Real-time In vivo Dual-color Imaging of Intracapillary ... · Yamamoto et al. (12) reported the...

8
Real-time In vivo Dual-color Imaging of Intracapillary Cancer Cell and Nucleus Deformation and Migration Kensuke Yamauchi, 1,2,3 Meng Yang, 1 Ping Jiang, 1 Norio Yamamoto, 1,2,3 Mingxu Xu, 1 Yasuyuki Amoh, 1,4 Kazuhiko Tsuji, 1,5 Michael Bouvet, 1,2 Hiroyuki Tsuchiya, 3 Katsuro Tomita, 3 A.R. Moossa, 1,2 and Robert M. Hoffman 1,2 1 AntiCancer, Inc., and 2 Department of Surgery, University of California, San Diego, California; 3 Department of Orthopedic Surgery, School of Medicine, Kanazawa University, Takaramachi, Kanazawa, Ishikawa; 4 Department of Dermatology, School of Medicine, Kitasato University, Kitasato, Sagamihara, Kanagawa; and 5 Department of Pharmacology and Toxicology, School of Medicine, Kyorin University, Shinkawa, Mitaka, Tokyo, Japan Abstract The mechanism of cancer cell deformation and migration in narrow vessels is incompletely understood. In order to visualize the cytoplasmic and nuclear dynamics of cells migrating in capillaries, red fluorescent protein was expressed in the cytoplasm, and green fluorescent protein, linked to histone H2B, was expressed in the nucleus of cancer cells. Immediately after the cells were injected in the heart of nude mice, a skin flap on the abdomen was made. With a color CCD camera, we could observe highly elongated cancer cells and nuclei in capillaries in the skin flap in living mice. The migration velocities of the cancer cells in the capillaries were measured by capturing images of the dual-color fluorescent cells over time. The cells and nuclei in the capillaries elongated to fit the width of these vessels. The average length of the major axis of the cancer cells in the capillaries increased to approximately four times their normal length. The nuclei increased their length 1.6 times in the capillaries. Cancer cells in capillaries over 8 Mm in diameter could migrate up to 48.3 Mm/hour. The data suggests that the minimum diameter of capillaries where cancer cells are able to migrate is approximately 8 Mm. The use of the dual-color cancer cells differentially labeled in the cytoplasm and nucleus and associated fluorescent imaging provide a powerful tool to understand the mechanism of cancer cell migration and deformation in small vessels. (Cancer Res 2005; 65(10): 4246-52) Introduction Animal models of cancer that use the stable expression of green fluorescent protein (GFP) have made it possible to directly observe cell behavior in primary tumors in live animals (1, 2). The interaction of cells with the extracellular matrix and entry of tumor cells into the circulation in the primary tumor are important factors in metastasis (1). Before the introduction of GFP and its derivatives, intravital- imaging studies were limited to the study of tumor cells that were transiently labeled with vital dyes. Stable fluorescent labeling via GFP expression vectors now allows direct imaging at the single-cell level in vivo (1–4). Initial studies of tumor biology that used stable GFP expression focused on static images and examination of metastases (1, 2). The first use of stable GFP expression to characterize cancer cells in vivo was by Chishima et al. (5). The first use of GFP to observe motility and shape changes of carcinoma cells in live intact tumors in vivo was described by Farina et al. (6). Cancer cells that escape from the primary site into the blood circulation eventually flow to the capillaries of the organs of the body (7). In vivo video microscopy has shown that both lung and liver capillaries are very efficient at arresting the flow of cancer cells (7). Most circulating cancer cells arrest by size restriction. Capillaries are small, typically 3 to 8 Am in diameter. Capillaries allow the passage of RBC which average 7 Am in diameter and are highly deformable (7). However, many cancer cells are large being 20 Am or more in diameter (7). Flow or arrest in capillaries is determined by physical factors, such as the relative sizes of the cells and the capillaries, the blood pressure in the organ and the deformability of the cell (7). Cancer cells can, under certain conditions, undergo adhesive arrest in the capillary vessels that are larger than the cell diameter. Chishima et al. (5) and Huang et al. (8) showed that GFP- transduced cancer cells allowed the imaging of tumor cells in blood vessels. To examine cell behavior during intravasation, Wyckoff et al. (9) have used GFP imaging to view these cells in time-lapse images within a single optical section using a confocal microscope. In vivo imaging of the primary tumors indicated that both metastatic and nonmetastatic cells are motile and show protrusive activity. Metastatic cells show greater orientation toward blood vessels. Nonmetastatic cells fragment when interacting with vessels. Naumov et al. (3), using GFP imaging, visualized fine cellular details such as pseudopodial projections, even after extended periods of in vivo growth. Mook et al. (10) visualized initial arrest of GFP colon cancer cells in sinusoids of the liver due to size restriction after portal vein injection. Tumor cells divided exclusively intravascularly during the first 4 days. Al-Mehdi et al. (11) observed the steps in early hematogenous metastasis of tumor cells expressing GFP in subpleural microvessels in intact, perfused mouse and rat lungs. Metastatic tumor cells attached to the endothelia of pulmonary precapillary arterioles and capillaries. Extravasation of tumor cells was rare. Early tumor colonies were observed growing entirely within the blood vessels. Yamamoto et al. (12) reported the genetic engineering of dual- color fluorescent cells with one color in the nucleus and the other in the cytoplasm. The dual-color cells enable real-time nuclear- cytoplasmic dynamics to be visualized in living cells in vivo as well as in vitro . To obtain the dual-color cells, red fluorescent protein (RFP) was expressed in the cytoplasm of HT-1080 human fibrosarcoma cells, and GFP linked to histone H2B was expressed in the nucleus. Nuclear GFP expression enabled visualization of Requests for reprints: Robert M. Hoffman, AntiCancer, Inc., 7917 Ostrow Street, San Diego, CA 92111. Phone: 858-654-2555; Fax: 858-268-4175; E-mail: [email protected]. I2005 American Association for Cancer Research. Cancer Res 2005; 65: (10). May 15, 2005 4246 www.aacrjournals.org Research Article Research. on August 14, 2020. © 2005 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Transcript of Real-time In vivo Dual-color Imaging of Intracapillary ... · Yamamoto et al. (12) reported the...

Page 1: Real-time In vivo Dual-color Imaging of Intracapillary ... · Yamamoto et al. (12) reported the genetic engineering of dual-color fluorescent cells with one color in the nucleus and

Real-time In vivo Dual-color Imaging of Intracapillary Cancer Cell and

Nucleus Deformation and Migration

Kensuke Yamauchi,1,2,3

Meng Yang,1Ping Jiang,

1Norio Yamamoto,

1,2,3Mingxu Xu,

1

Yasuyuki Amoh,1,4Kazuhiko Tsuji,

1,5Michael Bouvet,

1,2Hiroyuki Tsuchiya,

3

Katsuro Tomita,3A.R. Moossa,

1,2and Robert M. Hoffman

1,2

1AntiCancer, Inc., and 2Department of Surgery, University of California, San Diego, California; 3Department of Orthopedic Surgery, School ofMedicine, Kanazawa University, Takaramachi, Kanazawa, Ishikawa; 4Department of Dermatology, School of Medicine, Kitasato University,Kitasato, Sagamihara, Kanagawa; and 5Department of Pharmacology and Toxicology, School of Medicine, Kyorin University, Shinkawa,Mitaka, Tokyo, Japan

Abstract

The mechanism of cancer cell deformation and migration innarrow vessels is incompletely understood. In order tovisualize the cytoplasmic and nuclear dynamics of cellsmigrating in capillaries, red fluorescent protein was expressedin the cytoplasm, and green fluorescent protein, linked tohistone H2B, was expressed in the nucleus of cancer cells.Immediately after the cells were injected in the heart of nudemice, a skin flap on the abdomen was made. With a color CCDcamera, we could observe highly elongated cancer cells andnuclei in capillaries in the skin flap in living mice. Themigration velocities of the cancer cells in the capillaries weremeasured by capturing images of the dual-color fluorescentcells over time. The cells and nuclei in the capillarieselongated to fit the width of these vessels. The average lengthof the major axis of the cancer cells in the capillaries increasedto approximately four times their normal length. The nucleiincreased their length 1.6 times in the capillaries. Cancer cellsin capillaries over 8 Mm in diameter could migrate up to 48.3Mm/hour. The data suggests that the minimum diameter ofcapillaries where cancer cells are able to migrate isapproximately 8 Mm. The use of the dual-color cancer cellsdifferentially labeled in the cytoplasm and nucleus andassociated fluorescent imaging provide a powerful tool tounderstand the mechanism of cancer cell migration anddeformation in small vessels. (Cancer Res 2005; 65(10): 4246-52)

Introduction

Animal models of cancer that use the stable expression ofgreen fluorescent protein (GFP) have made it possible to directlyobserve cell behavior in primary tumors in live animals (1, 2). Theinteraction of cells with the extracellular matrix and entry of tumorcells into the circulation in the primary tumor are importantfactors in metastasis (1).Before the introduction of GFP and its derivatives, intravital-

imaging studies were limited to the study of tumor cells that weretransiently labeled with vital dyes. Stable fluorescent labeling viaGFP expression vectors now allows direct imaging at the single-celllevel in vivo (1–4).Initial studies of tumor biology that used stable GFP expression

focused on static images and examination of metastases (1, 2). The

first use of stable GFP expression to characterize cancer cells in vivowas by Chishima et al. (5). The first use of GFP to observe motilityand shape changes of carcinoma cells in live intact tumors in vivowas described by Farina et al. (6).Cancer cells that escape from the primary site into the blood

circulation eventually flow to the capillaries of the organs of the body(7). In vivo video microscopy has shown that both lung and livercapillaries are very efficient at arresting the flow of cancer cells (7).Most circulating cancer cells arrest by size restriction. Capillaries aresmall, typically 3 to 8 Am in diameter. Capillaries allow the passage ofRBC which average 7 Am in diameter and are highly deformable (7).However, many cancer cells are large being 20 Am or more indiameter (7). Flow or arrest in capillaries is determined by physicalfactors, such as the relative sizes of the cells and the capillaries, theblood pressure in the organ and the deformability of the cell (7).Cancer cells can, under certain conditions, undergo adhesive arrestin the capillary vessels that are larger than the cell diameter.Chishima et al. (5) and Huang et al. (8) showed that GFP-

transduced cancer cells allowed the imaging of tumor cells in bloodvessels. To examine cell behavior during intravasation, Wyckoffet al. (9) have used GFP imaging to view these cells in time-lapseimages within a single optical section using a confocal microscope.In vivo imaging of the primary tumors indicated that bothmetastatic and nonmetastatic cells are motile and show protrusiveactivity. Metastatic cells show greater orientation toward bloodvessels. Nonmetastatic cells fragment when interacting with vessels.Naumov et al. (3), using GFP imaging, visualized fine cellular

details such as pseudopodial projections, even after extendedperiods of in vivo growth.Mook et al. (10) visualized initial arrest of GFP colon cancer cells

in sinusoids of the liver due to size restriction after portal veininjection. Tumor cells divided exclusively intravascularly during thefirst 4 days.Al-Mehdi et al. (11) observed the steps in early hematogenous

metastasis of tumor cells expressing GFP in subpleural microvesselsin intact, perfused mouse and rat lungs. Metastatic tumor cellsattached to the endothelia of pulmonary precapillary arterioles andcapillaries. Extravasation of tumor cells was rare. Early tumorcolonies were observed growing entirely within the blood vessels.Yamamoto et al. (12) reported the genetic engineering of dual-

color fluorescent cells with one color in the nucleus and the otherin the cytoplasm. The dual-color cells enable real-time nuclear-cytoplasmic dynamics to be visualized in living cells in vivo as wellas in vitro . To obtain the dual-color cells, red fluorescent protein(RFP) was expressed in the cytoplasm of HT-1080 humanfibrosarcoma cells, and GFP linked to histone H2B was expressedin the nucleus. Nuclear GFP expression enabled visualization of

Requests for reprints: Robert M. Hoffman, AntiCancer, Inc., 7917 OstrowStreet, San Diego, CA 92111. Phone: 858-654-2555; Fax: 858-268-4175; E-mail:[email protected].

I2005 American Association for Cancer Research.

Cancer Res 2005; 65: (10). May 15, 2005 4246 www.aacrjournals.org

Research Article

Research. on August 14, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: Real-time In vivo Dual-color Imaging of Intracapillary ... · Yamamoto et al. (12) reported the genetic engineering of dual-color fluorescent cells with one color in the nucleus and

nuclear dynamics. Simultaneous cytoplasmic RFP expressionenabled visualization of nuclear-cytoplasmic ratios as well assimultaneous cell and nuclear shape changes. Thus, total cellulardynamics can be visualized in the living dual-color cells in realtime. Common carotid artery injection of dual-color cells and areversible skin flap enabled the external visualization of the dual-color cells in microvessels in the mouse brain where extremeelongation of the cell body as well as the nucleus occurred.In this report, we describe real-time imaging of the deformation

of cancer cells and their nuclei in vivo . In addition to thedeformability, we imaged migration of HT-1080-dual-color cells inmicrovessels and capillaries in real time. The capability to makesuch measurements in vivo should enable better understanding ofthe mechanism of metastasis.

Materials and Methods

Production of red fluorescent protein retroviral vector. For RFP

retrovirus production (13, 14), the HindIII/NotI fragment from pDsRed2

(Clontech Laboratories, Inc., Palo Alto, CA), containing the full-length RFP

cDNA, was inserted into the HindIII/NotI site of pLNCX2 (ClontechLaboratories) containing the neomycin resistance gene. PT67, an NIH3T3-

derived packaging cell line (Clontech Laboratories) expressing the 10 Al

viral envelope, was cultured in DMEM (Irvine Scientific, Santa Ana, CA)

supplemented with 10% heat-inactivated fetal bovine serum (Gemini Bio-Products, Calabasas, CA). For vector production, PT67 cells, at 70%

confluence, were incubated with a precipitated mixture of LipofectAMINE

reagent (Life Technologies, Inc., Grand Island, NY) and saturating amounts

of pLNCX2-DsRed2 plasmid for 18 hours. Fresh medium was replenished atthis time. The cells were examined by fluorescence microscopy 48 hours

post-transduction. For selection of a clone producing high amounts of RFP

retroviral vector (PT67-DsRed2), the cells were cultured in the presence of200 to 1,000 Ag/mL G418 (Life Technologies) for 7 days. The isolated clone

was termed PT67-DSRed2.

Production of histone H2B-green fluorescent protein vector. Thehistone H2B gene has no stop codon (15), thereby enabling the ligation ofthe H2B gene to the 5V-coding region of the GFP gene (Clontech

Laboratories). The histone H2B-GFP fusion gene was then inserted at the

HindIII/CalI site of the pLHCX (Clontech Laboratories) that has the

hygromycin resistance gene. To establish a packaging cell clone producinghigh amounts of histone H2B-GFP retroviral vector, the pLHCX histone

H2B-GFP plasmid was transfected in PT67 cells using the same methods

described above for PT67-DsRed2. The transfected cells were cultured in thepresence of 200 to 400 Ag/mL hygromycin (Life Technologies) for 15 days to

establish stable PT67 H2B-GFP packaging cells.

Red fluorescent protein and histone H2B-green fluorescent proteingene transduction of fibrosarcoma cells. For RFP and H2B-GFP genetransduction, 70% confluent human HT-1080 fibrosarcoma cells were used

(12). To establish dual-color cells, clones of HT-1080 expressing RFP in the

cytoplasm (HT-1080-RFP) were initially established. In brief, HT-1080 cells

were incubated with a 1:1 precipitated mixture of retroviral supernatants ofPT67-RFP cells and RPMI 1640 (Irvine Scientific) containing 10% fetal bovine

serum for 72 hours. Fresh medium was replenished at this time. Cells were

harvested with trypsin/EDTA 72 hours posttransduction and subcultured ata ratio of 1:15 into selective medium, which contained 200 Ag/mL G418. The

level of G418 was increased stepwise up to 800 Ag/mL. HT-1080-RFP cells

were isolated with cloning cylinders (Bel-Art Products, Pequannock, NJ)

using trypsin/EDTA and amplified by conventional culture methods.For establishing dual-color cells, HT-1080-RFP cells were then incubated

with a 1:1 precipitated mixture of retroviral supernatants of PT67 H2B-GFP

cells and culture medium. To select the double transformants, cells were

incubated with hygromycin 72 hours after transfection. The level ofhygromycin was increased stepwise up to 400 Ag/mL (Fig. 1).

Real-time visualization of deformation of HT-1080 dual-color cellsin vessels in live mice. To visualize cell dynamics in vessels in live mice,

cells were injected into the heart. Five nude mice were anesthetized with a

ketamine mixture (10 AL ketamine HCl, 7.6 AL xylazine, 2.4 AL acepromazine

maleate, and 10 AL H2O) via s.c. injection. A total of 200 AL of mediumcontaining 5 � 106 HT-1080-dual-color cells were injected into the heart. To

observe the shapes of the HT-1080-dual-color cells within the microvessels

before arrest, the epigastrica cranialis vein of the mouse was wired with a 6-0

suture (Ethicon Inc., Somerville, NJ) before cell injection. Immediately afterinjection, an arc-shaped incision was made in the abdominal skin, and then

s.c. connective tissue was separated to free the skin flap without injuring the

epigastrica cranialis artery and vein. The skin flap was spread and fixed onthe flat stand. The inside surface of the skin flap was directly observed under

fluorescence microscopy. After making the skin flap, HT-1080-dual-color

cells were imaged immediately and 2 hours later to determine the migration

velocities of the cells. During the interval between imaging, PBS (IrvineScientific) was occasionally sprayed on the inside of the skin flap to keep the

surface wet. The skin flap could be completely reversed.

Morphologic analysis of cancer cell deformation. For morphologic

analysis, we examined 30 cells in microvessels and 60 cells in capillaries infive mice. During the period of the measurement, the animal was kept under

anesthesia. The animal was kept warm and the skin flap was kept hydrated

with saline solution. Measurements were taken at the initial time and 2 hourslater. Images were taken at the initial time. The image included the cell in its

vessel as well as the surrounding vessels which were used as amap to relocate

the cell in its vessel 2 hours later when the next images were captured.

Usually, 10 to 20 cells were followed in a given experiment. The lengths of themajor and minor axes of the whole cells and the nuclei were measured using

Image ProPlus 3.1 software, where (A) was the length of the major axis and

(B) was the length of the minor axis of the whole cell. For the nuclei, (a) is the

length of the major axis and (b) is the length of the minor axis.Motility analysis in vivo. For motility analysis, the epigastrica cranialis

vein was not wired. All other procedures were the same as for morphologic

analyses. Motility analysis was done using Image ProPlus 3.1 software.Images that were taken immediately after injection and 2 hours after taking

the first images to determine migration velocities of 60 cells.

Fluorescent optical imaging and data analysis. Images were captured

directly with a Hamamatsu C5810 3CCD camera (Hamamatsu Photonics,Bridgewater, NJ). For microimaging, a Leica fluorescent stereo microscope

(model MZ16) was coupled with the Hamamatsu camera. High-resolution

images (1024 � 724 pixels) were captured directly on an IBM PC. Images

Figure 1. Stable high GFP- and RFP-expressing human fibrosarcoma cellsin vitro . GFP is localized in the nuclei due to linkage with histone H2B. RetroviralDsRed2 is expressed in the cytoplasm. See Materials and Methods for details;bar , 50 Am.

In vivo Real-time Imaging of Dual-Color Cells

www.aacrjournals.org 4247 Cancer Res 2005; 65: (10). May 15, 2005

Research. on August 14, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: Real-time In vivo Dual-color Imaging of Intracapillary ... · Yamamoto et al. (12) reported the genetic engineering of dual-color fluorescent cells with one color in the nucleus and

were processed for contrast and brightness and analyzed with the use ofImage ProPlus 3.1 software.

All animal studies were conducted in accordance with the principles and

procedures outlined in the NIH Guide for the Care and Use of Laboratory

Animals under assurance number A3873-1. Animals were kept in a barrierfacility under HEPA filtration. Mice were fed with autoclaved laboratory

rodent diet (Tecklad LM-485, Western Research Products, Orange, CA).

Results

Real-time observations were made of nuclear-cytoplasmicdeformability in capillaries of HT-1080-dual-color cells. The cellswere injected into the heart of mice whose epigastrica cranialis veinwas wired to close it off (Fig. 1). The skin flap was made withoutinjuring the epigastrica cranialis artery and vein, and then spreadand fixed on a stand. Because of this stable fixation, thousands ofcells could be easily visualized in capillaries and in microvessels inthe skin in the live mouse under fluorescence microscopy (Fig. 2).The wired epigastrica cranialis vein was clogged with round cells.Some cells were trapped due to size restriction in capillaries, wherethe cells and nuclei were highly deformed.We classified the shapes of the cells into four categories. One

included the cells in microvessels, where the cells were round andthe nuclei were oval (Fig. 3A). Another category included the cellsthat were elongated in capillaries, where the nuclei were alsoelongated (Fig. 3B). Another category was that of cells that wereelongated and bifurcated at the corner of the capillaries (Fig. 3C).The other category was that of cells that were so elongated that thecytoplasm disconnected. The nuclei, however, remained intactalthough they were very elongated (Fig. 3D).Quantitation of cancer cell and nuclear deformation. The

lengths of the major and minor axes of the whole cells and thenuclei were measured (Fig. 4; Table 1). The average lengths ofthe axes of the whole cells in capillaries were 3.97 times as long as

in microvessels. As for the nuclei, the average lengths were 1.64times as long. The lengths of the minor axes of the whole cells incapillaries were equal to those of the nuclei.Real-time observation of nuclear-cytoplasmic clasmatosis.

Clasmatosis or cytoplasmic fragmentation was observed in thecells immediately after injection (13). The cytoplasm was removedfrom many cells leaving naked nuclei (Fig. 5C and D). Nuclei werealso fragmented or stretched dramatically (Fig. 5A and B).Motility analysis in capillaries. Two hours after taking the

first images, the same cells were imaged again to determinethe migration velocities of the cells. The bifurcated corners of thecapillaries were used as markers to calculate the distancemigrated. The distances from the marker to the centers of thenuclei were measured and subtracted (Fig. 6). Approximately 25%of the cells observed bifurcated at the corner of the capillariesand could not migrate and were therefore excluded from ourcalculations. Of the 45 cells that were not at the corner of thecapillaries, 20 cells migrated and 25 cells did not migrate. Theaverage migration velocity of the 20 cells that migrated was13.2 Am/hour (1.1-48.3). A 1 Am difference in distance could bedistinguished from the captured images of the dual-colorfluorescent cells.The diameters of the capillaries were noted when calculating

migration velocity. The minimum diameter of the capillary thatallowed cell migration was approximately 8 Am. Sixteen out of 20cells in capillaries over 8 Am in diameter migrated up to 48.3 Am/hour. In contrast, 21 out of 25 cells in capillaries less than 8 Am indiameter could not migrate at all.

Discussion

Using the skin flap for observation of cells in capillaries hasimportant advantages. The skin can be spread stably on a stand,such that motion from the mouse’s heartbeat or breathing has no

Figure 2. Visualization of HT-1080-dual-color cells in microvessels and in capillaries in the skin in live mice. HT-1080-dual-color cells were injected into the heart of themice under anesthesia. A, immediately after injection, the cells were visualized in microvessels and in capillaries in the skin flap. The round or oval cells occupythe full diameter of the epigastrica cranialis artery, the outlines of which are indicated by arrowheads. The cells occupy the full diameter of the capillaries, the outlines ofwhich are indicated by the arrows. B, immediately after injection in a different mouse than in A , numerous round HT-1080 dual-color cells in the epigastricacranialis artery. Bar , 50 Am.

Cancer Research

Cancer Res 2005; 65: (10). May 15, 2005 4248 www.aacrjournals.org

Research. on August 14, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: Real-time In vivo Dual-color Imaging of Intracapillary ... · Yamamoto et al. (12) reported the genetic engineering of dual-color fluorescent cells with one color in the nucleus and

influence on imaging. Disturbance of the blood supply for the skindoes not occur during the skin flap procedure, because theepigastrica cranialis artery is not injured during the procedure. Inaddition to these advantages, the skin flap could be completely

reversed such that the mice need not be sacrificed. In our study, theskin flap was reversed after 24 hours.To observe the shape of the HT-1080-dual-color cells within

blood vessels, we wired the epigastrica cranialis vein closed before

Figure 4. Shapes of HT-1080-dual-color cells in different sizevessels. (Top ) cells in microvessels; (bottom ) cells in capillaries.

Figure 3. Classification of the deformation of HT-1080-dual-color cells in the vessels in the skin. A, nondeformed cells are within a microvessel. The cells in themicrovessel are round and the nuclei oval. The cells occupy the full diameter of the vessel. B, the cells and nuclei are elongated to fit a capillary. C, the cellsare arrested at the capillary bifurcation. Because of the difference of the deformability between cytoplasm and nucleus, only the cytoplasm is bifurcated. The nucleus isalso deformed but remains intact. D, cytoplasmic fragmentation in very thin capillary; bar , 50 Am.

In vivo Real-time Imaging of Dual-Color Cells

www.aacrjournals.org 4249 Cancer Res 2005; 65: (10). May 15, 2005

Research. on August 14, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: Real-time In vivo Dual-color Imaging of Intracapillary ... · Yamamoto et al. (12) reported the genetic engineering of dual-color fluorescent cells with one color in the nucleus and

injection of the cells into the heart. We observed many roundcancer cells in the microvessels in the skin. As for the cells incapillaries, almost all the cells and their nuclei deformed in orderto conform to the diameter of the capillaries. The cancer cells andtheir nuclei deformed into three-pronged forks when the cells werearrested at capillary bifurcations.There is an apparent limitation to cell and nuclear deformation

when the cells are arrested in capillaries. We found many cancercells whose cytoplasms seemed to fragment and separate fromnuclei, a process called clasmatosis. We also found that nucleicould fragment. When the cells arrested in very narrow capillaries,the cytoplasm of many cells as well as their nuclei were destroyed.

Morris et al. (13) examined clasmatosis of melanoma andmammary carcinoma cells in vivo . Morris suggested that thecells undergoing clasmatosis were damaged and would ultimatelydie. Clasmatosis may account in part for the inefficiency ofmetastasis.The nucleus seems to be less deformable than the cytoplasm.

This may be due to the difference between the cytoskeleton in thecytoplasm and the nucleus (16, 17). In our study, the lengths of themajor and minor axes of the nuclei and the whole cells deformed tofit the width of the capillaries.Bioluminescence imaging. magnetic resonance imaging,

positron emission tomography, and single photon emission

Table 1. Imaging cellular and nuclear morphometry in microvessels and capillaries in living mice

Whole cells (Am) Nuclei (Am)

Major axis Minor axis Major axis Minor axis

Cells in microvessels 23.3 (15.5-32.5) 19.3 (14.4-23.8) 16.8 (13.5-20.2) 12.2 (8.1-16.8)

Cells in capillaries 92.6 (36.8-228.8) 7.8 (4.9-10.5) 27.6 (14.1-51.8) 7.8 (4.9-10.5)

NOTE: HT1080 human fibrosarcoma cells were labeled with GFP in nuclei and RFP in cytoplasm. The cells were injected in the mice as described in

Materials and Methods. The dual-color cells were observed in both capillaries and microvessels in a skin flap in live mice under fluorescent microscopy

(as described in Materials and Methods). Morphometric measurements described in the table were made from digitally captured images of the dual-color cells.

Figure 5. Clasmatosis of HT-1080-dual-color cells in vivo . A, cells with nuclei andno cytoplasm remain in microvesselsexcept for one intact cell. One of the nucleiis deformed like a spoon and is fragmentedin two (arrow). B, a nucleus with thecytoplasm removed is deformed like acomet. C, fragmented cytoplasm remainsaround a nucleus. The remainingcytoplasm is stretched out dramatically.The nucleus is also deformed in a capillary.D, the cell is bifurcated and the cytoplasmis fragmented into two (arrow ). Anothercell has only a part of its cytoplasm but thenucleus is intact (arrowhead); bar , 50 Am.

Cancer Research

Cancer Res 2005; 65: (10). May 15, 2005 4250 www.aacrjournals.org

Research. on August 14, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: Real-time In vivo Dual-color Imaging of Intracapillary ... · Yamamoto et al. (12) reported the genetic engineering of dual-color fluorescent cells with one color in the nucleus and

computed tomography have allowed detailed and dynamic viewsof tissues. However, these methods lack the spatial and/ortemporal resolution to visualize single-cell dynamics in situ .Such information can be generated with fluorescence imaging.Newer techniques such as multiphoton microscopy can becombined with second harmonic generation imaging tovisualize cellular behavior in the interstitium of solid organs(18). The dual-colored cells described in this report should be apowerful tool when used with multiphoton microscopy infuture experiments.Acquisition of protease-independent amoeboid dissemination

was visualized for HT-1080 cells injected into the mouse dermis

monitored by intravital multiphoton time-lapse microscopy (19).The transition from proteolytic mesenchymal toward nonpro-teolytic amoeboid movement shows a plasticity mechanism in cellmigration (19). Such transitions in movement can now beimaged with the dual-colored cells described in the presentreport in order to visualize the role of the nucleus as well as thecytoplasm.The development of transgenic mice expressing GFP in specific

cells will also be important to visualize single-cell behavior in vivo .For example, T cells with a CD2 promoter driving GFP expressionallowed visualization of T cell rolling and adhesion in postca-pillary venules during inflammation in vivo (20). Transgenic GFP-

Figure 6. Migration velocity of HT-1080-dual-color cell in capillaries. A, cell immediately after injection. B, the same cell as in (A ) imaged 2 hours later. The corner ofthe capillary was regarded at the starting point of migration. C–F, migration of cells in capillaries from 2 to 14 hours; bar , 50 Am.

In vivo Real-time Imaging of Dual-Color Cells

www.aacrjournals.org 4251 Cancer Res 2005; 65: (10). May 15, 2005

Research. on August 14, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: Real-time In vivo Dual-color Imaging of Intracapillary ... · Yamamoto et al. (12) reported the genetic engineering of dual-color fluorescent cells with one color in the nucleus and

expressing mice can be developed to image many aspects of cellbiology, including cancer.The present study shows that HT-1080-dual-color cells are useful

for visualization of cellular and nuclear dynamics in vivo . Afterinjection of the cells into the heart, we could observe cancer celland nuclear deformation as well as clasmatosis in real time.Furthermore, we could calculate the migration velocities of tumorcells in capillaries.The dual-color cells, with GFP in the nucleus and RFP in the

cytoplasm, along with new approaches to in vivo imaging inappropriate hosts provide a powerful tool to understand the

mechanism of cancer cell migration and deformation in verysmall vessels in vivo . With this new information, we can betterunderstand the mechanism of metastasis.

Acknowledgments

Received 1/11/2005; revised 2/18/2005; accepted 3/1/2005.Grant support: Supported in part by grants CA099258, CA103563, and CA101600

from the National Cancer Institute.The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.

References1. Hoffman RM. Green fluorescent protein imaging oftumour growth, metastasis, and angiogenesis in mousemodels. Lancet Oncol 2002;3:546–56.

2. Condeelis J, Segall JE. Intravital imaging of cellmovement in tumours. Nat Rev Cancer 2003;3:921–30.

3. Naumov GN, Wilson SM, Macdonald IC, et al. Cellularexpression of green fluorescent protein, coupled withhigh-resolution in vivo videomicroscopy, to monitorsteps in tumor metastasis. J Cell Sci 1999;112:1835–42.

4. Yang M, Baranov E, Wang J-W, et al. Direct externalimaging of nascent cancer, tumor progression, angio-genesis, and metastasis on internal organs in thefluorescent orthotopic model. Proc Natl Acad Sci U S A2002;99:3824–9.

5. Chishima T, Miyagi Y, Wang X, et al. Cancer invasionand micrometastasis visualized in live tissue by greenfluorescent protein expression. Cancer Res 1997;57:2042–7.

6. Farina KL, Wyckoff JB, Rivera J, et al. Cell motility oftumor cells visualized in living intact primary tumorsusing green fluorescent protein. Cancer Res 1998;58:1528–32.

7. Chambers AF, Groom AC, MacDonald IC. Dissemina-tion and growth of cancer cells in metastatic sites. NatRev Cancer 2002;2:563–72.

8. Huang MS, Wang TJ, Liang CL, et al. Establishment offluorescent lung carcinoma metastasis model and itsreal-time microscopic detection in SCID mice. Clin ExpMetastasis 2002;19:359–68.

9. Wyckoff JB, Jones JG, Condeelis JS, Segall JE. Acritical step in metastasis: in vivo analysis of intra-vasation at the primary tumor. Cancer Res 2000;60:2504–11.

10. Mook ORF, Marle JV, Vreeling-Sindelarova H. Visuali-zation of early events in tumor formation of eGFP-transfected rat colon cancer cells in liver. Hepatology2003;38:295–304.

11. Al-Mehdi AB, Tozawa K, Fisher AB, Shientag L,Lee A, Muschel RJ. Intravascular origin of metastasisfrom the proliferation of endothelium-attached tumorcells: a new model for metastasis. Nat Med 2000;6:100–2.

12. Yamamoto N, Jiang P, Yang M, et al. Cellulardynamics visualized in live cells in vitro and in vivoby differential dual-color nuclear-cytoplasmic fluorescent-protein expression. Cancer Res 2004;64:4251–6.

13. Morris VL, Macdonald IC, Koop S, Schmidt EE,Chamber AF, Groom AC. Early interaction ofcancer cells with the microvasculature in mouseliver and muscle during hematogenous metastasis:videomicroscopic analysis. Clin Exp Metastasis 1993;11:377–90.

14. Yamamoto N, Yang M, Jiang P, et al. Real-timeimaging of individual fluorescent-protein color-codedmetastatic colonies in vivo . Clin Exp Metastasis 2003;20:633–8.

15. Kanda T, Sullivan KF, Wahl GM. Histone-GFP fusionprotein enables sensitive analysis of chromosomedynamics in living mammalian cells. Curr Biol 1998;8:377–85.

16. Erkell LJ, Ryd W, Hagmor B, et al. Comments onthe filter test for tumor cell deformability. InvasionMetastasis 1982;2:260–7.

17. Gabor H, Weiss L, Hangmar B. Survival of L1210and Ehrlich ascites cancer cells after mechanicaltrauma is a random event. Invasion Metastasis 1985;5:84–95.

18. Mempel TR, Scimone ML, Mora JR, von Andrian UH.In vivo imaging of leukocyte trafficking in blood vesselsand tissues. Curr Opin Immunol 2004;16:406–17.

19. Wolf K, Mazo I, Leung H, et al. Compensationmechanism in tumor cell migration: mesenchymal-amoeboid transition after blocking of pericellularproteolysis. J Cell Biol 2003;160:267–77.

20. Singbartl K, Thatte J, Smith ML, Wethmar K, Day K,Ley K. A CD2-green fluorescence protein-transgenicmouse reveals very late antigen-4-dependent CD8+

lymphocyte rolling in inflamed venules. J Immunol 2001;166:7520–6.

Cancer Research

Cancer Res 2005; 65: (10). May 15, 2005 4252 www.aacrjournals.org

Research. on August 14, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: Real-time In vivo Dual-color Imaging of Intracapillary ... · Yamamoto et al. (12) reported the genetic engineering of dual-color fluorescent cells with one color in the nucleus and

2005;65:4246-4252. Cancer Res   Kensuke Yamauchi, Meng Yang, Ping Jiang, et al.   Cell and Nucleus Deformation and Migration

Dual-color Imaging of Intracapillary CancerIn vivoReal-time

  Updated version

  http://cancerres.aacrjournals.org/content/65/10/4246

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/65/10/4246.full#ref-list-1

This article cites 20 articles, 7 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/65/10/4246.full#related-urls

This article has been cited by 24 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://cancerres.aacrjournals.org/content/65/10/4246To request permission to re-use all or part of this article, use this link

Research. on August 14, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from