Summary

鼠标中枢神经系统的光透明使用被动CLARITY

Published: June 30, 2016
doi:

Summary

Optical clearing techniques are revolutionizing the way tissues are visualized. In this report we describe modifications of the original Clear Lipid-exchanged Acrylamide-hybridized Rigid Imaging-compatible Tissue-hYdrogel (CLARITY) protocol that yields more consistent and less expensive results.

Abstract

Traditionally, tissue visualization has required that the tissue of interest be serially sectioned and imaged, subjecting each tissue section to unique non-linear deformations, dramatically hampering one’s ability to evaluate cellular morphology, distribution and connectivity in the central nervous system (CNS). However, optical clearing techniques are changing the way tissues are visualized. These approaches permit one to probe deeply into intact organ preparations, providing tremendous insight into the structural organization of tissues in health and disease. Techniques such as Clear Lipid-exchanged Acrylamide-hybridized Rigid Imaging-compatible Tissue-hYdrogel (CLARITY) achieve this goal by providing a matrix that binds important biomolecules while permitting light-scattering lipids to freely diffuse out. Lipid removal, followed by refractive index matching, renders the tissue transparent and readily imaged in 3 dimensions (3D). Nevertheless, the electrophoretic tissue clearing (ETC) used in the original CLARITY protocol can be challenging to implement successfully and the use of a proprietary refraction index matching solution makes it expensive to use the technique routinely. This report demonstrates the implementation of a simple and inexpensive optical clearing protocol that combines passive CLARITY for improved tissue integrity and 2,2′-thiodiethanol (TDE), a previously described refractive index matching solution.

Introduction

The ability to image complete neuroanatomical structures is immensely valuable for understanding the brain in health and disease. Traditionally, 3D imaging has required tissue sectioning to provide the axial resolution and to visualize deep anatomical structures. This approach can produce high-resolution data sets, but requires sophisticated image reconstruction techniques and is very labor intensive. As a result, it has been limited to the imaging of small volumes of tissue1-3. Optical sectioning, on the other hand, is well suited for the creation of high-resolution 3D images of fluorescently labeled tissues. Since optical sectioning is inherently three dimensional, it does not require extensive computation to produce a 3D image volume. However, light scattering and tissue opacity limit the depth at which tissues can be optically sectioned. The depth of imaging is limited to about 150 µm in laser scanning confocal microscopy and to less than 800 µm using two-photon excitation microscopy4-8.

In order to overcome these limitations, several optical clearing techniques have been recently developed and then further refined to permit the deep microscopic imaging of intact tissues. The use of benzyl alcohol and benzyl benzoate (BABB) to render fixed tissues transparent was among one of the earliest approaches9. However, this approach was limited by the quenching of fluorescence in the samples and incomplete clearing of highly myelinated structures10. Refinements of this technique, such as 3D Imaging of Solvent Cleared Organs (3DISCO), have led to very rapid and complete tissue clearing, but still suffer from rapid loss of fluorescent signals, especially yellow fluorescent protein (YFP)10. Water-based clearing solutions, such as Sca/e11 and SeeDB12, preserve fluorescent signals, but do not completely clear highly myelinated tissues. Clear, Unobstructed Brain Imaging Cocktails and Computational analysis (CUBIC) is a promising new optical clearing technology that appears to overcome the limitations of previously developed water-based clearing solutions13. In contrast with other optical clearing techniques, Clear Lipid-exchanged Acrylamide-hybridized Rigid Imaging-compatible Tissue-hYdrogel (CLARITY)14 embeds the brain in a porous matrix that provides structural integrity to proteins, nucleic acids and small molecules, while leaving lipids unbound. The lipids are then removed electrophoretically, culminating in an optically cleared brain that can be easily visualized and equally easily probed using commonly available techniques.

Electrophoretic tissue clearing (ETC) is used to remove lipids from hydrogel embedded tissues in CLARITY, however, ETC can be difficult to implement consistently and tissues subject to electrophoresis can exhibit tissue distortion, browning, loss of fluorescence and antibody reactivity. Passive lipid clearing avoids these limitations, but requires increased clearing times15-17. Passive clearing also permits the clearing of large numbers of samples in parallel, since it is not limited by the number of ETC apparatuses available. Decreasing the concentration of paraformaldehyde and excluding bis-acrylamide from the hydrogel have led to great increases in the speed of clearing at the expense of greater tissue expansion16. Less expensive alternatives to the refractive index matching solution used in the original CLARITY technique have been developed17-19. 2,2′-thiodiethanol (TDE) is an inexpensive and rapid brain clearing agent that reverses some of the tissue expansion that occurs during lipid removal18. This report incorporates passive clearing of hydrogel embedded tissue and the use of TDE as a refractive index matching solution to the original CLARITY technique to produce a highly reproducible and inexpensive protocol for the optical clearing of the mouse central nervous system.

Protocol

所有实验均按照机构动物护理和使用委员会(IACUC)的指导方针进行。 THY1-YFP,PLP-EGFP和PV-TdTomato小鼠用于这些实验,但表达的荧光蛋白的任何小鼠可以成功地清除并成像。 1.组织准备注意:多聚甲醛(PFA)和丙烯酰胺是有毒的和刺激物。执行利用这些试剂在通风橱,并配备适当的个人防护装备(实验室外套,手套和护目镜)的实验。 牺牲安乐死?…

Representative Results

可视化脑的结构组织为理解形态学和连通如何影响在健康和疾病的脑功能的关键。光透明的技术使得有可能在三维图像的细胞群在完整组织,使我们能够研究形态学和连接内聚。 小鼠表达荧光蛋白通过特异于亚群的启动子驱动的细胞允许一个梳理开神经连接的大脑中的复杂的图案。 THY1-YFP的转基因小鼠进行了广泛的光透明文?…

Discussion

水凝胶包埋组织(无源净度)的被动结算是用于清除大块组织的一个简单和廉价的方法。这种方法不要求专用设备,并且可以很容易地在一个温度控制的振荡器进行。在几个星期的跨度,即使是大的,高度髓组织,例如整个脑或脊髓,将成为透明,适用于显微镜。尽管这份报告集中在CNS组织中的清除,被动净度可应用于任何组织。

的光透明的技术的相对优点和弱点已在文献<s…

Divulgations

The authors have nothing to disclose.

Acknowledgements

这项工作是慷慨神经疾病和中风(NIH / NINDS)资助1R01NS086981和康拉德希尔顿N.基金会健康/国立全国学院的支持。我们感谢洛朗邦托利拉博士和马修SCHIBLER博士及其与激光共聚焦显微镜宝贵的援助。我们感谢那些给CLARITY论坛(http://forum.claritytechniques.org)作出了贡献。我们特别感谢卡尔戴瑟罗特博士打开了他的实验室,教这个迷人的技术。笔者是从脑成像医学研究组织,脑成像支持基金会皮尔森 – 洛夫莱斯基金会,基金会阿曼森,资本集团慈善基金会,威廉·M和琳达R. Dietel慈善基金,以及基金北极星的慷慨支持表示感谢。研究本出版物中报也部分由国家研究资源中心和民族的主任办公室的支持在奖项数量C06RR012169,C06RR015431和S10OD011939卫生研究院人。内容完全是作者的责任,并不一定代表美国国立卫生研究院的官方意见。

Materials

10X phosphate buffered saline (PBS) Fisher BP399-1 buffers
32% paraformaldehyde (PFA) Electron Microscopy Sciences 15714-5 perfusion and hydrogel
2,2'-thiodiethanol (TDE) Sigma-Aldrich 166782-500G refractive index matching solution
40% acrylamide Bio-Rad 161-0140 hydrogel
2% bis-acrylamide Bio-Rad 161-0142 hydrogel
VA-044 initiator Wako Pure Chemical Industries, Ltd. VA044 hydrogel
boric acid Sigma-Aldrich B7901 clearing buffer
sodium dodecyl sulfate (SDS) Fisher BP166-5 clearing buffer
sodium hydroxide Fisher 55255-1 buffers
sodium azide Sigma-Aldrich 52002-100G preservative
triton x-100 Sigma-Aldrich X100-500G buffers
heparin Sigma-Aldrich H3393-50KU perfusion
sodium nitrate Fisher BP360-500 perfusion
DAPI Molecular Probes D1306 nuclear stain
99.9% isoflurane Phoenix 57319-559-06 anesthetic
hydrocholoric acid Fisher A1445-500 buffers
glass bottom dish Willco HBSB-5040 Willco dishes
reusable adhesive Bostick 371351 Blu-Tack
silicon elastomer World Precision Instruments KWIK-CAST Kwik-Cast
lint-free wipe Kimberly-Clark 34120 KimWipe
mouse brain matrix Roboz SA-2175 sectioning tissue
matrix blades Roboz RS-9887 sectioning tissue
peristaltic pump Cole-Parmer 77122-24 pefusion
laser scanning confocal microscope Leica SP5 microscopy
imaging software Leica LAS AF microscopy

References

  1. Micheva, K. D., Smith, S. J. Array tomography: a new tool for imaging the molecular architecture and ultrastructure of neural circuits. Neuron. 55, 25-36 (2007).
  2. Kleinfeld, D., et al. Large-scale automated histology in the pursuit of connectomes. J Neurosci. 31, 16125-16138 (2011).
  3. MacKenzie-Graham, A., et al. A multimodal, multidimensional atlas of the C57BL/6J mouse brain. J Anat. 204, 93-102 (2004).
  4. Denk, W., Svoboda, K. Photon upmanship: why multiphoton imaging is more than a gimmick. Neuron. 18, 351-357 (1997).
  5. Denk, W., et al. Anatomical and functional imaging of neurons using 2-photon laser scanning microscopy. J Neurosci Methods. 54, 151-162 (1994).
  6. Zipfel, W. R., Williams, R. M., Webb, W. W. Nonlinear magic: multiphoton microscopy in the biosciences. Nat Biotechnol. 21, 1369-1377 (2003).
  7. Helmchen, F., Denk, W. Deep tissue two-photon microscopy. Nat Methods. 2, 932-940 (2005).
  8. Theer, P., Denk, W. On the fundamental imaging-depth limit in two-photon microscopy. J Opt Soc Am A Opt Image Sci. Vis. 23, 3139-3149 (2006).
  9. Dodt, H. U., et al. Ultramicroscopy: three-dimensional visualization of neuronal networks in the whole mouse brain. Nat Methods. 4, 331-336 (2007).
  10. Erturk, A., et al. Three-dimensional imaging of solvent-cleared organs using 3DISCO. Nat Protoc. 7, 1983-1995 (2012).
  11. Hama, H., et al. Scale: a chemical approach for fluorescence imaging and reconstruction of transparent mouse brain. Nat Neurosci. 14, 1481-1488 (2011).
  12. Ke, M. T., Fujimoto, S., Imai, T. SeeDB: a simple and morphology-preserving optical clearing agent for neuronal circuit reconstruction. Nat Neurosci. 16, 1154-1161 (2013).
  13. Susaki, E. A., et al. Whole-brain imaging with single-cell resolution using chemical cocktails and computational analysis. Cell. 157, 726-739 (2014).
  14. Chung, K., et al. Structural and molecular interrogation of intact biological systems. Nature. 497, 332-337 (2013).
  15. Spence, R. D., et al. Bringing CLARITY to gray matter atrophy. Neuroimage. 101, 625-632 (2014).
  16. Tomer, R., Ye, L., Hsueh, B., Deisseroth, K. Advanced CLARITY for rapid and high-resolution imaging of intact tissues. Nat Protoc. 9, 1682-1697 (2014).
  17. Yang, B., et al. Single-cell phenotyping within transparent intact tissue through whole-body clearing. Cell. 158, 945-958 (2014).
  18. Costantini, I., et al. A versatile clearing agent for multi-modal brain imaging. Sci Rep. 5, 9808 (2015).
  19. Zheng, H., Rinaman, L. Simplified CLARITY for visualizing immunofluorescence labeling in the developing rat brain. Brain Struct Funct. , (2015).
  20. Parra, S. G., et al. Multiphoton microscopy of cleared mouse brain expressing YFP. J Vis Exp. (67), e3848 (2012).
  21. Weinger, J. G., et al. Two-photon imaging of cellular dynamics in the mouse spinal cord. J Viz Exp. (96), e52580 (2015).
  22. Hama, H., et al. ScaleS: an optical clearing palette for biological imaging. Nat Neurosci. 18, 1518-1529 (2015).
  23. Treweek, J. B., et al. Whole-body tissue stabilization and selective extractions via tissue-hydrogel hybrids for high-resolution intact circuit mapping and phenotyping. Nat Protoc. 10, 1860-1896 (2015).
check_url/fr/54025?article_type=t

Play Video

Citer Cet Article
Roberts, D. G., Johnsonbaugh, H. B., Spence, R. D., MacKenzie-Graham, A. Optical Clearing of the Mouse Central Nervous System Using Passive CLARITY. J. Vis. Exp. (112), e54025, doi:10.3791/54025 (2016).

View Video