Open Access
30 November 2015 Optical reprogramming of human somatic cells using ultrashort Bessel-shaped near-infrared femtosecond laser pulses
Author Affiliations +
Abstract
We report a virus-free optical approach to human cell reprogramming into induced pluripotent stem cells with low-power nanoporation using ultrashort Bessel-shaped laser pulses. Picojoule near-infrared sub-20 fs laser pulses at a high 85 MHz repetition frequency are employed to generate transient nanopores in the membrane of dermal fibroblasts for the introduction of four transcription factors to induce the reprogramming process. In contrast to conventional approaches which utilize retro- or lentiviruses to deliver genes or transcription factors into the host genome, the laser method is virus-free; hence, the risk of virus-induced cancer generation limiting clinical application is avoided.

1.

Introduction

Induced pluripotent stem (iPS) cells closely resemble embryonic stem cells in many aspects such as differentiation potency, protein content and gene expression, proliferation, morphology, and embryoid body formation.1 Typically, iPS cells are generated through the reprogramming of somatic cells by forced expression of a combination of appropriate transcription factors.13 Conventional approaches utilize retro- or lentiviruses for gene delivery.1

iPS cells derived from patients’ cells have tremendous potential for the investigation of disease pathology and the development of pharmaceuticals,4 in particular, since they open the possibility of using the patient’s own cells for transplantation therapy and circumvent problems associated with immune compatibility.5

iPS cells were first derived from mouse fibroblasts1 by Yamanaka’s group in Japan in 2006 (Nobel Prize in 2012). It was shown that the viral introduction of the transcription factors Oct4, Klf4, Sox2, and c-Myc transferred fibroblasts into iPS cells. Although only one decade has passed since 2006, iPS cells have been generated from a range of tissue-specific stem cells and fully differentiated somatic cells such as peripheral blood cells, hepatocytes, keratinocytes, oral mucosa, and umbilical cord blood, as well as dental pulp cells.6

Alternatively, functional somatic cells have been generated from other fully differentiated cells as well as from committed, but not yet fully differentiated, cells (so-called progenitor cells) through “direct reprogramming” (also known as trans-differentiation) without reversion to an intermediate pluripotent state.79 This approach provides a faster method for the generation of functional cells and excludes the risk of generating potentially tumorigenic iPS cells7 due to their pluripotency.

Direct reprogramming was realized by the forced expression of the gene MyoD in dermal fibroblasts, retinal cells, and immature chondrocytes that converted them into muscle cells.10 The combination of transcription factors such as Ascl1, Brn2, and Myt1lo converted fibroblasts into neurons,11 whereas Gata4, Mef2c, and Tbx5 could reprogram mouse cardiac and dermal fibroblasts directly into functional cardiomyocytes.12,13

The delivery of transcription factors into the genome of a target cell is typically achieved through the use of retro- or lentiviruses.1 Clinical translation of iPS cells generated with these viral techniques is challenging due to insertional mutations caused by the random integration of genes carried by viruses.6 Moreover, the integration of viral genes into the cell genome may result in residual transgene sequences, thereby increasing the risk of cancer formation.14

Alternative strategies of reprogramming through nonviral gene vectors such as small interfering ribonucleic acid and plasmid deoxyribose nucleic acid (DNA) have also been employed.6 A variety of delivery approaches such as electroporation, magnet-assisted transfection, DNA guns, ultrasound, microinjection, and liposome-mediated transfection have been applied to assist penetration of the genes into the cytoplasm and further into the nucleus. However, the issues of cytotoxicity, contamination with chemicals, low posttransfection viability due to irreversible damage, and low transfection efficiency are still unsolved problems.

A virus-free delivery method is the targeted femtosecond laser transfection of adherent cells.15 Due to a cell’s innate self-repair mechanism, transient pores in the cell membrane can be created by highly focused femtosecond laser beams in the near-infrared (NIR) range to allow genes or vectors to enter the cell without any adverse effect on cellular viability. Femtosecond laser transfection is sterile and nontoxic, and it achieves high posttransfection viability. This method has been tested on a variety of mammalian cells including stem cells.16 Thus, femtosecond laser transfection has been employed for the insertion of the green fluorescence protein (GFP) gene in stem cells17 as well as to activate mouse stem cell differentiation through GATA6.18 NIR femtosecond lasers can create nanoholes in biological objects with a diameter of less than 80 nm.19

In this paper, we present a proof-of-principle study for virus-free optical reprogramming of human somatic cells by the introduction of a cocktail of four transcription factors as well as the GFP gene with femtosecond laser nanoporation.

2.

Materials and Methods

2.1.

Laser Scanning Microscope for Cell Detection and Nanoporation

Nanoporation was performed with a 10 fs multiphoton microscope (JenLab GmbH, Jena, Germany).17 The mode-locked Ti:sapphire laser Integral Pro 400 oscillator with a broad M-shaped emission spectrum centered around 800 nm, a repetition rate of 85 MHz, and a mean output power of 420 mW was employed. For cell nanoporation, the scanning mirrors of the microscope were kept fixed in the center position. A 40× NA1.3 objective (Zeiss EC Plan-Neofluar) was used. A motorized stage (Märzhäuser, Wetzlar, Germany) was employed to move the sample in xy directions, and a piezoelectric z-translator (nanoMipos400, Piezosystems Jena, Germany) was used to shift the objective along the z-direction. The laser illumination time was controlled by a mechanical shutter (Thorlabs Inc., New Jersey).

An axicon lens (apex angle of 0.5 deg, Thorlabs Inc., New Jersey) was used to elongate the laser focal region along the axial direction by creating a quasi-Bessel beam [Fig. 1(a)]. The lateral laser intensity pattern is described by a Bessel function of the first kind, that is, it consists of an intense central part with several concentric rings. Experimentally, however, only an approximation to a Bessel beam with the properties of the mathematical entity over a finite distance can be realized.20 This intensity pattern is here referred to as a quasi-Bessel beam. The quasi-Bessel beam extends along the optical axis leading to an effectively elongated focal region compared to a Gaussian beam shape. The elongated focal region relaxes the need for precise fine-focus positioning onto the cell membrane, which is necessary with Gaussian beam focal shape. Due to the enhanced focal length by a factor of 2 in our setup in the case of the quasi-Bessel beam, the exact positioning of the laser focal volume is less critical for the realization of efficient nanoporation.

Fig. 1

(a) Scheme of the femtosecond laser nanosurgery microscope with quasi-Bessel beam focal geometry. The dashed rectangles indicate the objects controlled by the automation software. (b) Interferometric autocorrelation trace of the femtosecond laser pulses in the focal plane of the NA1.3 objective. The width of about 27 fs of the autocorrelation trace indicates a sub-20 fs pulse width assuming Gaussian temporal shape. (c) The laser spectrum exhibits an FWHM bandwidth of 115 nm with two peaks at 770 and 830 nm.

JBO_20_11_115008_f001.png

Chirped mirrors were employed to precompensate for the group velocity dispersion (GVD) of all optics (Fig. 1). The in situ pulse duration behind the objective, that is, at the sample position, was determined by interferometric autocorrelation measurements. Figures 1(b) and 1(c) show an autocorrelation trace measured behind the objective as well as the pulse spectrum. Although the autocorrelation trace still indicates the presence of some uncompensated components of GVD, a pulse duration of 19fs (27/1.41) can be derived assuming pulses with Gaussian temporal shapes.

Typically, we used a mean power of 40 mW in situ for the nanoporation. Nanoporation was observed over an axial range of 4μm, that is, the acquired focusing precision for the focal volume on the cell membrane was about 4μm. This range was enough to potentially optoporate all adherent cells inside a culture dish and to balance different x-y-position-dependent vertical offsets due to slight horizontal misalignments of the cell dish in the microscope stage. While the axial focus region was extended, the lateral focal was not significantly widened compared to a Gaussian focal beam shape. Axial and lateral extensions of the focal region of the quasi-Bessel beam as determined from imaging a subresolution fluorescent sphere with 5 mW are shown in Figs. 2(a) and 2(b), respectively, together with the measurement data for the Gaussian-shaped focal volume (obtained with the axicon removed).

Fig. 2

Gaussian and axicon-generated quasi-Bessel beam focus dimensions along (a) axial (z) and (b) radial axes.

JBO_20_11_115008_f002.png

The optical reprogramming procedure was controlled with automated software that was developed in our laboratory.21 A CCD camera was used for online monitoring of the fibroblasts during the laser treatment. The software recognizes the cell positions by fast gray-level evaluation and steers the hardware components to center the periphery of the target cell on the laser beam position, and it controls the number and duration of laser shots per cell. Mean powers of 10 and 40 mW were required for the laser treatment in the case of the Gaussian and the quasi-Bessel beams, respectively.

2.2.

Cell Lines

Human dermal fibroblasts (passage 2) isolated from a healthy donor were purchased from LONZA (Adult Human Normal Dermal Fibroblasts #CC-2511, Lonza) and cultured in fibroblast growth medium containing high-glycose Dulbecco’s modified Eagle's medium (# 41965-039, Life Technologies), 15% fetal bovine serum (# 10270-106, Life Technologies), and 1% Gentamicin (# 1570, Gibco). Cells were passaged in a 1:4 ratio and split every three to four days upon reaching the cell density of about 80%.

Adherent cells in a 170-μm glass bottom dish with 500-μm grids and removal inserts were prepared one day before the transfection. The growth medium was aspirated from the flask, cells were washed once with Dulbecco’s phosphate-buffered saline medium free of Ca2+ and Mg2+ (# 10010, Life Technologies), and 1 ml of 0.25% trypsin–ethylenediaminetetraacetic acid (EDTA) (# 25200, Gibco) was added. The cells in the flask were incubated for about 3 to 4 min at 37°C (5% CO2) until all cells were completely detached. Subsequently, the trypsin–EDTA was inactivated by adding 3 to 5 ml fibroblast growth medium. Dislodged cells were transferred into a 15-ml conical tube and centrifuged at 200 rpm for 5 min. A defined number of cells were transferred into 500-μm-grid cell culture dishes (#81168 Ibidi) and incubated overnight in a growth medium. Cell inserts (# 80209 Ibidi) were applied to minimize the amount of plasmids. Before the laser treatment, the cell samples were washed with a serum-free medium and incubated in an 80 ml Opti-Mem medium (#11058-021, Life Technologies). Three microliters of genes were added into the medium before transfection. The inserts were removed after laser transfection to avoid overgrowth. After transfection, cells were incubated in fresh growth medium at 37°C (5% CO2) and monitored for GFP expression and colony formation for several days after laser exposure using a standard fluorescence microscope.

2.3.

Nonviral Minicircle Deoxyribose Nucleic Acid

Nonviral minicircle DNA, as described by Narsinh et al.,22 was used. The minicircle vector contained the reprogramming factors OCT4, SOX2, NANOG, and LIN28 as well as the GFP reporter gene.20

2.4.

Optoporation with Accelerated Control Software

In order to be able to treat a large number of cells, we implemented means to speed up the process of laser targeting, which is time-demanding if done solely by hand. Of particular importance, in this context, is the automatic identification of the target positions on the cells for laser illumination. The exact locations on the cell, namely, the cell membrane, cytoplasm, or nucleus, as well as the number of illuminated positions per cell, greatly influence the optoporation and the cell survival rate.21 Briefly, the identification is based on the contrast evaluation in bright-field microscope images of the sample. To determine a position for laser targeting, the camera images are subdivided into parallel lines in subsquare regions with edge lengths corresponding to the typical length of the targeted cells, for instance, 10 to 20μm. The cells are identified by determining image contrast changes along the horizontal square edge lines with the built-in LabView function IMAQ Rake 3 (National Instruments, LabView Vision Development Module), which evaluates changes in the image contrast to determine the so-called edge positions along a set of parallel search lines. Figure 3 shows a microscope bright-field image with overlaid search lines and identified “contrast edges,” that is, positions on the cell membranes. The positions are marked by filled circles (blue and yellow). The image contrast changes are only evaluated from left to right along the horizontal lines such that at most one edge position is identified per square, specifically, between neighboring vertical lines. The filled circles mark the automatically determined positions for later laser illumination, which correspond to the periphery of a cell. This results in laser poration of the cell’s membrane away from the nucleus, which is typically located at the center of the cell.

Fig. 3

Microscopic bright-field image of the cell sample with illustration of the cell position finding. Contrast changes in the image are evaluated along the horizontal lines. Between neighboring vertical lines, a maximum of positions is determined. The filled circles mark the found positions. Too close–lying circles (yellow) are removed from the position set.

JBO_20_11_115008_f003.png

Circles that lie very close to each other are in a further step removed from the set of illumination positions by the software to avoid stressing the cells by laser illumination (removed circles are indicated by the yellow color). The separation distance between the horizontal blue lines can be fine-tuned by the user to accommodate different cell sizes and alter the average number of shots per cell. The software for the automated cell optoporation further controls several hardware components such as the microscope stage and optical shutter. After identifying the positions for laser illumination, these are centered in the focal volume of the microscope objective (i.e., “laser focus”), and the mechanical shutter in the laser beam path is opened for a preset illumination duration. Afterward, the same procedure is repeated with all other identified positions inside the microscope field of view (FOV). After addressing all located cell positions inside a microscope FOV, the stage moves to the adjacent region in a meander shape such that a large sample area can be covered in a mosaic pattern.

2.5.

Atomic Force Microscopy

To visualize and characterize holes in cells induced with the quasi-Bessel beam focus geometry, laser-processed cells were imaged with an atomic force microscope (AFM) Dimension™ 3100 (Digital Instruments, Veeco Metrology Group) in tapping mode. The cantilevers (Micro Cantilever, OMCL-AC160TS-C2, Olympus) with silicon tips with a tip diameter of about 5 nm, a spring constant of 40N/m, and a resonance frequency of 300 kHz were used. WSxM 4.0 software was used to analyze the AFM (.stp) files.

2.6.

Fluorescence Microscopy

An Olympus IX70 fluorescence microscope equipped with a high-pressure mercury lamp was used to evaluate GFP expression after the laser procedures. An excitation wavelength of 435 nm (Bandpass 434/17) was used to induce the GFP fluorescence with a maximum at 530 nm. Detection filters BP510/42 and BG 39 were used for GFP fluorescence and bright-field images, respectively.

3.

Results

3.1.

Fast Cell Recognition and Low Power Nanoporation

The laser scanning microscope with automated cell recognition software and an elongated focus was employed to perform nanoporation on a high number of cells. At first, the destructive effects of the quasi-Bessel beam were studied using AFM. If the power and beam dwell times are too high, destructive effects occur not only in the central part of the beam but also in the microenvironment due to the symmetric pattern of a quasi-Bessel beam that consists of an intense central core surrounded with several concentric rings [Fig. 4(a)]. However, only the highly localized destructive effects of the central core are desired. The destructive effects of the less-intense “ring structure pattern” should be avoided [Fig. 4(b)]. When using a beam dwell time of 100 ms and a mean power of 40 mW, precise submicron holes in the cell’s membrane could be achieved. This corresponds to about 8 million laser pulses with a 12-ns pulse interval.

Fig. 4

Atomic force microscope (AFM) images of a cell nucleus, taken after illumination with pulses with a quasi-Bessel beam focal geometry at different mean powers. (a) The AFM image shows the destructive effects of a high-power quasi-Bessel beam consisting of an intense central core surrounded with several concentric rings. (b) Destructive effects occur in the central part of the beam only when using lower mean powers.

JBO_20_11_115008_f004.png

Cells were recognized by fast gray-level evaluation from images obtained by white light source illumination. The simple edge detection algorithm needs about 0.2 ms to identify a cell position. In fact, in our setup, the duration of moving the stage and the laser illumination time itself overwhelmingly determine the speed of the optoporation procedure. The microscope stage needs about 15 ms for a position change of 10μm. Therefore, with an illumination time of 100 ms, about 115 ms is required to identify, position, and laser illuminate a cell position. With an FOV of about 400×400μm2, up to 500 cells can be theoretically nanoporated within 1 min. However, we employed typically up to three positions per cell for laser exposure (Fig. 3). Not all cells were found to be perfectly recognized by the software. Therefore, a realistic value is about 100 nanoporated cells per minute.

In the experiments, the successfully nanoporated cells took up the minicircle DNA including the GFP reporter gene. The integration of the foreign DNA into the cell’s genome resulted in the formation of a green fluorescence as well as significant changes of the cell morphology. Human dermal fibroblasts have a spindle-shaped morphology and grow as monolayer adherent cells in vitro (Fig. 5). Only some of the laser-exposed cells started to change their morphology as well as their fluorescence behavior. In particular, iPS cell-like green-fluorescent dome-shaped colonies appeared as early as four to six days after a single optical surgery procedure (Fig. 5). The number of cell colonies varied from experiment to experiment. In a successful experiment, up to 20% to 30% of colonies per experiment were generated, which shows about 2% to 3% colony-forming efficiency. These colonies are typical for iPS cells and were found in all previous nonoptical iPS generation techniques such as lipofectamine and nucleofector transfection. Positive green fluorescence shows the successful synthesis of the reporter gene GFP and ensured coexpression of the whole set of reprogramming genes OCT4, SOX2, NANOG, and LIN28 in the genome. Figure 5 shows the procedure of virus-free optical reprogramming as well as images of GFP-expressing iPS cell-like colonies. Typically, 10 to 20 cells are estimated per colony.

Fig. 5

Optical reprogramming procedure and detection of induced pluripotent stem (iPS) cell-like colonies. Cell monolayers were prepared in removal inserts 24 h prior to the experiment. Plasmids with transcription factors were given to the medium shortly before the laser nanoporation. Laser holes were created with quasi-Bessel beams at 40 mW mean power at 85 MHz at sub-20 fs. Inserts were removed; cells were washed and then incubated for expansion. Green fluorescence protein (GFP)-expressing dome-shaped iPS cell-like colonies were expected on days 4 to 6. Transfected cells were monitored with a fluorescence microscope for GFP fluorescence. iPS cell-like colonies generated from human dermal fibroblasts as a result of optical reprogramming were detected, where strong GFP expression indicated successful integration of minicircle DNA comprising GFP and four reprogramming factors.

JBO_20_11_115008_f005.png

4.

Discussion

Reprogramming of somatic cells was achieved without the use of viruses. Approximately, 8 million femtosecond laser pulses were applied to the cell’s membrane to create transient holes. This allowed diffusion of the DNA minicircle containing a cocktail of four genes required to induce cell reprogramming as well as reporter gene GFP. Normally, the exact focusing of the laser beam to the cell’s membrane periphery takes a relatively long alignment time. When using quasi-Bessel beams with an elongated focus as demonstrated in this paper, this alignment procedure is no longer required. The fast software-assisted transfection system with cell recognition can realize a high-throughput transfection.

In conventional nonoptical methods, iPS generation requires from 3 to 27 repeated transfections of the same genes to achieve a sustained expression of introduced genes. Typically, the colony forms within 12 to 18 days after the transfection. Narsinh et al.22 have used minicircle DNA transfection of OCT4, SOX2, NANOG, and LIN 28 genes in human stem cells to induce iPS cell-like colonies with dome-like structures at approximately day 12 to 18 following nucleofection and repeated 2× lipofectamine transfection.

Interestingly, in this proof-of-principle study using femtosecond laser pulses, the bright GFP-expressing dome-shaped iPS cell-like colonies occurred even after one single optical nanosurgery procedure. Even more interestingly, the colonies were formed as early as four to six days after laser treatment. It remains challenging to know if and how ultrashort femtosecond laser NIR pulses accelerate the intricate iPS cell generation process. Nonetheless, most recent studies demonstrated that terahertz and low-level laser radiation increases stem cell proliferation and differentiation by the activation of transcription genes.2325 The full iPS cell reprogramming procedure is very complex and is still being studied. Additionally, understanding the role of the laser in colony formation is crucial in future studies, since the complex reprogramming mechanism is dependent not only on the transfection procedure but also on many other chemical (medium, bioactive molecules, factors of reprogramming, precise gene balance, and culture conditions), physical (oxygen pressure, pH, and mechanical forces), and biological (type of cells, age, and origin of cells) factors. Furthermore, a variety of gene cocktails should also be investigated.

5.

Conclusion

Optically generated iPS cells, such as immature pluripotent cells that are capable of developing into all types of cells in the body, are of great interest due to the avoidance of viruses or chemicals to realize gene transfer into the cell and would advance the clinical translation of iPS cells one step further. Our experimental finding demonstrates that the internalization of four reprogramming factors and the reporter gene GFP can be optically realized. In contrast to other transfection and reprogramming methods, the optical reprogramming method provides a contamination-free transfection environment, and physical contact is not required. Virus-free minicircle DNA has been delivered without using further chemicals. The application of ultrashort NIR femtosecond laser nanosurgery enhanced colony-forming efficacy. However, further studies are required to optimize the optical procedure in particular to enhance the iPS cell generation efficiency.

Acknowledgments

The authors wish to thank the German Science Foundation (DFG) for financial support within the Priority Program “SPP1327: optically induced sub-100-nm structures for biomedical and technical applications.”

References

1. 

K. Takahashi and S. Yamanaka, “Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors,” Cell, 126 (4), 663 –676 (2006). http://dx.doi.org/10.1016/j.cell.2006.07.024 CELLB5 0092-8674 Google Scholar

2. 

K. Takahashi et al., “Induction of pluripotent stem cells from adult human fibroblasts by defined factors,” Cell, 131 (5), 861 –872 (2007). http://dx.doi.org/10.1016/j.cell.2007.11.019 CELLB5 0092-8674 Google Scholar

3. 

J. Yu et al., “Induced pluripotent stem cell lines derived from human somatic cells,” Science, 318 (5858), 1917 –1920 (2007). http://dx.doi.org/10.1126/science.1151526 SCIEAS 0036-8075 Google Scholar

4. 

I. de Lazaro, A. Yilmazer and K. Kostarelos, “Induced pluripotent stem (iPS) cells: a new source for cell-based therapeutics?,” J. Controlled Release, 185 37 –44 (2014). http://dx.doi.org/10.1016/j.jconrel.2014.04.011 Google Scholar

5. 

P. Sachamitr, S. Hackett and P. J. Fairchild, “Induced pluripotent stem cells: challenges and opportunities for cancer immunotherapy,” Front. Immunol., 5 176 (2014). http://dx.doi.org/10.3389/fimmu.2014.00176 Google Scholar

6. 

S. Miyazaki et al., “Emerging methods for preparing iPS cells,” Jpn. J. Clin. Oncol., 42 (9), 773 –779 (2012). http://dx.doi.org/10.1093/jjco/hys108 Google Scholar

7. 

S. Kelaini, A. Cochrane and A. Margariti, “Direct reprogramming of adult cells: avoiding the pluripotent state,” Stem Cells Cloning, 7 19 –29 (2014). http://dx.doi.org/10.2147/SCCAA.S38006 Google Scholar

8. 

T. Graf, “Historical origins of transdifferentiation and reprogramming,” Cell Stem Cell, 9 (6), 504 –516 (2011). http://dx.doi.org/10.1016/j.stem.2011.11.012 Google Scholar

9. 

T. Graf and T. Enver, “Forcing cells to change lineages,” Nature, 462 (7273), 587 –594 (2009). http://dx.doi.org/10.1038/nature08533 Google Scholar

10. 

J. Choi et al., “MyoD converts primary dermal fibroblasts, chondroblasts, smooth muscle, and retinal pigmented epithelial cells into striated mononucleated myoblasts and multinucleated myotubes,” Proc. Natl. Acad. Sci. U. S. A., 87 (20), 7988 –7992 (1990). http://dx.doi.org/10.1073/pnas.87.20.7988 Google Scholar

11. 

T. Vierbuchen et al., “Direct conversion of fibroblasts to functional neurons by defined factors,” Nature, 463 (7284), 1035 –1041 (2010). http://dx.doi.org/10.1038/nature08797 Google Scholar

12. 

K. Inagawa et al., “Induction of cardiomyocyte-like cells in infarct hearts by gene transfer of Gata4, Mef2c, and Tbx5,” Circ. Res., 111 (9), 1147 –1156 (2012). http://dx.doi.org/10.1161/CIRCRESAHA.112.271148 Google Scholar

13. 

J. A. Efe et al., “Conversion of mouse fibroblasts into cardiomyocytes using a direct reprogramming strategy,” Nat. Cell Biol., 13 (3), 215 –222 (2011). http://dx.doi.org/10.1038/ncb2164 Google Scholar

14. 

K. Okita, T. Ichisaka and S. Yamanaka, “Generation of germline-competent induced pluripotent stem cells,” Nature, 448 (7151), 313 –317 (2007). http://dx.doi.org/10.1038/nature05934 Google Scholar

15. 

U. K. Tirlapur and K. König, “Cell biology—targeted transfection by femtosecond laser,” Nature, 418 (6895), 290 –291 (2002). http://dx.doi.org/10.1038/418290a Google Scholar

16. 

M. Antkowiak et al., “Femtosecond optical transfection of individual mammalian cells,” Nat. Protoc., 8 (6), 1216 –1233 (2013). http://dx.doi.org/10.1038/nprot.2013.071 Google Scholar

17. 

A. Uchugonova et al., “Targeted transfection of stem cells with sub-20 femtosecond laser pulses,” Opt. Express, 16 (13), 9357 –9364 (2008). http://dx.doi.org/10.1364/OE.16.009357 OPEXFF 1094-4087 Google Scholar

18. 

P. Mthunzi, K. Dholakia and F. Gunn-Moore, “Phototransfection of mammalian cells using femtosecond laser pulses: optimization and applicability to stem cell differentiation,” J. Biomed. Opt., 15 (4), 041507 (2010). http://dx.doi.org/10.1117/1.3430733 Google Scholar

19. 

A. Uchugonova et al., “Nanosurgery of cells and chromosomes using near-infrared twelve-femtosecond laser pulses,” J. Biomed. Opt., 17 (10), 101502 (2012). http://dx.doi.org/10.1117/1.JBO.17.10.101502 JBOPFO 1083-3668 Google Scholar

20. 

D. McGloin and K. Dholakia, “Bessel beams: diffraction in a new light,” Contemp. Phys., 46 (1), 15 –28 (2005). http://dx.doi.org/10.1080/0010751042000275259 CTPHAF 0010-7514 Google Scholar

21. 

H. G. Breunig et al., “Software-aided automatic laser optoporation and transfection of cells,” Sci. Rep., 5 11185 (2015). http://dx.doi.org/10.1038/srep11185 Google Scholar

22. 

K. H. Narsinh et al., “Generation of adult human induced pluripotent stem cells using nonviral minicircle DNA vectors,” Nat. Protoc., 6 (1), 78 –88 (2010). http://dx.doi.org/10.1038/nprot.2010.173 Google Scholar

23. 

J. Bock et al., “Mammalian stem cells reprogramming in response to terahertz radiation,” PLoS One, 5 (12), e15806 (2010). http://dx.doi.org/10.1371/journal.pone.0015806 POLNCL 1932-6203 Google Scholar

24. 

T. Kushibiki et al., “Low reactive level laser therapy for mesenchymal stromal cells therapies,” Stem Cells Int., 2015 974864 (2015). http://dx.doi.org/10.1155/2015/974864 Google Scholar

25. 

F. Ginani et al., “Effect of low-level laser therapy on mesenchymal stem cell proliferation: a systematic review,” Lasers Med. Sci., 30 (8), 2189 –2194 (2015). http://dx.doi.org/10.1007/s10103-015-1730-9 Google Scholar

Biography

Aisada Uchugonova received her master’s degree in biomedical sciences from the National University of Kyrgyzstan and his PhD degree from Saarland University. She served as a postdoc at the UCSD. Now she works in the Department of Biophotonics and Laser Technology, Saarland University, as a PI of the project “optical reprogramming” funded by the German Science Foundation (DFG). Her research interests include multiphoton imaging, optical characterization of stem cells, and nanosurgery.

Hans Georg Breunig received his master’s degree in physics from the University of Maryland at College Park and his PhD in physics from the University of Bremen, Germany. Afterwards, he worked on the control of competing chemical processes with ultrashort pulses at the Philipps-University Marburg. Now he works as a research scientist at JenLab GmbH.

Ana Batista obtained her master’s degree in biomedical engineering from the University of Coimbra. She enrolled in the biomedical engineering PhD program at the University of Coimbra and works curently as a PhD student at the Department of Biophotonics and Laser Technology, Saarland University.

Karsten König studied physics in Rostock, wrote his PhD and habilitation thesis in Jena, and worked at the Institute for Laser Technologies in Ulm, at the Beckman Laser Institute in Irvine, at the Institute for Molecular Biotechnology in Jena and at the University Jena. He is currently full professor for biophotonics and laser technology at Saarland University in Saarbrücken, Germany. He is co-founder of the spin-off company JenLab GmbH.

© 2015 Society of Photo-Optical Instrumentation Engineers (SPIE) 1083-3668/2015/$25.00 © 2015 SPIE
Aisada Uchugonova, Hans Georg Breunig, Ana Batista, and Karsten König "Optical reprogramming of human somatic cells using ultrashort Bessel-shaped near-infrared femtosecond laser pulses," Journal of Biomedical Optics 20(11), 115008 (30 November 2015). https://doi.org/10.1117/1.JBO.20.11.115008
Published: 30 November 2015
Lens.org Logo
CITATIONS
Cited by 6 scholarly publications.
Advertisement
Advertisement
RIGHTS & PERMISSIONS
Get copyright permission  Get copyright permission on Copyright Marketplace
KEYWORDS
Femtosecond phenomena

Microscopes

Green fluorescent protein

Luminescence

Biomedical optics

Stem cells

Atomic force microscopy

RELATED CONTENT

Femtosecond-laser assisted cell reprogramming
Proceedings of SPIE (February 16 2017)
Optical gene transfer by femtosecond laser pulses
Proceedings of SPIE (July 10 2003)

Back to Top