Expand this Topic clickable element to expand a topic
Skip to content
Optica Publishing Group

Light sheet-based fluorescence microscopy (LSFM) reduces phototoxic effects and provides new means for the modern life sciences

Not Accessible

Your library or personal account may give you access

Abstract

Most optical technologies are applied to flat, basically two-dimensional cellular systems. However, physiological meaningful information relies on the morphology, the mechanical properties and the biochemistry of a cell’s context. A cell requires the complex three-dimensional relationship to other cells. However, the observation of multi-cellular biological specimens remains a challenge. Specimens scatter and absorb light, thus, the delivery of the probing light and the collection of the signal light become inefficient; many endogenous biochemical compounds also absorb light and suffer degradation of some sort (photo-toxicity), which induces malfunction of a specimen. In conventional and confocal fluorescence microscopy, whenever a single plane, the entire specimen is illuminated. Recording stacks of images along the optical Z-axis thus illuminates the entire specimen once for each plane. Hence, cells are illuminated 10-20 and fish 100-300 times more often than they are observed. This can be avoided by changing the optical arrangement. The basic idea is to use light sheets, which are fed into the specimen from the side and overlap with the focal plane of a wide-field fluorescence microscope. In contrast to an epi-fluorescence arrangement, such an azimuthal fluorescence arrangement uses two independently operated lenses for illumination and detection. Optical sectioning and no photo-toxic damage or photo-bleaching outside a small volume close to the focal plane are intrinsic properties. Light sheet-based fluorescence microscopy (LSFM) takes advantage of modern camera technologies. LSFM can be operated with laser cutters and for fluorescence correlation spectroscopy. During the last few years, LSFM was used to record zebrafish development from the early 32-cell stage until late neurulation with sub-cellular resolution and short sampling periods (60-90 sec/stack). The recording speed was five 4-Megapixel large frames/sec with a dynamic range of 12-14 bit. We followed cell movements during gastrulation, revealed the development during cell migration processes and showed that an LSFM exposes an embryo to 200 times less energy than a conventional and 5,000 times less energy than a confocal fluorescence microscope. Most recently, we implemented incoherent structured illumination in our DSLM. The intensity modulated light sheets can be generated with dynamic frequencies and allow us to estimate the effect of the specimen on the image formation process at various depths in objects of different age.

© 2011 OSA/SPIE

PDF Article
More Like This
High resolution 3D imaging of primary and secondary tumor spheroids using multicolor multi-angle Light Sheet Fluorescence Microscopy (LSFM)

Stylianos E. Psycharakis, Evangelos Liapis, Athanasios Zacharopoulos, Mariam-Eleni Oraiopoulou, Chrysoula Aivalioti, Vangelis Sakkalis, Joseph Papamatheakis, Jorge Ripoll, and Giannis Zacharakis
11076_24 European Conference on Biomedical Optics (ECBO) 2019

The Zebrafish Digital Embryo: In Toto Reconstruction of Zebrafish Early Embryonic Development with Digital Scanned Laser Light Sheet Fluorescence Microscopy

Philipp J. Keller, Annette D. Schmidt, Joachim Wittbrodt, and Ernst H.K. Stelzer
7367_0G European Conference on Biomedical Optics (ECBO) 2009

3D fluorescence imaging of tumor cell spheroids

Verena Richter, Thomas Bruns, Petra Weber, Wolfgang S. L. Strauß, Rainer Wittig, and Herbert Schneckenburger
80890D European Conference on Biomedical Optics (ECBO) 2011

Select as filters


Select Topics Cancel
© Copyright 2024 | Optica Publishing Group. All rights reserved, including rights for text and data mining and training of artificial technologies or similar technologies.