Open Access
14 August 2018 Advances in nonlinear optical microscopy for biophotonics
Rui Li, Xinxin Wang, Yi Zhou, Huan Zong, Maodu Chen, Mengtao Sun
Author Affiliations +
Abstract
This article reviews recent advances in nonlinear optical microscopy for biophotonics, including stimulated Raman scattering (SRS), coherent anti-Stokes Raman scattering (CARS), two-photon excited fluorescence (TPEF), second harmonic generation (SHG), and sum frequency generation (SFG). We first introduce the principles and applications of SRS, CARS, TPEF, SHG, and SFG and their individual applications for biophotonics. We then introduce the combination of SRS, CARS, TPEF, SHG, and SFG microscopy for biophotonics. Our review not only summarizes the recent advances in nonlinear optical microscopy but also can deepen the understanding of the combination of these types of nonlinear optical microscopy for biophotonics.

1.

Introduction

Over the past decade, the nonlinear optical methods have become widely used tools for biomolecular detection, medical diagnosis in cells or tissues at the micrometer and nanometer level.1 Advancement of these optical methods promotes and enhances basic research in biology, pharmacy, and medicine.24 Compared with the optical method, radiation is usually applied for imaging modalities, including x-rays, computed tomography, plain radiography, magnetic resonance imaging, and other nuclear imaging methods.5 However, these techniques are costly and emit radiation. For disease detection and diagnoses and cells development process, optical imaging methods can provide molecular information on human tissues with noninvasive, real-time, accurate, sensitive, and economic properties, as discussed in recent reviews.6 When human diseases develop, cells and tissues are used to study the genetics, drug screening, and disease control via optical imaging techniques.79 Among optical imaging technologies, the nonlinear optical methods have great advantages such as noninvasiveness, depth penetration, high sensitivity, and ultrahigh resolution. The nonlinear signal is generated via nonlinear optical microscopy in a small area (measured in nanometers) in the focal plane of the objective lens using a pico- or femtosecond near-infrared pulsed excited laser. This technology noninvasively enables tissue depth penetration, image sensitivity, and high resolution, as discussed in recent reviews.1012

Combined various fluorescent dye labels13,14 and two-photon excited fluorescence (TPEF) microscopy are potentially used in laboratories and clinics to analyze live cells and accurately localize and completely resect live tumors.15 Live cells and tissues also can be imaged without external labeling by directly tracking different chemical bonds or proteins with nonlinear optical microscopes such as stimulated Raman scatting (SRS),1619 coherent anti-Stokes Raman scattering (CARS), TPEF, second harmonic generation (SHG), and sum frequency generation (SFG), among other techniques.

In this review, we will discuss several nonlinear methods began to be used in cells and tissues optics, such as SRS,16,20 CARS,8,21 TPEF,22 SHG,23 SFG,24 and multiple combinations of techniques. Although it is not comprehensive, an overview of nonlinear optics method applications with representative references is given in Table 1.

Table 1

Representative example applications of nonlinear optical microscopy.

Nonlinear optics methodTissues or cells
TPEFOvarian cancer25
Brain8,21
Mouse L929 fibroblastic7
Mouse sciatic nerve26
Mouse living27
Axonal myelin28
Neonatal cardiomyocyte23,29
HeLa cells22
C. elegans9
Oocyte, embryo, egg4
CARSBrain8,21
Mouse L929 fibroblastic7
C. elegans9
Axonal myelin28
Oocyte, embryo, egg4
Human stem cells30
Human mastoid cortex11
Breast and prostate cancer cells31
SRSBrain16,20
Mouse sciatic nerve26
Human lung cancer cells32
C. elegans33
Mouse skin32
HeLa cells22
Human macrophages34
SHGNeonatal cardiomyocyte29,33
Human mastoid cortex11
Cardiac myocytes23
SFGC. elegans24

2.

Single Experimental Methods

In both nonlinear microscopy and confocal laser scanning microscopy, a vibrating mirror scans the sample with focused laser beams.35 In confocal laser scanning microscopy, a high-resolution optical sectioning image is obtained with pinhole apertures.

In biology and medicine, optical microscopy is restricted by phototoxicity. Each measurement often necessitates the minimum average energy of an excited laser to avoid phototoxicity. Compared to linear processes, nonlinear processes can provide momentary extremely high pulsed excited light with low average energy on a live sample. Near-infrared excitation light is typically used in nonlinear excitation microscopy and can also decrease the phototoxicity. Using infrared light can minimize scattering in live cells and tissues. All of these effects increase the penetration depth of nonlinear microscopes.

The technology and theories of nonlinear optical microscopy have made significant contributions to biological research and medical diagnoses.36 The potential applications of these nonlinear optical microscopes are in the fields of biology and medicine. An energy-level diagram of a variety of nonlinear optical processes, including TPEF, SHG, SFG, CARS, and SRS, is shown in Fig. 1.

Fig. 1

Energy-level diagram for nonlinear optical processes of TPEF, SHG, SFG, CARS, and SRS process.

JNP_12_3_033007_f001.png

To accurately study nonlinear optical microscopy, the relationship between polarization P(t) and electric field strength is used to precisely describe nonlinear optical microscopy using the following equation:37

Eq. (1)

P(t)=ϵ0[χ(1)E(t)+χ(2)E2(t)+χ(3)E3(t)+],
where χ(1), χ(2), and χ(2) are the linear, second-order, and third-order susceptibility, respectively. ϵ0 is the permittivity of free space. The second-order nonlinear optics are described using the following equation:37

Eq. (2)

P(ω1+ω2)=2ϵ0χ(2)E1E2.

SHG and SFG are labeled as ω1=ω2 and ω1ω2.

The third-order nonlinear optics (CARS and SRS) are described using the following equations:37

Eq. (3)

P(ω1+ω2ω3)=6ϵ0χ(3)E1E2E3*,

Eq. (4)

P(2ω1ω2)=3ϵ0χ(3)E12E2*.

2.1.

Two-Photon Excited Fluorescence

TPEF microscopy is one of the traditional fluorescence imaging techniques. Due to its nonlinear optical effects, the penetration depth can reach 1 mm in live tissue with high resolution and high sensitivity. Combining TPEF microscopy with fluorescence materials can provide rapid techniques to diagnose and monitor a variety of diseases using encoded fluorescent proteins, exogenous dyes, and nanomaterials.25 A picosecond or femtosecond beam is focused on the sample using a scanning microscope to generate a fluorescence signal (Fig. 2). TPEF microscopy uses exogenous markers to detect and diagnose live cancer cells’ phenotypic changes, metabolic activity, and protein expression.38 Live ovarian cancer cells have been studied using TPEF microscopy to detect β-galactosidase with lysosome-targetable and two-photon fluorescent probe FC-βgal, which are important for the diagnosis of primary ovarian cancer (Fig. 3).25

Fig. 2

Diagram of a TPEF or SHG microscope with epi and forward channel.

JNP_12_3_033007_f002.png

Fig. 3

TPEF images of living ovarian cancer SKOV-3cells, (a) probe FC-βgal image of β-gal, (b) probe lysosome image of LysoTracker Red DND-99 by the tracker, (c) merged image of (a) and (b), and (d) intensity profile of regions of interest across SKOV-3 cells, adapted from Ref. 25.

JNP_12_3_033007_f003.png

2.2.

Second-Harmonic Generation

In general, for the second-order nonlinear optical techniques of three-wave mixing, the generation of a w3 photo has to emit one w1 and one w2 photon (w1+w2=w3). When w1 equals w2, it is defined as SHG (Fig. 1); otherwise, it is defined as SFG (Fig. 1).39 SHG occurs when an incident laser beam passes through a noncentrosymmetric and highly ordered medium such as tendons, axons, and striated muscle. Due to the nonabsorptive effects of nonlinear processes, SHG inhibits photobleaching.40 SHG was discovered by Franken in 1961.41 Because it uses the same laser scanning microscope and laser source with an additional proper narrow band filter, SHG can be combined with other nonlinear equipment. SHG is suitable for the collagen, microtubules, and muscles in live tissues and cells42 and is widely applied for biological research, medical diagnoses,43 and medicine.10,29,23,42,44 SHG microscopy has been used to measure the muscles’ contractile integrity via sarcomeric myosin imaging. Previous studies found that the muscle contractile integrity and neuromuscular health are strongly correlated in mice (Fig. 4).44

Fig. 4

SHG images of normal and diseased gastrocnemius muscle dissected, (a) normal muscle and (b) diseased muscle, adapted from Ref. 44.

JNP_12_3_033007_f004.png

2.3.

Coherent Anti-Stokes Raman Scattering

CARS is one of the most powerful Raman techniques for imaging molecular vibrations in live cells and tissues as discussed in recent reviews.3,45,4650 Similar to SRS, CARS is also generated by four-wave mixing with a three-order nonlinear effect, and these work together in the same process. However, unlike SRS, anti-Stokes signal is used for molecular imaging, and a variation in the pump intensity is applied in SRS microscopy.51 Collinear picosecond or femtosecond beams (pump and Stokes) are focused on the sample using a scanning microscope to generate anti-Stokes signals (Fig. 5). Similar to SRS, CARS microscopy is widely applied for imaging live cancer cells,31,52 probing the interactions between live cells and plasmas,53 investigating intracellular lipid storage and dynamics in live tissues,54 and assessing lipid uptake in live stem cells during differentiation,30 with the same source, ease of use, real-time, label-free, and ultrahigh resolution. CARS is used to image lipids and measure their number and size to analyze their effects on hormone-treated breast and prostate cancer cells as shown in Fig. 6.

Fig. 5

Diagram of a CARS or SFG microscope with epi and forward channel.

JNP_12_3_033007_f005.png

Fig. 6

CARS images of treated versus vehicle control living breast and prostate cancer cells. (a) Medroxyprogesterone acetate treated and (b) untreated breast cells, (c) synthetic androgen R1881treated, and (d) untreated prostate cells, scale bar 30  μm, adapted from Ref. 52.

JNP_12_3_033007_f006.png

2.4.

Stimulated Raman Scattering

SRS is a form of Raman scattering. Similar to Raman, it uses femtosecond or picosecond lasers to directly image the vibrational fingerprints of live cells and tissues. SRS is also a special case of four-wave mixing with the three-order nonlinear effect. SRS can provide high-speed, ultrahigh resolution, high-sensitivity, free label, real-time, and three-dimensional55 properties to image the distribution of specific molecules56 and monitor glucose metabolic activity,2 intracellular drug uptake,19,32,57,58 and image the vibration of newly synthesized proteins,18,59 image multiple proteins in situ,16,60,61 detect brain tumors,20 and probe the interactions between nanoparticles and live cancer cells.62 Based on the configuration of CARS microscopy, electro-optical modulators or acousto-optic modulators and lock-in amplifiers are added to the Stokes beam and photomultiplier tube, respectively, to generate an SRS signal (Fig. 7). Macrophages are among the most important white blood cells in the immune systems of animals. They phagocytose cancer cells and cell debris, among other functions. SRS microscopy was first used to characterize the different uptake kinetics of d31-palmitic acid by macrophages between individual cells as shown in Fig. 8.

Fig. 7

Diagram of an SRS microscope with epi and forward channel.

JNP_12_3_033007_f007.png

Fig. 8

SRS image of a fixed living human macrophage cells in d31-palmitic acid environment with the corresponding Raman shift being 2125  cm1. (a) Nonresonant background of CARS signal, (b) SRS image, (c) merge image of SRS (red), and CARS (gray), adapted from Ref. 34.

JNP_12_3_033007_f008.png

3.

Multiple Experimental Methods

Picosecond or femtosecond pulsed laser and multi- or two-photon scanning microscopes are used in SRS and CARS. Many useful signals can be applied to biological research and medical diagnosis, such as TPEF, SHG, and SFG.

3.1.

Two Methods

3.1.1.

TPEF and SHG

Both TPEF and SHG are second-order nonlinear processes with minimum device, single incident lasers, and the same optical system.10,29,23 The distribution of alpha-actinin with fluorescence drugs and sarcomeric structures in live embryonic cardiomyocytes is imaged using TPEF and SHG channels, respectively. This second-order nonlinear optical technique can effectively reveal long-term structural changes in the live DiO-stained myofibrillogenesis of a single cardiomyocyte using real-time, high-resolution, and high-speed (4 SPF) features as shown in Fig. 9.

Fig. 9

TPEF and SHG images of living cultured neonatal cardiomyocyte: (a) TPEF image, (b) SHG image of sarcomeric structure, and (c) merged of images (scale bars: 10  μm), adapted from Ref. 29.

JNP_12_3_033007_f009.png

3.1.2.

SRS and TPEF

TPEF and SRS microscopy are combined with new features to image live cells and tissues. They can provide similar quality images with the same organelles of live cells as shown in Fig. 10.22 Deferent organelles can also be imaged using TPEF and SRS, respectively. In a recent study, the distribution of lipid droplets and the endoplasmic reticulum was obtained in cancer cells in situ. The spatial–temporal dynamics of lipid droplets and the endoplasmic reticulum in live cancer cells can be monitored to study organelle dynamics and metabolism as shown in Fig. 10(d).63

Fig. 10

(a) TPEF image, (b) SRS image, and (c) merged image of living Hela cancer cells, adapted from Ref. 22. (d) SRS, TPEF, merged, and trajectories images of living Hela cancer cells, adapted from Ref. 63.

JNP_12_3_033007_f010.png

3.1.3.

CARS and TPEF

Due to the spectra overlap of TPEF and CARS signals, they cannot be easily separated. Using the correct wavelength of the pump and probe source solves this problem. As female gametocytes and germ cells are involved in reproduction, oocytes are the earliest stages of mammals. As with early embryos, they are widely used to study genetic diseases, cloned animals, genetic breeding, organ transplantation, and cell differentiation mechanisms. Lipid droplets store fatty acids and play a significant role in the preimplantation development of oocytes. Combined CARS and differential interference contrast (DIC) microscopy have been used to quantitatively image lipids in live mouse oocytes and early embryos at different stages of cell division as shown in Fig. 11.

Fig. 11

(a) DIC, (b) CARS, (c) TPEF, and (d) merged images in living early embryonic stages. The CARS images are shown with the corresponding Raman shift being 2850  cm1, adapted from Ref. 4.

JNP_12_3_033007_f011.png

3.1.4.

CARS and SHG

Stem cell-based bone engineering is a treatment for bone regeneration. Combined CARS and SHG microscopy has been used to image the apatite and collagen of live cells, respectively, as shown in Fig. 12.11

Fig. 12

CARS (magenta) and SHG (green) three-dimensionality images of calcium hydroxyapatite deposits embedded in collagen. The SHG image of collagen is shown with 532 nm, adapted from Ref. 11.

JNP_12_3_033007_f012.png

3.2.

Three Methods

With the appropriate filters, fluorescent labels, and excited wavelengths, three or more types of nonlinear optic microscopy can be combined in the same scanning microscope. The results indicate that these multiple processes are advantageous for imaging live tissues or animals.7,24 Caenorhabditis elegans, a free-living (nonparasitic) transparent nematode (roundworm) 1  mm in length, is widely used in genetics and developmental biology, behavior and neurobiology, aging and longevity, human genetic diseases, pathogen and biological interactions, drug screening, animal emergency response, and other fields. CARS, SFG, and TPEF have been used to study the muscle of live C. elegans using a scanning microscope with an excited laser as shown in Fig. 13.

Fig. 13

(a) CARS (Red), (b) SFG (Blue), and (c) TPEF (green fuorescent protein, Green) images of muscle of C. elegans for the 2860  cm1 vibration mode. Scale bar is 10 and 50  μm, adapted from Ref. 24.

JNP_12_3_033007_f013.png

4.

Conclusion

We have summarized the principles, applications, and advantages of individual and combination of several types of nonlinear optical microscopy, including SRS, TPEF, SHG, and SFG. The experimental results indicate that these multiple processes are of great advantage for imaging live tissues or animals, C. elegans. The nonlinear optical microscopy can monitor specific molecules and proteins inside cells and tissues in three dimensions with high sensitivity and ultrahigh resolution. Our review can promote further understanding of the advanced application of combination of these types of nonlinear optical microscopy for biophotonics.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (Grant Nos. 91436102, 11374353, 11474141, 51401239, and 11704058), Fundamental Research Funds for the Central Universities in USTB, and National Basic Research Program of China (Grant No. 2016YFA0200802).

References

1. 

L. Opilik, T. Schmid and R. Zenobi, “Modern Raman imaging: vibrational spectroscopy on the micrometer and nanometer scales,” Annu. Rev. Anal. Chem., 6 (1), 379 –398 (2013). https://doi.org/10.1146/annurev-anchem-062012-092646 SCIEAS 0036-8075 Google Scholar

2. 

W. Hong et al., “Antibiotic susceptibility determination within one cell cycle at single-bacterium level by stimulated Raman metabolic imaging,” Anal. Chem., 90 3737 –3743 (2018). https://doi.org/10.1021/acs.analchem.7b03382 ANCHAM 0003-2700 Google Scholar

3. 

R. E. Kast et al., “Emerging technology: applications of Raman spectroscopy for prostate cancer,” Cancer Metastasis Rev., 33 (2), 673 –693 (2014). https://doi.org/10.1007/s10555-013-9489-6 Google Scholar

4. 

J. Bradley et al., “Quantitative imaging of lipids in live mouse oocytes and early embryos using CARS microscopy,” Development, 143 (12), 2238 –2247 (2016). https://doi.org/10.1242/dev.129908 Google Scholar

5. 

S. Ayyachamy and V. S. Manivannan, “Distance measures for medical image retrieval,” Int. J. Imaging Syst. Technol., 23 (1), 9 –21 (2013). https://doi.org/10.1002/ima.v23.1 IJITEG 0899-9457 Google Scholar

6. 

R. H. Wilson, K. Vishwanath and M. Mycek, “Optical methods for quantitative and label-free sensing in living human tissues: principles, techniques, and applications,” Adv. Phys., 1 (4), 523 –543 (2016). https://doi.org/10.1080/23746149.2016.1221739 Google Scholar

7. 

H. Segawa et al., “Multimodal imaging of living cells with multiplex coherent anti-Stokes Raman scattering (CARS), third-order sum frequency generation (TSFG) and two-photon excitation fluorescence (TPEF) using a nanosecond white-light laser source,” Anal. Sci., 31 (4), 299 –305 (2015). https://doi.org/10.2116/analsci.31.299 ANSCEN 0910-6340 Google Scholar

8. 

C. L. Evans et al., “Chemical imaging of tissue in vivo with video-rate coherent anti-Stokes Raman scattering microscopy,” Proc. Natl. Acad. Sci. U. S. A., 102 (46), 16807 –16812 (2005). https://doi.org/10.1073/pnas.0508282102 Google Scholar

9. 

T. Hellerer et al., “Monitoring of lipid storage in Caenorhabditis elegans using coherent anti-Stokes Raman scattering (CARS) microscopy,” Proc. Natl. Acad. Sci. U. S. A., 104 (37), 14658 –14663 (2007). https://doi.org/10.1073/pnas.0703594104 Google Scholar

10. 

P. J. Campagnola et al., “High-resolution nonlinear optical imaging of live cells by second harmonic generation,” Biophys. J., 77 (6), 3341 –3349 (1999). https://doi.org/10.1016/S0006-3495(99)77165-1 BIOJAU 0006-3495 Google Scholar

11. 

A. D. Hofemeier et al., “Label-free nonlinear optical microscopy detects early markers for osteogenic differentiation of human stem cells,” Sci. Rep., 6 26716 (2016). https://doi.org/10.1038/srep26716 SRCEC3 2045-2322 Google Scholar

12. 

W. Min et al., “Coherent nonlinear optical imaging: beyond fluorescence microscopy,” Annu. Rev. Phys. Chem., 62 (1), 507 –530 (2011). https://doi.org/10.1146/annurev.physchem.012809.103512 ARPLAP 0066-426X Google Scholar

13. 

K. Svoboda and R. Yasuda, “Principles of two-photon excitation microscopy and its applications to neuroscience,” Neuron, 50 (6), 823 –839 (2006). https://doi.org/10.1016/j.neuron.2006.05.019 NERNET 0896-6273 Google Scholar

14. 

H. W. Liu et al., “Molecular engineering of two-photon fluorescent probes for bioimaging applications,” Methods Appl. Fluores., 5 (1), 012003 (2017). https://doi.org/10.1088/2050-6120/aa61b0 Google Scholar

15. 

C. Vinegoni et al., “Measurement of drug-target engagement in live cells by two-photon fluorescence anisotropy imaging,” Nat. Protoc., 12 1472 –1497 (2017). https://doi.org/10.1038/nprot.2017.043 1754-2189 Google Scholar

16. 

F. Hu et al., “Bioorthogonal chemical imaging of metabolic activities in live mammalian hippocampal tissues with stimulated Raman scattering,” Sci. Rep., 6 39660 (2016). https://doi.org/10.1038/srep39660 SRCEC3 2045-2322 Google Scholar

17. 

L. Zhang and W. Min, “Bioorthogonal chemical imaging of metabolic changes during epithelial–mesenchymal transition of cancer cells by stimulated Raman scattering microscopy,” J. Biomed. Opt., 22 1 –7 (2017). https://doi.org/10.1117/1.JBO.22.10.106010 JBOPFO 1083-3668 Google Scholar

18. 

L. Wei et al., “Vibrational imaging of newly synthesized proteins in live cells by stimulated Raman scattering microscopy,” Proc. Nat. Acad. Sci. U. S. A., 110 (28), 11226 –11231 (2013). https://doi.org/10.1073/pnas.1303768110 Google Scholar

19. 

B. G. Saar et al., “Video-rate molecular imaging in vivo with stimulated Raman scattering,” Science, 330 (6009), 1368 –1370 (2010). https://doi.org/10.1126/science.1197236 SCIEAS 0036-8075 Google Scholar

20. 

M. Ji et al., “Rapid, label-free detection of brain tumors with stimulated Raman scattering microscopy,” Sci. Transl. Med., 5 (201), 201ra119 (2013). https://doi.org/10.1126/scitranslmed.3005954 STMCBQ 1946-6234 Google Scholar

21. 

B. F. M. Romeike et al., “Coherent anti-Stokes Raman scattering and two photon excited fluorescence for neurosurgery,” Clin. Neurol. Neurosurg., 131 42 –46 (2015). https://doi.org/10.1016/j.clineuro.2015.01.022 CNNSBV 0303-8467 Google Scholar

22. 

X. Li et al., “Mitochondrial imaging with combined fluorescence and stimulated Raman scattering microscopy using a probe of the aggregation-induced emission characteristic,” J. Am. Chem. Soc., 139 (47), 17022 –17030 (2017). https://doi.org/10.1021/jacs.7b06273 JACSAT 0002-7863 Google Scholar

23. 

S. Awasthi et al., “Multimodal SHG-2PF imaging of microdomain Ca2+-contraction coupling in live cardiac myocytes,” Circ. Res., 118 (2), e19 (2016). https://doi.org/10.1161/CIRCRESAHA.115.307919 CIRUAL 0009-7330 Google Scholar

24. 

H. Kim et al., “Coherent Raman imaging of live muscle sarcomeres assisted by SFG microscopy,” Sci. Rep., 7 (1), 9211 (2017). https://doi.org/10.1038/s41598-017-09571-w SRCEC3 2045-2322 Google Scholar

25. 

J. Huang et al., “A lysosome-targetable and two-photon fluorescent probe for imaging endogenous β-galactosidase in living ovarian cancer cells,” Sens. Actuators B, 246 833 –839 (2017). https://doi.org/10.1016/j.snb.2017.02.158 SABCEB 0925-4005 Google Scholar

26. 

F. Tian et al., “Monitoring peripheral nerve degeneration in ALS by label-free stimulated Raman scattering imaging,” Nat. Commun., 7 13283 (2016). https://doi.org/10.1038/ncomms13283 NCAOBW 2041-1723 Google Scholar

27. 

L. Zhou et al., “Molecular engineering of d-A-d-based non-linearity fluorescent probe for quick detection of thiophenol in living cells and tissues,” Sens. Actuators B, 244 958 –964 (2017). https://doi.org/10.1016/j.snb.2017.01.079 SABCEB 0925-4005 Google Scholar

28. 

H. Wang et al., “Coherent anti-Stokes Raman scattering imaging of axonal myelin in live spinal tissues,” Biophys. J., 89 (1), 581 –591 (2005). https://doi.org/10.1529/biophysj.105.061911 BIOJAU 0006-3495 Google Scholar

29. 

H. Liu et al., “Myofibrillogenesis in live neonatal cardiomyocytes observed with hybrid two-photon excitation fluorescence-second harmonic generation microscopy,” J. Biomed. Opt., 16 (12), 126012 (2011). https://doi.org/10.1117/1.3662457 JBOPFO 1083-3668 Google Scholar

30. 

C. Di Napoli et al., “Quantitative spatiotemporal chemical profiling of individual lipid droplets by hyperspectral CARS microscopy in living human adipose-derived stem cells,” Anal. Chem., 88 (7), 3677 –3685 (2016). https://doi.org/10.1021/acs.analchem.5b04468 ANCHAM 0003-2700 Google Scholar

31. 

M. C. Potcoava et al., “Raman and coherent anti-Stokes Raman scattering microscopy studies of changes in lipid content and composition in hormone-treated breast and prostate cancer cells,” J. Biomed. Opt., 19 (11), 111605 (2014). https://doi.org/10.1117/1.JBO.19.11.111605 Google Scholar

32. 

C. W. Freudiger et al., “Label-free biomedical imaging with high sensitivity by stimulated Raman scattering microscopy,” Science, 322 (5909), 1857 –1861 (2008). https://doi.org/10.1126/science.1165758 Google Scholar

33. 

C. W. Freudiger et al., “Highly specific label-free molecular imaging with spectrally tailored excitation-stimulated Raman scattering (STE-SRS) microscopy,” Nat. Photonics, 5 103 –109 (2011). https://doi.org/10.1038/nphoton.2010.294 PSSABANPAHBY 1862-63001749-4885 Google Scholar

34. 

C. Stiebing et al., “Real-time Raman and SRS imaging of living human macrophages reveals cell-to-cell heterogeneity and dynamics of lipid uptake,” J. Biophotonics, 10 (9), 1217 –1226 (2017). https://doi.org/10.1002/jbio.201600279 Google Scholar

35. 

R. Li et al., “Optical characterizations of two-dimensional materials using nonlinear optical microscopies of CARS, TPEF, and SHG,” Nanophotonics, 7 (5), 873 –881 (2018). https://doi.org/10.1515/nanoph-2018-0002 Google Scholar

36. 

W. W. Mantulin, B. R. Masters and P. T. C. So, “Handbook of biomedical nonlinear optical microscopy,” J. Biomed. Opt., 14 (1), 019901 (2009). https://doi.org/10.1117/1.3077566 JBOPFO 1083-3668 Google Scholar

37. 

R. W. Boyd, Chapter 1: The Nonlinear Optical Susceptibility, in Nonlinear Optics, 1 –67 3rdAcademic Press, Burlington (2008). Google Scholar

38. 

Z. Liu et al., “A new fluorescent probe with a large turn-on signal for imaging nitroreductase in tumor cells and tissues by two-photon microscopy,” Biosens. Bioelectron., 89 853 –858 (2017). https://doi.org/10.1016/j.bios.2016.09.107 BBIOE4 0956-5663 Google Scholar

39. 

A. V. Petukhov et al., “Energy exchange in second-order nonlinear optics in centrosymmetric media,” Phys. Status Solidi A, 170 (2), 417 –422 (1999). https://doi.org/10.1002/(ISSN)1521-396X Google Scholar

40. 

A. H. Reshak and C. R. Sheue, “Second harmonic generation imaging of the deep shade plant Selaginella erythropus using multifunctional two-photon laser scanning microscopy,” J. Microsc., 248 (3), 234 –244 (2012). https://doi.org/10.1111/jmi.2012.248.issue-3 JRSPAFJMICAR 0377-04860022-2720 Google Scholar

41. 

A. E. Hill et al., “Generation of optical harmonics,” Phys. Rev. Lett., 7 (4), 118 –119 (1961). https://doi.org/10.1103/PhysRevLett.7.118 PRLTAO 0031-9007 Google Scholar

42. 

X. Chen et al., “Second harmonic generation microscopy for quantitative analysis of collagen fibrillar structure,” Nat. Protoc., 7 654 –669 (2012). https://doi.org/10.1038/nprot.2012.009 1754-2189 Google Scholar

43. 

C. Macias-Romero et al., “Probing rotational and translational diffusion of nanodoublers in living cells on microsecond time scales,” Nano Lett., 14 (5), 2552 –2557 (2014). https://doi.org/10.1021/nl500356u NALEFD 1530-6984 Google Scholar

44. 

S. V. Plotnikov et al., “Measurement of muscle disease by quantitative second-harmonic generation imaging,” J. Biomed. Opt., 13 (4), 044018 (2008). https://doi.org/10.1117/1.2967536 Google Scholar

45. 

J. Cheng and X. S. Xie, “Coherent anti-Stokes Raman scattering microscopy: instrumentation, theory, and applications,” J. Phys. Chem. B, 108 (3), 827 –840 (2004). https://doi.org/10.1021/jp035693v JPCBFK 1520-6106 Google Scholar

46. 

C. L. Evans and X. S. Xie, “Coherent Anti-Stokes Raman scattering microscopy: chemical imaging for biology and medicine,” Annu. Rev. Anal. Chem., 1 (1), 883 –909 (2008). https://doi.org/10.1146/annurev.anchem.1.031207.112754 Google Scholar

47. 

H. Kano et al., “Hyperspectral coherent Raman imaging: principle, theory, instrumentation, and applications to life sciences,” J. Raman Spectrosc., 47 (1), 116 –123 (2015). https://doi.org/10.1002/jrs.4853 Google Scholar

48. 

J. Cheng et al., “An epi-detected coherent anti-Stokes raman scattering (E-CARS) microscope with high spectral resolution and high sensitivity,” J. Phys. Chem. B, 105 (7), 1277 –1280 (2001). https://doi.org/10.1021/jp003774a JRSPAFJPCBFK 0377-04861520-6106 Google Scholar

49. 

J. Cheng et al., “Laser-scanning coherent anti-Stokes Raman scattering microscopy and applications to cell biology,” Biophys. J., 83 (1), 502 –509 (2002). https://doi.org/10.1016/S0006-3495(02)75186-2 BIOJAU 0006-3495 Google Scholar

50. 

C. Krafft, B. Dietzek and J. Popp, “Raman and CARS microspectroscopy of cells and tissues,” Analyst, 134 (6), 1046 –1057 (2009). https://doi.org/10.1039/b822354h ANLYAG 0365-4885 Google Scholar

51. 

M. Müller and A. Zumbusch, “Coherent anti-Stokes Raman scattering microscopy,” ChemPhysChem, 8 (15), 2156 –2170 (2007). https://doi.org/10.1002/(ISSN)1439-7641 CPCHFT 1439-4235 Google Scholar

52. 

K. Ishitsuka et al., “Identification of intracellular squalene in living algae, Aurantiochytrium mangrovei with hyper-spectral coherent anti-Stokes Raman microscopy using a sub‐nanosecond supercontinuum laser source,” J. Raman Spectrosc., 48 (1), 8 –15 (2016). https://doi.org/10.1002/jrs.v48.1 Google Scholar

53. 

R. Furuta et al., “Intracellular-molecular changes in plasma-irradiated budding yeast cells studied using multiplex coherent anti-Stokes Raman scattering microscopy,” Phys. Chem. Chem. Phys., 19 (21), 13438 –13442 (2017). https://doi.org/10.1039/C7CP00489C PPCPFQ 1463-9076 Google Scholar

54. 

S. Daemen et al., “Microscopy tools for the investigation of intracellular lipid storage and dynamics,” Mol. Metab., 5 (3), 153 –163 (2016). https://doi.org/10.1016/j.molmet.2015.12.005 Google Scholar

55. 

X. Chen et al., “Volumetric chemical imaging by stimulated Raman projection microscopy and tomography,” Nat. Commun., 8 15117 (2017). https://doi.org/10.1038/ncomms15117 NCAOBW 2041-1723 Google Scholar

56. 

R. Long et al., “Two-color vibrational imaging of glucose metabolism using stimulated Raman scattering,” Chem. Commun., 54 (2), 152 –155 (2018). https://doi.org/10.1039/C7CC08217G Google Scholar

57. 

W. J. Tipping et al., “Imaging drug uptake by bioorthogonal stimulated Raman scattering microscopy,” Chem. Sci., 8 (8), 5606 –5615 (2017). https://doi.org/10.1039/C7SC01837A 1478-6524 Google Scholar

58. 

T. Ito, Y. Obara and K. Misawa, “Single-beam phase-modulated stimulated Raman scattering microscopy with spectrally focused detection,” J. Opt. Soc. Am. B, 34 (5), 1004 –1015 (2017). https://doi.org/10.1364/JOSAB.34.001004 JOBPDE 0740-3224 Google Scholar

59. 

L. Wei et al., “Live-cell imaging of alkyne-tagged small biomolecules by stimulated Raman scattering,” Nat. Methods, 11 410 –412 (2014). https://doi.org/10.1038/nmeth.2878 1548-7091 Google Scholar

60. 

X. Zhang et al., “Label-free live-cell imaging of nucleic acids using stimulated Raman scattering microscopy,” ChemPhysChem, 13 (4), 1054 –1059 (2012). https://doi.org/10.1002/cphc.201100890 CPCHFT 1439-4235 Google Scholar

61. 

W. Yang et al., “Simultaneous two-color stimulated Raman scattering microscopy by adding a fiber amplifier to a 2 ps OPO-based SRS microscope,” Opt. Lett., 42 (3), 523 –526 (2017). https://doi.org/10.1364/OL.42.000523 OPLEDP 0146-9592 Google Scholar

62. 

L. Zhang et al., “Label-free, quantitative imaging of MoS2-nanosheets in live cells with simultaneous stimulated Raman scattering and transient absorption microscopy,” Adv. Biosyst., 1 (4), 1700013 (2017). https://doi.org/10.1002/adbi.201700013 Google Scholar

63. 

C. Zhang et al., “Quantification of lipid metabolism in living cells through the dynamics of lipid droplets measured by stimulated Raman scattering imaging,” Anal. Chem., 89 (8), 4502 –4507 (2017). https://doi.org/10.1021/acs.analchem.6b04699 ANCHAM 0003-2700 Google Scholar

Biography

Rui Li received his PhD from the Department of Physics of Dalian University, China, in 2014. From 2009 to 2012, he worked as an exchanged student at the Department of Electronic Engineering, University of Texas at Arlington. Since 2014, he has worked as a lecturer at the Department of Physics of Dalian University of Technology, China. His current research interests focus on surface enhancement Raman scattering and coherent anti-stokes Raman scattering microscopy.

Xinxin Wang is the PhD candidate supervised by Professor Mengtao Sun, at the School of Mathematics and Physics, University of Science and Technology Beijing, China. Her current research interests focus on one and two photon absorptions, fluorescence, and Raman spectra.

Yi Zhou is the PhD candidate supervised by Professor Rui Li, at Dalian University of Technology. His current research interests focus on one and two photon absorptions, fluorescence, and Raman spectra.

Huan Zong is a PhD candidate supervised by Professor Mengtao Sun, at the School of Mathematics and Physics, University of Science and Technology Beijing, China. Her current research interests focus on one and two photon absorptions, fluorescence, and Raman spectra.

Maodu Chen received his PhD in 2003 from Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS). From 2003 to 2005, he worked as a postdoc at the Department of Chemical Physics, Northwestern University. Since 2005, he has worked as an associate professor at the Department of Physics of Dalian University of Technology. In 2010, he became a full professor at Dalian University of Technology, China.

Mengtao Sun received his PhD in 2003 from the Dalian Institute of Chemical Physics, CAS. From 2003 to 2006, he worked as a postdoc at the Department of Chemical Physics, Lund University. Since 2006, he has worked as an associate professor at the Institute of Physics, CAS. In 2016, he became a full professor at the University of Science and Technology Beijing. His current research interests focus on nonlinear optical microscopy, such as CARS, TPEF, and SHG, applied in biophtonics and two-dimensional materials.

© 2018 Society of Photo-Optical Instrumentation Engineers (SPIE) 1934-2608/2018/$25.00 © 2018 SPIE
Rui Li, Xinxin Wang, Yi Zhou, Huan Zong, Maodu Chen, and Mengtao Sun "Advances in nonlinear optical microscopy for biophotonics," Journal of Nanophotonics 12(3), 033007 (14 August 2018). https://doi.org/10.1117/1.JNP.12.033007
Received: 6 June 2018; Accepted: 30 July 2018; Published: 14 August 2018
Lens.org Logo
CITATIONS
Cited by 27 scholarly publications.
Advertisement
Advertisement
RIGHTS & PERMISSIONS
Get copyright permission  Get copyright permission on Copyright Marketplace
KEYWORDS
Microscopy

Second-harmonic generation

Optical microscopy

Biomedical optics

Tissues

Microscopes

Luminescence

Back to Top