supporting information - pnas...ab 200 μm baseline after irradiation 350 μm c baseline after...

6
Supporting Information Choi et al. 10.1073/pnas.1018790108 SI Materials and Methods Histology. Mice were killed under deep anesthesia by transcardial perfusion with PBS, followed by xation with 4% formalin in phosphate buffer. Brains were removed, immersed in 4% for- malin in phosphate buffer overnight, and then transferred to a 30% sucrose solution for at least 3 d. Target regions were navigated by stereotaxic coordinates and uorescence, followed by sectioning into 30-μm coronal sections on a freezing sliding microtome. The sections were stained using the Nissl method or immunostaining and mounted on glass slides for microscopic evaluation. Dorsal Skinfold Chamber Model. A small dorsal skinfold chamber kit (SM100) was purchased from APJ Trading, and surgery was performed as described previously (1). In brief, mice were an- esthetized with an i.p. injection of ketaminexylazine, the dorsal skin was extended, and two mirror-image titanium frames were mounted. One layer of skin was excised, leaving the striated muscle of the opposite side intact, and then replaced with a glass coverslip mounted into the frame. In Vivo Skull Imaging Preparation. After anesthesia with an i.p. injection of ketamine-xylazine, the scalp was removed, and the mouse head was xed to a stereotaxic frame. In Vivo Ear Imaging Preparation. After anesthesia with an i.p. in- jection of ketamine-xylazine, the ear was glued to a glass and placed on a custom-designed mount. Intravenous Administration. All dyes were injected i.v. through the lateral tail vein or the retro-orbital venous sinus. An i.v. catheter was used when timely injection was necessary. For the plasma marker, 100 μL of 2% wt/wt dextran-conjugated dye was ad- ministered as indicated. To determine the dose of dye for i.v. injection, we multiplied the dilution factor for plasma by the conventional dose of the dye used for tissue staining and modi- ed it experimentally. The following dyes were used: OGB-1:00 AM (Invitrogen; 50 μL of 100 μg), Hoechst (20 mg/kg), SR101 (Invitrogen; 10 μL of 0.5 mg/mL), adenovirus (100 μL of 1 × 10 9 pfu of adenoviral vector with CMV promoter-driven GFP ex- pression), Quantum Dot (Q21021MP, Molecular Probes; 20 μL of 2 μM), TAMRA-MION (50 μL of 10 μM), and siRNA (60 μL of 125 pmol mixed with 60 μL of 2% in vivo jetPEI; Polyplus). 1. Isaka N, Padera TP, Hagendoorn J, Fukumura D, Jain RK (2004) Peritumor lymphatics induced by vascular endothelial growth factor-C exhibit abnormal function. Cancer Res 64:44004404. Choi et al. www.pnas.org/cgi/content/short/1018790108 1 of 6

Upload: others

Post on 17-Feb-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Supporting Information - PNAS...AB 200 μm Baseline After irradiation 350 μm C Baseline After irradiation Extravasation Fig. S3. Feasibility of laser-induced extravasation in the

Supporting InformationChoi et al. 10.1073/pnas.1018790108SI Materials and MethodsHistology.Mice were killed under deep anesthesia by transcardialperfusion with PBS, followed by fixation with 4% formalin inphosphate buffer. Brains were removed, immersed in 4% for-malin in phosphate buffer overnight, and then transferred toa 30% sucrose solution for at least 3 d. Target regions werenavigated by stereotaxic coordinates and fluorescence, followedby sectioning into 30-μm coronal sections on a freezing slidingmicrotome. The sections were stained using the Nissl methodor immunostaining and mounted on glass slides for microscopicevaluation.

Dorsal Skinfold Chamber Model. A small dorsal skinfold chamberkit (SM100) was purchased from APJ Trading, and surgery wasperformed as described previously (1). In brief, mice were an-esthetized with an i.p. injection of ketamine–xylazine, the dorsalskin was extended, and two mirror-image titanium frames weremounted. One layer of skin was excised, leaving the striatedmuscle of the opposite side intact, and then replaced with a glasscoverslip mounted into the frame.

In Vivo Skull Imaging Preparation. After anesthesia with an i.p.injection of ketamine-xylazine, the scalp was removed, and themouse head was fixed to a stereotaxic frame.

In Vivo Ear Imaging Preparation. After anesthesia with an i.p. in-jection of ketamine-xylazine, the ear was glued to a glass andplaced on a custom-designed mount.

Intravenous Administration. All dyes were injected i.v. through thelateral tail vein or the retro-orbital venous sinus. An i.v. catheterwas used when timely injection was necessary. For the plasmamarker, 100 μL of 2% wt/wt dextran-conjugated dye was ad-ministered as indicated. To determine the dose of dye for i.v.injection, we multiplied the dilution factor for plasma by theconventional dose of the dye used for tissue staining and modi-fied it experimentally. The following dyes were used: OGB-1:00AM (Invitrogen; 50 μL of 100 μg), Hoechst (20 mg/kg), SR101(Invitrogen; 10 μL of 0.5 mg/mL), adenovirus (100 μL of 1 × 109

pfu of adenoviral vector with CMV promoter-driven GFP ex-pression), Quantum Dot (Q21021MP, Molecular Probes; 20 μLof 2 μM), TAMRA-MION (50 μL of 10 μM), and siRNA (60 μLof 125 pmol mixed with 60 μL of 2% in vivo jetPEI; Polyplus).

1. Isaka N, Padera TP, Hagendoorn J, Fukumura D, Jain RK (2004) Peritumor lymphaticsinduced by vascular endothelial growth factor-C exhibit abnormal function. Cancer Res64:4400–4404.

Choi et al. www.pnas.org/cgi/content/short/1018790108 1 of 6

Page 2: Supporting Information - PNAS...AB 200 μm Baseline After irradiation 350 μm C Baseline After irradiation Extravasation Fig. S3. Feasibility of laser-induced extravasation in the

A 2 sec 10 sec 1 minBaseline

10 sec 1 min 2 minBaseline

Arte

ryC

apilla

ry

Baseline 20 sec 40 sec

60 sec 120 sec 180 sec

C D

B

0 40 80 120 160 20060

80

100

120

Time (s)Fl

uore

scen

ce (r

. f. u

.)

Fig. S1. (A) Laser-induced extravasation in an artery. Arterial stimulation accompanied arterial contraction. The dashed line demarcates the baseline lumenarea. (Scale bar: 50 μm.) (B) Laser-induced extravasation in a capillary. Capillaries were vulnerable to hemorrhage when irradiated. Note the loss of dark streaks,indicating red blood cell movement; a clot formed around the irradiated region (red arrow). (Scale bar: 10 μm.) (C) Transient clot formation after laser ir-radiation. During laser-induced vascular permeabilization, transient clot formation (white arrow) was observed with disturbed downstream blood flow. Thered dot indicates the position of laser irradiation. (Scale bar: 20 μm.) (D) Temporal dynamics of fluorescence intensity in the boxed region shown in C. The arrowindicates the time of laser irradiation.

A B

0 1 2 3 4 5

0

20

40

60

80

100

Energy (mJ)

Succ

essf

ul e

xtra

vasa

tion

(%)

0.5 1.0 2.0 3.0 4.0 5.0

0

10

20

30

40

50Transient clot formationStable clot formation

Energy (mJ)

Res

pons

e (%

)

Fig. S2. (A) Relationship between laser energy and the probability of successful extravasation (n = 18 for each dose of laser energy in three mice). (B) De-pendence of clot formation on laser energy (n = 18 for each dose of laser energy in three mice).

Choi et al. www.pnas.org/cgi/content/short/1018790108 2 of 6

Page 3: Supporting Information - PNAS...AB 200 μm Baseline After irradiation 350 μm C Baseline After irradiation Extravasation Fig. S3. Feasibility of laser-induced extravasation in the

A B

200

μm

Baseline After irradiation

350

μm

Baseline After irradiation ExtravasationC

Fig. S3. Feasibility of laser-induced extravasation in the deep cortical layer and to a large area. (A) 3D view of vascular structure in the cortex. The skull anddura layer were removed, 2 MDa of FITC-dextran was i.v. injected, and images were obtained at 3-μm intervals to a depth of 450 μm. (B) Laser-induced ex-travasation in the deep cortical layers. (Scale bar: 20 μm.) (C) Molecular delivery to a large area by irradiation of multiple blood vessels. Here 70 kDa of FITC-dextran was injected i.v. in the thinned-skull window model, and three extravasations were induced by irradiating three regions in the pial venules. (Scale bar:100 μm.)

A

C 5 sec 20 secBaseline 1 min

B 5 sec 20 secBaseline 1 min

Fig. S4. Red blood cells (RBCs) did not leak during laser-induced vascular permeabilization. RBCs were stained and injected i.v. with TRITC-dextran. (A) StainedRBCs (green) in TRITC-dextran (red) solution. (Scale bar: 10 μm.) (B) RBCs did not leak during laser-induced vascular permeabilization. Green streaks representmovement of stained RBCs. (Scale bar: 20 μm.) (C) RBC leakage during severe arterial contraction by laser irradiation. Severe contraction was induced by RBCleakage after laser irradiation to the artery, as denoted by the clear circular green signals outside of the lumen. (Scale bar: 20 μm.)

Choi et al. www.pnas.org/cgi/content/short/1018790108 3 of 6

Page 4: Supporting Information - PNAS...AB 200 μm Baseline After irradiation 350 μm C Baseline After irradiation Extravasation Fig. S3. Feasibility of laser-induced extravasation in the

A B

D

C

F

0 10 20 30 40 500.0

0.2

0.4

0.6

0.8

1.0

Lumen diameter (μm)

Circ

ular

ity

E

0 10000 20000 30000 40000 500000.0

0.2

0.4

0.6

0.8

1.0

Extravasation area (μm2)

Circ

ular

ity

0 1 2 3 40.0

0.2

0.4

0.6

0.8

1.0

Blood flow (mm/s)

Circ

ular

ity

0 10 20 30 40 500

1

2

3

4

Lumen diameter (μm)

Bloo

d flo

w (m

m/s

)

0 1 2 3 4 50

10000

20000

30000

40000

50000

Blood flow (mm/s)

Extr

avas

atio

n ar

ea (μ

m2 )

Extr

avas

atio

n ar

ea (μ

m2 )

Small venule Large venule0

10000

20000

30000

40000

50000

60000 p = 0.0003

Fig. S5. Characterization of laser-induced extravasation. Lumen diameter and blood flow were measured at baseline, and the extravasation area and cir-cularity of the extravasation area were quantified based on an image obtained at 1 min. (A) Lumen diameter and blood flow in venules showed a strongpositive correlation (R2 = 0.5612; P < 0.0001; n = 37). (B) Compared with large venules (lumen diameter <25 μm; n = 28), small venules (lumen diameter >25 μm;n = 9) showed significantly less extravasation area (P = 0.0003). (C) Blood flow had a nonsignificant negative correlation with extravasation area (R2 = 0.0718;P = 0.1089) (D and E) Smaller venules or venules with slower blood flow had more circular extravasation (R2 = 0.4721; P < 0.0001 vs. R2 = 0.1988; P = 0.0057). (F)The extravasation area was positively correlated with the circularity of extravasation (R2 = 0.2232; P = 0.0032).

Choi et al. www.pnas.org/cgi/content/short/1018790108 4 of 6

Page 5: Supporting Information - PNAS...AB 200 μm Baseline After irradiation 350 μm C Baseline After irradiation Extravasation Fig. S3. Feasibility of laser-induced extravasation in the

Intact skull Thin skull Thin skull + Extravasation

GFA

PIb

a-1

A

B

Open skull

Thin skull Thin skull + Extravasation

Fig. S6. Assessment of delayed cell death and immune response at 2 d after laser-induced extravasation. (A) Nissl staining. Thinned-skull windows (∼1 mm indiameter) were made in each hemisphere, and extravasation was induced in the right hemisphere. (Insets) Magnified views of the thinned-skull windowsregions. (Scale bar: 500 μm.) (B) Immunostaining of astrocyte (GFAP) and microglial (Iba-1) activation. (Scale bar: 100 μm.)

Baseline During extravasation 10 min after extravasation

20 s

10 %∆F/F

Fig. S7. Functional calcium activity of astrocytes during laser-induced plasma leakage. A calcium indicator (OGB-1:00 AM) and astrocyte marker (SR101) wereloaded in the cortex using a conventional bath-loading method. Rhodamine was administered i.v. as a plasma indicator. Astrocyte calcium activity wasmeasured with a 2-Hz acquisition rate. Spontaneous calcium activity in astrocytes is shown.

Choi et al. www.pnas.org/cgi/content/short/1018790108 5 of 6

Page 6: Supporting Information - PNAS...AB 200 μm Baseline After irradiation 350 μm C Baseline After irradiation Extravasation Fig. S3. Feasibility of laser-induced extravasation in the

7 sec 30 sec 1 minBaseline

Qdo

t

B

A

3 sec 8 sec 30 secBaseline

MIO

N

Fig. S8. Laser-induced permeabilization of nanoparticles. (A) Quantum dot. (B) TAMRA- conjugated MiONs.

Head frameThinned Skull

Dental cement

CorticalVasculature

aCSF

20X 1.0 NA

Head frameXYtranslation

stage

A BThinned skull surgery

Intravenous injection of probes

Localization of the target venule

Laser irradiation to the vascular wall

Imaging

Objective

Fig. S9. Experimental setup. (A) Procedure for the laser-induced extravasation experiments. (B) Schematic diagram of the experimental setup.

Choi et al. www.pnas.org/cgi/content/short/1018790108 6 of 6