Buy ebook Bioluminescence Sung Bae Kim cheap price
Buy ebook Bioluminescence Sung Bae Kim cheap price
Buy ebook Bioluminescence Sung Bae Kim cheap price
com
https://ebookmeta.com/product/bioluminescence-sung-bae-kim/
OR CLICK HERE
DOWLOAD NOW
https://ebookmeta.com/product/live-cell-imaging-sung-bae-kim/
ebookmeta.com
https://ebookmeta.com/product/mechanics-of-solids-and-fracture-4th-
edition-ho-sung-kim/
ebookmeta.com
https://ebookmeta.com/product/recent-developments-in-neuroscience-
research-on-human-motivation-1st-edition-sung-il-kim/
ebookmeta.com
https://ebookmeta.com/product/you-can-conquer-cancer-a-new-way-of-
living-ian-gawler/
ebookmeta.com
Principles of Neural Science 6th Edition Eric R. Kandel
(Editor)
https://ebookmeta.com/product/principles-of-neural-science-6th-
edition-eric-r-kandel-editor/
ebookmeta.com
https://ebookmeta.com/product/process-safety-for-engineers-an-
introduction-2nd-edition-ccps-center-for-chemical-process-safety/
ebookmeta.com
https://ebookmeta.com/product/kiss-the-girl-1st-edition-zoraida-
cordova/
ebookmeta.com
https://ebookmeta.com/product/a-lion-s-pride-catherine-hapka/
ebookmeta.com
Methods in
Molecular Biology 2525
Bioluminescence
Methods and Protocols
Volume 2
Fourth Edition
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire Hatfield,
Hertfordshire, UK
Fourth Edition
Edited by
Sung-Bae Kim
Environmental Management Research Institute, National Institute of Advanced Industrial
Science and Technology (AIST), Tsukuba, Ibaraki, Japan
Editor
Sung-Bae Kim
Environmental Management Research
Institute
National Institute of Advanced Industrial
Science and Technology (AIST)
Tsukuba, Ibaraki, Japan
© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Science+Business Media, LLC, part
of Springer Nature 2022
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher, whether the whole or part
of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation,
broadcasting, reproduction on microfilms or in any other physical way, and transmission or information storage and
retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter
developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not imply,
even in the absence of a specific statement, that such names are exempt from the relevant protective laws and regulations
and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in this book are believed to
be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty,
expressed or implied, with respect to the material contained herein or for any errors or omissions that may have been
made. The publisher remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Cover Illustration Caption: Longitudinal BLI analysis of Australian bat lyssavirus (ABLV)-luc replication in the same
mouse on Day 3, Day 5, and Day 10 post-infection. For each image, BLI data are overlaid on a CT image of one B6
Albino mouse infected with ABLV-Luc. Scale bars indicate range of the pseudocolor intensity scale that is used to show
mean luminescence intensity (MLI) values. Intensity of viral replication is greatest in the brain.
This Humana imprint is published by the registered company Springer Science+Business Media, LLC, part of Springer
Nature.
The registered company address is: 1 New York Plaza, New York, NY 10004, U.S.A.
Preface
v
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi
vii
viii Contents
PART IV INSTRUMENTATION
23 ATP Sensing Paper with Smartphone Bioluminescence-Based Detection . . . . . . 297
Maria Maddalena Calabretta, Ruslan Alvarez-Diduk, Elisa Michelini,
and Arben Merkoçi
Contents ix
PART V SOFTWARE
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 425
Contributors
KOJI ABE • National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
BYEONG-CHEOL AHN • BK21 FOUR KNU Convergence Educational Program of
Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School
of Medicine, Kyungpook National University, Daegu, Republic of Korea; Department of
Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of
Korea; Department of Nuclear Medicine, Kyungpook National University Hospital,
Daegu, Republic of Korea
RUSLAN ALVAREZ-DIDUK • Nanobioelectronics and Biosensors Group, Catalan Institute of
Nanoscience and Nanotechnology (ICN2), CSIC, The Barcelona Institute of Science and
Technology, Barcelona, Spain
DANILO T. AMARAL • Departamento de Biologia, Centro de Ciências Humanas e Biologicas,
Universidade Federal de São Carlos (UFSCar), Sorocaba, Brazil; Programa de Pos
Graduação em Biologia Comparada, Faculdade de Filosofia, Ciências e Letras de Ribeirão
Preto, Universidade de São Paulo (USP), Ribeirão Preto, Brazil
RABAH AZOUANI • EBI-Ecole de Biologie Industrielle, Cergy, France
YUJI BABA • Graduate School of Bionics, Tokyo University of Technology, Tokyo, Japan
ANURUP GOHAIN BARUA • Department of Physics, Gauhati University, Guwahati, India
KEN BERGLUND • Department of Neurosurgery, Emory University School of Medicine,
Atlanta, GA, USA
ISABEL A. S. BONATELLI • Departamento de Ecologia e Biologia Evolutiva, Universidade
Federal de São Paulo (UNIFESP), São Paulo, Brazil
MARIA MADDALENA CALABRETTA • Department of Chemistry “Giacomo Ciamician”,
University of Bologna, Bologna, Italy; Center for Applied Biomedical Research (CRBA),
Azienda Ospedaliero-Universitaria Policlinico S. Orsola-Malpighi, Bologna, Italy
ZHENG CAO • Department of Chemical and Biomolecular Engineering, School of
Engineering, University of California, Los Angeles, Los Angeles, CA, USA
ROXANNE CASTILLO • Department of Chemical and Biomolecular Engineering, School of
Engineering, University of California, Los Angeles, Los Angeles, CA, USA
PETER L. CHOYKE • Molecular Imaging Branch, Center for Cancer Research, National
Cancer Institute, National Institutes of Health, Bethesda, MD, USA
RANIA DADI • LSPM-CNRS, Laboratoire des Sciences des Procédés et des Matériaux,
Université Paris 13, Sorbonne Paris Cité, Villetaneuse, France
ABHIJIT DE • Molecular Functional Imaging Lab, ACTREC, Tata Memorial Centre, Navi
Mumbai, India
SEBASTIÁN A. DÍAZ • Center for Bio/Molecular Science and Engineering, U.S. Naval
Research Laboratory Code 6900, Washington, DC, USA
JEFFREY F. DIBERTO • Department of Pharmacology, University of North Carolina at Chapel
Hill School of Medicine, Chapel Hill, NC, USA
MARTIN M. EDREIRA • Universidad de Buenos Aires, Facultad de Ciencias Exactas y
Naturales, Departamento de Quı́mica Biologica, Buenos Aires, Argentina; CONICET-
Universidad de Buenos Aires, Instituto de Quı́mica Biologica de la Facultad de Ciencias
Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina; Department of
xi
xii Contributors
YUKA IWASAKI • Graduate School of Bionics, Tokyo University of Technology, Tokyo, Japan
TAKASHI JIN • RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
RYOICHIRO KAGEYAMA • Institute for Frontier Life and Medical Sciences, Kyoto University,
Kyoto, Japan; Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University,
Kyoto, Japan; RIKEN Center for Brain Science, Wako, Japan
MASAMITSU KANADA • Molecular Imaging Program at Stanford, Stanford University School
of Medicine, Palo Alto, CA, USA; Canary Center at Stanford for Cancer Early Detection,
Stanford University School of Medicine, Palo Alto, CA, USA
ANDREI KANAEV • LSPM-CNRS, Laboratoire des Sciences des Procédés et des Matériaux,
Université Paris 13, Sorbonne Paris Cité, Villetaneuse, France
SUNG-BAE KIM • Environmental Management Research Institute, National Institute of
Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
SHINAE KIZAKA-KONDOH • School of Life Science and Technology, Tokyo Institute of
Technology, Yokohama, Japan
ULLA G. KNAUS • Conway Institute, School of Medicine, University College Dublin, Dublin,
Ireland
HISATAKA KOBAYASHI • Molecular Imaging Branch, Center for Cancer Research, National
Cancer Institute, National Institutes of Health, Bethesda, MD, USA
LIWEI LANG • Department of Oral Biology and Diagnostic Sciences, Georgia Cancer Center,
Augusta University, Augusta, GA, USA
ALEX LEMARCHAND • LSPM-CNRS, Laboratoire des Sciences des Procédés et des Matériaux,
Université Paris 13, Sorbonne Paris Cité, Villetaneuse, France
QI LI • Department of Pharmaceutics, College of Pharmacy, Third Military Medical
University (Army Medical University), Chongqing, China; Department of Biomedical
Engineering and Medical Imaging, Third Military Medical University (Army Medical
University), Chongqing, China
YA-LI LIU • Research Center for Translational Medicine at East Hospital, School of Life
Sciences and Technology, Tongji University, Shanghai, China
YI LIU • Molecular Imaging Program at Stanford, Stanford University School of Medicine,
Palo Alto, CA, USA; Canary Center at Stanford for Cancer Early Detection, Stanford
University School of Medicine, Palo Alto, CA, USA
YUNFENG LU • Department of Chemical and Biomolecular Engineering, School of
Engineering, University of California, Los Angeles, Los Angeles, CA, USA
TARIK F. MASSOUD • Molecular Imaging Program at Stanford, Stanford University School of
Medicine, Palo Alto, CA, USA; Canary Center at Stanford for Cancer Early Detection,
Stanford University School of Medicine, Palo Alto, CA, USA
SHINJI MASUDA • Department of Life Science and Technology, Tokyo Institute of Technology,
Yokohama, Kanagawa, Japan
DIVITA MATHUR • Center for Bio/Molecular Science and Engineering, U.S. Naval Research
Laboratory Code 6900, Washington, DC, USA; College of Science, George Mason
University, Fairfax, VA, USA
IGOR L. MEDINTZ • Center for Bio/Molecular Science and Engineering, U.S. Naval Research
Laboratory Code 6900, Washington, DC, USA
ARBEN MERKOÇI • Nanobioelectronics and Biosensors Group, Catalan Institute of
Nanoscience and Nanotechnology (ICN2), CSIC, The Barcelona Institute of Science and
Technology, Barcelona, Spain; Catalan Institution for Research and Advanced Studies
(ICREA), Barcelona, Spain
xiv Contributors
Heterogeneous Conjugates
Chapter 1
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a cell-specific cancer therapy in which antibody–photo-
absorber conjugates (APCs) are activated by NIR light to induce rapid immunogenic cell death with
minimal off-target effects. In preclinical settings, bioluminescence imaging (BLI) is useful to quantitatively
assess the efficacy of NIR-PIT for both in vitro and in vivo experiments, especially in the early phase of
testing. Here, we describe the detailed methods of the experiments for NIR-PIT and evaluation of its
efficacy using BLI.
1 Introduction
3
4 Ryuhei Okada et al.
2 Materials
Fig. 1 NIR light sources. Either LED or laser light source of 689 nm is used. Laser
diffusers consist of frontal and cylindrical diffusers. The light from a frontal
diffuser can be changed to a parallel beam with a collimator. For interstitial light
exposure, including endoscopic procedures, cylindrical diffusers are used
2.2 In Vitro PIT 1. A431-luc cell line (hEGFR- and FLuc-expressing cells).
2. Culture medium: add 50 mL of fetal bovine serum (FBS) and
5 mL of 100 antibiotics (penicillin–streptomycin; P/S) to
500 mL of RPMI 1640 medium with or without phenol red.
3. 0.05% (w/v) trypsin/EDTA solution: the diluted solution of
50 mL of 0.5% trypsin/EDTA with 500 mL of PBS.
4. Trypan blue stain solution.
5. NIR light source (e.g., LED or laser) (see Note 1, Fig. 1).
3 Methods
Fig. 2 Purification of APC. (a) PBS is used for the purification of the APC. APC can be confirmed as a blue band
(solid arrow), while unconjugated dye remains at a higher level than the APC band (dashed arrow). (b) Just
before the APC collection. (c) Collection of the APC
3.2 In Vitro NIR-PIT 1. Harvest exponentially growing A431-luc cells with 0.05%
(w/v) trypsin/EDTA solution and centrifuge the cells for
3–5 min at 1500 RPM, at 4 C.
2. Count the cell number with trypan blue staining, and re-plate
the cells at appropriate density (2–3 105 cells) in each well of
the 12-well microplate.
3. Incubate 1 day in a CO2 incubator to allow the cell attachment
to the bottom of the well.
4. Aspirate the cell culture medium and add a fresh medium
containing mAb–IR700 conjugate (final concentration:
10 μg/mL). The optimal concentration of the mAb–IR700
conjugate will vary for each mAb; however, 10 μg/mL is
generally sufficient.
5. Incubate for 1 h at 4 or 37 C in a dark space (see Note 5).
6. Aspirate the cell culture medium, wash the cells with PBS, and
add phenol red-free complete medium (see Note 6).
7. Expose the cells to NIR light without the microplate cover.
8. Incubate for 1 h at 37 C.
9. Add 300 μg of D-luciferin (20 μL of D-luciferin solution) per
1 mL medium and incubate for 2 min (see Note 7).
10. Count the photon number for 10–30 s with a BL imager (see
Note 8, Fig. 3).
3.3 In Vivo NIR-PIT 1. Harvest exponentially growing A431-luc cells with 0.05%
(w/v) trypsin, and collect the cells into a centrifuge tube with
the same volume of RPMI 1640 medium as the trypsin/EDTA
solution.
2. Centrifuge the cells for 3–5 min at 1500 rpm, at 4 C.
3. Resuspend the cell pellet with cold PBS and quantify the cells
with a hemocytometer or a cell counter and dilute cell suspen-
sion with PBS so that two million cells are contained in 100 μL
(see Note 9).
4. Keep the cell suspension on ice before use.
5. Anesthetize the mouse with 3% (v/v) isoflurane.
6. Mix the cell suspension thoroughly before injection.
7. Aspirate the cell suspension with a 1-mL syringe and 25–27 G
needle, and inject 100 μL of the cells subcutaneously into the
right dorsum of the mice. Withdraw the needle slowly.
8. Monitor the mouse daily and examine the tumor volume peri-
odically by caliper. Calculate tumor volume as (major axis of
tumor) (minor axis of tumor)2 0.5.
9. Once the tumor volume reaches ~100 mm3, assign the tumor-
bearing mice randomly to either the treatment group or non-
treatment control group (see Note 10).
10. Anesthetize the mice with isoflurane and slowly inject mAb–
IR700 conjugate into the tail vein with a 1-mL syringe and
27–30 G needle (see Note 11).
11. On the following day, confirm mAb–IR700 accumulation in
the tumor with in vivo FLFL imaging (see Note 12), followed
by anesthesia of the mice with isoflurane and intraperitoneal
(i.p.) injection of pentobarbital (~750 μg/body).
12. Expose the entire tumor to NIR light under deep anesthesia
(see Note 13, Fig. 4). Upon external NIR light exposure, place
a piece of aluminum foil with a hole of approximately half-inch
diameter (corresponding to the tumor) over the mouse to
ensure that the NIR light exposure is limited to the tumor
site. When performing interstitial light exposure, cylindrical
diffusers are placed through an i.v. catheter or endoscope.
13. Place the mice on heating pads and observe until the recovery
from anesthesia.
14. Monitor the mouse daily and examine the tumor volume
periodically by caliper.
15. Analyze treatment effects and survival outcomes by comparing
them with the control tumors (see Note 14).
Bioluminescence Imaging in NIR-PIT Experiments 9
Fig. 4 Light exposure in in vivo NIR-PIT. (a–c) An example of external light exposure. (a) Mouse is located
under the laser collimator (*). The black arrow represents the location of the tumor. (b) Upon NIR light
exposure, a piece of aluminum foil with a hole is placed over the mouse to shield the rest of the mouse. (c) NIR
light is applied onto the tumor site through the hole. (d) An example of interstitial light exposure. A cylindrical
diffuser is inserted through the intravenous (i.v.) catheter to allow deeper penetration of interstitial light
3.4 BL Assessment 1. Inject D-luciferin solution (15 mg/mL, 200 μL) i.p. before
of the Efficacy of In and after NIR-PIT under anesthesia with isoflurane.
Vivo NIR-PIT 2. Obtain cumulative images for 2–3 min with a BL imager (Pho-
ton Imager) 10–15 min after the injection of the luciferin
solution (see Note 15). Perform this imaging before and after
NIR-PIT (see Fig. 5).
3. For quantitative analysis, place regions of interest (ROIs) over
the entire tumor using analysis software that comes with the
imager or separate software.
4. Calculate the photon counts per minute of relative light units
(RLU).
5. Calculate %RLU as (RLU of post-treatment)/(RLU of
pre-treatment) 100.
6. Analyze the treatment effects by comparing them with the
control tumors.
10 Ryuhei Okada et al.
Fig. 5 In vivo imaging before and after the NIR-PIT. Luciferase-expressing A431 cell line and Panitumumab–
IR700 conjugate are exemplified. Top: The 700-nm FL images. The FL of IR700 is immediately quenched after
the NIR light exposure. Bottom: The BL images with luciferase/luciferin reaction. The luciferase activity
dramatically decreases 1 day after the light exposure
4 Notes
Acknowledgments
References
1. Mitsunaga M, Ogawa M, Kosaka N, Rosen- associated regulatory T cells with near-infrared
blum LT, Choyke PL, Kobayashi H (2011) photoimmunotherapy. Sci Transl Med 8(352):
Cancer cell-selective in vivo near infrared 352ra110. https://do i.org/10.1126/
photoimmunotherapy targeting specific mem- scitranslmed.aaf6843
brane molecules. Nat Med 17(12): 8. Okada R, Maruoka Y, Furusawa A, Inagaki F,
1685–1691. https://doi.org/10.1038/nm. Nagaya T, Fujimura D, Choyke PL, Kobayashi
2554 H (2019) The effect of antibody fragments on
2. Kobayashi H, Choyke PL (2019) Near-infrared CD25 targeted regulatory T cell near-infrared
photoimmunotherapy of cancer. Acc Chem photoimmunotherapy. Bioconjug Chem
Res 52(8):2332–2339. https://doi.org/10. 30(10):2624–2633. https://doi.org/10.
1021/acs.accounts.9b00273 1021/acs.bioconjchem.9b00547
3. Kobayashi H, Griffiths GL, Choyke PL (2020) 9. Okada R, Kato T, Furusawa A, Inagaki F,
Near-infrared photoimmunotherapy: photoac- Wakiyama H, Choyke PL, Kobayashi H
tivatable antibody-drug conjugates (ADCs). (2021) Local depletion of immune checkpoint
Bioconjug Chem 31(1):28–36. https://doi. ligand CTLA4 expressing cells in tumor beds
org/10.1021/acs.bioconjchem.9b00546 enhances antitumor host immunity. Adv Ther
4. Ogawa M, Tomita Y, Nakamura Y, Lee MJ, (Weinh) 4(5):2000269. https://doi.org/10.
Lee S, Tomita S, Nagaya T, Sato K, 1002/adtp.202000269
Yamauchi T, Iwai H, Kumar A, Haystead T, 10. Maruoka Y, Furusawa A, Okada R, Inagaki F,
Shroff H, Choyke PL, Trepel JB, Kobayashi Fujimura D, Wakiyama H, Kato T, Nagaya T,
H (2017) Immunogenic cancer cell death Choyke PL, Kobayashi H (2020) Combined
selectively induced by near infrared photoim- CD44- and CD25-targeted near-infrared
munotherapy initiates host tumor immunity. photoimmunotherapy selectively kills cancer
Oncotarget 8(6):10425–10436. https://doi. and regulatory T cells in syngeneic mouse can-
org/10.18632/oncotarget.14425 cer models. Cancer Immunol Res 8(3):
5. Sato K, Ando K, Okuyama S, Moriguchi S, 3 4 5 – 3 5 5 . h t t p s : // d o i . o r g / 1 0 . 1 1 5 8 /
Ogura T, Totoki S, Hanaoka H, Nagaya T, 2326-6066.Cir-19-0517
Kokawa R, Takakura H, Nishimura M, 11. Okada R, Furusawa A, Vermeer DW, Inagaki F,
Hasegawa Y, Choyke PL, Ogawa M, Kobayashi Wakiyama H, Kato T, Nagaya T, Choyke PL,
H (2018) Photoinduced ligand release from a Spanos WC, Allen CT, Kobayashi H (2021)
silicon phthalocyanine dye conjugated with Near-infrared photoimmunotherapy targeting
monoclonal antibodies: a mechanism of cancer human-EGFR in a mouse tumor model simu-
cell cytotoxicity after near-infrared photoim- lating current and future clinical trials. EBio-
munotherapy. ACS Cent Sci 4(11): Medicine 67:103345. https://doi.org/10.
1559–1569. https://doi.org/10.1021/ 1016/j.ebiom.2021.103345
acscentsci.8b00565 12. Nagaya T, Friedman J, Maruoka Y, Ogata F,
6. Ogata F, Nagaya T, Okuyama S, Maruoka Y, Okuyama S, Clavijo PE, Choyke PL, Allen C,
Choyke PL, Yamauchi T, Kobayashi H (2017) Kobayashi H (2019) Host immunity following
Dynamic changes in the cell membrane on near-infrared photoimmunotherapy is
three dimensional low coherent quantitative enhanced with PD-1 checkpoint blockade to
phase microscopy (3D LC-QPM) after treat- eradicate established antigenic tumors. Cancer
ment with the near infrared photoimmu- Immunol Res 7(3):401–413. https://doi.org/
notherapy. Oncotarget 8(61): 10.1158/2326-6066.Cir-18-0546
104295–104302. https://doi.org/10. 13. Maruoka Y, Furusawa A, Okada R, Inagaki F,
18632/oncotarget.22223 Fujimura D, Wakiyama H, Kato T, Nagaya T,
7. Sato K, Sato N, Xu B, Nakamura Y, Nagaya T, Choyke PL, Kobayashi H (2020) Near-infrared
Choyke PL, Hasegawa Y, Kobayashi H (2016) photoimmunotherapy combined with CTLA4
Spatially selective depletion of tumor- checkpoint blockade in syngeneic mouse
Bioluminescence Imaging in NIR-PIT Experiments 13
cancer models. Vaccines (Basel) 8(3):528. Paus R (2001) A comprehensive guide for the
https://doi.org/10.3390/vaccines8030528 accurate classification of murine hair follicles in
14. Maruoka Y, Nagaya T, Nakamura Y, Sato K, distinct hair cycle stages. J Invest Dermatol
Ogata F, Okuyama S, Choyke PL, Kobayashi H 117(1):3–15. https://doi.org/10.1046/j.
(2017) Evaluation of early therapeutic effects 0022-202x.2001.01377.x
after near-infrared photoimmunotherapy 17. Okuyama S, Nagaya T, Ogata F, Maruoka Y,
(NIR-PIT) using luciferase-luciferin photon- Sato K, Nakamura Y, Choyke PL, Kobayashi H
counting and fluorescence imaging. Mol (2017) Avoiding thermal injury during near-
Pharm 14(12):4628–4635. https://doi.org/ infrared photoimmunotherapy (NIR-PIT): the
10.1021/acs.molpharmaceut.7b00731 importance of NIR light power density. Onco-
15. Sato K, Watanabe R, Hanaoka H, Nakajima T, target 8(68):113194–113201. https://doi.
Choyke PL, Kobayashi H (2016) Comparative org/10.18632/oncotarget.20179
effectiveness of light emitting diodes (LEDs) 18. O’Neill K, Lyons SK, Gallagher WM, Curran
and lasers in near infrared photoimmunother- KM, Byrne AT (2010) Bioluminescent imag-
apy. Oncotarget 7(12):14324–14335. https:// ing: a critical tool in pre-clinical oncology
doi.org/10.18632/oncotarget.7365 research. J Pathol 220(3):317–327. https://
16. Müller-Röver S, Handjiski B, van der Veen C, doi.org/10.1002/path.2656
Eichmüller S, Foitzik K, McKay IA, Stenn KS,
Chapter 2
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains a deadly disease despite concerted efforts to
improve its diagnosis and treatment in recent decades. Metastasis of advanced HNSCC nearly always occurs
first in neck lymph nodes before the development of distant metastasis. However, the development of
preclinical animal models and therapeutic treatments for metastatic HNSCC is lagged from bench to clinic.
In this protocol, we exemplify an orthotopic tongue tumor model that can recapitulate the cervical
lymphatic metastases of HNSCC and the application to study the effect of novel saracatinib-loaded
nanoparticles (Nano-Sar). By taking advantage of bioluminescence imaging (BLI), the present protocol
reveals the strong anti-metastatic efficacy of Nano-Sar in the experimental setup. Collectively, the protocol
with a novel metastatic mouse model shows great potential to evaluate treatments on metastatic diseases
with the aid of bioluminescent technology.
Key words HNSCC, The orthotopic model, Lymphatic metastases, Saracatinib, Nanoparticles,
Bioluminescence (BL)
1 Introduction
15
16 Liwei Lang and Yong Teng
2 Materials
2.3 Animals Six-week-old NSG mice (Jackson Laboratory, Bar Harbor, ME,
USA).
3 Methods
3.1 Generation of 1. Seed HN12 cells in a 35-mm culture dish with 90% confluency,
HN12-Luc2 Cells (See and incubate cells at 37 C under 5% (v/v) CO2 overnight.
Note 4) 2. Transfect 1 μg of pGL4.50 into HN12 cells using
Lipofectamine 3000.
3. After 24 h following transfection, seed the cells in five 96-well
black-frame microplates using limiting dilution to obtain
monoclones.
4. Incubate the cells with 600 μg/mL of hygromycin B and select
hygromycin-resistant cells over 3 weeks.
5. Subculture single hygromycin-resistant resistant clones for
expansion.
6. Determine the FLuc expression in each clone using Steady-
Glo® Luciferase Reporter Assay system. Get the reading of
relative light unit with a GloMax® 20/20 luminometer.
3.2 Establishing the 1. Harvest HN12-Luc2 cells and prepare cell suspension with
Orthotopic Tongue DMEM medium/Matrigel (v/v, 3:1) mixture in a concentra-
Tumor Model in NSG tion of 4 105 cells.
Mice (See Note 5) 2. Anaesthetize NSG mice with isoflurane using an EZ-AF9000
auto flow anesthesia system, and inject 50 μL of HN12-Luc2
18 Liwei Lang and Yong Teng
Fig. 1 Nano-Sar inhibits lymph node metastasis of orthotopic tongue tumor in NSG mice. (a) The expression of
FLuc in HN12-Luc2 cells. The reading of relative light unit (RLU) is measured in 10,000 cells. (b) Lymph node
metastasis is monitored on Day 20 after indicated drug treatment by examining BLI using an IVIS-200 in vivo
imaging system. Representative BL images and quantitative data are respectively shown in the left and right
panels, **p < 0.01
4 Notes
Noin tunnin kuluttua nousi eräs mies vitkaan ylös. Kekäle kädessä
tarkasteli hän jokaista ruumista.
— Veljenne varmaankin?
11.
UIVA SAARI.
Samaan aikaan kokoontuivat intiaanit nuotion ääreen
neuvottelemaan, miten uudelleen hyökkäisivät kullanetsijäin leiriin;
mutta tällä välin palatkaamme me noiden kolmen miehen seuraan,
jotka pitkäksi ajaksi olemme syrjään jättäneet.
Oli noin neljän aika iltapäivällä. Erämaassa oli hiljaista, usva alkoi
hitaasti nousta joesta, jossa se saari oli, mihin Rosenholz, Fabian ja
José olivat asettuneet.
— Kas niin, mitä sanoin. Etkö kuule tuota ilkeää ulvontaa? Hirvi
rukka!
Ääretön aavikko, joka vielä äsken oli autio, oli äkkiä muuttunut
mellastuksen ja hämmingin näyttämöksi. Tuon ahdinkoon pakotetun
hirven oli uudelleen täytynyt palata rannalle, josta se tuulen
nopeudella pakeni, susien ulvoen ajaessa sitä takaa. Kesyttömät
hevoset juoksivat intiaanien edessä, joiden huuto ei ollut vähempi
kuin petojenkaan. Kaiku toisti susien ja apahien kamalaa kiljuntaa.
Mutta hetkinen, jonka hän oli epäröinyt minne pakenisi, oli riittänyt
päästämään intiaanit lähemmäksi.
— Tuo onneton on hukassa, yrittäköön mitä tahansa, huudahti
Rosenholz; hän ei ehdi enää joen ylitse.
12.
MUSTALINTU.
— Yhtä helppoa on, alkoi hän taasen, seurata valkoisten jälkiä kuin
aavikkojen puhvelin. Puhvelin jäljet ilmaisevat intiaanille sen ijän,
lihavuuden tai laihuuden, sen juoksun nopeuden vieläpä päivänkin,
milloin se on paikalla ollut. Uivan kätkyen, kaislikon takana on siis
mies, yhtä väkevä kuin puhveli, ja pitempi kuin pisin pyssy; hänen
kanssaan on puoleksi etelästä, puoleksi pohjoisesta kotoisin oleva
soturi, sekä nuori soturi etelän rotua, mutta näiden liitto
ensinmainitun kanssa todistaa, että he ovat etelän valkoisten
vihollisia, sillä heikompi etsii aina väkevämmän ystävyyttä ja on sen
puolella.
— Kas vaan! sanoi José; intiaani puhuu selvää kieltä, hän sanoo,
mitä on sydämellään; eipä ollut hullumpaa, että tahtoi meistä
liittolaisia.
Juuri kun José oli lausunut nämä lohduttavat sanat, palasivat nuo
viisi ratsastajaa, jotka olivat poistuneet päällikön käskystä; he olivat
kiireestä kantapäähän varustetut, heillä oli pyssyjä ja jousia. He
olivat uudestaan noutaneet aseensa, jotka olivat panneet pois,
voidaksensa helpommin ajaa takaa hevosia, ja taasen poistui viisi
ratsastajaa.
— Niinkuin tahdot.
— Ei, minä jään tähän, sanoi Fabian päättävästi. Minä olen nuorin;
en saa peljätä vaaraa, vaan koetan suojella teitä.
— Ei ole ennen kolme miestä ollut niin suuressa vaarassa kuin me;
vihollisemme ovat seitsemän kertaa lukuisammat meitä. Vaikka
jokainen meistä kaataisikin kuusi noita koiria, ovat he sittenkin lähes
yhtä lukuisat kuin mekin.