16
www.sciencetranslationalmedicine.org/cgi/content/full/3/99/99ra87/DC1 Supplementary Materials for Lysophosphatidic Acid Signaling May Initiate Fetal Hydrocephalus Yun C. Yung, Tetsuji Mutoh, Mu-en Lin, Kyoko Noguchi, Richard R. Rivera, Ji Woong Choi, Marcy A. Kingsbury, Jerold Chun* *To whom correspondence should be addressed. E-mail: [email protected] Published 7 September 2011, Sci. Transl. Med. 3, 99ra87 (2011) DOI: 10.1126/scitranslmed.3002095 This PDF file includes: Fig. S1. Serum and plasma but not RBCs produce LPA receptor–dependent cortical wall disruption. Fig. S2. Experimental parameters of LPA cortical exposure model. Fig. S3. Lack of fronto-occipital changes and survival curve of LPA-injected animals that develop hydrocephalus over time. Fig. S4. Bilaterally increased ventricular area and mitotic displacement following LPA exposure. Fig. S5. LPA exposure induces the formation of denuded cell clusters that originate from the ventricular zone of the developing cortex. Fig. S6. LPA induces RhoA and Rac1 activation. Fig. S7. Expression of Lpar1 in the developing embryonic brain at E13.5. Fig. S8. Expression of Lpar2 in the developing embryonic brain at E13.5. Fig. S9. LPA-induced cortical disruption and mitotic displacement are abrogated in double-null mutant mice. Table S1. Shared pathological features of hydrocephalus in FH patients and mouse models. Table S2. Histological features associated with hydrocephalus are abrogated in LPA 1 and LPA 2 double-null animals. Table S3. Average ± SD, n, and P values of data graphed in Figs. 1, 4, and 5. References Descriptions for Videos S1 and S2 Other Supplementary Material for this manuscript includes the following: (available at www.sciencetranslationalmedicine.org/cgi/content/full/3/99/99ra87/DC1) Video S1 (.avi format). Ciliated ependymal cells are maintained at the apical ventricular surface after vehicle exposure.

Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

  • Upload
    others

  • View
    7

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

www.sciencetranslationalmedicine.org/cgi/content/full/3/99/99ra87/DC1

Supplementary Materials for

Lysophosphatidic Acid Signaling May Initiate Fetal Hydrocephalus

Yun C. Yung, Tetsuji Mutoh, Mu-en Lin, Kyoko Noguchi, Richard R. Rivera, Ji Woong Choi, Marcy A. Kingsbury, Jerold Chun*

*To whom correspondence should be addressed. E-mail: [email protected]

Published 7 September 2011, Sci. Transl. Med. 3, 99ra87 (2011)

DOI: 10.1126/scitranslmed.3002095

This PDF file includes:

Fig. S1. Serum and plasma but not RBCs produce LPA receptor–dependent cortical wall disruption. Fig. S2. Experimental parameters of LPA cortical exposure model. Fig. S3. Lack of fronto-occipital changes and survival curve of LPA-injected animals that develop hydrocephalus over time. Fig. S4. Bilaterally increased ventricular area and mitotic displacement following LPA exposure. Fig. S5. LPA exposure induces the formation of denuded cell clusters that originate from the ventricular zone of the developing cortex. Fig. S6. LPA induces RhoA and Rac1 activation. Fig. S7. Expression of Lpar1 in the developing embryonic brain at E13.5. Fig. S8. Expression of Lpar2 in the developing embryonic brain at E13.5. Fig. S9. LPA-induced cortical disruption and mitotic displacement are abrogated in double-null mutant mice. Table S1. Shared pathological features of hydrocephalus in FH patients and mouse models. Table S2. Histological features associated with hydrocephalus are abrogated in LPA1 and LPA2 double-null animals. Table S3. Average ± SD, n, and P values of data graphed in Figs. 1, 4, and 5. References Descriptions for Videos S1 and S2

Other Supplementary Material for this manuscript includes the following: (available at www.sciencetranslationalmedicine.org/cgi/content/full/3/99/99ra87/DC1)

Video S1 (.avi format). Ciliated ependymal cells are maintained at the apical ventricular surface after vehicle exposure.

Page 2: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

Video S2 (.avi format). Ciliated ependymal cells are lost in an incomplete fashion at the apical ventricular surface after LPA exposure.

Page 3: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

3

Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption. Wildtype (A, B, and C) or LPA1 and LPA2 double null mutant E13.5 embryos (D, E, and F) were injected with plasma, serum, or RBCs and analyzed at E14.5. (A and B) Injection of serum or plasma produced disruptions of the apical ventricular surface (indicated by arrows and shown in the mag-nified boxed inset). (C) RBC injected cortices retained a smooth apical ventricular zone. Abundant eryt-hrocytes (pink) are seen within the lateral ventricle. (D-F) Injection of serum, plasma, or RBCs into LPA1

-/- LPA2-/- double null mutant embryos did not produce cortical disruption. Tissue was stained with

hematoxylin and eosin. A = anterior, D = dorsal. CP = cortical plate, IZ = intermediate zone, VZ = ven-tricular zone. Scale bars, 200 µm [(A-F)] and 50 µm [A, inset].

-

Page 4: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

4

Fig. S2. Experimental parameters of LPA cortical exposure model. Embryos injected at E13.5 were examined for total brain weight and 18:1 LPA levels at E13.5 (1 h later), at E14.0 (12 h later), and E15.5 (48 h later). (A) Total brain weights between vehicle (white bars) and LPA injected (black bars) embryos were not statistically different up to 48 hours post-exposure (n ≥ 3 embryos per condition, mean ± s.d., unpaired t-test; 1 h, P = 0.77; 12 h, P = 0.44; 48 h, P = 0.55). (B) Wildtype brain 18:1 LPA amount remained relatively stable (between 0.32-0.35 pmol/mg) from E13.5 to E15.5 in vehicle injected embryos (white bars). LPA cortical injection resulted in an initially elevated amount (approximately 10.75 pmol/mg 18:1 LPA) at 1 h post-exposure, declining by 48 h post-exposure to approximately 1.79 pmol/mg 18:1 LPA (black bars). Although it is unclear what fraction of total LPA is available for signal-ing towards the development of FH (e.g., concentration at the receptors), these levels were consistent with total LPA concentrations that could be attained under pathophysiological conditions (up to 30 µM in se-rum) (1-3). (n ≥ 3 embryos per condition, average ± s.d., unpaired t-test; 1 h, * P = 0.03; 12 h, ** P = 0.005; 48 h, * P = 0.01)

Page 5: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

5

Fig. S3. Lack of fronto-occipital changes and survival curve of LPA injected animals develop hydrocephalus over time. (A) Embryos exposed to LPA (n = 7) (red lines) that develop hy-drocephalus with macrocephaly and increased interaural and mandibular-rostral dimensions (see also Fig. 1) do not display statistically different fronto-occipital distances compared with vehicle-injected embryos (n = 10) (blue lines) (average ± s.d., Mann-Whitney test). (B) Survival curve of postnatal animals ex-posed at E13.5 to vehicle (veh, blue line) or LPA (red line).

- that

Page 6: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

6

Page 7: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

7

Fig. S4. Bilaterally increased ventricular area and mitotic displacement following LPA ex-posure. E13.5 littermates exposed to LPA for 24 h developed ventricular dilation at all levels in both left and right cortices (B, D, F, H, J, L), in contrast to matched vehicle exposed cortices (A, C, E, G, I, K). Dotted white lines indicate representative areas measured for vehicle and LPA sample cohorts. Quantification of measured ipsilateral and contralateral areas is found in Fig. 2C. Note the increased PHH3+ mitotic displacement (red) in the LPA-exposed brains (D, arrows), compared with brains exposed to vehicle (C, arrowheads) (DAPI, blue nuclear counterstain). Insets show magnified cortical regions de-noted by arrows and arrowheads. Ctx = cortex. Dorsal is to the top of the page. Scale bar = 400 µm [(A-L)].

Page 8: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

8

Fig. S5. LPA exposure induces the formation of denuded cell clusters that originate from the ventricular zone of the developing cortex. (A, B) In situ hybridization with an Lpar1 DIG la-beled probe shows expression along the apical ventricular surface in vehicle (A), and LPA injected (B) E13.5 embryos exposed to LPA for 24 h. Note the smooth apical ventricular surface in embryos exposed to vehicle versus LPA. (D, E) Immunohistochemistry shows nestin positive cells along the apical ventri-cular surface in embryos exposed to vehicle (D) or LPA (E) for 24 hours. (G, H) BrdU staining of ve-hicle (G) and LPA (H) injected E13.5 embryos’ apical ventricle surfaces. LPA exposed cortices had un-dulating, disrupted apical surfaces that manifested as nodules (compare dotted outlines); in many cases, these cells appeared to detach and become free-floating cell clusters in the ventricles. (C, F, I) Cells within these clusters were Lpar1+ (C), nestin+ (F), and BrdU+ (I). R = rostral, D = dorsal, VZ = ventricu-lar zone, v = lateral ventricle. Scale bars = 50 µm [(A), (B), (D), (E), (G), (H)] and 20 µm [(C), (F), (I)].

Page 9: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

9

Fig. S6. LPA induces RhoA and Rac1 activation. (A, B) E13.5 embryonic cortical hemispheres were exposed to vehicle (Veh) alone (A) or LPA (B) for 1, 3, 5, 10 15, 30, 60, 120, or 180 minutes, then immediately microdissected on ice to remove the ganglionic eminences (GE) and enrich for the overlying cortex that is known to express LPA1 and which demonstrated disruption by LPA exposure (tissue region above white dotted line). (C) Time course of RhoA (red line) and Rac1 (blue line) activation upon LPA exposure by ELISA assay showed a nonlinear trend with rapid overactivation, followed by inhibition compared with control hemispheres. These initial trend lines were used to guide time point selection for further analysis (Fig. 3J and K). Scale bar = 300 µm [(A) and (B)].

Page 10: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

10

Fig. S7. Expression of Lpar1 in the developing embryonic brain at E13.5. (A-D), In situ hybridization using anti-sense and sense strand DIG-labeled probes for Lpar1 using cryo-sections from freshly-frozen tissue can be seen in both the coronal view (A, B) and horizontal view (C, D). Lpar1 was expressed in the ventricular zone (VZ) layer (A, C, arrows) of the lateral cortical wall at E13.5, more lightly at the medial wall, and was also present within the developing 3rd ventricle (C, ar-rowheads). The development of 3rd ventricular occlusions from LPA exposure (see Fig. 2) is consistent with this Lpar1 spatiotemporal expression. (E-L) Lpar1 in situ hybridization was also performed in pa-raffin-embedded tissue. Strong Lpar1 expression was present in the VZ (indicated by arrows), the mar-ginal zone (future layer 1) / meninges of the cortex (indicated by arrowheads), and the medial septal wall (SW) between the lateral ventricles (E, G, I, K). Adjacent sections were stained with hematoxylin and eosin (F, H, J, L). v = lateral ventricle, IZ = intermediate zone, CP = cortical plate, Ctx = cortex, M = marginal zone / meninges, Sk = developing skull, ChPl = choroid plexus, D = dorsal, L = lateral, R = ro-stral, A = anterior. Scale bar = 200 µm [(A-L)].

Page 11: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

11

Fig. S8. Expression of Lpar2 in the developing embryonic brain at E13.5. (A, B) In situ hybridization with DIG-labeled Lpar2 antisense (A) and sense (B) riboprobes shows that LPA2 was more diffusely expressed throughout both the medial (arrowheads) and lateral cortical wall (ar-rows) compared with Lpar1 (see Fig. S7). VZ = ventricular zone, IZ = intermediate zone, CP = cortical plate, v = ventricle, D = dorsal, L = lateral. Scale bar = 200 µm [(A) and (B)].

Page 12: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

12

Fig. S9. LPA-induced cortical disruption and mitotic displacement are abrogated in double-null mutant mice. (A, D) Control mice (LPA1

+/- LPA2-/-) injected with LPA showed altered β-

III-tub expression, consistent with cortical disruption and heterotopia formation, in contrast to the double null mice (LPA1

-/- LPA2-/-). (B, E) Cortical disruption resulted in phospho-histone H3 (PHH3) positive

mitotic cells moving basally away from the ventricular surface (indicated by arrows), which was not present in double null mice. (C, F) Merged images of β-III-tub (green), PHH3 (red), and nuclear coun-terstaining DAPI (blue). (G) Quantification of mitotic displacement (%) (n ≥ 4 embryos per condition, average ± s.d., unpaired t-test, * P = 0.016). VZ = ventricular zone, SVZ = subventricular zone, IZ = in-termediate zone, CP = cortical plate, v = ventricle, Scale bar = 50 µm [(A-F)].

Page 13: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

13

Principal features of FH

Clinical evidence References* Evidence from animal models References* Exposure of mouse embryos to LPA or blood (Yung et al., this issue)

Intracranial hemorrhage

Strongest co-morbidity factor for FH in epidemiological studies (occurs in 30% of cases)

1, 23, 34 Injection of neonatal rats with whole or citrated blood results in ventricular dilation and hydrocephalus

11, 35 Exposure to the blood-borne lipid LPA or blood components initiates FH in a new mouse model

Ventricular dilation Defined as an atrial width >3 stan-dard deviations above mean; typi-cally precedes FH in epidemiological studies. Often but not always associ-ated with intracranial hemorrhage

1, 3, 25, 34 Reported in genetic null mice lacking proteins related to cilia, cell polarity, or cell adhesion

11,16-20, 24, 26-29, 35

Mouse embryos exposed to LPA or blood developed cerebral ventricular di-lation within 24 hours

Neural progenitor cell dysfunction

Neonatal babies with FH have clus-ters of β-III-tubulin+ neuroblasts that protrude into the cerebral ventricles

12, 25 Reported in N-cadherin and hyh mu-tant mice. Embryonic cortex showed misplacement of neural progenitor cells from the apical to the basal sur-face of the lateral ventricles

14, 15, 19, 29, 45

Exposure of embryonic mice to LPA or blood components caused mitotic cell displacement, as seen in N-cadherin null mutant mice

Loss of ependymal cells

Partial loss within lateral ventricles at 16 weeks of gestation; progressive loss by 40 weeks with concomitant gliosis. Cell loss within 4th ventricle at sites of hemorrhage reported

23, 25 Loss in genetic null mice lacking pro-teins related to cilia, cell polarity, or cell adhesion

16, 18, 20, 24, 29, 35

Loss seen by early postnatal develop-ment (indicated by loss of S100β+ cells lining the ventricles)

Presence of neurorosettes

These aberrant, radially-shaped structures were found localized near areas of ependymal cell loss and hemorrhage

23, 25 Seen in cortical walls of N-cadherin null mutant mice, after administration of N-cadherin antibodies, and in ge-netic null mutant mice lacking cell polarity proteins

14, 15, 19, 20, 29

Observed in the cortical walls of the VZ layer or more basally in mouse embryos exposed to LPA or blood

Presence of mislocalized cells (heterotopia)

Misplacement of β-III-tubulin+ cells within the glial layer, and gray mat-ter nodules protruding into lateral ventricles in FH neonates

12, 23 Columns of β-III-tubulin+ cells in mutant mice with dysfunctional cell adhesion. Abnormal protrusion of neurons into the 4th ventricle and clo-sure of the spinal canal in mice lack-ing myosin

17, 19 β-III tubulin+ cells protrude into the lat-eral and 3rd ventricles in mouse em-bryos exposed to LPA or blood. Cells form nodules that cause partial occlu-sion of the 3rd ventricle

Presence of defects in cilia

Loss of ciliated ependymal cells may partly explain altered CSF dynamics in FH

25 Mutant mice with loss of cilia or ependymal cells develop FH

26-28, 30 LPA or blood exposure causes partial loss of S100β+ ciliated ependymal cells with concomitant loss of cilia along cerebral ventricular walls

3rd ventricle occlu-sion or aqueductal stenosis

Occlusion or stenosis due to protru-sions of cells from the ventricular wall into the ventricular spaces

23, 25, 34 Genetic deletion of cilia proteins or myosin II-B and IXa causes stenosis of the Sylvian aqueduct (between the 3rd and 4th ventricles) and the spinal canal. Also seen in the H-Tx rat model of hydrocephalus

17, 18, 27, 45

Exposure to LPA or blood causes nodu-lar protrusions within the lateral ventri-cle and 3rd ventricle, producing partial occlusion of these structures

Table S1. Shared pathological features of hydrocephalus in FH patients and mouse models. *References are in main text. Only key references are cited.

Page 14: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

14

Table S2. Histological features associated with hydrocephalus are abrogated in LPA1 and LPA2 double null animals. Frank hydrocephalus was absent in ~ 90% of the double null mutants. The presence of mild histological features, markedly reduced from controls, supports the involvement of LPA1 and LPA2 in these histological changes, and suggest that one or more of the other 4 remaining LPA receptors may further contribute to these phenotypes.

-

Page 15: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

15

Table S3. Average ± , n, and P values of data graphed in Figs. 1, 4, and 5. * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001, * P ≤ 0.05 was considered significant. # denotes decreased animal number in this vehicle-exposed cohort due to intentional sacrifice for histology (see survival curve in Fig. S3B). mm = millimeters.

SD

Page 16: Supplementary Materials for...3 Fig. S1. Serum and plasma but not RBCs produce LPA receptor dependent cortical wall disruption.Wildtype (A, B , and C) or LPA1 and LPA 2 double null

16

References accompanying supplementary figures S1. A. Tokumura, M. Iimori, Y. Nishioka, M. Kitahara, M. Sakashita, S. Tanaka, Lysophosphatidic acids

induce proliferation of cultured vascular smooth muscle cells from rat aorta. Am J Physiol 267, C204-210 (1994).

S2. D.L. Baker, D.M. Desiderio, D.D. Miller, B. Tolley, G.J. Tigyi, Direct quantitative analysis of lysophosphatidic acid molecular species by stable isotope dilution electrospray ionization liquid chromatography-mass spectrometry. Anal Biochem 292, 287-295 (2001).

S3. J. Aoki, A. Taira, Y. Takanezawa, Y. Kishi, K. Hama, T. Kishimoto, K. Mizuno, K. Saku, R. Taguchi, H. Arai, Serum lysophosphatidic acid is produced through diverse phospholipase pathways. J Biol Chem 277, 48737-48744 (2002).

Video S1. Ciliated ependymal cells are maintained at the apical ventricular surface after vehicle exposure. S100β+ ependymal cells (red) line the apical ventricular surface (up) and are ciliated (AcTub, green). Nuclei are counterstained with DAPI (blue).

Video S2. Ciliated ependymal cells are lost in an incomplete fashion at the apical ventricu-lar surface after LPA exposure. Some S100β+ ependymal cells (red) that line the apical ventricular surface (up) and are ciliated (AcTub, green) are lost after LPA exposure; these areas have cells devoid of both S100β and AcTub immunoreactivity. Nuclei are counterstained with DAPI (blue).