Near field coherent anti stokes raman scattering (CARS) microscopy for bioimaging

135 398 0
Near field coherent anti stokes raman scattering (CARS) microscopy for bioimaging

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

Near-field Coherent Anti-Stokes Raman Scattering (CARS) Microscopy for Bioimaging LIN JIAN NATIONAL UNIVERSITY OF SINGAPORE 2012 NEAR-FIELD COHERENT ANTI-STOKES RAMAN SCATTERING (CARS) MICROSCOPY FOR BIOIMAGING LIN JIAN A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DEPARTMENT OF BIOENGINEERING NATIONAL UNIVERSITY OF SINGAPORE 2012 DECLARATION I hereby declare that the thesis is my original work and it has been written by me in its entirety. I have duly acknowledged all the sources of information which have been used in the thesis. This thesis has also not been submitted for any degree in any university previously. LIN JIAN 16 August 2012 Acknowledgements First and foremost, I want to thank my advisor Assistant Professor Huang Zhiwei, who provided me the opportunity to pursue the PhD degree in his group. I appreciate his professional and patient guidance, from the study of basic theory and the design of experiments to the development of new ideas, as well as the financial support throughout my four-year study. I want to acknowledge Dr. Wang Haifeng in Data Storage Institute of Singapore (DSI), who taught me the finite-difference time-domain method for simulating electromagnetic waves. I would like to thank Professor Colin Sheppard and Associate Professor Chen Nanguang, who gave me much valuable advice on my research work. I am grateful to Professor Hanry Yu and his group members, who taught me much knowledge on biology and the experimental skills in animal studies and sample preparations. I also appreciate the friendship and great support of my coworkers and group members. Special thanks go to Dr. Lu Fake for his selfless help to my experiments and constructive discussions. I would like to extend my thanks to other coworkers and group members in Optical Bioimaging Laboratory: Dr. Zheng Wei, Dr. Yuen Clement, Mo Jianhua, Teh Seng Knoon, Shao Xiaozhuo, Lin Kan and Shiyamala Duraipandian for their kind discussions, suggestions and guidance on my research work, and Dr. Lim Chin Seong in DSI for his kind help to coat gold on the aperture-less probes. Finally, I wish to thank my dear parents for their unconditional love and my wife, Dr. Xia Yijie, for her endless support and loving care. I Table of Contents Acknowledgements I Table of Contents . II Abstract . IV List of Figures . VI List of Abbreviations . VIII Chapter Introduction 1.1 Overview 1.2 Motivations 1.3 Research Objectives . 1.4 Thesis Organization . Chapter Literature Review . 2.1 Basic Theory 2.1.1 Raman scattering . 2.1.2 Fundamental theory of CARS . 2.2 Numerical simulation method for CARS microscopy . 12 2.2.1 FDTD method 12 2.2.2 Near-field CARS simulation using the FDTD method 17 2.2.3 Far-field CARS simulation 17 2.3 Instrumentations of CARS Microscopy . 18 2.3.1 Laser sources for CARS microscopy . 18 2.3.2 Laser scanning CARS microscope 19 2.3.3 Near-field CARS microscopy 20 2.3.4 Integrated CARS and multimodal nonlinear optical microscopy 24 2.4 Suppression of Nonresonant Background in CARS Microscopy 25 2.4.1 Backward (Epi-) detection CARS . 26 2.4.2 Focus-engineered CARS . 27 2.4.3 Polarization-sensitive CARS . 27 2.4.4 Time-resolved CARS . 29 2.4.5 Interferometric CARS . 29 2.5 CARS Applications in Life Sciences . 30 2.5.1 Cellular imaging 30 2.5.2 Tissue imaging . 31 Chapter Numerical Study of Near-field CARS 33 3.1 Effects of light polarization, scatterer sizes and orientations on near-field CARS . 34 3.1.1 Simulation method . 35 3.1.2   Influence   of   scatterers’   orientations   on   excitation   fields   and   near-field CARS signals . 37 3.1.3 Influence of the excitation light polarization on near-field CARS signals 40 3.1.4  Effect  of  the  scatterer’  s  size  on near-field CARS signals . 42 3.1.5 Summary . 45 II 3.2  Effects   of  scatterers’  sizes  on  near-field CARS under tightly focused radially and linearly polarized light excitation 47 3.2.1 Simulation method . 47 3.2.2 Results and discussion . 48 3.2.3 Summary . 53 Chapter Near-field CARS Imaging 55 4.1 Introduction 55 4.2 Sample Preparation 57 4.3 Experimental Setup 58 4.4 Fast Positioning of the Tip . 61 4.5 AFM and Aperture-less NSOM imaging . 62 4.6 Near-field CARS imaging 65 4.7 Summary 67 Chapter Annular aperture-detected CARS microscopy for high contrast vibrational imaging 69 5.1 Annular-aperture Detected Radially Polarized CARS . 69 5.1.1 Method . 69 5.1.2 Results and Discussions 71 5.1.3 Summary . 75 5.2 Annular-aperture Detected Linearly Polarized CARS . 75 5.2.1 Method . 75 5.2.2 Results and discussions . 76 5.2.3 Summary . 81 Chapter Assessment of liver Disease Using Integrated CARS and Multiphoton Microscopy 82 6.1 Introduction 82 6.2 Method . 84 6.2.1 Animal model and tissue preparations . 84 6.2.2 Histopathological method 85 6.2.3 Multimodal nonlinear optical microscopy . 85 6.2.4 Image acquisition and data processing 87 6.3 Results and Discussions . 89 6.3.1 Comparison between multimodal and histopathological images . 89 6.3.2 Qualitative assessment of liver diseases 90 6.3.3 Quantitative assessment of liver diseases 95 6.3.4 Considerations for in vivo applications of multimodal nonlinear optical microscopy 97 Chapter Conclusions and Future Directions 100 7.1 Conclusions 100 7.2 Future Directions . 103 List of Publications . 106 References 108 III Abstract Coherent anti-Stokes Raman scattering (CARS) microscopy is a nonlinear Raman imaging technique that has received great attention for biological and biomedical imaging due to its ability of real-time, nonperturbative chemical mapping of live unstained cells and tissue based on molecular vibrations. However, some challenges in CARS microscopy still remain unsolved for its wider biomedical applications. For instance, the strong nonresonant background in CARS imaging deteriorates the image contrast which ultimately limits the sensitivity of CARS technique in bioimaging. The spatial resolution of CARS is limited by the light diffraction (~ submicron scales in resolution), which is unsuited for imaging the inter-/intra- cellular fine structures at nanoscales. Hence, the main objectives of this thesis work are to explore the near-filed CARS microscopy technique for nanoimaging, and to develop new technique for effectively suppressing the nonresonant background for high contrast imaging in biomedical systems. To achieve these aims, we have employed the finite-difference time-domain (FDTD) technique as a numerical approach to studying the effects of different nanoparticle configurations and polarizations of excitation light on near-field CARS imaging. It was found that scatterers with diameters of less than three-eighths of the pump field wavelength (λp) are preferable to be oriented along the polarization direction of the excitation light fields due to the stronger electric field enhancement than that with other orientations, and the perpendicular polarization component of the induced near-field CARS radiations  from  scatterers’  smaller  than  half  a  wavelength  is   localized  within  a  spatial  dimension  of  λp/16 due to the light scattering by the sample, which may be useful for high sensitive and high contrast molecular imaging in cells with nanoscale resolutions. It was also found that the signal to background ratio of near-field radially polarized CARS (RP-CARS) is 4.5 times higher than near-field linearly polarized CARS (LP-CARS) with the presence of scatterers in water, while the full width at half maximum and the depth of focus of near-field RP-CARS are 23% narrower and 39% shorter than near-field LP-CARS. These results indicate the ability of RP-CARS for high-contrast and high-resolution nano-scale vibrational imaging. With the aid of theoretically modeling results, we have developed a radially polarized tip-enhanced near-field scanning CARS (TE-CARS) microscope for nano-scale vibrational imaging. Fast and precise positioning of the fiber tip at the focal region of the excitation light was realized based on the laser-scanning confocal IV imaging ability integrated in the system. Radially polarized light was used to improve the excitation efficiency and image contrast in cell imaging. We have also developed a unique annular-aperture detection scheme to effectively suppress the solvent background for high contrast CARS imaging. The results show that the resonant CARS signal to nonresonant background ratio varies with both the scatterers’  sizes  and  the  annular  aperture  diameters  used,  and  can  be  improved  by  20   folds in LP-CARS and 115 folds in RP-CARS by using an annular aperture. Finally, we have also developed an integrated femto-/pico-second switchable CARS/SHG/TPEF multimodal nonlinear optical microscopy imaging technique for biomedical imaging. High-quality CARS/SHG/TPEF images were acquired on the same platform for qualitative and quantitative diagnosis of liver diseases in a bile-duct-ligation (BDL) rat model by analyzing the cell morphology and biochemical changes with time after the BDL surgery. This research has systematically studied the near-field effects of nanoparticle sizes, orientations, polarization of excitation light on near-field CARS imaging using FDTD technique. We have developed a radially polarized near-field TE-CARS system for high-resolution vibrational imaging and a unique annular-aperture detection scheme for suppressing the solvent background. The novel tip-enhanced near-field CARS technique as well as the multimodal nonlinear optical microscopy imaging (CARS/SHG/TPEF) platform developed in the thesis work have great potential to provide new insights into better understanding of morphological, biochemical and biomolecular changes associated with tissue and cell pathologic transformation at the tissue, cellular and molecular levels without labeling. V List of Figures Fig. 2.1 Fig. 2.2 Fig. 2.3 Fig. 2.4 Fig. 2.5 Fig. 2.6 Fig. 2.7 Fig. 3.1 Fig. 3.2 Fig. 3.3 Fig. 3.4 Fig. 3.5 Fig. 3.6 Fig. 3.7 Fig. 3.8 Fig. 3.9 Fig. 3.10 Fig. 3.11 Fig. 4.1 Fig. 4.2 Fig. 4.3 Fig. 4.4 Fig. 4.5 Fig. 4.6 Fig. 4.7 Energy  diagram  of  light  scattering.……………………………… …….9 Energy diagram of resonant and nonresonant CARS processes……… .11 Phase-matching condition of CARS ……………………… … 12 Illustration of excitation and CARS radiation from a spherical scatterer…………………………………… .……………….… 18 Schematic of a laser-scanning CARS microscope………….… .20 Oscillation of electrons in a metallic tip structure… … .……22 Schematic of polarization vectors in CARS microscopy 29 Schematic of CARS simulation for three nanoparticle configurations .35 Distributions of the focused pump field for two scatterer orientations .37 Distributions of the Px and Py components of CARS polarization for two scatterer orientations .………………… 38 Distributions of the P x component of CARS polarization under x-polarized excitations … 41 Distributions of major component of CARS polarizations for different scatterers’ sizes 42 Distributions of perpendicular component of CARS polarizations for different scatterers’ sizes .44 Schematic of near-field LP-CARS or RP-CARS field generation 47 Comparison of near-field CARS intensity distributions between RP- and LP-CARS generated from pure water .48 Near-field intensity distributions of RP-CARS and LP-CARS for scatterers with different diameters………… .…….49 FWHM and DOF of LP- and RP-CARS in water .51 Comparison of signal to background ratio of RP- and LP-CARS for different scatterers’  sizes .52 Schematic of the radially polarized TE-CARS microscope .….60 A confocal image showing the process to make the tip and focal spot overlap with each other .62 AFM image of 200-nm GaAs grating sample .62 NSOM image of a 300-nm polystyrene bead under linearly and radially polarized excitations 64 Measurement of TE-CARS intensity with tip-sample distance 65 Comparison between radially polarized and linearly polarized near-field TE-CARS images 66 Near-field TE-CARS image of mitochondria 67 VI Fig. 5.1 Fig. 5.2 Fig. 5.3 Fig. 5.4 Fig. 5.5 Fig. 5.6 Fig. 5.7 Fig. 5.8 Fig. 6.1 Fig. 6.2 Fig. 6.3 Fig. 6.4 Illustration of the annular-aperture detected RP-CARS microscopy 70 Calculated Far-field RP-CARS radiation patterns.……… .…… 71 Calculated intensity and SBR of forward-detected RP-CARS for different scatterer diameters and annular aperture sizes 73 Calculated intensity and SBR of backward-detected RP-CARS for different scatterer diameters and annular aperture sizes………… … .73 Illustration of the annular-aperture detected LP-CARS microscopy 76 Calculated intensity and SBR of forward-detected LP-CARS for different scatterer diameters and annular aperture sizes 78 Comparison of conventional and annular aperture-detected CARS images of 300 nm, 800 nm and 1100 nm polystyrene beads immersed in D2O .79 Comparison of conventional and annular aperture-detected CARS images of human epithelial cells in aqueous environment 80 Schematic diagram of the integrated fs/ps swappable CARS and multiphoton (SHG/TPEF) microscopy .……… .87 Comparisons between stained tissue images and the corresponding CARS/SHG/TPEF images sectioned liver tissues 90 Comparison of multimodal images of the normal and pathologic liver tissues 93 Changes CARS/SHG/TPEF intensities in a BDL rat liver tissue with different time durations after BDL and correlation of SHG intensities with conventional histopathological scores of liver fibrosis .97 VII References 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. W. David, Bad medicine: doctors doing harm since Hippocrates (Oxford University Press, Oxford, 2006). M. R. Arnison, K. G. Larkin, C. J. R. Sheppard, N. I. Smith, and C. J. Cogswell, “Linear phase imaging using differential interference contrast microscopy,” J. Microsc. 214, 7-12 (2004). R. D. Allen, N. S. Allen, and J. L. Travis, “Video-enhanced contrast, differential interference contrast (AVEC-DIC) microscopy - a new method capable of analyzing microtubule-related motility in the recticulopodial network of allogromia laticollaris,” Cell Motil. Cytoskeleton 1, 291-302 (1981). J. P. Rigaut and J. Vassy, “High resolution 3-dimensional images from confocal scanning laser microscoy - Quantitative study and mathematical correlation of the effects from bleaching and fluorescence attenuation in depth,” Anal. Quant. Cytol. Histol. 13, 223-232 (1991). M. Chalfie, Y. Tu, G. Euskirchen, W. Ward, and D. Prasher, “Green fluorescent protein as a marker for gene expression,” Science 263, 802-805 (1994). K. Kneipp, H. Kneipp, I. Itzkan, R. R. Dasari, and M. S. Feld, “Ultrasensitive chemical analysis by Raman spectroscopy,” Chem. Rev. 99, 2957-2975 (1999). Z. Huang, A. McWilliams, H. Lui, D. I. McLean, S. Lam, and H. Zeng, “Near-infrared Raman spectroscopy for optical diagnosis of lung cancer,” Int. J. Cancer 107, 1047-1052 (2003). S. K. Teh, W. Zheng, D. P. Lau, and Z. Huang, “Spectroscopic diagnosis of laryngeal carcinoma using near-infrared Raman spectroscopy and random recursive partitioning ensemble techniques,” Analyst 134, 1232-1239 (2009). C. Yuen, W. Zheng, and Z. Huang, “Improving surface-enhanced Raman scattering effect using gold-coated hierarchical polystyrene bead substrates modified with postgrowth microwave treatment,” J. Biomed. Opt. 13, 064040 (2008). B. R. Masters and P. T. C. So, Handbook of biomedical nonlinear optical microscopy (Oxford University Press, Oxford, 2008). A. Zoumi, A. Yeh, and B. J. Tromberg, “Imaging cells and extracellular matrix in vivo by using second-harmonic generation and two-photon excited fluorescence,” Proc. Natl. Acad. Sci. U. S. A. 99, 11014-11019 (2002). W. R. Zipfel, R. M. Williams, R. Christie, A. Y. Nikitin, B. T. Hyman, and W. W. Webb, “Live tissue intrinsic emission microscopy using multiphoton-excited native fluorescence and second harmonic generation,” Proc. Natl. Acad. Sci. U. S. A. 100, 7075-7080 (2003). R. Carriles, D. N. Schafer, K. E. Sheetz, J. J. Field, R. Cisek, V. Barzda, A. W. Sylvester, and J. A. Squier, “Invited Review Article: Imaging techniques for harmonic and multiphoton absorption fluorescence microscopy,” Rev. Sci. Instrum. 80, 081101 (2009). 108 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. P. J. Campagnola, M. D. Wei, A. Lewis, and L. M. Loew, “High-resolution nonlinear optical imaging of live cells by second harmonic generation,” Biophys. J. 77, 3341-3349 (1999). P. J. Campagnola, A. C. Millard, M. Terasaki, P. E. Hoppe, C. J. Malone, and W. A. Mohler, “Three-dimensional high-resolution second-harmonic generation imaging of endogenous structural proteins in biological tissues,” Biophys. J. 82, 493-508 (2002). D. Debarre, A. M. Pena, W. Supatto, T. Boulesteix, M. Strupler, M. P. Sauviat, J. L. Martin, M. C. Schanne-Klein, and E. Beaurepaire, “Second- and third-harmonic generation microscopies for the structural imaging of intact tissues,” Med. Sci. (Paris) 22, 845-850 (2006). J. A. Squier, M. Muller, G. J. Brakenhoff, and K. R. Wilson, “Third harmonic generation microscopy,” Opt. Express 3, 315-324 (1998). D. Debarre, W. Supatto, A. M. Pena, A. Fabre, T. Tordjmann, L. Combettes, M. C. Schanne-Klein, and E. Beaurepaire, “Imaging lipid bodies in cells and tissues using third-harmonic generation microscopy,” Nat. Methods 3, 47-53 (2006). L. S. Friedman and E. B. Keeffe, Handbook of liver disease, 2nd edition (Churchill Livingstone, 2004). A. Zumbusch, G. R. Holtom, and X. S. Xie, “Three-dimensional vibrational imaging by coherent anti-Stokes Raman scattering,” Phys. Rev. Lett. 82, 4142-4145 (1999). J. Zhu, B. Lee, K. K. Buhman, and J. X. Cheng, “A dynamic, cytoplasmic triacylglycerol pool in enterocytes revealed by ex vivo and in vivo coherent anti-Stokes Raman scattering imaging,” J. Lipid Res. 50, 1080-1089 (2009). P. D. Maker and R. W. Terhune, “Study of optical effects due to an induced polarization third order in the Electric Field strength,” Phys. Rev. A 137, 801-818 (1965). R. F. Begley, A. B. Harvey, and R. L. Byer, “Coherent anti‐Stokes Raman spectroscopy,” Appl. Phys. Lett. 25, 387-390 (1974). M. D. Duncan, J. Reintjes, and T. J. Manuccia, “Scanning coherent anti-Stokes Raman scattering microscope,” Opt. Lett. 7, 350-352 (1982). J. X. Cheng, A. Volkmer, L. D. Book, and X. S. Xie, “An Epi-Detected Coherent Anti-Stokes Raman Scattering (E-CARS) Microscope with High Spectral Resolution and High Sensitivity,” J. Phys. Chem. B 105, 1277-1280 (2001). F. M. Kamga and M. G. Sceats, “Pulse-sequenced coherent anti-Stokes Raman scattering spectroscopy: a method for suppression of the nonresonant background,” Opt. Lett. 5, 126-128 (1980). J. X. Cheng, L. D. Book, and X. S. Xie, “Polarization coherent anti-Stokes Raman scattering microscopy,” Opt. Lett. 26, 1341-1343 (2001). F. C. V. Raman, “A new radiation,” Indian J. Phys. 2, 12 (1928). A. Y. Tu, Raman spectroscopy in biology: principles & Applications (Wiley, 1982). 109 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. Z. Huang, H. Lui, X. K. Chen, A. Alajlan, D. I. McLean, and H. Zeng, “Raman spectroscopy of in vivo cutaneous melanin,” J. Biomed. Opt. 9, 1198-1205 (2004). Z. Huang, H. Lui, D. I. McLean, M. Korbelik, and H. Zeng, “Raman spectroscopy in combination with background near-infrared autofluorescence enhances the in vivo assessment of malignant tissues,” Photochem. Photobiol. 81, 1219-1226 (2005). D. P. Lau, Z. Huang, H. Lui, D. W. Anderson, K. Berean, M. D. Morrison, L. Shen, and H. Zeng, “Raman spectroscopy for optical diagnosis in the larynx: preliminary findings,” Lasers Surg. Med. 37, 192-200 (2005). S. K. Teh, W. Zheng, K. Y. Ho, M. Teh, K. G. Yeoh, and Z. Huang, “Diagnosis of gastric cancer using near-infrared Raman spectroscopy and classification and regression tree techniques,” J. Biomed. Opt. 13, 034013 (2008). M. Bergholt, W. Zheng, K. Lin, K. Ho, M. Teh, K. Yeoh, J. So, and Z. Huang, “Raman endoscopy for in vivo differentiation between benign and malignant ulcers in the stomach,” Analyst 135, 3162-3168 (2010). M. Bergholt, W. Zheng, K. Lin, K. Ho, M. Teh, K. Yeoh, J. So, and Z. Huang, “In vivo diagnosis of gastric cancer using Raman endoscopy and ant colony optimization techniques,” Int. J. Cancer 128, 2673-2680 (2011). N. Lim, Z. Hamed, C. Yeow, C. Chan, and Z. Huang, “Early detection of biomolecular changes in disrupted p.orcine cartilage using polarized Raman spectroscopy,” J. Biomed. Opt. 16, 017003 (2011). M. Bergholt, W. Zheng, K. Lin, K. Ho, M. Teh, K. Yeoh, J. So, and Z. Huang, “Characterizing variability in in vivo Raman spectra of different anatomical sites in the upper gastrointestinal tract toward cancer detection,” J. Biomed. Opt. 16, 037003 (2011). M. Bergholt, W. Zheng, K. Lin, K. Ho, M. Teh, K. Yeoh, J. So, and Z. Huang, “In vivo diagnosis of esophageal cancer using image-guided Raman endoscopy and biomolecular modeling,” Technol. Cancer Res. Treat. 10, 103-112 (2011). M. Bergholt, W. Zheng, K. Lin, K. Ho, M. Teh, K. Yeoh, J. So, and Z. Huang, “Combining near-infrared-excited autofluorescence and Raman spectroscopy improves in vivo diagnosis of gastric cancer,” Biosensors Bioelectron. 26, 4104-4110 (2011). S. Duraipandian, W. Zheng, J. Ng, J. Low, A. Ilanacheran, and Z. Huang, “In vivo diagnosis of cervical precancer using Raman spectroscopy and genetic algorithm techniques,” Analyst 136 4328-4336 (2011). R. L. Sutherland, Handbook of Nonlinear Optics (Marcel Dekker, New York, 2003). M. D. Levenson, Introduction to nonlinear laser spectroscopy (Academic Press, New York, 1982). C. Flytzanis, Quantum Electronics: A Treatise, eds. H. Rabin and C. L. Tang. (Academic Press, New York, San Francisco, London, 1975). 110 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. J. X. Cheng, A. Volkmer, and X. S. Xie, “Theoretical and experimental characterization of coherent anti-Stokes Raman scattering microscopy,” J. Opt. Soc. Am. B 19, 1363-1375 (2002). K. S. Yee, “Numerical solution of initial boundary value problem involving Maxwell equations in isotropic media,” IEEE Trans. Antennas Propag. 14, 302-307 (1966). A. Taflove, Computational Electrodynamics: The Finite-Difference Time-Domain Method (Artech House, Boston, 1995). K. S. Kunz and R. J. Luebbers, The Finite Difference Time Domain Method for Electromagnetics (Boca Raton, FL: CRC Press, 1993). J. P. Berenger, “A perfectly matched layer for the absorption of electromagnetic waves,” J. Comput. Phys. 114, 185-200 (1994). J. X. Cheng and X. S. Xie, “Coherent Anti-Stokes Raman Scattering Microscopy: Instrumentation, Theory, and Applications,” J. Phys. Chem. B 108, 827-840 (2003). D. A. Kleinman, “Nonlinear Dielectric Polarization in Optical Media,” Phys. Rev. 126,1977 (1962). A. Jorio, C. Fantini, M. S. S. Dantas, M. A. Pimenta, A. G. Souza Filho, Ge. G. Samsonidze, V. W. Brar, G. Dresselhaus, M. S. Dresselhaus, A. K. Swan, M. S. Unlu, B. B. Goldberg, and R. Saito, “Linewidth of the Raman features of individual single-wall carbon nanotubes,” Phys. Rev. B 66, 115411 (2002). K. Ishii and H. Hamaguchi, “Picosecond time-resolved multiplex CARS spectroscopy using optical Kerr gating,” Chem. Phys. Lett. 367, 672-677 (2003). F. Benabid, J. C. Knight, G. Antonopoulos, and P. S. Russell, “Stimulated Raman scattering in hydrogen-filled hollow-core photonic crystal fiber,” Science 298, 399-402 (2002). A. Volkmer, J. X. Cheng, and X. S. Xie, “Vibrational imaging with high sensitivity via epidetected coherent anti-Stokes Raman scattering microscopy,” Phys. Rev. Lett. 87, 023901 (2001). R. D. Schaller, J. Ziegelbauer, L. F. Lee, L. H. Haber and R. J. Saykally, “Chemically selective imaging of subcellular structure in human hepatocytes with coherent anti-Stokes Raman scattering (CARS) near-field scanning optical microscopy (NSOM),” J. Phys. Chem. B 106, 8489-8492 (2002). R. D. Schaller, J. C. Johnson, K. R. Wilson, L. F. Lee, L. H. Haber and R. J. Saykally, “Nonlinear chemical imaging nanomicroscopy: from second and third harmonic generation to multiplex (broad-bandwidth) sum frequency generation near-field scanning optical microscopy,” J. Phys. Chem. B, 106, 5143-5154 (2002). S. Kawata, Near-Field Optics and Surface Plasmon Polaritons (Springer, New York, 2001). R. K. Chang and T. E. Furtak, Surface Enhanced Raman Scattering (Plenum, New York, 1981). 111 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. C. Y. Chen, A. R. B. de Castro, and Y. R. Shen, “Surface-enhanced second-harmonic generation,” Phys. Rev. Lett. 46, 145-148 (1981). K. Kneipp, H. Kneipp, I. Itzkan, R. R. Dasari and M. S. Feld, “Surface-enhanced non-linear Raman scattering at the single-molecule level,” Chem. Phys. 247, 155-162 (1999). H. Chew, D. S.   Wang   and   M.   Kerker,   “Surface   enhancement   of   coherent   anti-Stokes Raman scattering by colloidal spheres,”  J.  Opt.  Soc.  Am.   B   1,  56   (1984). E. J. Liang, A. Weippert, J. M. Funk, A. Materny, and W. Kiefer, “Experimental observation of surface-enhanced coherent anti-Stokes Raman scattering,” Chem. Phys. Lett. 227, 115-120 (1994). T. Ichimura, N. Hayazawa, M. Hashimoto, Y. Inouye, and S. Kawata, “Local enhancement of coherent anti-Stokes Raman scattering by isolated gold nanoparticles,” J. Raman. Spectrosc. 34, 651-654 (2003). A. Hartschuh, E. J. Sánchez, X. S. Xie, and L. Novotny, “High-resolution near-field Raman microscopy of single-walled carbon nanotubes,” Phys. Rev. Lett. 90, 095503 (2003). I. Yasushi and K. Satoshi, “Near-field scanning optical microscope with a metallic probe tip,” Opt. Lett. 19, 159-161 (1994). R. J. H. Clark and R. E. Hester, Advances in non-linear spectroscopy (Wiley, 1988). A. Bouhelier, M. Beversluis, A. Hartschuh, and L. Novotny, “Near-field second-harmonic generation induced by local field enhancement,” Phys. Rev. Lett. 90, 013903 (2003). N. Hayazawa, Y. Inouye, Z. Sekkat, and S. Kawata, “Metallized tip amplification of near-field Raman scattering,” Opt. Commun. 183, 333-336 (2000). T. Ichimura, N. Hayazawa, M. Hashimoto, Y. Inouye, and S. Kawata, “Tip-enhanced coherent anti-Stokes Raman scattering for vibrational nano-imaging,” Phys. Rev. Lett. 92, 220801 (2004). N. Hayazawa, K. Furusawa, A. Taguchi, S. Kawata, and H. Abe, “Tip-enhanced two-photon excited fluorescence microscopy with a silicon tip,” Appl. Phys. Lett. 94, 193112 (2009). A. F. Pegoraro, A. Ridsdale, D. J. Moffatt, Y. Jia, J. P. Pezacki, and A. Stolow, “Optimally chirped multimodal CARS microscopy based on a single Ti:sapphire oscillator,” Opt. Express 17, 2984-2996 (2009). H. Chen, H. Wang, M. N. Slipchenko, Y. Jung, Y. Shi, J. Zhu, K. K. Buhman, and J. X. Cheng, “A multimodal platform for nonlinear optical microscopy and microspectroscopy,” Opt. Express 17, 1282-1290 (2009). H. W. Wang, T. T. Le, and J. X. Cheng, “Label-free imaging of arterial cells and extracellular matrix using a multimodal CARS microscope,” Opt. Comm. 281, 1813-1822 (2008). 112 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. T. T. Le, I. M. Langohr, M. J. Locker, M. Sturek, and J. X. Cheng, “Label-free molecular imaging of atherosclerotic lesions using multimodal nonlinear optical microscopy,” J. Biomed. Opt. 12, 054007 (2007). C. P. Pfeffer, B. R. Olsen, F. Ganikhanov, and F. Legare, “Multimodal nonlinear optical imaging of collagen arrays,” J. Struct. Biol. 164, 140-145 (2008). M. Zimmerley, R. A. McClure, B. Choi, and E. O. Potma, “Following dimethyl sulfoxide skin optical clearing dynamics with quantitative nonlinear multimodal microscopy,” Appl. Opt. 48, D79-D87 (2009). M. D. Duncan, J. Reintjes, and T. J. Manuccia, “Imaging biological compounds using the coherent anti-Stokes Raman scattering microscope,” Opt. Eng. 24, 352-355 (1985). F. Ganikhanov, C. L. Evans, B. G. Saar, and X. S. Xie, “High-sensitivity vibrational imaging with frequency modulation coherent anti-Stokes Raman scattering (FM CARS) microscopy,” Opt. Lett. 31, 1872-1874 (2006). J. L. Oudar, R. W. Smith, and Y. R. Shen, “Polarization-sensitive coherent anti-Stokes Raman spectroscopy,” Appl. Phys. Lett. 34, 758-760 (1979). A. Y. Chikishev, G. W. Lucassen, N. I. Koroteev, C. Otto, and J. Greve, “Polarization sensitive coherent anti-Stokes Raman scattering spectroscopy of the amide I band of proteins in solutions,” Biophys. J. 63, 976-985 (1992). A. Y. Chikishev, N. I. Koroteev, C. Otto, and J. Greve, “Polarization-sensitive CARS of the amide I band of pure and liganded chymotrypsin,” J. Raman Spectra. 27, 893-896 (1996). A. Pott, T. Doerk, J. Uhlenbusch, J. Ehlbeck, J. Hoschele, and J. Steinwandel, “Polarization-sensitive coherent anti-Stokes Raman scattering applied to the detection of NO in a microwave discharge for reduction of NO,” J. Phys. D 31, 2485-2498 (1998). M. Nissum, J. M. Funk, and W. Kiefer, “Polarization-sensitive resonance CARS spectroscopy of magnesium octaethylporphine,” J. Raman Spectra. 30, 605-610 (1999). Y. Saito, T. Ishibashi, and H. Hamaguchi, “Polarization-resolved coherent anti-Stokes Raman scattering (CARS) spectroscopy: a new probe of molecular symmetry through accurate determination of the Raman depolarization ratio,” J. Raman Spectra. 31, 725-730 (2000). P. P. Aung, K. M. Cosert, L. R. Weisel, T. F. Schulz, M. W. Gealy, and D. J. Ulness, “Effects of beam polarization on coherent anti-Stokes Raman scattering using noisy light,” J. Raman Spectra. 36, 409-419 (2005). L. R. Weisel, T. Ta, E. C. Booth, and D. J. Ulness, “Polarization coherent anti-stokes Raman scattering using noisy light,” JRSp 38, 11-20 (2007). J. Koster and S. Schlucker, “Polarization-sensitive CARS spectroscopy on free-base porphyrins: coproporphyrin I tetramethyl ester,” J. Raman Spectra. 39, 942-952 (2008). 113 88. 89. 90. 91. 92. 93. 94. 95. 96. 97. 98. 99. 100. 101. J. X. Cheng, Y. K. Jia, G. Zheng, and X. S. Xie, “Laser-scanning coherent anti-Stokes Raman scattering microscopy and applications to cell biology,” Biophys. J. 83, 502-509 (2002). J. X. Cheng, E. Potma, and S. X. Xie, “Coherent anti-Stokes Raman scattering correlation spectroscopy: Probing dynamical processes with chemical selectivity,” J. Phys. Chem. A 106, 8561-8568 (2002). H. X. Xu, E. J. Bjerneld, M. Kall, and L. Borjesson, “Spectroscopy of single hemoglobin molecules by surface enhanced Raman scattering,” Phys. Rev. Lett. 83, 4357-4360 (1999). Z. Huang, H. Zeng, I. Hamzavi, D. I. McLean, and H. Lui, “Rapid near-infrared Raman spectroscopy system for real-time in vivo skin measurements,” Opt. Lett. 26, 1782-1784 (2001). Z. Huang, A. McWilliams, S. Lam, J. English, D. I. McLean, H. Lui, and H. Zeng, “Effect of formalin fixation on the near-infrared Raman spectroscopy of normal and cancerous human bronchial tissues,” Int. J. Oncol. 23, 649-655 (2003). S. K. Teh, W. Zheng, K. Y. Ho, M. Teh, K. G. Yeoh, and Z. Huang, “Near-infrared Raman spectroscopy for optical diagnosis in the stomach: Identification of Helicobacter-pylori infection and intestinal metaplasia,” Int. J. Cancer 126, 1920-1927(2010). D. Gachet, F. Billard, and H. Rigneault, “Coherent anti-Stokes Raman scattering in a microcavity,” Opt. Lett. 34, 1789-1791 (2009). C. L. Evans, E. O. Potma, M. Puoris'haag, D. Cote, C. P. Lin, and X. S. Xie, “Chemical imaging of tissue in vivo with video-rate coherent anti-Stokes Raman scattering microscopy,” Proc. Natl. Acad. Sci. USA. 102, 16807-16812 (2005). E. O. Potma, D. J. Jones, J. X. Cheng, X. S. Xie, and J. Ye, “High-sensitivity coherent anti-Stokes Raman scattering microscopy with two tightly synchronized picosecond lasers,” Opt. Lett. 27, 1168-1170 (2002). F. Lu, W. Zheng, C. Sheppard, and Z. Huang, “Interferometric polarization coherent anti-Stokes Raman scattering (IP-CARS) microscopy,” Opt. Lett. 33, 602-604 (2008). F. Ganikhanov, S. Carrasco, X. Sunney Xie, M. Katz, W. Seitz, and D. Kopf, “Broadly tunable dual-wavelength light source for coherent anti-Stokes Raman scattering microscopy,” Opt. Lett. 31, 1292-1294 (2006). D. A. Zheltikova and A. M. Zheltikov, “Toward all-fiber coherent anti-Stokes Raman scattering in the gas phase,” Appl. Phys. B 83, 11-16 (2006). E. R. Andresen, C. K. Nielsen, J. Thogersen, and S. R. Keiding, “Fiber laser-based light source for coherent anti-Stokes Raman scattering microspectroscopy,” Opt. Express 15, 4848-4856 (2007). G. Krauss, T. Hanke, A. Sell, D. Trautlein, A. Leitenstorfer, R. Selm, M. Winterhalder, and A. Zumbusch, “Compact coherent anti-Stokes Raman scattering microscope based on a picosecond two-color Er:fiber laser system,” Opt. Lett. 34, 2847-2849 (2009). 114 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. 113. 114. 115. S. A. Akhmanov, A. F. Bunkin, S. G. Ivanov, and N. I. Koroteev, “Coherent ellipsometry of Raman scattering of light,” JETP Lett. 25, 416-420 (1977). G. B. Hadjichristov, P. P. Kircheva, and N. Kirov, “Multiplex CARS spectroscopy of Rochelle salt crystal,” J. Mol. Struct. 382, 33-37 (1996). C. Otto, A. Voroshilov, S. G. Kruglik, and J. Greve, “Vibrational bands of luminescent zinc(II)-octaethyl-porphyrin using a polarization-sensitive 'microscopic' multiplex CARS technique,” J. Raman Spectra. 32, 495-501 (2001). S. Schlucker, J. Koster, M. Nissum, J. Popp, and W. Kiefer, “Structural investigations on octaethylporphyrin using density functional theory and polarization-sensitive resonance coherent anti-Stokes Raman scattering spectroscopy,” J. Phys. Chem. A 105, 9482-9488 (2001). M. Muller and J. M. Schins, “Imaging the thermodynamic state of lipid membranes with multiplex CARS microscopy,” J. Phys. Chem. B 106, 3715-3723 (2002). K. P. Knutsen, J. C. Johnson, A. E. Miller, P. B. Petersen, and R. J. Saykally, “High spectral resolution multiplex CARS spectroscopy using chirped pulses,” Chem. Phys. Lett. 387, 436-441 (2004). H. N. Paulsen, K. M. Hilligsoe, J. Thogersen, S. R. Keiding, and J. J. Larsen, “Coherent anti-Stokes Raman scattering microscopy with a photonic crystal fiber based light source,” Opt. Lett. 28, 1123-1125 (2003). H. Kano and H. Hamaguchi, “Femtosecond coherent anti-Stokes Raman scattering spectroscopy using supercontinuum generated from a photonic crystal fiber,” Appl. Phys. Lett. 85, 4298-4300 (2004). H. Kano and H. Hamaguchi, “Ultrabroadband (> 2500 cm-1) multiplex coherent anti-Stokes Raman scattering microspectroscopy using a supercontinuum generated from a photonic crystal fiber,” Appl. Phys. Lett. 86, 121113 (2005). H. Kano and H. Hamaguchi, “Near-infrared coherent anti-Stokes Raman scattering microscopy using supercontinuum generated from a photonic crystal fiber,” Appl. Phys. B 80, 243-246 (2005). E. R. Andresen, V. Birkedal, J. Thogersen, and S. R. Keiding, “Tunable light source for coherent anti-Stokes Raman scattering microspectroscopy based on the soliton self-frequency shift,” Opt. Express 31, 1328-1330 (2006). H. Kano and H. Hamaguchi, “Dispersion-compensated supercontinuum generation for ultrabroadband multiplex coherent anti-Stokes Raman scattering spectroscopy,” J. Raman Spectra. 37, 411-415 (2006). H. Kano and H. O. Hamaguchi, “In-vivo multi-nonlinear optical imaging of a living cell using a supercontinuum light source generated from a photonic crystal fiber,” Opt. Express 14, 2798-2804 (2006). H. Kano and H. O. Hamaguchi, “Three-dimensional vibrational imaging of a microcrystalline J-aggregate using supercontinuum-based ultra-broadband multiplex coherent anti-stokes Raman scattering microscopy,” J. Phys. Chem. B 110, 3120-3126 (2006). 115 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. H. Wang, T. B. Huff, and J. X. Cheng, “Coherent anti-Stokes Raman scattering imaging with a laser source delivered by a photonic crystal fiber,” Opt. Lett. 31, 1417-1419 (2006). S. Murugkar, C. Brideau, A. Ridsdale, M. Naji, P. K. Stys, and H. Anis, “Coherent anti-Stokes Raman scattering microscopy using photonic crystal fiber with two closely lying zero dispersion wavelengths,” Opt. Express 15, 14028-14037 (2007). M. Okuno, H. Kano, P. Leproux, V. Couderc, and H. O. Hamaguchi, “Ultrabroadband (>2000 cm-1) multiplex coherent anti-Stokes Raman scattering spectroscopy using a subnanosecond supercontinuum light source,” Opt. Lett. 32, 3050-3052 (2007). K. B. Shi, P. Li, and Z. W. Liu, “Broadband coherent anti-Stokes Raman scattering spectroscopy in supercontinuum optical trap,” Appl. Phys. Lett. 90, 141116 (2007). M. Okuno, H. Kano, P. Leproux, V. Couderc, and H. O. Hamaguchi, “Ultrabroadband multiplex CARS microspectroscopy and imaging using a subnanosecond supercontinuum light source in the deep near infrared,” Opt. Lett. 33, 923-925 (2008). K. Tada and N. Karasawa, “Broadband Coherent Anti-Stokes Raman Scattering Spectroscopy Using Pulse-Shaper-Controlled Variable-Wavelength Soliton Pulses from a Photonic Crystal Fiber,” Jpn. J. Appl. Phys. 47, 8825-8828 (2008). K. Tada and N. Karasawa, “Broadband coherent anti-Stokes Raman scattering spectroscopy using soliton pulse trains from a photonic crystal fiber,” Opt. Comm. 282, 3948-3952 (2009). G. W. Wurpel, H. A. Rinia, and M. Muller, “Imaging orientational order and lipid density in multilamellar vesicles with multiplex CARS microscopy,” J. Microsc. 218, 37-45 (2005). G. W. Wurpel, J. M. Schins, and M. Muller, “Chemical specificity in three-dimensional imaging with multiplex coherent anti-Stokes Raman scattering microscopy,” Opt. Lett. 27, 1093-1095 (2002). E. M. Vartiainen, H. A. Rinia, M. Muller, and M. Bonn, “Direct extraction of Raman line-shapes from congested CARS spectra,” Opt. Express 14, 3622-3630 (2006). H. A. Rinia, M. Bonn, M. Muller, and E. M. Vartiainen, “Quantitative CARS spectroscopy using the maximum entropy method: The main lipid phase transition,” Chemphyschem 8, 279-287 (2007). I. W. Schie, T. Weeks, G. P. McNerney, S. Fore, J. K. Sampson, S. Wachsmann-Hogiu, J. C. Rutledge, and T. Huser, “Simultaneous forward and epi-CARS microscopy with a single detector by time-correlated single photon counting,” Opt. Express 16, 2168-2175 (2008). J. M. Funk and A. Materny, “Analysis of the third-order susceptibilities of triplet excited molecules using nanosecond time-resolved resonance CARS,” J. Raman Spectra. 29, 1071-1078 (1998). 116 129. 130. 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. 141. 142. M. Schenk, A. Thumann, T. Seeger, and A. Leipertz, “Pure rotational coherent anti-Stokes Raman scattering: comparison of evaluation techniques for determining single-shot simultaneous temperature and relative N2 O2 concentration,” Appl. Opt. 37, 5659-5671 (1998). M. Karavitis, R. Zadoyan, and V. A. Apkarian, “Time resolved coherent anti-Stokes Raman scattering of I-2 isolated in matrix argon: Vibrational dynamics on the ground electronic state,” J. Chem. Phys. 114, 4131-4140 (2001). A. Volkmer, L. D. Book, and X. S. Xie, “Time-resolved coherent anti-Stokes Raman scattering microscopy: Imaging based on Raman free induction decay,” Appl. Phys. Lett. 80, 1505-1507 (2002). J. M. Funk, C. Eichhorn, W. Kiefer, and A. Materny, “Nanosecond time-resolved resonance CARS spectroscopy of triplet excited polycyclic aromatic compounds,” Spectrosc. Lett. 36, 1-23 (2003). T. Kiviniemi, J. Aumanen, P. Myllyperkio, V. A. Apkarian, and M. Pettersson, “Time-resolved coherent anti-Stokes Raman-scattering measurements of I-2 in solid Kr: Vibrational dephasing on the ground electronic state at 2.6-32 K,” J. Chem. Phys. 123, 064509 (2005). S. Roy, T. R. Meyer, and J. R. Gord, “Time-resolved dynamics of resonant and nonresonant broadband picosecond coherent anti-Stokes Raman scattering signals,” Appl. Phys. Lett. 87, 264103 (2005). M. Schenk, T. Seeger, and A. Leipertz, “Time-resolved CO2 thermometry for pressures as great as MPa by use of pure rotational coherent anti-Stokes Raman scattering,” Appl. Opt. 44, 6526-6536 (2005). S. Oguchi, A. Matsuda, K. Kondo, and K. G. Nakamura, “Time-resolved coherent anti-Stokes Raman scattering of cyclohexane under shock compression,” Jpn. J. Appl. Phys., Part 45, 5817-5820 (2006). D. A. Sidorov-Biryukov, E. E. Serebryannikov, and A. M. Zheltikov, “Time-resolved coherent anti-Stokes Raman scattering with a femtosecond soliton output of a photonic-crystal fiber,” Opt. Lett. 31, 2323-2325 (2006). B. von Vacano and M. Motzkus, “Time-resolved two color single-beam CARS employing supercontinuum and femtosecond pulse shaping,” Opt. Comm. 264, 488-493 (2006). S. Postma, A. C. van Rhijn, J. P. Korterik, P. Gross, J. L. Herek, and H. L. Offerhaus, “Application of spectral phase shaping to high resolution CARS spectroscopy,” Opt. Express 16, 7985-7996 (2008). A. C. W. van Rhijn, S. Postma, J. P. Korterik, J. L. Herek, and H. L. Offerhaus, “Chemically selective imaging by spectral phase shaping for broadband CARS around 3000 cm-1,” J. Opt. Soc. Am. B 26, 559-563 (2009). C. Vinegoni, J. Bredfeldt, D. Marks, and S. Boppart, “Nonlinear optical contrast enhancement for optical coherence tomography,” Opt. Express 12, 331-341 (2004). E. O. Potma, C. L. Evans, and X. S. Xie, “Heterodyne coherent anti-Stokes Raman scattering (CARS) imaging,” Opt. Lett. 31, 241-243 (2006). 117 143. 144. 145. 146. 147. 148. 149. 150. 151. 152. 153. 154. 155. 156. A. Nikolaenko, V. V. Krishnamachari, and E. O. Potma, “Interferometric switching of coherent anti-Stokes Raman scattering signals in microscopy,” Phys. Rev. A 79, 013823 (2009). D. L. Marks, C. Vinegoni, J. S. Bredfeldt, and S. A. Boppart, “Interferometric differentiation between resonant coherent anti-Stokes Raman scattering and nonresonant four-wave-mixing processes,” Appl. Phys. Lett. 85, 5787-5789 (2004). D. L. Marks and S. A. Boppart, “Nonlinear interferometric vibrational imaging,” Phys. Rev. Lett. 92, 123905 (2004). E. S. Lee, J. Y. Lee, and Y. S. Yoo, “Nonlinear optical interference of two successive coherent anti-Stokes Raman scattering signals for biological imaging applications,” J. Biomed. Opt. 12, 024010 (2007). M. Cui, M. Joffre, J. Skodack, and J. P. Ogilvie, “Interferometric Fourier transform coherent anti-Stokes Raman scattering,” Opt. Express 14, 8448-8458 (2006). V. V. Krishnamachari and E. O. Potma, “Imaging chemical interfaces perpendicular to the optical axis with focus-engineered coherent anti-Stokes Raman scattering microscopy,” Chem. Phys. 341, 81-88 (2007). V. V. Krishnamachari and E. O. Potma, “Focus-engineered coherent anti-Stokes Raman scattering microscopy: a numerical investigation,” J. Opt. Soc. Am. A 24, 1138-1147 (2007). X. Nan, J. X. Cheng, and X. S. Xie, “Vibrational imaging of lipid droplets in live fibroblast cells with coherent anti-Stokes Raman scattering microscopy,” J. Lipid Res. 44, 2202-2208 (2003). C. L. Evans, E. O. Potma, and X. S. Xie, “Coherent anti-stokes raman scattering spectral interferometry: determination of the real and imaginary components of nonlinear susceptibility chi(3) for vibrational microscopy,” Opt. Lett. 29, 2923-2925 (2004). C. Müller, T. Buckup, B. von Vacano, and M. Motzkus, “Heterodyne single-beam CARS microscopy”, J. Raman Spectra. 37, 809-816 (2009). E. O. Potma and X. S. Xie, “Detection of single lipid bilayers with coherent anti-Stokes Raman scattering (CARS) microscopy,” J. Raman Spectra. 34, 642-650 (2006). H. A. Rinia, K. N. Burger, M. Bonn, and M. Muller, “Quantitative label-free imaging of lipid composition and packing of individual cellular lipid droplets using multiplex CARS microscopy,” Biophys. J. 95, 4908-4914 (2008). J. X. Cheng, S. Pautot, D. A. Weitz, and X. S. Xie, “Ordering of water molecules between phospholipid bilayers visualized by coherent anti-Stokes Raman scattering microscopy,” Proc. Natl. Acad. Sci. USA. 100, 9826-9830 (2003). X. Nan, E. O. Potma, and X. S. Xie, “Nonperturbative chemical imaging of organelle transport in living cells with coherent anti-stokes Raman scattering microscopy,” Biophys. J. 91, 728-735 (2006). 118 157. 158. 159. 160. 161. 162. 163. 164. 165. 166. 167. 168. 169. 170. F. Lu, W. Zheng, and Z. Huang, “Elliptically polarized coherent anti-Stokes Raman scattering microscopy,” Opt. Lett. 33, 2842-2844 (2008). D. Pestov, M. H. Zhi, Z. E. Sariyanni, N. G. Kalugin, A. Kolomenskii, R. Murawski, Y. V. Rostovtsev, V. A. Sautenkov, A. V. Sokolov, and M. O. Scully, “Femtosecond CARS of methanol-water mixtures,” J. Raman Spectra. 37, 392-396 (2006). E. Potma, W. P. de Boeij, P. J. van Haastert, and D. A. Wiersma, “Real-time visualization of intracellular hydrodynamics in single living cells,” Proc. Natl. Acad. Sci. USA. 98, 1577-1582 (2001). J. Moger, B. D. Johnston, and C. R. Tyler, “Imaging metal oxide nanoparticles in biological structures with CARS microscopy,” Opt. Express 16, 3408-3419 (2008). Y. Jung, H. Chen, L. Tong, and J. X. Cheng, “Imaging Gold Nanorods by Plasmon-Resonance-Enhanced Four Wave Mixing,” J. Phys. Chem. C 113, 2657-2663 (2009). I. Baltog, M. Baibarac, and S. Lefrant, “Coherent anti-Stokes Raman scattering on single-walled carbon nanotube thin films excited through surface plasmons,” Phys. Rev. B 72, 245402 (2005). I. Baltog, M. Baibarac, and S. Lefrant, “Coherent anti-Stokes Raman scattering on single-walled carbon nanotubes and copper phthalocyanine thin films excited through surface plasmons,” J. Opt. A 7, 632-639 (2005). I. Baltog, M. Baibarac, L. Mihut, and S. Lefrant, “Abnormal anti-Stokes Raman spectra of single walled carbon nanotubes raised from coherent anti-Stokes Raman scattering and optical cooling processes,” Dig. J. Nanomater. Bios. 2, 185-198 (2007). S. Lefrant, J. P. Buisson, J. Y. Mevellec, L. Baltog, and M. Baibarac, “Single-beam pumped coherent anti-Stokes Raman scattering on carbon nanotubes,” Phys. Status Solidi B 245, 2221-2224 (2008). Y. Fu, T. B. Huff, H. W. Wang, H. Wang, and J. X. Cheng, “Ex vivo and in vivo imaging of myelin fibers in mouse brain by coherent anti-Stokes Raman scattering microscopy,” Opt. Express 16, 19396-19409 (2008). Y. Fu, W. Sun, Y. Shi, R. Shi, and J. X. Cheng, “Glutamate excitotoxicity inflicts paranodal myelin splitting and retraction,” PLoS One 4, e6705 (2009). Y. Fu, T. M. Talavage, and J. X. Cheng, “New imaging techniques in the diagnosis of multiple sclerosis,” Expert. Opin. Med. Diagn. 2, 1055-1065 (2008). Y. Fu, H. Wang, T. B. Huff, R. Shi, and J. X. Cheng, “Coherent anti-Stokes Raman scattering imaging of myelin degradation reveals a calcium-dependent pathway in lyso-PtdCho-induced demyelination,” J. Neurosci. Res. 85, 2870-2881 (2007). Y. Fu, H. Wang, R. Shi, and J. X. Cheng, “Second harmonic and sum frequency generation imaging of fibrous astroglial filaments in ex vivo spinal tissues,” Biophys. J. 92, 3251-3259 (2007). 119 171. 172. 173. 174. 175. 176. 177. 178. 179. 180. 181. 182. 183. 184. Y. Fu, H. F. Wang, R. Y. Shi, and J. X. Cheng, “Noninvasive molecular imaging of intact myelin sheath by coherent anti-stokes raman scattering microscopy,” Am. Lab. 39, 12-14 (2007). F. P. Henry, D. Cote, M. A. Randolph, E. A. Rust, R. W. Redmond, I. E. Kochevar, C. P. Lin, and J. M. Winograd, “Real-time in vivo assessment of the nerve microenvironment with coherent anti-Stokes Raman scattering microscopy,” Plast. Reconstr. Surg. 123, 123S-130S (2009). T. B. Huff and J. X. Cheng, “In vivo coherent anti-Stokes Raman scattering imaging of sciatic nerve tissue,” J. Microsc. 225, 175-182 (2007). A. P. Kennedy, J. Sutcliffe, and J. X. Cheng, “Molecular composition and orientation in myelin figures characterized by coherent anti-stokes Raman scattering microscopy,” Langmuir 21, 6478-6486 (2005). H. Wang, Y. Fu, P. Zickmund, R. Shi, and J. X. Cheng, “Coherent anti-stokes Raman scattering imaging of axonal myelin in live spinal tissues,” Biophys. J. 89, 581-591 (2005). Y. M. Wu, H. C. Chen, W. T. Chang, J. W. Jhan, H. L. Lin, and I. Liau, “Quantitative Assessment of Hepatic Fat of Intact Liver Tissues with Coherent Anti-Stokes Raman Scattering Microscopy,” Anal. Chem. 81, 1496-1504 (2009). A. Dogariu, A. Goltsov, and M. O. Scully, “Real-time monitoring of blood using coherent anti-Stokes Raman spectroscopy,” J. Biomed. Opt. 13, 054004 (2008). M. Zimmerley, C. Y. Lin, D. C. Oertel, J. M. Marsh, J. L. Ward, and E. O. Potma, “Quantitative detection of chemical compounds in human hair with coherent anti-Stokes Raman scattering microscopy,” J. Biomed. Opt. 14, 044019 (2009). C. L. Evans, X. Xu, S. Kesari, X. S. Xie, S. T. Wong, and G. S. Young, “Chemically-selective imaging of brain structures with CARS microscopy,” Opt. Express 15, 12076-12087 (2007). J. X. Cheng and X. S. Xie, “Green’s   function   formulation   for   third-harmonic generation microscopy,” J. Opt. Soc. Am. B 19, 1604-1610 (2002). Y. Saito, M. Motohashi, N. Hayazawa, M. Iyoki, and S. Kawata, “Nanoscale characterization of strained silicon by tip-enhanced Raman spectroscope in reflection mode,” Appl. Phys. Lett. 88, 143109 (2006). B. H. Jia, X. S. Gan, and M. Gu, “Direct observation of a pure focused evanescent field of a high numerical aperture objective lens by scanning near-field optical microscopy,” Appl. Phys. Lett. 86, 131110 (2005). D. Courjon and C. Bainier, “Near field microscopy and near field optics,” Rep. Prog. Phys. 57, 989-1028 (1994). B. H. Jia, X. S. Gan, and M. Gu, “Direct measurement of a radially polarized focused evanescent field facilitated by a single LCD,” Opt. Express 13, 6821-6827 (2005). 120 185. 186. 187. 188. 189. 190. 191. 192. 193. 194. 195. 196. 197. 198. 199. C. Liu, Z. Huang, F. Lu, W. Zheng, D. W. Hutmacher, and C. Sheppard, “Near-field effects on coherent anti-Stokes Raman scattering microscopy imaging,” Opt. Express 15, 4118-4131 (2007). A. Volkmer, “Vibrational imaging and microspectroscopies based on coherent anti-Stokes Raman scattering microscopy,” J. Phys. D: Appl. Phys. 38, 59-81 (2005). K. Takeda, Y. Ito, and C. Munakata, “Simultaneous measurement of size and refractive index of a fine particle in flowing liquid,” Meas. Sci. Technol. 3, 27-32 (1992). M. Born and E. Wolf, Principles of Optics (7th edition, Cambridge University Press, Cambridge, 1999). S. Kawata, I. Yasushi, and I. Taro, “Near-field optics and spectroscopy for molecular nano-imaging,” Sci. Progress 87, 25-49 (2004). W. P. Beeker, P. Groß, C. J. Lee, C. Cleff, H. L. Offerhaus, C. Fallnich, J. L. Herek, and K.-J. Boller, “A route to sub-diffraction-limited CARS microscopy,” Opt. Express 17, 22632-22638 (2009). W. P. Beeker, C. J. Lee, K.-J. Boller, P. Groß, C. Cleff, C. Fallnich, H. L. Offerhaus, and J. L. Herek, “Spatially dependent Rabi oscillations: An approach to sub-diffraction-limited coherent anti-Stokes Raman-scattering microscopy,” Phys. Rev. A 81, 012507 (2010). W. Liu and H. Niu, “Diffraction barrier breakthrough in coherent anti-Stokes Raman scattering microscopy by additional probe-beam-induced phonon depletion,” Phys. Rev. A 81, 023830 (2011). S. W. Hell and J. Wichmann, “Breaking the diffraction resolution limit by stimulated emission: stimulated-emission-depletion fluorescence microscopy,” Opt. Lett. 19, 780-782 (1994). M. J. Rust, M. Bates, and X. Zhuang, “Sub-diffraction-limit Imaging by Stochastic Optical Reconstruction Microscopy (STORM),” Nat. Methods 3, 793-795 (2006). H. F. Wang, L. P. Shi, B. Lukyanchuk, C. Sheppard, and C. T. Chong, “Creation of a needle of longitudinally polarized light in vacuum using binary optics,” Nature Photon. 2, 501-505 (2008). M. Stalder and M. Schadt, “Linearly polarized light with axial symmetry generated by liquid-crystal polarization converters,” Opt. Lett. 21, 1948-1950 (1996). F. Lu, W. Zheng, and Z. Huang, “Coherent anti-Stokes Raman scattering microscopy using tightly focused radially polarized light,” Opt. Lett. 34, 1870-1872 (2009). J. Lin, H. Wang, W. Zheng, F. Lu, C. Sheppard, and Z. Huang, “Numerical study of effects of light polarization, scatterer sizes and orientations on near-field coherent anti-Stokes Raman scattering microscopy,” Opt. Express 17, 2423-2434 (2009). E. Bjornsson and P. Angulo, “Non-alcoholic fatty liver disease,” Scand. J. Gastroenterol. 42, 1023-1030 (2007). 121 200. 201. 202. 203. 204. 205. 206. 207. 208. 209. 210. 211. 212. P.Angulo, “Nonalcoholicfattyliverdisease,” N. Engl. J. Med. 346, 1221-1231 (2002). R. Bataller and D. A. Brenner, “Liver fibrosis,” J. Clin. Invest. 115, 209-218 (2005). A. Wieckowska, A. J. McCullough, and A. E. Feldstein, “Noninvasive diagnosis and monitoring of nonalcoholic steatohepatitis: present and future,” Hepatology. 46, 582-589 (2007). F. Lu, W. Zheng, J. Lin, Z. Huang, “Integrated coherent anti-Stokes Raman scattering and multiphoton microscopy for biomolecular imaging using spectral filtering of a femtosecond laser,” Appl. Phys. Lett. 96, 133701 (2010). E. Brown, T. McKee, E. diTomaso, A. Pluen, B. Seed, Y. Boucher, and R. K. Jain, “Dynamic imaging of collagen and its modulation in tumors in vivo using second-harmonic generation,” Nat. Med. 9, 796-800 (2003). D. C. S. Tai, N. Tan, S. Xu, C. H. Kang, S. M. Chia, C. L. Cheng, A. Wee, C. L. Wei, A. M. Raja, G. Xiao, S. Chang, J. C. Rajapakse, P. T. C. So, H. Tang, C. S. Chen, and H. Yu, “Fibro-C-Index: comprehensive, morphology-based quantification of liver fibrosis using second har- monic generation and two-photon microscopy,” J. Biomed. Opt. 14, 044013 (2009). S. R. De Vriese, J. L. Savelii, J. P. Poisson, M. Narce, I. Kerremans, R. Lefebvre, W. S. Dhooge, W. De Greyt, and A. B. Christophe, “Fat absorption and metabolism in bile duct ligated rats,” Ann. Nutr. Metab. 45, 209-216 (2001). S. F. Assimakopoulos and C. E. Vagianos, “Bile duct ligation in rats: A reliable model of hepatorenal syndrome?” World J. Gastroenterol. 15, 121-123 (2009). M. J. Ruwart, K. F. Wilkinson, B. D. Rush, T. J. Vidmar, K. M. Peters, K. S. Henley, H. D. Appelman, K. Y. Kim, D. Schuppan, and E. G. Hahn, “The integrated value of serum procollagen-III peptide over time predicts hepatic hydroxyproline content and staniable collagen in a model of dietart cirrhosis in the rat,” J. Hepatol. 10, 801-806 (1989). G. Boigk, L. Stroedter, H. Herbst, J. Waldschmidt, E. O. Riecken, and D. Schuppan, “Silymarin retards collagen accumulation in early and advanced biliary fibrosis secondary to complete bile duct obliteration in rats,” J. Hepatol. 26, 643-649 (1997). G. Boigk, L. Stroedter, H. Herbst, J. Waldschmidt, E. O. Riecken, and D. Schuppan, “Silymarin retards collagen accumulation in early and advanced biliary fibrosis secondary to complete bile duct obliteration in rats,” J. Hepatol. 26, 643-649 (1997). C. Xu and W. W. Webb, “Measurement of two-photon excitation cross sections of molecular fluorophores with data from 690 to 1050 nm,” J. Opt. Soc. Am. B 13, 481-491 (1996). M. Zimmerley, C. Y. Lin, D. C. Oertel, J. M. Marsh, J. L. Ward, and E. O. Potma, “Quantitative detection of chemical compounds in human hair with coherent anti-Stokes Raman scattering microscopy,” J. Biomed. Opt. 14, 044019 (2009). 122 213. 214. 215. 216. 217. 218. 219. 220. 221. 222. 223. http://dx.doi.org/10.1117/1.3655353.1. http://dx.doi.org/10.1117/1.3655353.2. W. Cheong, S. A. Prahl, and A. J. Welch, “A review of the opti- cal properties of biological tissues,” IEEE J. Quantum Elect. 26, 2166-2185 (1990). J. Mo, W. Zheng, and Z. Huang, “Fiber-optic Raman probe couples ball lens for depth-selected Raman measurements of epithelial tissue,” Biomed. Opt. Express 1, 17-30 (2010). A. Chong, J. Buckley, W. Renninger, and F. Wise, “All-normal-dispersion femtosecond fiber laser,” Opt. Express 14, 10095-10100 (2006). Z. Huang, S. Teh, W. Zheng, J. Mo, K. Lin, X. Shao, K. Ho, M. Teh, and K. Yeoh, “Integrated Raman spectroscopy and tri- modal wide-field imaging techniques for real-time in vivo tissue Raman measurements at endoscopy,” Opt. Lett. 34, 758-760 (2009). L. Fu and M. Gu, “Fibre-optic nonlinear optical microscopy and endoscopy,” J. Microsc. 226, 195-206 (2007). A. Cuschieri, “Minimal access surgery and the future of interventional laparoscopy,” Am. J. Surg. 161, 404-407 (1991). http://dx.doi.org/10.1117/1.3655353.3. H. F. Wang, L. P. Shi, B. Lukyanchuk, C. Sheppard, and C. T. Chong, “Creation of a needle of longitudinally polarized light in vacuum using binary optics,” Nature Photon. 2, 501-505 (2008). M. Stalder and M. Schadt, “Linearly polarized light with axial symmetry generated by liquid-crystal polarization converters,” Opt. Lett. 21, 1948-1950 (1996). 123 [...]... probability of Raman process is several orders lower than that of Rayleigh scattering At thermal equilibrium, most molecules resides at the lowest energy state following the Boltzmann distribution, and thus the Stokes Raman scattering is much stronger than the anti- Stokes Raman scattering 8 Virtual anergy states Vibrational energy states Rayleigh scattering Stokes Raman scattering Anti- Stokes Raman scattering. .. nonlinear susceptibility and thus can be used to image interfaces and inhomogeneities in the sample As one of the two-beam modalities, coherent anti- Stokes Raman scattering (CARS) is a third-order nonlinear optical process, which has become a valuable tool for tissue and cell imaging based on the Raman- active vibrations of biomolecules [19-21] The observation of the CARS process was reported for the... (FDTD) algorithm and use this method to study the near- field effects of CARS microscopy, including near- field CARS generation from two adjacent scatterers as well as the effects of scatterers’ sizes and the excitation polarizations on CARS microscopy 2) To extend the numerical simulation method from near- field to far -field and based on the analysis of the far -field CARS radiation patterns, develop a novel... reports on the numerical study of CARS microscopy in the near- field region, including near- field CARS generation from two adjacent scatterers and the effects of   scatterers’   sizes   on   near- field CARS under tightly focused radially and linearly polarized light excitations In Chapter 4, a radially polarized tip-enhanced CARS microscope is developed and demonstrated for nano-scale high-contrast vibrational... waist in the following studies 2.2.2 Near- field CARS simulation using the FDTD method After the electric fields have been computed by the FDTD method, the near- field CARS polarization can be calculated by Eq (2.10) 2.2.3 Far -field CARS simulation As the near- field  CARS  distribution  is  known,  according  to  the  Green’s  function,  the   CARS radiation in the far -field can be expressed as [44]: ... nonlinear susceptibility and thus can be used to image interfaces and inhomogeneities in the sample A NLO microscope may include many modalities including two-photon excitation fluorescence (TPEF), second harmonic generation (SHG), third harmonic generation (THG), sum-frequency generation (SFG), and coherent anti- Stokes Raman scattering (CARS) [71-76] NLO microscopy has many attractive properties for. .. process was reported for the first time by Maker and Terhune at the Ford Motor Company in 1965 [22], while the name of coherent anti- Stokes Raman spectroscopy was assigned by Begley et al at Stanford University in 1974 [23] The first CARS microscope was reported by Duncan et al in 1982 [24] However, the non-collinear configuration of pump and Stokes beams deteriorated the spatial resolution and the excitations... approaches for CARS microscopy need to be developed to improve the contrast of small scatterers 3) It is known that near- field CARS microscopy can achieve higher resolution than conventional CARS microscopy, but only very limited work has been done to either improve this technique or explore its applications Furthermore, previous research work only used linearly polarized excitations Therefore, it is... background; which is the reason for the long spectrum or image acquisition time The weak Raman signal can be enhanced up to several orders by stimulated instead of spontaneous process, in which coherent anti- Stokes Raman scattering (CARS) is a widely used technique 2.1.2 Fundamental theory of CARS The interaction   of   light   field   with   materials   is   described   by   the   Maxwell’s   equation   Without... Bile duct ligation CARS = Coherent anti- Stokes Raman scattering DIC = Differential inference contrast DOF = Depth of focus E-CARS = Epi-detected CARS F-CARS = Forward-detected CARS FDTD = Finite-difference time-domain FOV = Field of view fs = Femtosecond FWHM = Full width at half maximum HCC = Hepatocellular carcinoma HG = Hermite-Gaussian LG = Laguerre-Gaussian LP-CARS = Linearly polarized CARS MT . Near-field Coherent Anti-Stokes Raman Scattering (CARS) Microscopy for Bioimaging LIN JIAN NATIONAL UNIVERSITY OF SINGAPORE 2012 NEAR-FIELD COHERENT ANTI-STOKES. References 108 IV Abstract Coherent anti-Stokes Raman scattering (CARS) microscopy is a nonlinear Raman imaging technique that has received great attention for biological and biomedical imaging. SINGAPORE 2012 NEAR-FIELD COHERENT ANTI-STOKES RAMAN SCATTERING (CARS) MICROSCOPY FOR BIOIMAGING LIN JIAN A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DEPARTMENT

Ngày đăng: 09/09/2015, 18:54

Từ khóa liên quan

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan