3 &
  • Zhanjun Gu   ORCID: orcid.org/0000-0003-3717-2423 1 , 2  

Nature Nanotechnology ( 2024 ) Cite this article

Metrics details

  • Biomaterials
  • Nanotechnology in cancer

Residual tumours that persist after radiotherapy often develop acquired radiation resistance, increasing the risk of recurrence and metastasis while providing obstacles to re-irradiation. Using samples from patients and experimental mice, we discovered that FDX1 and LIAS, key regulators of cuproptosis, were up-regulated in residual tumours following radiotherapy, conferring the increased sensitivity to cuproptosis. Therefore, we proposed a novel radiosensitization strategy focused on cuproptosis, using a copper-containing nanocapsule-like polyoxometalate as a paradigm. In an initial demonstration, we showed that the nanocapsule released copper ions in a controlled manner upon exposure to ionizing radiation. Furthermore, radiation-triggered cuproptosis overcame acquired radiation resistance even at clinically relevant radiation doses and activated a robust abscopal effect, with a 40% cure rate in both radioresistant and re-irradiation tumour models. Collectively, targeting cuproptosis is a compelling strategy for addressing acquired radiation resistance, optimizing the local antitumour effects of radiotherapy while simultaneously activating systemic antitumour immunity.

This is a preview of subscription content, access via your institution

Access options

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

$29.99 / 30 days

cancel any time

Subscribe to this journal

Receive 12 print issues and online access

$259.00 per year

only $21.58 per issue

Buy this article

  • Purchase on SpringerLink
  • Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

laser scanning confocal microscopy experiments

Data availability

All data supporting the findings of this study are included in the paper and its Supplementary Information . Source data are provided with this paper.

Sung, H. et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71 , 209–249 (2021).

Article   PubMed   Google Scholar  

Begg, A. C., Stewart, F. A. & Vens, C. Strategies to improve radiotherapy with targeted drugs. Nat. Rev. Cancer 11 , 239–253 (2011).

Article   CAS   PubMed   Google Scholar  

Pitroda, S. P., Chmura, S. J. & Weichselbaum, R. R. Integration of radiotherapy and immunotherapy for treatment of oligometastases. Lancet Oncol. 20 , e434–e442 (2019).

Nishiga, Y. et al. Radiotherapy in combination with CD47 blockade elicits a macrophage-mediated abscopal effect. Nat. Cancer 3 , 1351–1366 (2022).

Article   CAS   PubMed   PubMed Central   Google Scholar  

Petroni, G., Cantley, L. C., Santambrogio, L., Formenti, S. C. & Galluzzi, L. Radiotherapy as a tool to elicit clinically actionable signalling pathways in cancer. Nat. Rev. Clin. Oncol. 19 , 114–131 (2022).

Andratschke, N. et al. European Society for Radiotherapy and Oncology and European Organisation for Research and Treatment of Cancer consensus on re-irradiation: definition, reporting, and clinical decision making. Lancet Oncol. 23 , e469–e478 (2022).

Fattahi, S. et al. Reirradiation for locoregional recurrent breast cancer. Adv. Radiat. Oncol. 6 , 100640 (2021).

De Ruysscher, D., Faivre-Finn, C., Le Pechoux, C., Peeters, S. & Belderbos, J. High-dose re-irradiation following radical radiotherapy for non-small-cell lung cancer. Lancet Oncol. 15 , e620–e624 (2014).

Beddok, A. et al. Clinical and technical challenges of cancer reirradiation: words of wisdom. Crit. Rev. Oncol. Hematol. 174 , 103655 (2022).

Liu, Y.-P. et al. Endoscopic surgery compared with intensity-modulated radiotherapy in resectable locally recurrent nasopharyngeal carcinoma: a multicentre, open-label, randomised, controlled, phase 3 trial. Lancet Oncol. 22 , 381–390 (2021).

Xiang, G. et al. Re-irradiation for local primary-recurrence esophageal squamous cell carcinoma treated with IMRT/VMAT. Radiat. Oncol. 18 , 114 (2023).

Mahantshetty, U. et al. Reirradiation with advanced brachytherapy techniques in recurrent GYN cancers. Brachytherapy 22 , 753–760 (2023).

Huang, R.-X. & Zhou, P.-K. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduct. Target. Ther. 5 , 60 (2020).

Wang, K. & Tepper, J. E. Radiation therapy-associated toxicity: etiology, management, and prevention. CA Cancer J. Clin. 71 , 437–454 (2021).

Yang, Y. et al. ZBP1-MLKL necroptotic signaling potentiates radiation-induced antitumor immunity via intratumoral STING pathway activation. Sci. Adv. 7 , eabf6290 (2021).

Tam, S. Y., Wu, V. W. C. & Law, H. K. W. Influence of autophagy on the efficacy of radiotherapy. Radiat. Oncol. 12 , 57 (2017).

Article   PubMed   PubMed Central   Google Scholar  

Lei, G. et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 30 , 146–162 (2020).

Han, C. et al. The AIM2 and NLRP3 inflammasomes trigger IL-1–mediated antitumor effects during radiation. Sci. Immunol. 6 , eabc6998 (2021).

Chen, H. et al. Radiotherapy modulates tumor cell fate decisions: a review. Radiat. Oncol. 17 , 196 (2022).

Tsvetkov, P. et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science 375 , 1254–1261 (2022).

Chen, L., Min, J. & Wang, F. Copper homeostasis and cuproptosis in health and disease. Signal Transduct. Target. Ther. 7 , 378 (2022).

Cobine, P. A. & Brady, D. C. Cuproptosis: cellular and molecular mechanisms underlying copper-induced cell death. Mol. Cell 82 , 1786–1787 (2022).

Jin, X.-K. et al. Orchestrated copper-based nanoreactor for remodeling tumor microenvironment to amplify cuproptosis-mediated anti-tumor immunity in colorectal cancer. Mater. Today 68 , 108–124 (2023).

Hasegawa, K., Saga, R. & Hosokawa, Y. Radiotherapeutic review: acquisition of radioresistance and cancer stem cell properties via irradiation. Radiat. Environ. Med. 12 , 25–36 (2023).

Google Scholar  

Gray, M. et al. Development and characterisation of acquired radioresistant breast cancer cell lines. Radiat. Oncol. 14 , 64 (2019).

Zhang, P., et al. Cancer nanomedicine toward clinical translation: obstacles, opportunities, and future prospects. Med 4 , 147–167 (2022).

Liu, L., Zhao, C., Miller, J. T. & Li, Y. Mechanistic study of CO 2 photoreduction with H 2 O on Cu/TiO 2 nanocomposites by in situ X-ray absorption and infrared spectroscopies. J. Phys. Chem. C 121 , 490–499 (2017).

Article   CAS   Google Scholar  

Wang, D. et al. In situ formed Z-scheme graphdiyne heterojunction realizes NIR-photocatalytic oxygen evolution and selective radiosensitization for hypoxic tumors. ACS Nano 16 , 21186–21198 (2022).

Ding, Z. et al. Radiotherapy reduces N -oxides for prodrug activation in tumors. J. Am. Chem. Soc. 144 , 9458–9464 (2022).

Guo, Z. et al. Radiotherapy-induced cleavage of quaternary ammonium groups activates prodrugs in tumors. Angew. Chem. Int. Ed. 61 , e202205014 (2022).

Zhang, C. et al. Tumor microenvironment-responsive Cu 2 (OH)PO 4 nanocrystals for selective and controllable radiosentization via the X-ray-triggered Fenton-like reaction. Nano Lett. 19 , 1749–1757 (2019).

Wang, L., Jiang, N., Xu, H., Luo, Y. & Zhang, T. Trace Cu(II)-mediated selective oxidation of benzothiazole: the predominance of sequential Cu(II)–Cu(I)–Cu(III) valence transition and dissolved oxygen. Environ. Sci. Technol. 57 , 12523–12533 (2023).

Zeng, L., Miller, E. W., Pralle, A., Isacoff, E. Y. & Chang, C. J. A selective turn-on fluorescent sensor for imaging copper in living cells. J. Am. Chem. Soc. 128 , 10–11 (2006).

Liu, X. et al. Actin cytoskeleton vulnerability to disulfide stress mediates disulfidptosis. Nat. Cell Biol . 25 , 404–414 (2023).

Xu, W. et al. Tumor microenvironment responsive hollow nanoplatform for triple amplification of oxidative stress to enhance cuproptosis-based synergistic cancer therapy. Adv. Healthc. Mater. 12 , 2202949 (2023).

Zhou, J. et al. Photothermally triggered copper payload release for cuproptosis‐promoted cancer synergistic therapy. Angew. Chem. Int. Ed. 62 , e202213922 (2023).

Guo, B. et al. Cuproptosis induced by ROS responsive nanoparticles with elesclomol and copper combined with αPD-L1 for enhanced cancer immunotherapy. Adv. Mater . 35 , 2212267 (2023).

Tang, D., Chen, X. & Kroemer, G. Cuproptosis: a copper-triggered modality of mitochondrial cell death. Cell Res. 32 , 417–418 (2022).

Zhao, B. et al. Photocatalysis-mediated drug-free sustainable cancer therapy using nanocatalyst. Nat. Commun. 12 , 1345 (2021).

Ning, S. et al. Type-I AIE photosensitizer loaded biomimetic system boosting cuproptosis to inhibit breast cancer metastasis and rechallenge. ACS Nano 17 , 10206–10217 (2023).

Chan, L. et al. Cuproptosis‐driven enhancement of thermotherapy by sequentially response Cu 2‐ x Se via copper chemical transition. Adv. Funct. Mater . 33 , 2302054 (2023).

Xu, W. et al. A hollow amorphous bimetal organic framework for synergistic cuproptosis/ferroptosis/apoptosis anticancer therapy via disrupting intracellular redox homeostasis and copper/iron metabolisms. Adv. Funct. Mater. 32 , 2205013 (2022).

Demaria, O. et al. Harnessing innate immunity in cancer therapy. Nature 574 , 45–56 (2019).

Kroemer, G., Galassi, C., Zitvogel, L. & Galluzzi, L. Immunogenic cell stress and death. Nat. Immunol. 23 , 487–500 (2022).

Fucikova, J. et al. Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis. 11 , 1013 (2020).

Liao, Y. et al. Piezoelectric materials for synergistic piezo- and radio-catalytic tumor therapy. Nano Today 44 , 101510 (2022).

Ni, K. et al. Synergistic checkpoint-blockade and radiotherapy–radiodynamic therapy via an immunomodulatory nanoscale metal–organic framework. Nat. Biomed. Eng. 6 , 144–156 (2022).

Huang, Z. et al. Nanoscale coordination polymers induce immunogenic cell death by amplifying radiation therapy mediated oxidative stress. Nat. Commun. 12 , 145 (2021).

Min, Y. et al. Antigen-capturing nanoparticles improve the abscopal effect and cancer immunotherapy. Nat. Nanotechnol. 12 , 877–882 (2017).

Melo, L. M. N., Lesner, N. P., Sabatier, M., Ubellacker, J. M. & Tasdogan, A. Emerging metabolomic tools to study cancer metastasis. Trends Cancer 8 , 988–1001 (2022).

Li, H. et al. Nanomedicine embraces cancer radio-immunotherapy: mechanism, design, recent advances, and clinical translation. Chem. Soc. Rev . 52 , 47–96 (2023).

Twyman-Saint Victor, C. et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 520 , 373–377 (2015).

Franken, N. A. P., Rodermond, H. M., Stap, J., Haveman, J. & Van Bree, C. Clonogenic assay of cells in vitro. Nat. Protoc. 1 , 2315–2319 (2006).

Download references

Acknowledgements

We greatly acknowledge the financial support from the National High Level Hospital Clinical Research Funding (2022-PUMCH-B-052, to F.Z., J.Y. and Z.G.), Strategic Priority Research Program of Chinese Academy of Sciences (XDB36000000, to Z.G.), National Basic Research Program of China (2021YFA1201200 and 2020YFA0710702, to Z.G.), the Directional Institutionalized Scientific Research Project, which relies on the Beijing Synchrotron Radiation Facility of Chinese Academy of Sciences (E12982U810 to Z.G.), the National Natural Science Foundation of China (22375205, to Z.G.) and the Beijing Natural Science Foundation (2222087, to Z.G.).

Author information

These authors contributed equally: You Liao, Dongmei Wang.

Authors and Affiliations

CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China

You Liao, Dongmei Wang, Chenglu Gu, Xue Wang, Shuang Zhu & Zhanjun Gu

College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, China

Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China

Ziye Zheng, Fuquan Zhang & Junfang Yan

You can also search for this author in PubMed   Google Scholar

Contributions

Y.L. and Z.G. conceived and designed the experiments. Y.L. and D.W. performed the experiments. Y.L. and D.W. synthesized and characterized PWCu polyoxometalates. Y.L. and D.W. performed the cell and animal experiments. C.G., X.W., S.Z. and Z.Z. assisted with the mouse radiation experiment. Y.L., D.W. and Z.G. analysed the data. Y.L. and Z.G. contributed materials and analysis tools. Z.G., J.Y. and F.Z. supervised the project. Y.L., D.W., J.Y. and Z.G. wrote the paper. Y.L. and D.W. revised the paper. All authors discussed the results and edited and approved the paper before submission.

Corresponding authors

Correspondence to Junfang Yan or Zhanjun Gu .

Ethics declarations

Competing interests.

The authors declare no competing interests.

Peer review

Peer review information.

Nature Nanotechnology thanks Xin Jiang, Fudi Wang and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended data fig. 1 up-regulation of fdx1 and lias in experimental tumours after ebrt..

a , Treatment scheme of tumours from mouse models before and after IR irradiation. b , Representative confocal images of tumour lesions before and after IR irradiation (scale bar, 50 μm). c , d , Normalized fluorescence signals of FDX1 and LIAS (CT26, n  = 5 mice per group; Hepa 1-6, n  = 3 mice per group; HeLa, n  = 3 mice per group; MDA-MB-231, n  = 4 mice per group). Data in c , d are shown as mean ± s.d. P values were calculated by two-tailed unpaired t -test ( c , d ). The experiments shown in b were repeated independently at least two times with similar results. Panel a created with BioRender.com .

Source data

Extended data fig. 2 the intratumoral distribution of pwcu after intratumoral injection..

a , 3D reconstruction images of synchrotron radiation micro-CT. b , H&E staining images (scale bar, 500 μm) and LA-ICP-TOFMS element mapping images (scale bar, 500 μm) of tumour tissue. LA-ICP-TOFMS, laser ablation-inductively coupled plasma-time of flight mass spectrometry. The experiments in b were repeated independently at least two times with similar results.

Extended Data Fig. 3 Comparison of radiosensitizing effect between HfO 2 and PWCu.

a , Schematic diagram of orthotopic 4T1-R breast cancer establishment and radiotherapy. b , c , Growth curves of tumours. Results are expressed as mean tumour volume ± s.d. with % TGI and P value relative to NT (on day 23) as determined by two-tailed unpaired t -test. n  = 10 biologically independent mice per group. CR, complete response. d , Tumour growth inhibition rates of 2 Gy × 3 IR on 4T1-R tumours. n  = 10 biologically independent mice per group. e , Representative photographs of tumours. Data in d are represented as box and whiskers (centre line, median; box limits, 25th and 75th percentiles; whiskers, minimum and maximum). The P value was calculated by two-tailed unpaired t -test ( d ).

Supplementary information

Supplementary information.

Supplementary Figs. 1–27, Tables 1–5 and Methods.

Reporting Summary

Source data fig. 1.

Statistical source data.

Unprocessed western blots.

Source Data Fig. 2

Source data fig. 3, source data fig. 4, source data fig. 5, source data fig. 6, source data extended data fig. 1, source data extended data fig. 3, rights and permissions.

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Cite this article.

Liao, Y., Wang, D., Gu, C. et al. A cuproptosis nanocapsule for cancer radiotherapy. Nat. Nanotechnol. (2024). https://doi.org/10.1038/s41565-024-01784-1

Download citation

Received : 25 September 2023

Accepted : 08 August 2024

Published : 19 September 2024

DOI : https://doi.org/10.1038/s41565-024-01784-1

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

laser scanning confocal microscopy experiments

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

Principles and practices of laser scanning confocal microscopy

Affiliation.

  • 1 Department of Molecular Biology, University of Wisconsin, 1525 Linden Drive, Madison, Wisconsin 53706, USA. [email protected]
  • PMID: 11131973
  • DOI: 10.1385/MB:16:2:127

The laser scanning confocal microscope (LSCM) is an essential tool for many biomedical imaging applications at the level of the light microscope. The basic principles of confocal microscopy and the evolution of the LSCM into today's sophisticated instruments are outlined. The major imaging modes of the LSCM are introduced including single optical sections, multiple wavelength images, three-dimensional reconstructions, and living cell and tissue sequences. Practical aspects of specimen preparation, image collection, and image presentation are included along with a primer on troubleshooting the LSCM for the novice.

PubMed Disclaimer

  • Methods Mol Biol. 1999;122:373-84 - PubMed
  • Biophys J. 1999 Nov;77(5):2871-86 - PubMed
  • Methods Mol Biol. 1999;122:403-20 - PubMed
  • J Cell Biol. 1987 Jul;105(1):41-8 - PubMed
  • Curr Biol. 1996 Dec 1;6(12):1595-8 - PubMed

Publication types

  • Search in MeSH

LinkOut - more resources

Full text sources.

  • Ovid Technologies, Inc.

Other Literature Sources

  • The Lens - Patent Citations
  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

  • Open access
  • Published: 17 September 2024

Impact of different activation procedures on sodium hypochlorite penetration into dentinal tubules after endodontic retreatment via confocal laser scanning microscopy

  • Betul Gunes 1 ,
  • Kübra Yeşildal Yeter 1 &
  • Yasin Altay 2  

BMC Oral Health volume  24 , Article number:  1103 ( 2024 ) Cite this article

Metrics details

Infected dentinal tubules are a possible source of bacteria that are responsible for the failure of root canal treatment. Therefore, disinfection of dentinal tubules by increasing the penetration of the irrigation solution is important for success in retreatment cases. This study utilized confocal laser scanning microscopy (CLSM) to assess and compare the impact of XPR, ultrasonic irrigation (UI) and sonic activation (SA) on NaOCl penetration into dentinal tubules following endodontic retreatment.

A total of forty mandibular premolars were enrolled in this investigation. Following root canal preparation up to ProTaper X3 file (30/0.07), root canals were obturated with gutta-percha and bioceramic root canal sealer with single cone technique. The root canal filling materials were removed using ProTaper nickel-titanium rotary retreatment files until the working length was reached. The retreatment procedure was finalized using the ProTaper Next X4 (40/0.06). The teeth were divided into four groups based on the irrigation activation technique: control (conventional needle irrigation), SA, UI and XPR. During the final irrigation procedure, Rhodamine B dye was introduced to 5% NaOCl for visualization via CLSM. Subsequent to image acquisition, the maximum penetration, penetration percentage, and penetration area were calculated. Data were statistically analyzed using the Kruskal-Wallis, Friedman, and Bonferroni Dunn multiple comparison tests through R software ( p  < 0.05).

In the middle third, UI yielded a significantly higher penetration percentage than the control group ( p  < 0.05). The UI and XPR groups showed increased penetration percentages in the coronal and middle thirds compared with the apical third ( P  < 0.05). Maximum penetration was notably reduced in the apical third than in comparison with the coronal and middle thirds in all groups ( p  < 0.05). In the control, SA and XP groups, the penetration area was ranked in descending order as coronal, middle and apical ( p  < 0.05). Conversely, in the ultrasonic group, the penetration area was significantly lower in the apical third than in the middle and coronal thirds ( p  < 0.05).

Conclusions

UI enhanced the penetration percentage in the middle third of the root compared with that in the control group. XPR and SA showed no significant effect on NaOCl penetration following retreatment.

Peer Review reports

Introduction

Endodontic treatment failures can result from various factors, including coronal leakage, missed canals and procedural errors; a systematic review was reported that the success rate of primary root canal treatment ranged between 68% and 85% [ 1 ]. In such cases, clinicians must select an appropriate retreatment option to ensure long-term success [ 2 ]. The primary choice for addressing endodontic failures is non-surgical endodontic retreatment, involving the complete removal of existing filling materials and the eradication of microorganisms from the root canal system [ 3 ]. However, the complete removal of root canal filling materials is the most challenging step in non-surgical retreatment. Residual root canal filling materials can impede the penetration of irrigation solutions and root canal sealers into dentinal tubules [ 4 , 5 , 6 , 7 ], potentially leading to incomplete healing or new inflammation. Numerous methods have been proposed for root canal retreatment, using hand and/or rotary instruments with or without different solvents. However, neither instruments nor solvents can entirely eliminate root canal filling material [ 8 , 9 , 10 ]. Therefore, supplementary procedures are recommended to reduce the remaining root canal filling material [ 11 , 12 , 13 , 14 ].

Infected dentinal tubules are a possible source of bacteria responsible for post-treatment diseases [ 15 ]. Therefore, the complete removal of root canal filling material and the disinfection of dentinal tubules are vital for long-term success in retreatment cases. Previous studies have primarily focused on the impact of supplementary procedures such as sonic, ultrasonic and laser assisted irrigation activation techniques, as well as specially designed instruments (XP Endo Finisher R), on the removal of root canal filling material from the root canal walls [ 9 , 10 , 11 , 12 , 13 , 14 ].

Ultrasonic irrigation (UI) generates cavitation and acoustic streaming that enhances cleaning of the root canal wall surface by increasing the shear stress [ 16 ]. Prior researches have investigated the effectiveness of UI in removing root canal filling materials during retreatment [ 17 , 18 , 19 ] and enhancing the penetration of irrigation solutions into dentinal tubules during primary root canal treatment [ 12 , 20 , 21 ]; these studies suggest that UI is a viable method for both procedures. Notably, irrigation activation using sonic devices operates at lower frequencies than that using UI devices. Nevertheless, existing studies examining the efficacy of sonic activation (SA) in promoting the penetration of irrigation solutions during primary root canal treatment have produced conflicting results [ 18 , 19 ].

Recently, the XP-endo Finisher R (XPR) (FKG, La Chaux-de-Fonds, Switzerland) file, crafted from a MaxWire alloy with a core diameter of ISO #30 (distinct from its precursor, the XP-endo Finisher (XPF), which has an ISO #25 core diameter) and featuring no taper, has been introduced. XPR can alter its shape by adjusting its temperature during the transition from the martensite to the austenite phase. At body temperature, XPR adopts a spoon-shaped configuration, enabling it to eliminate debris and root canal filling material from otherwise inaccessible regions without modifying the canal shape [ 22 , 23 ]. A recent systematic review and meta-analysis of in vitro studies involving XPF and XPR concluded that both files offer benefits in removing root canal filling materials from the root canal walls [ 24 ]. Notably, to the best of our knowledge, no previous study has examined the impact of XPR on the penetration of sodium hypochlorite (NaOCl) following the removal of root canal filling materials.

The objective of this in vitro study was to compare the effects of XPR, UI, and SA on the penetration of NaOCl into the dentinal tubules after retreatment using confocal laser scanning microscopy (CLSM). The null hypothesis proposed that there would be no significant difference in the penetration of NaOCl into the dentinal tubules between the groups.

Materials and methods

Ethical compliance of this in vitro study was granted by the University Ethics Committee (18/24.09.2019). The sample size was calculated based on a previous study [ 25 ] with a similar methodology in the literature. As a result of the power analysis performed using G*Power 3.1 Software (Heinrich Heine University, Düsseldorf, Germany), following these input conditions: effect size = 0.58; α err = 0.05; power = 0.95, total sample size was determined to be at least 33. Forty freshly extracted human mandibular premolars with a single root and root canal, confirmed through radiographs, fully developed roots and no signs of root cracks or resorption, were selected for this study. The teeth were preserved in a saline solution at room temperature until they underwent the experimental procedure. Prior to root canal preparation teeth were decoronated at 16 mm from the apex for standardization.

Preparation and obturation of the root canals

A #10 K-file was introduced into the root canal until the tip of the file was visible from the apex; then, 1 mm was subtracted from this length to determine the working length for each specimen. Root canals were prepared using X1, X2 and X3 ProTaper Next rotary system files (Dentsply Maillefer, Balleigues, Switzerland) following the manufacturer’s recommendations. During the preparation, the root canals were irrigated with 5% NaOCl. Final irrigation was carried out using 17% ethylenediaminetetraacetic acid (EDTA), 5% NaOCl and distilled water. After the final irrigation procedure, the root canals were dried with paper points and filled with a bioceramic-based root canal sealer (BioRoot RCS; Septogon, Saint Maur Des Fosses, France) and #25/0.06 tapered ProTaper X3 gutta-percha (Dentsply Maillefer, Ballaigues, Switzerland) using the single-cone obturation technique. Coronal access to the root canal was sealed using temporary filling material (Cavit G; 3 M Espe, Seefeld, Germany). Specimens were stored at 37 °C under 100% humidity for 2 weeks to allow the sealer to completely set.

Retreatment of root canal filling materials

After removing the temporary filling material, the coronal 3 mm of the root canal filling was removed using a Gates-Glidden #2. Retreatment of the root canal filling materials was performed using D1, D2 and D3 ProTaper retreatment files until the working length was reached. The retreatment procedure was finalized using the ProTaper Next X4 (40/0.06). No solvent was used during retreatment procedure. Root canals were irrigated with NaOCl during the retreatment procedure. The specimens were randomly divided into four groups ( n  = 10) according to the final irrigation activation procedure.

Conventional needle irrigation (control)

The root canals were irrigated with 17% EDTA and 5% NaOCl solutions for 1 min each. For irrigation, a 31-gauge closed end, two-sided vented irrigation needle (NaviTip Sideport; Ultradent Products. Inc., South Jordan, UT, USA) was placed in the canal 2 mm short of the working length and used in a back-and-forth motion.

The root canals were irrigated with 17% EDTA and activated with a sonic irrigation activation device (EndoActivator, Dentsply Advanced Endodontics, Santa Barbara, CA, USA) using a 25/0.04 polymer tip placed 2 mm short of the working length. Activation was performed for 20 s at 10,000 cpm according to the manufacturer’s recommendations. This procedure was repeated three times for a total of activation time 1 min. The same procedure was repeated to activate the 5% NaOCl solution.

Root canals were irrigated with 17% EDTA and then activated using an ultrasonic device (Newtron P5 XS BLED, Satelec/Acteon, Merignac, France) with an Irrisafe 20/0.01 tip (Satelec/Acteon, Merignac, France) placed 2 mm short of the working length. Activation was carried out for 20 s at a power setting of 6, following the manufacturer’s recommendations. This procedure was repeated three times for a total of activation time of 1 min. The same procedure was repeated to activate the 5% NaOCl solution.

Root canals were irrigated with 17% EDTA and then activated with the XP Endo Finisher R (30/0.00) retreatment file at 800 rpm and with a torque of 1 Ncm. Activation involved a 20-s up-and-down motion over a range of 7–8 mm, following the manufacturer’s recommendations. This procedure was repeated three times for a total of activation time of 1 min. The same procedure was repeated to activate the 5% NaOCl solution. Notably, the XPR file is activated at body temperature, and preparations for this group was carried out in a 37 °C water bath.

A 0.1% rhodamine fluorescent dye was added to the NaOCl solution before the irrigation procedure to enable the examination of solution penetration using CLSM. During the final irrigation procedure, each solution was applied at a volume of 2.5 ml for each specimen. At the conclusion of the final irrigation procedure, the root canals were irrigated with 2.5 ml of distilled water. All endodontic procedures were performed by an experienced endodontist.

Assessment of penetration of NaOCl

Following the final irrigation procedure, the specimens were embedded in auto-polymerizing acrylic, and ∼  1 mm thick horizontal sections were obtained at 3 mm (apical), 8 mm (middle) and 12 mm (coronal) from the apex using a diamond cutting disc (Isomed1000, Buehler, Lake Bluff, IL) under water cooling. The apical, middle and coronal thirds were scanned using CLSM at x5 magnification with a laser wave-length of 561 nm (LSM 800; Zeiss, Jena, Germany) (Fig. 1 ). Subsequently, after acquiring the images, the following three parameters were calculated for each third.

figure 1

Representative confocal laser scanning images of the coronal, middle and apical root thirds of each experimental group

1) Maximum penetration depth : This parameter was defined as the distance from the root canal wall to the deepest point of penetration at four standardized points with 90° angles. The total value of these four measurements was divided by 4 to calculate the mean maximum penetration depth (μm) [ 26 ] (Fig.  2 a).

figure 2

( a ) Measurement of maximum dentinal tubule penetration at four standardized points. ( b ) Measurement of sealer penetration (yellow lines) and circumference of the root canal wall (white line) to calculate percentage of sealer. ( c ) Measurement of the area of penetrations

2) Penetration percentage : The penetration percentage was calculated by dividing the length to which the irrigation solution penetrated the dentinal tubules along the root canal walls by the circumference of the root canal wall and multiplying this result by 100 (as %) [ 25 ] (Fig.  2 b).

3) Area of penetration : To calculate the area of penetration, the total area covered by penetration of the irrigation solution was measured (μm 2 ) [ 27 ] (Fig.  2 c).

The investigator who performed the measurements on the CLSM images was blinded to the treatment groups. The images were analyzed using the Zeiss Zen software 2.3 (Carl Zeiss).

Statistical analysis

The study was conducted with an average of 10 replicates, following a 4 × 3 factorial experimental design. The analysis aimed to determine whether a statistical difference existed between the means of the activation methods (Control, SA, UI and XPR) and the root thirds (apical, middle and coronal). Statistical analysis was performed using the Anderson Darling test to assess normal distribution and Levene test to evaluate the homogeneity of group variances. However, these tests did not meet the prerequisites for parametric tests ( P  < 0.005). The Kruskal–Wallis test was employed to compare the medians of the activation methods in each root third, whereas the Friedman test was used for the median comparisons of the root third for each activation method. Statistical analyses were performed using R software (R Core Team, 2020). To determine which activation method and root third exhibited statistically significant differences between medians, the Bonferroni–Dunn multiple comparison test was applied at a significance level of 5%.

The mean, standard error and p - values obtained by comparing the data of root regions for each experimental group are shown in Table  1 . When examining each activation method for the different root thirds, the SA and control groups had no significant impact on the penetration percentage in all root thirds ( p  > 0.05). In contrast, the UI and XPR groups displayed a higher penetration percentage in the coronal and middle thirds than in the apical third ( p  < 0.05). The maximum penetration value was significantly lower in the apical third than in the coronal and middle thirds in all groups ( p  < 0.05). In the control, sonic and XP groups, the irrigation solution penetration area was ranked from highest to lowest as coronal, middle and apical, respectively ( p  < 0.05). However, in the ultrasonic group, the penetration area was significantly smaller in the apical third than in the middle and coronal thirds ( p  < 0.05).

The mean, standard error and p - values obtained by comparing the data of experimental groups for each root region are shown in Table  2 . When comparing the irrigation activation methods for each root third, no statistically significant differences were observed in the penetration area, penetration percentage, or maximum penetration parameters in the apical and coronal thirds ( p  > 0.05). In the middle third, the UI group exhibited a higher percentage of penetration than the control group ( p  < 0.05), whereas the penetration area and maximum penetration parameters were statistically similar for all experimental groups ( p  > 0.05).

After primary root canal therapy, the presence of bacteria within the dentinal tubules is considered as a primary cause of apical periodontitis [ 15 ]. Thus, effective irrigation during and after the removal of root canal filling materials is crucial to achieving optimal disinfection in retreatment cases. In this study, we evaluated the impact of various final irrigation activation techniques on the penetration of NaOCl into the dentinal tubules using CLSM. According to our results, UI demonstrated a significantly higher penetration percentage in the middle third of the root than in the control and SA group, where the UI and XPR performed similar. In the apical root third, NaOCl exhibited less penetration than in the coronal and middle thirds, except for the SA and control groups, where these changes remained statistically insignificant. As a result, we partially rejected null hypothesis.

It has been reported that the failure of endodontic treatment may be due to the persistence of microorganisms in the root canal system, especially in the apical part and even in well-treated teeth [ 28 ]. The persistence of microorganisms in the root canal system after retreatment may cause periradicular inflammation to continue. Therefore, optimum disinfection of the root canal system is important to increase the success rate of retreatment. In a previous study examining bacterial penetration in root filled teeth, bacteria were most frequently detected in the cervical third and inner dentin adjacent to the root canal [ 29 ]. In our study, we aimed to evaluate the penetration of NaOCl after retreatment by calculating the penetration percentage parameter around the root canal circumference and the maximum penetration and penetration area parameters in the region between the root canal and edge of the root in the horizontal root section for the coronal, middle and apical thirds.

Previous studies examining the effects of different activation systems on irrigation penetration solution into dentinal tubules primarily focused on primary root canal treatment [ 17 , 19 , 30 , 31 , 32 ]. These studies varied in terms of specimen type (human or bovine teeth), irrigation solution (NaOCl, CHX, Irritol), irrigation activation system (UI, SA, Laser-assisted activation) and observation method (Stereomicroscope, CLSM), making direct comparisons challenging. To the best of our knowledge, this study is the first to evaluate the penetration of irrigation solutions into dentinal tubules after retreatment. Therefore, directly comparing the results of our study with those reported in the literature is not appropriate.

According to our results, the only difference between the activation methods was observed between the percentage of penetration of the irrigation solution in UI and control groups in the middle third of the root. UI increased the penetration percentage of NaOCl compared with that in the control group. This result is in accordance with the results of the study of Akcay et al. [ 19 ] that observed irrigation penetration during primary root canal treatment. The UI provides cavitation and acoustic streaming [ 16 ] and produces sufficient shear forces to dislodge debris in instrumented canals [ 33 ]. Greater cleanliness of the canal walls may result in increased irrigation penetration. The effect of SA on the penetration of irrigation solutions has been studied, but the results are controversial [ 19 , 31 , 33 ]. Some studies reported that SA was superior to conventional needle irrigation, but these studies evaluated different types of irrigation solutions [ 31 , 33 ]. Similar to our study, Akcay et al. [ 19 ] reported no significant difference between SA and conventional needle irrigation and the same irrigation solution was used in our study. SA does not have the ability to allow sufficient streaming because of its low energy level and long wavelength [ 34 ]. The efficacy of XPR in the removal of root canal filling materials has been previously studied [ 11 , 23 , 35 , 36 ]. The results of these studies demonstrated that XPR was more effective than UI and SA for the removal of root canal filling materials. However, our results do not directly correlate with those of the aforementioned studies. In this study, no statistically significant differences were observed among XPR, UI and SA.

While bacterial penetration can be seen in every region of the root canal, more bacterial presence can be seen in the apical third because it is more difficult to reach this region via preparation or irrigation procedures [ 37 ]. The importance of bacterial presence in the apical region in root canal treatment failure was previously emphasized [ 28 ]. Studies that evaluated the penetration of irrigation solution into dentinal tubules showed lower penetration values for the apical third than for the middle and coronal thirds [ 19 , 31 ]. This result can be explained by the presence of narrower, sclerotic and fewer dentinal tubules at the apical root third [ 38 ]. Furthermore, the flow of the irrigation solutions to the apical third may be inefficient at removing the smear layer and debris [ 39 ]. Consistent with these studies, our results showed less penetration for the apical third than for the middle and coronal thirds for the UI and XPR groups for all of the parameters. In the SA and control groups, the apical root third showed less penetration than the middle and coronal thirds in terms of the penetration area and maximum penetration parameters. However, the SA and control groups showed similar percentages of penetration in all of the root thirds. The percentage of penetration, a circumferential measurement of the root canal wall, may be more affected by dentin tubule density. The inability to establish standardization in specimens may have revealed this contrasting result. In terms of clinical significance, future studies on the penetration of irrigation solution penetration should focus on alternative methods to enhance the penetration of NaOCl in the apical region.

The penetration of irrigation solutions into the dentinal tubules was observed using various microscopy techniques, including stereomicroscopy, scanning electron microscopy (SEM) and CLSM [ 17 , 19 , 31 , 33 , 40 , 41 ]. Detailed images of dentinal tubules can be obtained using CLSM, providing observations and measurements of penetration into the dentinal tubules in a single image along the circumference of the root canal walls [ 41 ]. Moreover, CLSM is a non-destructive method and does not dehydrate the specimen [ 42 ]. In contrast, SEM can visualize the dentinal tubules in only one plane, necessitating the further reconstruction of high magnification images to obtain a detailed final image. Acquiring images via SEM involves additional procedures, such as coating the specimens with gold and working under a vacuum, which can be time-consuming and can introduce artefacts during the preparation of the specimens [ 43 ]. Stereomicroscopy, on the other hand, is not capable of providing sufficiently detailed images of the dentinal tubules. Given these considerations, CLSM imaging was chosen as the preferred method for evaluating NaOCl penetration into the dentinal tubules in this study.

Every effort was made to select teeth that met the inclusion criteria. Notably, aging can lead to dentinal tubule sclerosis, especially in the apical third. This sclerosis can alter the number and diameter of the dentinal tubules, directly influencing the penetration of the irrigation solutions into them [ 44 ]. This variation can introduce considerable variability in experimental results and should be recognized as a limitation. Furthermore, to maintain standardization, the number and volume of irrigants, as well as the duration of the irrigation and activation procedures, were kept consistent for all experimental groups.

In this in vitro study, the following conclusions were drawn within the established limitations: in the middle third of the root, UI increased the penetration percentage compared with the control group, while XPR and SA showed no notable effect on NaOCl penetration following retreatment. Future studies are needed to explore the impact of various irrigation activation techniques on penetration of irrigation solutions into dentinal tubules after the retreatment procedure to provide definitive insights.

Data availability

The data of this research are available upon reasonable request from the corresponding author.

Abbreviations

  • Confocal laser scanning microscopy

Ethylenediaminetetraacetic acid

Sodium hypochlorite

  • Sonic activation

Scanning electron microscopy

  • Ultrasonic irrigation

XP-endo Finisher

XP endo Finisher R

Ng Y-L, et al. Outcome of primary root canal treatment: systematic review of the literature–part 1. Effects of study characteristics on probability of success. Int Endod J. 2007;40:921–39.

Article   PubMed   Google Scholar  

Ruddle CJ. Nonsurgical endodontic retreatment. J Calif Dent Assoc. 1997;25:769–96.

CAS   PubMed   Google Scholar  

Siqueira JF, Rôças IN, Ricucci D, et al. Causes and management of post-treatment apical periodontitis. Br Dent J. 2014;216:305–12.

Endo MS, Ferraz CCR, Zaia AA, et al. Quantitative and qualitative analysis of microorganisms in root-filled teeth with persistent infection: monitoring of the endodontic retreatment. Eur J Dent. 2013;7:302–9.

Article   PubMed   PubMed Central   Google Scholar  

Kok D, Da Rosa RA, Barreto MS, et al. Penetrability of AH plus and MTA fillapex after endodontic treatment and retreatment: a confocal laser scanning microscopy study. Microsc Res Tech. 2014;77:467–71.

Article   CAS   PubMed   Google Scholar  

Ricucci D, Siqueira JF, Bate AL, et al. Histologic investigation of root canal-treated teeth with apical periodontitis: a retrospective study from twenty-four patients. J Endod. 2009;35:493–502.

Good ML, McCammon A. Removal of gutta-percha and root canal sealer: a literature review and an audit comparing current practice in dental schools. Dent Update. 2012;39:703–8.

Bueno CE, da Delboni S, de Araújo MG, et al. Effectiveness of rotary and hand files in gutta-percha and sealer removal using chloroform or chlorhexidine gel. Braz Dent J. 2006;17:139–43.

Monguilhott Crozeta B, Damião de Sousa-Neto M, Bianchi Leoni G, et al. A micro-computed tomography assessment of the efficacy of rotary and reciprocating techniques for filling material removal in root canal retreatment. Clin Oral Investig. 2016;20:2235–40.

Zuolo AS, Mello JE, Cunha RS, et al. Efficacy of reciprocating and rotary techniques for removing filling material during root canal retreatment. Int Endod J. 2013;46:947–53.

Sinsareekul C, Hiran-us S. Comparison of the efficacy of three different supplementary cleaning protocols in root-filled teeth with a bioceramic sealer after retreatment—a micro-computed tomographic study. Clin Oral Investig. 2022;26:3515–21.

Grischke J, Müller-Heine A, Hülsmann M. The effect of four different irrigation systems in the removal of a root canal sealer. Clin Oral Investig. 2014;18:1845–51.

Machado AG, Guilherme BPS, Provenzano JC, et al. Effects of preparation with the self-adjusting file, TRUShape and XP-endo shaper systems, and a supplementary step with XP-endo finisher R on filling material removal during retreatment of mandibular molar canals. Int Endod J. 2019;52:709–15.

Jiang S, Zou T, Li D, et al. Effectiveness of sonic, ultrasonic, and photon-induced photoacoustic streaming activation of NaOCl on filling material removal following retreatment in oval canal anatomy. Photomed Laser Surg. 2016;34:3–10.

Vieira AR, Siqueira JF, Ricucci D, et al. Dentinal tubule infection as the cause of recurrent disease and late endodontic treatment failure: a case report. J Endod. 2012;38:250–4.

Boutsioukis C, Maria Teresa Arias-Moliz. Present status and future directions–irrigants and irrigation methods. Int Endod J. 2022;55:588–612.

Ghorbanzadeh A, Aminsobhani M, Sohrabi K, et al. Penetration depth of sodium hypochlorite in dentinal tubules after conventional irrigation, passive ultrasonic agitation and nd: YAG laser activated irrigation. J Lasers Med Sci. 2016;7:105–11.

Galler KM, Grubmüller V, Schlichting R, et al. Penetration depth of irrigants into root dentine after sonic, ultrasonic and photoacoustic activation. Int Endod J. 2019;52:1210–7.

Akcay M, Arslan H, Mese M, et al. Effect of photon-initiated photoacoustic streaming, passive ultrasonic, and sonic irrigation techniques on dentinal tubule penetration of irrigation solution: a confocal microscopic study. Clin Oral Investig. 2017;21:2205–12.

Lee TY, Kahm SH, Kim KJ et al. The retrievability of calcium silicate-based sealer during retreatment and the effectiveness of additional passive ultrasonic irrigation: a microcomputed tomographic study. Scanning. 2022;3933305.

Jain M, Singhal A, Gurtu A, et al. Influence of ultrasonic irrigation and chloroform on cleanliness of dentinal tubules during endodontic retreatment-an invitro SEM study. J Clin Diagn Res. 2015;9:ZC11–5.

PubMed   PubMed Central   Google Scholar  

Alves FRF, Marceliano-Alves MF, Sousa JCN, et al. Removal of root canal fillings in curved canals using either reciprocating single- or rotary multi-instrument systems and a supplementary step with the XP-Endo finisher. J Endod. 2016;42:1114–9.

Hassan R, El Zahar S. Cleaning efficiency of XP finisher, XP finisher R and passive ultrasonic irrigation following retreatment of teeth obturated with TotalFill HiFlow bioceramic sealer. Eur Endod J. 2022;7:143–9.

Uzunoglu-Özyürek E, Küçükkaya Eren S, Karahan S. Contribution of XP‐Endo files to the root canal filling removal: a systematic review and meta‐analysis of in vitro studies. Aust Endod J. 2021;47:703–14.

Küçük M, Kermeoğlu F. Efficacy of different irrigation methods on dentinal tubule penetration of chlorhexidine, qmix and irritrol: a confocal laser scanning microscopy study. Aust Endod J. 2019;45:202–8.

Gharib SR, Tordik PA, Imamura GM, et al. A confocal laser scanning microscope investigation of the epiphany obturation system. J Endod. 2007;33:957–61.

Machado R, Cruz ATG, de Araujo BM. Tubular dentin sealer penetration after different final irrigation protocols: a confocal laser scanning microscopy study. Microsc Res Tech. 2018;81:649–54.

Siqueira F Jr. Aetiology of root canal treatment failure: why well-treated teeth can fail. Int Endod J. 2001;34:1–10.

Kwang S, Abbott P. The presence and distribution of bacteria in dentinal tubules of root filled teeth. Int Endod J. 2014;47:600–10.

Rajakumaran A, Ganesh A. Comparative evaluation of depth of penetration of root canal irrigant after using manual, passive ultrasonic, and diode laser-assisted irrigant activation technique. J Pharm Bioallied Sci. 2019;11:S216–220.

Article   CAS   PubMed   PubMed Central   Google Scholar  

Keskin NB, Bozkurt DA, Yusufoğlu Sİ. Evaluation of dentinal tubule penetration of irritrol and chlorhexidine irrigating solutions activated using EDDY and photon-initiated photoacoustic streaming. Photodiagnosis Photodyn Ther. 2022;39:102925.

Sabins RA, Johnson JD, Hellstein JW. A comparison of the cleaning efficacy of short-term sonic and ultrasonic passive irrigation after hand instrumentation in molar root canals. J Endod. 2003;29:674–8.

Jain S, Patni PM, Jain P, et al. Comparison of dentinal tubular penetration of intracanal heated and preheated sodium hypochlorite through different agitation techniques. J Endod. 2023;49:686–91.

Al-Jadaa A, Paqué F, Attin T, et al. Acoustic hypochlorite activation in simulated curved canals. J Endod. 2009;35:1408–11.

Da Silva V, Loroño G, de Valencia O, et al. The supplementary use of XP-endo finisher R after reciproc enhances the removal of a bioceramic sealer in oval root canals. Aust Endod J. 2022;49(Suppl 1):156–61.

PubMed   Google Scholar  

Volponi A, Pelegrine RA, Kato AS, et al. Micro–computed tomographic assessment of supplementary cleaning techniques for removing bioceramic sealer and gutta-percha in oval canals. J Endod. 2020;46:1901–6.

Peters LB, Wesselink PR, Moorer WR. The fate and the role of bacteria left in root dentinal tubules. Int Endod J. 1995;28:95–9.

Paqué F, Luder HU, Sener B, et al. Tubular sclerosis rather than the smear layer impedes dye penetration into the dentine of endodontically instrumented root canals. Int Endod J. 2006;39:18–25.

Teixeira CS, Felippe MCS, Felippe WT. The effect of application time of EDTA and NaOCI on intracanal smear layer removal: an SEM analysis. Int Endod J. 2005;38:285–90.

de Oliveira RA, Weissheimer T, Só GB, et al. Dentinal tubule penetration of sodium hypochlorite in root canals with and without mechanical preparation and different irrigant activation methods. Restor Dent Endod. 2023;48:e1.

Giardino L, Pedullà E, Cavani F, et al. Comparative evaluation of the penetration depth into dentinal tubules of three endodontic irrigants. Materials. 2021;14:5853.

Van Meerbeek B, Vargas M, Perdigao J, et al. Microscopy investigations. Techniques, results, limitations. Am J Dent. 2000;13(Spec No):D3–18.

Google Scholar  

Mamootil K, Messer HH. Penetration of dentinal tubules by endodontic sealer cements in extracted teeth and in vivo. Int Endod J. 2007;40:873–81.

Montoya C, Arango-Santander S, Peláez-Vargas A, et al. Effect of aging on the microstructure, hardness and chemical composition of dentin. Arch Oral Biol. 2015;60:1811–20.

Download references

Acknowledgements

This study was supported by the Eskisehir Osmangazi University Scientific Research Projects Coordination Unit (Project number: 202111002).

Author information

Authors and affiliations.

Faculty of Dentistry, Department of Endodontics, Eskisehir Osmangazi University, Eskişehir, 26040, Turkey

Betul Gunes & Kübra Yeşildal Yeter

Faculty of Agriculture, Department of Animal Science Biometry and Genetic, Eskisehir Osmangazi University, Eskişehir, Turkey

Yasin Altay

You can also search for this author in PubMed   Google Scholar

Contributions

B. G.: Conceptualization, Investigation, writing- reviewing&editing. K. Y. Y.: Conceptualization, writing-original draft preparation, writing- reviewing&editing, visualization, project administration. Y. A.: Formal analyses, writing- reviewing&editing.

Corresponding author

Correspondence to Kübra Yeşildal Yeter .

Ethics declarations

Ethical approval and consent to participate.

Ethical compliance of this in-vitro study was approved by the Eskişehir Osmangazi University Ethics Committee (18/24.09.2019). All participants gave informed consent to participate.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/ .

Reprints and permissions

About this article

Cite this article.

Gunes, B., Yeter, K.Y. & Altay, Y. Impact of different activation procedures on sodium hypochlorite penetration into dentinal tubules after endodontic retreatment via confocal laser scanning microscopy. BMC Oral Health 24 , 1103 (2024). https://doi.org/10.1186/s12903-024-04891-6

Download citation

Received : 18 July 2024

Accepted : 10 September 2024

Published : 17 September 2024

DOI : https://doi.org/10.1186/s12903-024-04891-6

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Endodontic retreatment
  • Irrigation penetration
  • XP-endo finisher R

BMC Oral Health

ISSN: 1472-6831

laser scanning confocal microscopy experiments

Mindat.org localities near to this feature

LocalityLatitudeLongitudeDistanceBearing
56° 46' 19" N60° 38' 30" E9.1km (5.6 miles)
56° 53' 59" N60° 46' 59" E11.7km (7.3 miles)
56° 46' 23" N60° 23' 25" E16.1km (10.0 miles)
56° 53' 8" N60° 54' 47" E18.7km (11.6 miles)
56° 39' 17" N60° 43' 23" E22.9km (14.2 miles)
56° 39' 47" N60° 46' 55" E23.4km (14.6 miles)
56° 38' 21" N60° 36' 17" E23.6km (14.7 miles)
56° 37' 27" N60° 36' 32" E25.3km (15.7 miles)
56° 37' 19" N60° 36' 45" E25.5km (15.9 miles)
56° 37' 27" N60° 43' 13" E26.1km (16.2 miles)
56° 56' 25" N60° 10' 49" E28.0km (17.4 miles)
57° 3' 47" N60° 54' 40" E29.7km (18.4 miles)
56° 34' 26" N60° 40' 41" E31.2km (19.4 miles)
56° 42' 36" N60° 5' 24" E35.5km (22.1 miles)
56° 32' 44" N60° 24' 28" E36.2km (22.5 miles)
56° 30' 15" N60° 22' 54" E41.2km (25.6 miles)
56° 36' 5" N61° 7' 1" E41.5km (25.8 miles)
56° 28' 47" N60° 20' 9" E44.7km (27.8 miles)
57° 14' 39" N60° 46' 22" E44.7km (27.8 miles)
57° 4' 59" N59° 55' 0" E49.4km (30.7 miles)
56° 57' 44" N59° 49' 20" E49.5km (30.8 miles)
56° 27' 56" N60° 11' 30" E50.1km (31.1 miles)
57° 0' 47" N61° 29' 32" E56.3km (35.0 miles)
57° 6' 52" N61° 24' 26" E56.3km (35.0 miles)
57° 4' 59" N61° 30' 0" E59.6km (37.1 miles)
57° 24' 14" N60° 57' 47" E64.9km (40.4 miles)
56° 17' 39" N60° 17' 21" E65.1km (40.4 miles)
57° 25' 33" N61° 1' 27" E68.5km (42.6 miles)
57° 25' 33" N61° 1' 27" E68.5km (42.6 miles)
57° 25' 49" N61° 6' 7" E70.8km (44.0 miles)

Minerals recorded nearby (within 20 km)

ⓘ 'Amphibole Supergroup'

ⓘ Anorthite

ⓘ Chalcopyrite

ⓘ 'Chlorite Group'

ⓘ Cooperite

ⓘ Cuproiridsite

ⓘ Cuprorhodsite

ⓘ Erlichmanite

ⓘ 'Fayalite-Forsterite Series'

ⓘ Forsterite

ⓘ Geversite

ⓘ Heazlewoodite

ⓘ Hollingworthite

ⓘ Isoferroplatinum

ⓘ Kashinite

ⓘ Magnesiochromite

ⓘ Magnetite

ⓘ var: Titanium-bearing Magnetite

ⓘ Millerite

ⓘ Pargasite

ⓘ Pentlandite

ⓘ 'Prassoite'

ⓘ Pyrrhotite

ⓘ Sperrylite var: Platarsite

ⓘ Tetra-auricupride

ⓘ Tetraferroplatinum

ⓘ Tolovkite

ⓘ Tulameenite

Rock types recorded nearby (within 20 km)

ⓘ Chromitite

ⓘ Clinopyroxenite

Regions in mindat.org that contain this locality

RegionType

Regions in GADM Database

RegionGADM number
188.66.1915.2032.0.0
Russia, Sverdlovsk188.66.0.0.0.0
Russia, Sverdlovsk, Ekaterinburg gorsovet188.66.1915.0.0.0

Mindat Discussions

  • Places - European, Western and Northern Russia

YEKATERINBURG: FACTORIES, URAL SIGHTS, YELTSIN AND THE WHERE NICHOLAS II WAS KILLED

Sverdlovsk oblast.

Sverdlovsk Oblast is the largest region in the Urals; it lies in the foothills of mountains and contains a monument indicating the border between Europe and Asia. The region covers 194,800 square kilometers (75,200 square miles), is home to about 4.3 million people and has a population density of 22 people per square kilometer. About 83 percent of the population live in urban areas. Yekaterinburg is the capital and largest city, with 1.5 million people. For Russians, the Ural Mountains are closely associated with Pavel Bazhov's tales and known for folk crafts such as Kasli iron sculpture, Tagil painting, and copper embossing. Yekaterinburg is the birthplace of Russia’s iron and steel industry, taking advantage of the large iron deposits in the Ural mountains. The popular Silver Ring of the Urals tourist route starts here.

In the summer you can follow in the tracks of Yermak, climb relatively low Ural mountain peaks and look for boulders seemingly with human faces on them. You can head to the Gemstone Belt of the Ural mountains, which used to house emerald, amethyst and topaz mines. In the winter you can go ice fishing, ski and cross-country ski.

Sverdlovsk Oblast and Yekaterinburg are located near the center of Russia, at the crossroads between Europe and Asia and also the southern and northern parts of Russia. Winters are longer and colder than in western section of European Russia. Snowfalls can be heavy. Winter temperatures occasionally drop as low as - 40 degrees C (-40 degrees F) and the first snow usually falls in October. A heavy winter coat, long underwear and good boots are essential. Snow and ice make the sidewalks very slippery, so footwear with a good grip is important. Since the climate is very dry during the winter months, skin moisturizer plus lip balm are recommended. Be alert for mud on street surfaces when snow cover is melting (April-May). Patches of mud create slippery road conditions.

Yekaterinburg

Yekaterinburg (kilometer 1818 on the Trans-Siberian Railway) is the fourth largest city in Russia, with of 1.5 million and growth rate of about 12 percent, high for Russia. Located in the southern Ural mountains, it was founded by Peter the Great and named after his wife Catherine, it was used by the tsars as a summer retreat and is where tsar Nicholas II and his family were executed and President Boris Yeltsin lived most of his life and began his political career. The city is near the border between Europe and Asia.

Yekaterinburg (also spelled Ekaterinburg) is located on the eastern slope of the Ural Mountains in the headwaters of the Iset and Pyshma Rivers. The Iset runs through the city center. Three ponds — Verkh-Isetsky, Gorodskoy and Nizhne-Isetsky — were created on it. Yekaterinburg has traditionally been a city of mining and was once the center of the mining industry of the Urals and Siberia. Yekaterinburg remains a major center of the Russian armaments industry and is sometimes called the "Pittsburgh of Russia.". A few ornate, pastel mansions and wide boulevards are reminders of the tsarist era. The city is large enough that it has its own Metro system but is characterized mostly by blocky Soviet-era apartment buildings. The city has advanced under President Vladimir Putin and is now one of the fastest growing places in Russia, a country otherwise characterized by population declines

Yekaterinburg is technically an Asian city as it lies 32 kilometers east of the continental divide between Europe and Asia. The unofficial capital of the Urals, a key region in the Russian heartland, it is second only to Moscow in terms of industrial production and capital of Sverdlovsk oblast. Among the important industries are ferrous and non-ferrous metallurgy, machine building and metalworking, chemical and petrochemicals, construction materials and medical, light and food industries. On top of being home of numerous heavy industries and mining concerns, Yekaterinburg is also a major center for industrial research and development and power engineering as well as home to numerous institutes of higher education, technical training, and scientific research. In addition, Yekaterinburg is the largest railway junction in Russia: the Trans-Siberian Railway passes through it, the southern, northern, western and eastern routes merge in the city.

Accommodation: There are two good and affordable hotels — the 3-star Emerald and Parus hotels — located close to the city's most popular landmarks and main transport interchanges in the center of Yekaterinburg. Room prices start at RUB 1,800 per night.

History of Yekaterinburg

Yekaterinburg was founded in 1723 by Peter the Great and named after his wife Catherine I. It was used by the tsars as a summer retreat but was mainly developed as metalworking and manufacturing center to take advantage of the large deposits of iron and other minerals in the Ural mountains. It is best known to Americans as the place where the last Tsar and his family were murdered by the Bolsheviks in 1918 and near where American U-2 spy plane, piloted by Gary Powers, was shot down in 1960.

Peter the Great recognized the importance of the iron and copper-rich Urals region for Imperial Russia's industrial and military development. In November 1723, he ordered the construction of a fortress factory and an ironworks in the Iset River Valley, which required a dam for its operation. In its early years Yekaterinburg grew rich from gold and other minerals and later coal. The Yekaterinburg gold rush of 1745 created such a huge amount of wealth that one rich baron of that time hosted a wedding party that lasted a year. By the mid-18th century, metallurgical plants had sprung up across the Urals to cast cannons, swords, guns and other weapons to arm Russia’s expansionist ambitions. The Yekaterinburg mint produced most of Russia's coins. Explorations of the Trans-Baikal and Altai regions began here in the 18th century.

Iron, cast iron and copper were the main products. Even though Iron from the region went into the Eiffel Tower, the main plant in Yekaterinburg itself was shut down in 1808. The city still kept going through a mountain factory control system of the Urals. The first railway in the Urals was built here: in 1878, the Yekaterinburg-Perm railway branch connected the province's capital with the factories of the Middle Urals.

In the Soviet era the city was called Sverdlovsk (named after Yakov Sverdlov, the man who organized Nicholas II's execution). During the first five-year plans the city became industrial — old plants were reconstructed, new ones were built. The center of Yekaterinburg was formed to conform to the historical general plan of 1829 but was the layout was adjusted around plants and factories. In the Stalin era the city was a major gulag transhipment center. In World War II, many defense-related industries were moved here. It and the surrounding area were a center of the Soviet Union's military industrial complex. Soviet tanks, missiles and aircraft engines were made in the Urals. During the Cold War era, Yekaterinburg was a center of weapons-grade uranium enrichment and processing, warhead assembly and dismantlement. In 1979, 64 people died when anthrax leaked from a biological weapons facility. Yekaterinburg was a “Closed City” for 40 years during the Cold Soviet era and was not open to foreigners until 1991

In the early post-Soviet era, much like Pittsburgh in the 1970s, Yekaterinburg had a hard struggle d to cope with dramatic economic changes that have made its heavy industries uncompetitive on the world market. Huge defense plants struggled to survive and the city was notorious as an organized crime center in the 1990s, when its hometown boy Boris Yeltsin was President of Russia. By the 2000s, Yekaterinburg’s retail and service was taking off, the defense industry was reviving and it was attracting tech industries and investments related to the Urals’ natural resources. By the 2010s it was vying to host a world exhibition in 2020 (it lost, Dubai won) and it had McDonald’s, Subway, sushi restaurants, and Gucci, Chanel and Armani. There were Bentley and Ferrari dealerships but they closed down

Transportation in Yekaterinburg

Getting There: By Plane: Yekaterinburg is a three-hour flight from Moscow with prices starting at RUB 8,000, or a 3-hour flight from Saint Petersburg starting from RUB 9,422 (direct round-trip flight tickets for one adult passenger). There are also flights from Frankfurt, Istanbul, China and major cities in the former Soviet Union.

By Train: Yekaterinburg is a major stop on the Trans-Siberian Railway. Daily train service is available to Moscow and many other Russian cities.Yekaterinburg is a 32-hour train ride from Moscow (tickets RUB 8,380 and above) or a 36-hour train ride from Saint Petersburg (RUB 10,300 and above). The ticket prices are round trip for a berth in a sleeper compartment for one adult passenger). By Car: a car trip from Moscow to Yekateringburg is 1,787 kilometers long and takes about 18 hours. The road from Saint Petersburg is 2,294 kilometers and takes about 28 hours.

Regional Transport: The region's public transport includes buses and suburban electric trains. Regional trains provide transport to larger cities in the Ural region. Buses depart from Yekaterinburg’s two bus stations: the Southern Bus Station and the Northern Bus Station.

Regional Transport: According the to Association for Safe International Road Travel (ASIRT): “Public transportation is well developed. Overcrowding is common. Fares are low. Service is efficient. Buses are the main form of public transport. Tram network is extensive. Fares are reasonable; service is regular. Trams are heavily used by residents, overcrowding is common. Purchase ticket after boarding. Metro runs from city center to Uralmash, an industrial area south of the city. Metro ends near the main railway station. Fares are inexpensive.

“Traffic is congested in city center. Getting around by car can be difficult. Route taxis (minivans) provide the fastest transport. They generally run on specific routes, but do not have specific stops. Drivers stop where passengers request. Route taxis can be hailed. Travel by bus or trolleybuses may be slow in rush hour. Trams are less affected by traffic jams. Trolley buses (electric buses) cannot run when temperatures drop below freezing.”

Entertainment, Sports and Recreation in Yekaterinburg

The performing arts in Yekaterinburg are first rate. The city has an excellent symphony orchestra, opera and ballet theater, and many other performing arts venues. Tickets are inexpensive. The Yekaterinburg Opera and Ballet Theater is lavishly designed and richly decorated building in the city center of Yekaterinburg. The theater was established in 1912 and building was designed by architect Vladimir Semyonov and inspired by the Vienna Opera House and the Theater of Opera and Ballet in Odessa.

Vaynera Street is a pedestrian only shopping street in city center with restaurants, cafes and some bars. But otherwise Yekaterinburg's nightlife options are limited. There are a handful of expensive Western-style restaurants and bars, none of them that great. Nightclubs serve the city's nouveau riche clientele. Its casinos have closed down. Some of them had links with organized crime. New dance clubs have sprung up that are popular with Yekaterinburg's more affluent youth.

Yekaterinburg's most popular spectator sports are hockey, basketball, and soccer. There are stadiums and arenas that host all three that have fairly cheap tickets. There is an indoor water park and lots of parks and green spaces. The Urals have many lakes, forests and mountains are great for hiking, boating, berry and mushroom hunting, swimming and fishing. Winter sports include cross-country skiing and ice skating. Winter lasts about six months and there’s usually plenty of snow. The nearby Ural Mountains however are not very high and the downhill skiing opportunities are limited..

Sights in Yekaterinburg

Sights in Yekaterinburg include the Museum of City Architecture and Ural Industry, with an old water tower and mineral collection with emeralds. malachite, tourmaline, jasper and other precious stone; Geological Alley, a small park with labeled samples of minerals found in the Urals region; the Ural Geology Museum, which houses an extensive collection of stones, gold and gems from the Urals; a monument marking the border between Europe and Asia; a memorial for gulag victims; and a graveyard with outlandish memorials for slain mafia members.

The Military History Museum houses the remains of the U-2 spy plane shot down in 1960 and locally made tanks and rocket launchers. The fine arts museum contains paintings by some of Russia's 19th-century masters. Also worth a look are the History an Local Studies Museum; the Political History and Youth Museum; and the University and Arboretum. Old wooden houses can be seen around Zatoutstovsya ulitsa and ulitsa Belinskogo. Around the city are wooded parks, lakes and quarries used to harvest a variety of minerals. Weiner Street is the main street of Yekaterinburg. Along it are lovely sculptures and 19th century architecture. Take a walk around the unique Literary Quarter

Plotinka is a local meeting spot, where you will often find street musicians performing. Plotinka can be described as the center of the city's center. This is where Yekaterinburg holds its biggest events: festivals, seasonal fairs, regional holiday celebrations, carnivals and musical fountain shows. There are many museums and open-air exhibitions on Plotinka. Plotinka is named after an actual dam of the city pond located nearby (“plotinka” means “a small dam” in Russian).In November 1723, Peter the Great ordered the construction of an ironworks in the Iset River Valley, which required a dam for its operation. “Iset” can be translated from Finnish as “abundant with fish”. This name was given to the river by the Mansi — the Finno-Ugric people dwelling on the eastern slope of the Northern Urals.

Vysotsky and Iset are skyscrapers that are 188.3 meters and 209 meters high, respectively. Fifty-story-high Iset has been described by locals as the world’s northernmost skyscraper. Before the construction of Iset, Vysotsky was the tallest building of Yekaterinburg and Russia (excluding Moscow). A popular vote has decided to name the skyscraper after the famous Soviet songwriter, singer and actor Vladimir Vysotsky. and the building was opened on November 25, 2011. There is a lookout at the top of the building, and the Vysotsky museum on its second floor. The annual “Vysotsky climb” (1137 steps) is held there, with a prize of RUB 100,000. While Vysotsky serves as an office building, Iset, owned by the Ural Mining and Metallurgical Company, houses 225 premium residential apartments ranging from 80 to 490 square meters in size.

Boris Yeltsin Presidential Center

The Boris Yeltsin Presidential Center (in the city center: ul. Yeltsina, 3) is a non-governmental organization named after the first president of the Russian Federation. The Museum of the First President of Russia as well as his archives are located in the Center. There is also a library, educational and children's centers, and exposition halls. Yeltsin lived most of his life and began his political career in Yekaterinburg. He was born in Butka about 200 kilometers east of Yekaterinburg.

The core of the Center is the Museum. Modern multimedia technologies help animate the documents, photos from the archives, and artifacts. The Yeltsin Museum holds collections of: propaganda posters, leaflets, and photos of the first years of the Soviet regime; portraits and portrait sculptures of members of Politburo of the Central Committee of the Communist Party of various years; U.S.S.R. government bonds and other items of the Soviet era; a copy of “One Day in the Life of Ivan Denisovich” by Alexander Solzhenitsyn, published in the “Novy Mir” magazine (#11, 1962); perestroika-era editions of books by Alexander Solzhenitsyn, Vasily Grossman, and other authors; theater, concert, and cinema posters, programs, and tickets — in short, all of the artifacts of the perestroika era.

The Yeltsin Center opened in 2012. Inside you will also find an art gallery, a bookstore, a gift shop, a food court, concert stages and a theater. There are regular screenings of unique films that you will not find anywhere else. Also operating inside the center, is a scientific exploritorium for children. The center was designed by Boris Bernaskoni. Almost from the its very opening, the Yeltsin Center has been accused by members of different political entities of various ideological crimes. The museum is open Tuesday to Sunday, from 10:00am to 9:00pm.

Where Nicholas II was Executed

On July, 17, 1918, during this reign of terror of the Russian Civil War, former-tsar Nicholas II, his wife, five children (the 13-year-old Alexis, 22-year-old Olga, 19-year-old Maria and 17-year-old Anastasia)the family physician, the cook, maid, and valet were shot to death by a Red Army firing squad in the cellar of the house they were staying at in Yekaterinburg.

Ipatiev House (near Church on the Blood, Ulitsa Libknekhta) was a merchant's house where Nicholas II and his family were executed. The house was demolished in 1977, on the orders of an up and coming communist politician named Boris Yeltsin. Yeltsin later said that the destruction of the house was an "act of barbarism" and he had no choice because he had been ordered to do it by the Politburo,

The site is marked with s cross with the photos of the family members and cross bearing their names. A small wooden church was built at the site. It contains paintings of the family. For a while there were seven traditional wooden churches. Mass is given ay noon everyday in an open-air museum. The Church on the Blood — constructed to honor Nicholas II and his family — was built on the part of the site in 1991 and is now a major place of pilgrimage.

Nicholas and his family where killed during the Russian civil war. It is thought the Bolsheviks figured that Nicholas and his family gave the Whites figureheads to rally around and they were better of dead. Even though the death orders were signed Yakov Sverdlov, the assassination was personally ordered by Lenin, who wanted to get them out of sight and out of mind. Trotsky suggested a trial. Lenin nixed the idea, deciding something had to be done about the Romanovs before White troops approached Yekaterinburg. Trotsky later wrote: "The decision was not only expedient but necessary. The severity of he punishment showed everyone that we would continue to fight on mercilessly, stopping at nothing."

Ian Frazier wrote in The New Yorker: “Having read a lot about the end of Tsar Nicholas II and his family and servants, I wanted to see the place in Yekaterinburg where that event occurred. The gloomy quality of this quest depressed Sergei’s spirits, but he drove all over Yekaterinburg searching for the site nonetheless. Whenever he stopped and asked a pedestrian how to get to the house where Nicholas II was murdered, the reaction was a wince. Several people simply walked away. But eventually, after a lot of asking, Sergei found the location. It was on a low ridge near the edge of town, above railroad tracks and the Iset River. The house, known as the Ipatiev House, was no longer standing, and the basement where the actual killings happened had been filled in. I found the blankness of the place sinister and dizzying. It reminded me of an erasure done so determinedly that it had worn a hole through the page. [Source: Ian Frazier, The New Yorker, August 3, 2009, Frazier is author of “Travels in Siberia” (2010)]

“The street next to the site is called Karl Liebknecht Street. A building near where the house used to be had a large green advertisement that said, in English, “LG—Digitally Yours.” On an adjoining lot, a small chapel kept the memory of the Tsar and his family; beneath a pedestal holding an Orthodox cross, peonies and pansies grew. The inscription on the pedestal read, “We go down on our knees, Russia, at the foot of the tsarist cross.”

Books: The Romanovs: The Final Chapter by Robert K. Massie (Random House, 1995); The Fall of the Romanovs by Mark D. Steinberg and Vladimir Khrustalëv (Yale, 1995);

See Separate Article END OF NICHOLAS II factsanddetails.com

Execution of Nicholas II

According to Robert Massie K. Massie, author of Nicholas and Alexandra, Nicholas II and his family were awakened from their bedrooms around midnight and taken to the basement. They were told they were to going to take some photographs of them and were told to stand behind a row of chairs.

Suddenly, a group of 11 Russians and Latvians, each with a revolver, burst into the room with orders to kill a specific person. Yakob Yurovsky, a member of the Soviet executive committee, reportedly shouted "your relatives are continuing to attack the Soviet Union.” After firing, bullets bouncing off gemstones hidden in the corsets of Alexandra and her daughters ricocheted around the room like "a shower of hail," the soldiers said. Those that were still breathing were killed with point black shots to the head.

The three sisters and the maid survived the first round thanks to their gems. They were pressed up against a wall and killed with a second round of bullets. The maid was the only one that survived. She was pursued by the executioners who stabbed her more than 30 times with their bayonets. The still writhing body of Alexis was made still by a kick to the head and two bullets in the ear delivered by Yurovsky himself.

Yurovsky wrote: "When the party entered I told the Romanovs that in view of the fact their relatives continued their offensive against Soviet Russia, the Executive Committee of the Urals Soviet had decided to shoot them. Nicholas turned his back to the detachment and faced his family. Then, as if collecting himself, he turned around, asking, 'What? What?'"

"[I] ordered the detachment to prepare. Its members had been previously instructed whom to shoot and to am directly at the heart to avoid much blood and to end more quickly. Nicholas said no more. he turned again to his family. The others shouted some incoherent exclamations. All this lasted a few seconds. Then commenced the shooting, which went on for two or three minutes. [I] killed Nicholas on the spot."

Nicholas II’s Initial Burial Site in Yekaterinburg

Ganina Yama Monastery (near the village of Koptyaki, 15 kilometers northwest of Yekaterinburg) stands near the three-meter-deep pit where some the remains of Nicholas II and his family were initially buried. The second burial site — where most of the remains were — is in a field known as Porosyonkov (56.9113628°N 60.4954326°E), seven kilometers from Ganina Yama.

On visiting Ganina Yama Monastery, one person posted in Trip Advisor: “We visited this set of churches in a pretty park with Konstantin from Ekaterinburg Guide Centre. He really brought it to life with his extensive knowledge of the history of the events surrounding their terrible end. The story is so moving so unless you speak Russian, it is best to come here with a guide or else you will have no idea of what is what.”

In 1991, the acid-burned remains of Nicholas II and his family were exhumed from a shallow roadside mass grave in a swampy area 12 miles northwest of Yekaterinburg. The remains had been found in 1979 by geologist and amateur archeologist Alexander Avdonin, who kept the location secret out of fear that they would be destroyed by Soviet authorities. The location was disclosed to a magazine by one his fellow discovers.

The original plan was to throw the Romanovs down a mine shaft and disposes of their remains with acid. They were thrown in a mine with some grenades but the mine didn't collapse. They were then carried by horse cart. The vats of acid fell off and broke. When the carriage carrying the bodies broke down it was decided the bury the bodies then and there. The remaining acid was poured on the bones, but most of it was soaked up the ground and the bones largely survived.

After this their pulses were then checked, their faces were crushed to make them unrecognizable and the bodies were wrapped in bed sheets loaded onto a truck. The "whole procedure," Yurovsky said took 20 minutes. One soldiers later bragged than he could "die in peace because he had squeezed the Empress's -------."

The bodies were taken to a forest and stripped, burned with acid and gasoline, and thrown into abandoned mine shafts and buried under railroad ties near a country road near the village of Koptyaki. "The bodies were put in the hole," Yurovsky wrote, "and the faces and all the bodies, generally doused with sulfuric acid, both so they couldn't be recognized and prevent a stink from them rotting...We scattered it with branches and lime, put boards on top and drove over it several times—no traces of the hole remained.

Shortly afterwards, the government in Moscow announced that Nicholas II had been shot because of "a counterrevolutionary conspiracy." There was no immediate word on the other members of the family which gave rise to rumors that other members of the family had escaped. Yekaterinburg was renamed Sverdlov in honor of the man who signed the death orders.

For seven years the remains of Nicholas II, Alexandra, three of their daughters and four servants were stored in polyethylene bags on shelves in the old criminal morgue in Yekaterunburg. On July 17, 1998, Nicholas II and his family and servants who were murdered with him were buried Peter and Paul Fortress in St. Petersburg along with the other Romanov tsars, who have been buried there starting with Peter the Great. Nicholas II had a side chapel built for himself at the fortress in 1913 but was buried in a new crypt.

Near Yekaterinburg

Factory-Museum of Iron and Steel Metallurgy (in Niznhy Tagil 80 kilometers north of Yekaterinburg) a museum with old mining equipment made at the site of huge abandoned iron and steel factory. Officially known as the Factory-Museum of the History of the Development of Iron and Steel Metallurgy, it covers an area of 30 hectares and contains a factory founded by the Demidov family in 1725 that specialized mainly in the production of high-quality cast iron and steel. Later, the foundry was renamed after Valerian Kuybyshev, a prominent figure of the Communist Party.

The first Russian factory museum, the unusual museum demonstrates all stages of metallurgy and metal working. There is even a blast furnace and an open-hearth furnace. The display of factory equipment includes bridge crane from 1892) and rolling stock equipment from the 19th-20th centuries. In Niznhy Tagil contains some huge blocks of malachite and

Nizhnyaya Sinyachikha (180 kilometers east-northeast of Yekaterinburg) has an open air architecture museum with log buildings, a stone church and other pre-revolutionary architecture. The village is the creation of Ivan Samoilov, a local activist who loved his village so much he dedicated 40 years of his life to recreating it as the open-air museum of wooden architecture.

The stone Savior Church, a good example of Siberian baroque architecture. The interior and exterior of the church are exhibition spaces of design. The houses are very colorful. In tsarist times, rich villagers hired serfs to paint the walls of their wooden izbas (houses) bright colors. Old neglected buildings from the 17th to 19th centuries have been brought to Nizhnyaya Sinyachikha from all over the Urals. You will see the interior design of the houses and hear stories about traditions and customs of the Ural farmers.

Verkhoturye (330 kilometers road from Yekaterinburg) is the home a 400-year-old monastery that served as 16th century capital of the Urals. Verkhoturye is a small town on the Tura River knows as the Jerusalem of the Urals for its many holy places, churches and monasteries. The town's main landmark is its Kremlin — the smallest in Russia. Pilgrims visit the St. Nicholas Monastery to see the remains of St. Simeon of Verkhoturye, the patron saint of fishermen.

Ural Mountains

Ural Mountains are the traditional dividing line between Europe and Asia and have been a crossroads of Russian history. Stretching from Kazakhstan to the fringes of the Arctic Kara Sea, the Urals lie almost exactly along the 60 degree meridian of longitude and extend for about 2,000 kilometers (1,300 miles) from north to south and varies in width from about 50 kilometers (30 miles) in the north and 160 kilometers (100 miles) the south. At kilometers 1777 on the Trans-Siberian Railway there is white obelisk with "Europe" carved in Russian on one side and "Asia" carved on the other.

The eastern side of the Urals contains a lot of granite and igneous rock. The western side is primarily sandstone and limestones. A number of precious stones can be found in the southern part of the Urals, including emeralds. malachite, tourmaline, jasper and aquamarines. The highest peaks are in the north. Mount Narodnaya is the highest of all but is only 1884 meters (6,184 feet) high. The northern Urals are covered in thick forests and home to relatively few people.

Like the Appalachian Mountains in the eastern United States, the Urals are very old mountains — with rocks and sediments that are hundreds of millions years old — that were one much taller than they are now and have been steadily eroded down over millions of years by weather and other natural processes to their current size. According to Encyclopedia Britannica: “The rock composition helps shape the topography: the high ranges and low, broad-topped ridges consist of quartzites, schists, and gabbro, all weather-resistant. Buttes are frequent, and there are north–south troughs of limestone, nearly all containing river valleys. Karst topography is highly developed on the western slopes of the Urals, with many caves, basins, and underground streams. The eastern slopes, on the other hand, have fewer karst formations; instead, rocky outliers rise above the flattened surfaces. Broad foothills, reduced to peneplain, adjoin the Central and Southern Urals on the east.

“The Urals date from the structural upheavals of the Hercynian orogeny (about 250 million years ago). About 280 million years ago there arose a high mountainous region, which was eroded to a peneplain. Alpine folding resulted in new mountains, the most marked upheaval being that of the Nether-Polar Urals...The western slope of the Urals is composed of middle Paleozoic sedimentary rocks (sandstones and limestones) that are about 350 million years old. In many places it descends in terraces to the Cis-Ural depression (west of the Urals), to which much of the eroded matter was carried during the late Paleozoic (about 300 million years ago). Found there are widespread karst (a starkly eroded limestone region) and gypsum, with large caverns and subterranean streams. On the eastern slope, volcanic layers alternate with sedimentary strata, all dating from middle Paleozoic times.”

Southern Urals

The southern Urals are characterized by grassy slopes and fertile valleys. The middle Urals are a rolling platform that barely rises above 300 meters (1,000 feet). This region is rich in minerals and has been heavily industrialized. This is where you can find Yekaterinburg (formally Sverdlovsk), the largest city in the Urals.

Most of the Southern Urals are is covered with forests, with 50 percent of that pine-woods, 44 percent birch woods, and the rest are deciduous aspen and alder forests. In the north, typical taiga forests are the norm. There are patches of herbal-poaceous steppes, northem sphagnous marshes and bushy steppes, light birch forests and shady riparian forests, tall-grass mountainous meadows, lowland ling marshes and stony placers with lichen stains. In some places there are no large areas of homogeneous forests, rather they are forests with numerous glades and meadows of different size.

In the Ilmensky Mountains Reserve in the Southern Urals, scientists counted 927 vascular plants (50 relicts, 23 endemic species), about 140 moss species, 483 algae species and 566 mushroom species. Among the species included into the Red Book of Russia are feather grass, downy-leaved feather grass, Zalessky feather grass, moccasin flower, ladies'-slipper, neottianthe cucullata, Baltic orchis, fen orchis, helmeted orchis, dark-winged orchis, Gelma sandwart, Krasheninnikov sandwart, Clare astragalus.

The fauna of the vertebrate animals in the Reserve includes 19 fish, 5 amphibian and 5 reptile. Among the 48 mammal species are elks, roe deer, boars, foxes, wolves, lynxes, badgers, common weasels, least weasels, forest ferrets, Siberian striped weasel, common marten, American mink. Squirrels, beavers, muskrats, hares, dibblers, moles, hedgehogs, voles are quite common, as well as chiropterans: pond bat, water bat, Brandt's bat, whiskered bat, northern bat, long-eared bat, parti-coloured bat, Nathusius' pipistrelle. The 174 bird bird species include white-tailed eagles, honey hawks, boreal owls, gnome owls, hawk owls, tawny owls, common scoters, cuckoos, wookcocks, common grouses, wood grouses, hazel grouses, common partridges, shrikes, goldenmountain thrushes, black- throated loons and others.

Activities and Places in the Ural Mountains

The Urals possess beautiful natural scenery that can be accessed from Yekaterinburg with a rent-a-car, hired taxi and tour. Travel agencies arrange rafting, kayaking and hiking trips. Hikes are available in the taiga forest and the Urals. Trips often include walks through the taiga to small lakes and hikes into the mountains and excursions to collect mushrooms and berries and climb in underground caves. Mellow rafting is offered in a relatively calm six kilometer section of the River Serga. In the winter visitor can enjoy cross-mountains skiing, downhill skiing, ice fishing, dog sledding, snow-shoeing and winter hiking through the forest to a cave covered with ice crystals.

Lake Shartash (10 kilometers from Yekaterinburg) is where the first Ural gold was found, setting in motion the Yekaterinburg gold rush of 1745, which created so much wealth one rich baron of that time hosted a wedding party that lasted a year. The area around Shartash Lake is a favorite picnic and barbecue spot of the locals. Getting There: by bus route No. 50, 054 or 54, with a transfer to suburban commuter bus route No. 112, 120 or 121 (the whole trip takes about an hour), or by car (10 kilometers drive from the city center, 40 minutes).

Revun Rapids (90 kilometers road from Yekaterinburg near Beklenishcheva village) is a popular white water rafting places On the nearby cliffs you can see the remains of a mysterious petroglyph from the Paleolithic period. Along the steep banks, you may notice the dark entrance of Smolinskaya Cave. There are legends of a sorceress who lived in there. The rocks at the riverside are suited for competitive rock climbers and beginners. Climbing hooks and rings are hammered into rocks. The most fun rafting is generally in May and June.

Olenii Ruchii National Park (100 kilometers west of Yekaterinburg) is the most popular nature park in Sverdlovsk Oblast and popular weekend getaway for Yekaterinburg residents. Visitors are attracted by the beautiful forests, the crystal clear Serga River and picturesque rocks caves. There are some easy hiking routes: the six-kilometer Lesser Ring and the 15-kilometer Greater Ring. Another route extends for 18 km and passes by the Mitkinsky Mine, which operated in the 18th-19th centuries. It's a kind of an open-air museum — you can still view mining an enrichment equipment here. There is also a genuine beaver dam nearby.

Among the other attractions at Olenii Ruchii are Druzhba (Friendship) Cave, with passages that extend for about 500 meters; Dyrovaty Kamen (Holed Stone), created over time by water of Serga River eroding rock; and Utoplennik (Drowned Man), where you can see “The Angel of Sole Hope”., created by the Swedish artist Lehna Edwall, who has placed seven angels figures in different parts of the world to “embrace the planet, protecting it from fear, despair, and disasters.”

Image Sources: Wikimedia Commons

Text Sources: Federal Agency for Tourism of the Russian Federation (official Russia tourism website russiatourism.ru ), Russian government websites, UNESCO, Wikipedia, Lonely Planet guides, New York Times, Washington Post, Los Angeles Times, National Geographic, The New Yorker, Bloomberg, Reuters, Associated Press, AFP, Yomiuri Shimbun and various books and other publications.

Updated in September 2020

  •  Facebook
  •  Twitter
  •  Google+
  •  e-mail

 Page Top

This site contains copyrighted material the use of which has not always been authorized by the copyright owner. Such material is made available in an effort to advance understanding of country or topic discussed in the article. This constitutes 'fair use' of any such copyrighted material as provided for in section 107 of the US Copyright Law. In accordance with Title 17 U.S.C. Section 107, the material on this site is distributed without profit. If you wish to use copyrighted material from this site for purposes of your own that go beyond 'fair use', you must obtain permission from the copyright owner. If you are the copyright owner and would like this content removed from factsanddetails.com, please contact me.

IMAGES

  1. Confocal Laser Scanning Microscopy in 3 Simple Steps

    laser scanning confocal microscopy experiments

  2. Laser scanning confocal microscopy showing the results of...

    laser scanning confocal microscopy experiments

  3. Representations of confocal laser scanning microscopy to visualize

    laser scanning confocal microscopy experiments

  4. High Resolution Confocal Laser Scanning Microscopy

    laser scanning confocal microscopy experiments

  5. Carl Zeiss 710 Spectral Confocal Microscope

    laser scanning confocal microscopy experiments

  6. Confocal Laser Scanning Microscopy

    laser scanning confocal microscopy experiments

VIDEO

  1. ZEISS LSM 800: Drawing the ZEISS logo with "Interactive Bleaching" software feature

  2. The Confocal Microscope: A Brief Introduction Pt 1 🔬 #shorts

  3. Confocal Laser Scanning Microscopy

  4. Confocal Scanning Laser Ophthalmoscopy (CSLO) AK Khurana Optical Instruments and Techniques

  5. ZEISS Quasar Detector: Four-color live imaging of organelles in Marchantia polymorpha thallus cells

  6. A revolutionary 3D-microscope

COMMENTS

  1. Confocal Microscopy: Principles and Modern Practices

    As the name implies, in a laser scanning confocal microscope (LSCM), a laser beam is swept over the sample by means of scanning galvanometer mirrors. ... It is crucial in a confocal microscopy experiment to choose the correct technique, objective, fluorophores, mounting medium, and optical components to achieve the best images. Table 1 ...

  2. Tutorial: guidance for quantitative confocal microscopy

    Figure 2a shows a schematic of the main components and the lightpath in the classic confocal laser-scanning microscope (CLSM). A laser beam is focused into a specimen, where it excites fluorescent ...

  3. PDF Confocal Laser Scanning Microscopy

    The prototype of the LSM 44 series is now on display in the Deutsches Museum in Munich. The LSM 10 - a confocal system with two fluorescence channels. The LSM 310 combines confocal laser scanning microscopy with state-of- the-art computer technology. The LSM 410 is the first inverted micro scope of the LSM family.

  4. Advanced Microscopy: Laser Scanning Confocal Microscopy

    This chapter introduces the reader to the methodology of setting up basic experiments for use with a laser scanning confocal microscope. ... Isshiki, M., Sawada, H., and Segawa, A. (2001) Quantitative comparison of anti-fading mounting media for confocal laser scanning microscopy, J Histochem Cytochem 49, 305-312. Harris, J., Schwinn, N ...

  5. PDF Principles and practices of laser scanning confocal microscopy

    The laser scanning confocal microscope (LSCM) is an essential tool for many biomedical imaging appli-cations at the level of the light microscope. The basic principles of confocal microscopy and the evolution of the LSCM into today's sophisticated instruments are outlined. The major imaging modes of the LSCM are introduced including single ...

  6. Seeing beyond the limit: A guide to choosing the right super-resolution

    STED is a confocal laser scanning-based technique, with super-resolution achieved through the addition of high-powered torus-shaped STED lasers. The STED lasers are aligned with the excitation beam and deplete the emission of fluorescent molecules in the overlapping region (Fig. 1 Ai) (8, 9). This depletion is achieved through interruption of ...

  7. Laser Scanning Confocal Microscopy: History ...

    The laser scanning confocal microscope continues to be chosen for most routine work although a number of instruments have been developed for more specific applications. Significant improvements have been made to all areas of the confocal approach, not only to the instruments themselves, but also to the protocols of specimen preparation, to the ...

  8. Laser Scanning Confocal Microscopy

    Laser scanning confocal microscopes employ a pair of pinhole apertures to limit the specimen focal plane to a confined volume approximately a micron in size. Relatively thick specimens can be imaged in successive volumes by acquiring a series of sections along the optical (z) axis of the microscope. This tutorial explores imaging of specimens ...

  9. Laser Scanning Confocal Microscopy

    Choosing Fluorophore Combinations for Confocal Microscopy - In planning multiple label fluorescence staining protocols for widefield and laser scanning confocal fluorescence microscopy experiments, the judicious choice of probes is paramount in obtaining the best target signal while simultaneously minimizing bleed-through artifacts. This ...

  10. Laser Scanning Confocal Microscopy: History, Applications, and Related

    2.1 Marvin Minsky's Microscope. The development of confocal microscopes was, and continues to be, driven by the desire to image biological events as they occur in vivo. The invention of the confocal microscope is attributed to Marvin Minsky who built a working scanning optical microscope in 1955 with the goal of imaging neural networks in unstained preparations of living brains.

  11. Using laser scanning confocal microscopy combined with saturated oil

    Accordingly, the laser scanning confocal microscopy (LSCM) combined with saturated oil experiment is used to quantitatively characterize the pseudo in-situ occurrence characteristics of light and heavy components of shale oil in sub-micron scale in Fengcheng Formation of Mahu Sag.

  12. Specimen Preparation and Imaging

    Figure 5 - Objectives for Laser Scanning Confocal Microscopy. During scanning of the specimen an image averaging routine is usually employed to reduce random noise from the detection system and to enhance the constant (nonrandom) features in the image. ... When this occurs with confocal imaging experiments, there may be an initial reflex to ...

  13. Laser-scanning confocal vibrometer microscope: Theory and experiments

    Therefore, the laser-scanning confocal laser-Doppler vibrometer microscope (CVM) can also be used as common confocal microscope to image and measure geometries of three-dimensional structures. The focus of this article is on the analysis of the transverse resolution, which cannot be adopted from other techniques.

  14. Systematic evaluation of FRAP experiments performed in a confocal laser

    Abstract. The diffusion coefficient as well as the dimensionality of the diffusion process can be determined by straightforward and facile data analysis, when fluorescence recovery after photobleaching (FRAP) is measured as a function of time and space by means of confocal laser scanning microscopy. Experiments representing one-dimensional ...

  15. Biofilms in Space—Confocal Laser Scanning Microscopy Helps NASA

    Preparing for the Spaceflight Experiment with Confocal Microscopy To prep for the spaceflight experiment, Bob and his team used an Olympus FV1000 confocal laser scanning microscope and a 60X water immersion objective at Texas State to capture 3D images of bacterial biofilms on flight hardware relevant to the ISS water system.

  16. FV4000

    The FV4000 confocal microscope uses our advanced, silicon-based SilVIR™ detector that makes it easier than ever to acquire precise, reproducible data. SilVIR Next-Generation Detector Technology. The SilVIR detector combines two advanced technologies—a silicon photomultiplier (SiPM) and our patented* fast signal processing design.

  17. Fluorescence Microscopy: A Concise Guide to Current Imaging Methods

    Modern Confocal Microscopy. The laser scanning confocal microscope (LSCM) remains a key piece of equipment in most imaging laboratories. Most modern LSCM systems offer a variety of advantages and are equipped with software to perform complex 3D (z-stack), 4D (z-stack over time), or even 5D (z-stack over time including spectral imaging) experiments.

  18. A cuproptosis nanocapsule for cancer radiotherapy

    The 4T1 cells were seeded on confocal laser scanning microscopy dishes at 10 × 10 4 cells per well. After 12 h, PBS or PWCu (10 μM) was added to the wells and incubated for 6 h.

  19. ZEISS Microscopy Online Campus

    Systematic evaluation of FRAP experiments performed in a confocal laser scanning microscope Journal of Microscopy 220: 20-30 (2005). A systematic analysis of FRAP measurements using highly defined model systems in confocal microscopy. ... 14-28 (2003). The authors examine the experimental design, mathematical modeling, and analysis of FRAP ...

  20. Principles and practices of laser scanning confocal microscopy

    The laser scanning confocal microscope (LSCM) is an essential tool for many biomedical imaging applications at the level of the light microscope. The basic principles of confocal microscopy and the evolution of the LSCM into today's sophisticated instruments are outlined. The major imaging modes of the LSCM are introduced including single ...

  21. Impact of different activation procedures on sodium hypochlorite

    Background Infected dentinal tubules are a possible source of bacteria that are responsible for the failure of root canal treatment. Therefore, disinfection of dentinal tubules by increasing the penetration of the irrigation solution is important for success in retreatment cases. This study utilized confocal laser scanning microscopy (CLSM) to assess and compare the impact of XPR, ultrasonic ...

  22. Surface integrity and high-cycle fatigue life of direct laser metal

    The authors would like to acknowledge the use of the optical microscope, scanning electron microscope, and micro-indenter provided by Microscopy Australia, a facility that is co-funded by the University of South Australia, the State Government of South Australia and the Australian Federal Government, and the use of laser confocal microscopy ...

  23. Deposition of PbS Films by Pyrolysis of Atomized Solutions of Lead

    Atomic force microscopy results show that the roughness and rms-surface slope of the samples obtained from bath-I (PbS-I) were higher than those of bath-II (PbS-II) samples.

  24. Yekaterinburg, Sverdlovskaya Oblast', Russia

    Locality Latitude Longitude Distance Bearing; Uktus complex, Yekaterinburg, Sverdlovsk Oblast, Russia: 56° 46' 18" N: 60° 38' 30" E: 9.1km (5.6 miles) Berezovsk deposit, Beryozovsky, Sverdlovsk Oblast, Russia

  25. YEKATERINBURG: FACTORIES, URAL SIGHTS, YELTSIN AND ...

    SVERDLOVSK OBLAST. Sverdlovsk Oblast is the largest region in the Urals; it lies in the foothills of mountains and contains a monument indicating the border between Europe and Asia.

  26. Yekaterinburg & Sverdlovsk Oblast

    Sverdlovvsk Oblast, like most of the Urals region, possesses abundant natural resources. It is one of Russia's leaders in mineral extraction. Sverdlovsk produces 70% of Russia's bauxite, 60% of asbestos, 23% of iron, 97% of vanadium, 6% of copper and 2% of nickel. Forests cover 65% of the oblast. It also produces 6% of Russia's timber and ...