throbber
i\
`t;~
`
`PNAS
`
`w,
`
`.
`
`.
`
`. . Proceedings of the National Academy of Sciences
`\...:~""'·"'"'
`of the United States of America
`.
`DOES NOT LEA VE
`THE LIBRARY
`
`April 10, 2001 I vol. 98 I no. 8 I pp. 4277-4816 I www.pnas.org
`
`univ. of Minn.
`Bio~Medical
`Library
`
`Where the poly(A)-binding protein binds translation factors
`
`Ancient carbon cycle tells of past biological diversity
`
`New World patterns of human migration
`
`Retrovirus linked with schizophrenia
`
`Pharmaceutical intervention for weight control
`
`Breckenridge Exhibit 1096
`Wen
`Page 001
`
`

`

`4581 Selective ablation of retinoid X receptor a in
`~ hepatocytes impairs their lifespan and
`regenerative capacity
`Takeshi Imai, Ming Jiang, Philippe· Kastner, Pierre Cbambon,
`and Daniel Metzger
`
`IMMUNOLOGY
`
`4587 Localization of co4+ T cell epitope hotspots to exposed
`~ strands of HIV envelope glycoprotein suggests
`structural influences on antigen processing
`Sherri Surman, Timothy D. Lockey, Kar en S. Slobod,
`Bart Jones, Janice M. Riberdy, Stephe n W. While,
`Peter C. Doherty, and Julia L. Hurw itz
`4593 lmmucillin H, a powerful transition-state analog
`~ inhibitor of purine nucleoside phosphorylase,
`selectively inhibits human T lymphocytes
`Greg A. Kicska, Li Long, H eidi Ha rig, Craig Fairchild,
`Peter C. Tyler, Richard H. Furneaux, Vern L. Schramm
`and Howard L. Kaufman
`
`4599 Role of MEKK2-MEK5 in the regulation of TNF-a gene
`~ expression and MEKK2-MKK7 in the activation of
`c-Jun N-terminal kinase in mast cells
`Kosuke Chayama, Philip J. Papst, Timothy P. Garrington,
`Joanne C. Pratt, Tamotsu Ishizuka, Saipbone Webb,
`Soula Ganiatsas, Leonard I. Zon, Weiyong Sun,
`Gary L. Johnson, and E rwin W. Gelfand
`
`MEDICAL SCIENCES
`
`4605 Comparative evaluation of the antitumor activity of
`~ antiangiogenic proteins delivered by gene transfer
`Calvin J. Kuo, Filip Farnebo, Evan Y. Yu,
`Rolf Christofferson, Rebecca A. ·swearingen, Robert Carter,
`Horst A. von Recum, Jenny Yuan, Junne Kamihara,
`Evelyn Flynn, Robert D'Amato, Judah Folkman,
`and Richard C. Mulligan
`4611 An important function of Nrf2 in combating oxidative
`~ stress: Detoxification of acetaminophen
`Kaimin Chan, Xiao-Dong Han, and Yuet Wai Kan
`4617 Correlation of breath ammonia with blood urea
`nitrogen and creatinine during hemodialysis
`L. R. Narasimhan, William Goodman, and C. Kumar N. Patel
`
`4622 PTEN controls tumor-induced angiogenesis
`~ Shenghua Wen, Javor Stolarov, Michael P. Myers,
`Jing Dong Su, Michael H. Wigler, Nicholas K. Tonks,
`and Donald L. Durden
`4628 Role of tumor-host interactions in interstitial diffusion
`~ of macromolecules: Cranial vs. subcutaneous tumors
`A lain Pluen, Yves Boucher, Saroja Ra manujan,
`Trevor D. McKee, Takeshi Gohongi, Emmanuelle di Tomaso,
`Edward B. Brown, Yotaro Izumi, Robert B. Campbell,
`David A. Berk, and R akesh K. Jain
`4634 Retroviral RNA identified in the cerebrospinal fluids and
`brains of individuals with schizophrenia
`Hllkan Karlsson, Silke Bachmann, Johannes Schroder,
`Justin McArthur, E. Fuller Torrey, and Robert H. Yolken
`-t See commentary on page 4293
`4640 A phosphatidylinositol 3-kinase/ Akt/ mTOR pathway
`~ mediates and PTEN antagonizes tumor necrosis factor
`inhibition of insulin signaling through insulin
`receptor substrate-1
`Osman Nidai Ozes, H akan Akca, Lindsey D. Mayo,
`Jason A. Gustin, Tomohiko Maehama,
`Jack E. Dixon, and David B. Donner
`
`vi
`
`I www.pnas.org
`
`4646 Antisense-mediated depletion of p300 in human cells
`leads to premature G, exit and up-regulation of c-MYC
`Sivanagarani Kolli, Ann Marie Buchmann, Justin Williams,
`Sigmund Weitzman, and Bayar Thimmapaya
`
`4652 Ciliary neurotrophic factor activates leptin-like
`·~ pathways and reduces body fat, without cachexia or
`rebound weight gain, even in leptin-resistant obesity
`P. D. Lambert, K. D. A nderson, M. W. Sleeman, V. Wong,
`J. Tan, A. Hijarunguru, T. L. Corcoran, J. D. Murray,
`K. E. T habet, G.D. Yancopoulos, and S. J. Wiegand
`-t See commentary 011 page 4279
`
`MICROBIOLOGY
`
`4658 Complete genome sequence of an M1 strain of
`Streptococcus pyogenes
`Joseph J. Ferretti, William M. McShan, Dragana Ajdic,
`Dragutin J. Savic, Gorana Savic, Kevin Lyon,
`Charles Primeaux, Steven Sezate, Alexander N. Suvorov,
`Steve Kenton, Hong Shing Lai, Shao Ping Lin, Yudong Qian,
`Hong Gui J ia, Fares Z. Najar, Q un Ren, Hua Zhu, Lin Song,
`Jim White, Xiling Yuan, Sandra W. Clifton, Bruce A. Roe,
`and Robert McLaughlin
`
`4664 Purification and characterization of an autoregulatory
`~ substance capable of regulating the morphological
`transition in Candida albicans
`Ki-Bong Oh, Hiroshi Miyazawa, Tosbimichi Naito,
`and Hideaki Matsuoka
`4669 Polymerization of a single protein of the pathogen
`~ Yersinia enterocofitica into needles punctures
`eukaryotic cells
`Egbert Hoiczyk and Gunter Blobel
`4675 Epstein-Barr virus latent-infection membrane proteins
`are palmitoylated and raft-associated: Protein 1
`binds to the cytoskeleton through TNF receptor
`cytoplasmic factors
`Masaya Higuchi, Kenneth M. Izumi, and Elliott Kieff
`4681 Proteomic analysis of the bacterial cell cycle
`~ Bjorn Griinenfelder, Gabriele Rummel, J iri Vohradsky,
`Daniel Roder, Hanno Langen, and Urs Jena!
`
`NEUROBlOLOGY
`
`4687 Mechanisms of migraine aura revealed by functional
`~ MRI in human visual cortex
`Nouchine Hadjikhani, Margarita Sanchez de! Rio, Ona Wu,
`Denis Schwartz, Dick Bakker, Bruce Fischl,
`Kenneth K. Kwong, F. Michael Cutrer, Bruce R. Rosen,
`Roger B. H. Toolell, A. Gregory Sorensen,
`and Michael A. Moskowitz
`4693 Rethinking the role of phosducin: Light-regulated
`~ binding of phosducin to 14-3-3 in rod inner segments
`Koichi Nakano, Jing Chen, George E. Tarr, Tatsuro Yoshida,
`Julia M. F lynn, and Mark W. Bitensky
`4699 Allosteric modulation of Ca2+ channels by G proteins,
`voltage-dependent facilitation, protein kinase C, and
`CavfJ subunits
`Stefan Herlitze, Huijun Zhong, Todd Scheuer,
`and William A. Catterall
`4705 Control of gating mode by a single amino acid residue
`in transmembrane segment 153 of the N-type
`Ca2 + channel
`Huijun Zhong, Bin Li, Todd Scheuer,
`and William A. Catterall
`
`4710 Net
`sut
`the
`Ku:
`So1
`
`471 6 Lac
`~ 11,
`kn1
`lea
`Jo}
`Ca
`an<
`
`4722 F-s
`~ me
`Ve
`
`4728 Eft
`~ pe
`M1
`Joi
`an
`4734 {J(cid:173)
`~ ai
`hi1
`Qi
`
`4740 Di
`~ an
`in·
`El
`M
`Cl
`ar
`4746 G·
`o1
`y
`K
`a1
`4752 Tl
`o'
`D
`4758 R
`p
`A
`4764 H
`h
`H
`J.
`I
`I
`
`4770 J
`~ ~
`1
`
`4776 (
`~ 1
`
`Breckenridge Exhibit 1096
`Wen
`Page 002
`
`

`

`PTEN controls tumor-induced angiogenesis
`
`Shenghua Wen*, Javor Stolarov†, Michael P. Myers†, Jing Dong Su*, Michael H. Wigler†, Nicholas K. Tonks†,
`and Donald L. Durden*‡
`
`*Section of HematologyyOncology, Department of Pediatrics, Herman B Wells Center for Pediatric Research, Department of Biochemistry and Molecular
`Biology, Indiana School of Medicine, Indianapolis, IN 46202; and †Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
`
`Contributed by Michael H. Wigler, February 7, 2001
`
`Mutations of the tumor suppressor PTEN, a phosphatase with
`specificity for 3-phosphorylated inositol phospholipids, accom-
`pany progression of brain tumors from benign to the most malig-
`nant forms. Tumor progression, particularly in aggressive and
`malignant tumors, is associated with the induction of angiogene-
`sis, a process termed the angiogenic switch. Therefore, we tested
`whether PTEN regulates tumor progression by modulating angio-
`genesis. U87MG glioma cells stably reconstituted with PTEN cDNA
`were tested for growth in a nude mouse orthotopic brain tumor
`model. We observed that the reconstitution of wild-type PTEN had
`no effect on in vitro proliferation but dramatically decreased tumor
`growth in vivo and prolonged survival in mice implanted intracra-
`nially with these tumor cells. PTEN reconstitution diminished
`phosphorylation of AKT within the PTEN-reconstituted tumor,
`induced thrombospondin 1 expression, and suppressed angiogenic
`activity. These effects were not observed in tumors reconstituted
`with a lipid phosphatase inactive G129E mutant of PTEN, a result
`that provides evidence that the lipid phosphatase activity of PTEN
`regulates the angiogenic response in vivo. These data provide
`evidence that PTEN regulates tumor-induced angiogenesis and the
`progression of gliomas to a malignant phenotype via the regula-
`tion of phosphoinositide-dependent signals.
`
`The reversible phosphorylation of proteins and lipids is critical
`
`to the control of signal transduction in mammalian cells and
`is regulated by kinases and phosphatases (1). The product of the
`tumor suppressor gene PTENyMMAC (hereafter termed
`PTEN) was identified as a dual specificity phosphatase and has
`been shown to dephosphorylate inositol phospholipids (2–9).
`The PTEN gene is mutated in 40–50% of high-grade gliomas as
`well as prostate, endometrial, breast, lung, and other tumors (2,
`3, 10). In addition, PTEN is mutated in several rare autosomal
`dominant cancer predisposition syndromes, including Cowden
`disease, Lhermitte-Duclos disease, and Bannayan-Zonana syn-
`drome (11–14). The phenotype of PTEN-knockout mice reveal
`a requirement for this phosphatase in normal development and
`confirm its role as a tumor suppressor (15–17).
`PTEN is a 55-kDa protein comprising an N-terminal catalytic
`domain, identified as a segment with homology to the cytoskel-
`etal protein tensin and containing the sequence HC(X)5R, which
`is the signature motif of members of the protein tyrosine
`phosphatase family, and a C-terminal C2 domain with lipid-
`binding and membrane-targeting functions (18). The sequence
`of the extreme C terminus of PTEN suggests a function in
`binding PDZ domain-containing proteins. PTEN is a dual
`specificity phosphatase that displays a pronounced preference
`for acidic substrates (5). Importantly, PTEN possesses lipid
`phosphatase activity, preferentially dephosphorylating phos-
`phoinositides at the D3 position of the inositol ring. It is the only
`enzyme known to dephosphorylate the D3 position in inositol
`phospholipids, suggesting that PTEN may function as a direct
`antagonist of phosphatidylinositol 3-kinase (PI3-kinase) and
`phosphatidylinositol 3,4,5-trisphosphate [PtdIns(3,4,5)P3]-
`dependent signaling (7, 13). Reconstitution of PTEN in tumor
`cells that carry mutations in the PTEN gene have established
`that this phosphatase regulates the PI3-kinase-dependent acti-
`vation of AKT, a major player in cell survival (6, 14). However,
`despite progress in understanding the biochemistry of PTEN,
`
`the role of this phosphatase in tumor progression, as it relates to
`its diverse effects on cell growth, angiogenesis, andyor survival,
`remains unclear.
`Mutations in PTEN accompany progression of brain tumors
`from grade IyII to malignant grade III and IV in vivo (19, 20).
`Tumor progression is associated with angiogenesis, the formation
`of new blood vessels from existing vascular structures, with in-
`creases in microvessel density (MVD) and increased invasion of
`tumor cells into brain parenchyma (21–23). For tumor growth to
`occur, tumor dormancy must be broken, an event termed the
`angiogenic switch. During angiogenesis endothelial cells are in-
`duced to degrade the basement membrane of existing vessels, break
`away, and migrate to the site of the tumor, where they proliferate
`to form linear structures that differentiate to form blood vessels.
`Factors that control angiogenesis include growth factors, matrix
`metalloproteinases, plasminogen activators, thrombospondins, in-
`tegrins avb3, avb5, and a5b1, etc. (23–25). The angiogenic switch
`involves a shift in the balance of angiogenic stimulators and
`angiogenic inhibitors. Stimulators include the growth factors, vas-
`cular endothelial growth factor and basic fibroblast growth factor,
`and the induction of matrix remodeling via matrix metal-
`loproteinases (26). Inhibitors include thrombospondin 1 (TSP-1),
`angiostatin, endostatin, tissue inhibitors of metalloproteinases, and
`others (24, 27). It has been observed that neovascularization and
`PTEN mutations are associated with high-grade gliomas and are
`not observed in low-grade glial tumors, leading to the hypothesis
`that these two events may be causally linked.
`Regulation of PI3-kinase-dependent signals, including activa-
`tion of AKT by vascular endothelial growth factor and its
`receptors, the protein tyrosine kinases Flt-1 and KDR, have been
`implicated in brain tumor angiogenesis (28). Data generated in
`the chicken chorioallantoic membrane model suggests that PI3-
`kinase-dependent pathways may regulate angiogenesis and vas-
`cular endothelial growth factor expression in endothelial cells
`(29). Furthermore, correlative studies in prostate tumor speci-
`mens have demonstrated that tumors containing PTEN muta-
`tions have higher microvessel counts than tumors expressing
`wild-type (WT) PTEN (30). However, whether PTEN is causally
`linked to induction of angiogenesis by the tumor cell remains
`unproven. These and other observations led us to hypothesize
`that PTEN may control tumor-induced angiogenesis and con-
`tribute to the high mortality associated with malignant brain
`tumors.
`
`Materials and Methods
`Cell Culture, Constructs, and Reagents. WT PTEN or mutant PTEN
`(G129E, R130 M) cDNAs were subcloned into the pBabe-puro
`retroviral expression vector. Stable clones of U87MG cells
`expressing WT PTEN (WT.E1, WT.C7) or mutant PTEN
`(G129E, R130 M) were established under puromycin selection
`
`Abbreviations: PI3-kinase, phosphatidylinositol 3-kinase; PtdIns(3,4,5)P3, phosphatidylino-
`sitol 3,4,5-trisphosphate; TSP-1, thrombospondin 1; WT, wild type; MVD, microvessel
`density.
`‡To whom reprint requests should be addressed. E-mail: ddurden@iupui.edu.
`
`The publication costs of this article were defrayed in part by page charge payment. This
`article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C.
`§1734 solely to indicate this fact.
`
`4622– 4627 u PNAS u April 10, 2001 u vol. 98 u no. 8
`
`www.pnas.orgycgiydoiy10.1073ypnas.081063798
`
`Breckenridge Exhibit 1096
`Wen
`Page 003
`
`

`

`MEDICALSCIENCES
`
`Stable expression of PTEN and PTEN mutants in U87MG cells regulates
`Fig. 1.
`AKT. (A) Cell lysates from the U87MG (U87) cell line and U87 cells infected with
`a retroviral vector encoding PTEN (pBabe-Puro-PTEN) or mutants of PTEN
`(pBabe-Puro-PTEN-G129E or R130 M) were resolved by SDSyPAGE, and equal
`amounts of proteins were loaded per lane, immunoblotted with antisera to
`PTEN, phospho-AKT, and total AKT, and visualized by enhanced chemilumi-
`nescence. The basal levels of PTEN (Top), phosphorylated AKT (Ser-473) (Mid-
`dle), and total AKT (Bottom) are shown. The status of the PTEN gene in each
`stable cell line was designated as: WT.E1 and WT.C7, two separate clones
`expressing WT PTEN; R130 M and G129E are mutated PTEN, R130 M is inert as
`both a protein and a lipid phosphatase. The G129E PTEN can dephosphorylate
`acidic phosphopeptides, but cannot dephosphorylate lipid substrate,
`PtdIns(3,4,5)P3. The U87MG (U87) cell line is the parental cell line isolated from
`a human glioblastoma multiforme patient. (B) Comparison of in vitro growth
`of U87MG cells transduced with mutants of PTEN. Equal number of cells (1 3
`105) were incubated in RPMI 1 10% FBS for different times, and cell numbers
`were quantitated by direct cell counting.
`
`determined for five tumors per experimental group and analyzed
`by Student’s t test.
`
`Results
`Effect of PTEN Reconstitution on in Vitro Growth of Tumor. To
`determine whether PTEN exerts control over angiogenesis and
`the growth of glial tumors, we developed an orthotopic brain
`tumor model in which PTEN-deficient tumor cells were genet-
`ically manipulated in vitro and then stereotactically injected into
`the skin or frontal cerebral cortex of nude mice. The U87MG cell
`line is derived from a patient diagnosed with glioblastoma
`multiforme, a highly malignant and uniformly fatal brain tumor.
`This tumor and other human glioblastomas and glioblastoma cell
`lines contain a mutation in both PTEN alleles, resulting in a null
`genotype. In light of these observations, we reconstituted the
`PTEN gene in the parental U87MG (U87) cells.
`Stable derivatives of the parental U87 cells were generated
`after transduction with retroviruses encoding cDNA for WT
`PTEN or specific mutants of this phosphatase. In particular, we
`used missense mutations in the PTP signature motif to ascertain
`the importance of the enzymatic activity of PTEN to its tumor
`suppressor function. This included R130 M, in which all phos-
`phatase activity is abrogated (5, 33), and G129E, which has been
`identified in Cowden disease and endometrial cancer and in
`which the activity toward inositol phospholipids is severely
`attenuated but protein phosphatase activity is essentially intact.
`Tumor cells were characterized biochemically for levels of
`activated AKT (phospho-S473-AKT), growth in vitro, and PTEN
`expression (Fig. 1). Anti-PTEN blots confirmed that parental
`
`(2 mgyml) (6). Muristirone-induced expression of PTEN in
`U87MG cells was performed as described by J. Stolarov (31).
`Antibodies were obtained specific for PTEN (6), AKT and
`phospho-S473-AKT (New England Biolabs, #9270), TSP-1 (Cal-
`biochem, #605230), and anti-BrdUrd mAb clone BU33 (Sigma,
`#B9285).
`
`Tumor Implantation. Cells were cultured in fresh medium for 24 h
`and harvested, adjusting the cell concentration to 1 3 106 in 10
`ml of RPMI medium. Mice, under general anesthesia were placed
`into the stereotactic device (model 963, Kopf Instruments,
`Tujunga, CA). Stereotactically controlled drill assembly was
`used to provide a hole 0.3 mm deep and of 0.8 mm diameter in
`cranium at a position 0.5 mm anterior and 1.2 mm lateral to the
`landmark. Tumor cells (1 3 106) were
`bregmal anatomical
`introduced slowly through a 10-ml Hamilton syringe at a depth
`of 2.5 mm at a rate of 2 mlymin. We then slowly removed the
`needle at a rate of 0.5 mmymin. After needle removal we sealed
`the hole with bone wax and closed the incision with a wound clip.
`In some of the mice, 5 3 106 tumor cells were implanted s.c. into
`the right flank to monitor tumor volume and to perform
`biochemical and immunohistochemical analysis of tumor tissue.
`All animal experiments performed were approved by the Animal
`Care Committee at Indiana University School of Medicine.
`
`Biochemical Analysis. Immunoblots were performed on cell lysates
`obtained from U87 cells grown in tissue culture or from multiple
`cryostat sections of s.c. tumor tissues. A Bradford assay was
`performed to determine protein concentration of each lysate.
`Equivalent amounts of protein were resolved by SDSyPAGE and
`transferred to nitrocellulose. Membranes were probed with
`antisera specific for PTEN, AKT, phospho-S473-AKT, or TSP-1.
`The RNase protection assay was performed by using a RPA III
`kit from (Ambion) according to the manufacturer’s specifica-
`tions. Briefly, 20 mg of total RNA was precipitated and resus-
`pended in 10 ml of hybridization buffer containing the radioac-
`tive probe. The RNA then was heated to 95°C for 10 min and
`hybridized for 16 h at 42°C. A total of 150 ml of this mixture was
`treated with 1:100 dilution of RNase in RNase buffer for 30 min.
`RNase was inactivated, and RNA was reprecipitated and re-
`solved on 5% acrylamide gel. RNA probes were synthesized by
`using MAXI SCRIPT using PCR templates and T7 polymerase. The
`glyceraldehyde-3-phosphate dehydrogenase probe was provided
`in the kit, and TSP-1 probe represents a 590-nt sequence in the
`39 untranslated region of TSP-1 sequence. All probes were
`sequenced.
`
`Immunohistochemical Analysis. MVD was determined for each s.c.
`and brain tumors by CD31 staining, and a proliferative index was
`determined by using anti-BrdUrd mAb staining performed on
`cryostat sections (7 mm), fixed in acetone, blocked in 1% goat
`serum, and stained with anti-CD31 antibody (PharMingen,
`#01951D). Antibody staining was visualized with peroxidase-
`conjugated anti-mouse and counterstained with hematoxylin. A
`negative control was performed on each tumor tissue stained
`with mouse IgG. Two sections from each tumor were scanned
`under low-power magnification (340) to identify areas of highest
`CD31-positive vessel density (32), followed by digitization of
`three fields from this area. For in vivo BrdUrd labeling, mice
`received 100 ml of BrdUrd in PBS (10 mM) injected into the tail
`vein 1 h before tumor harvesting. The digitized images repre-
`senting one 320 field were counted for the number of CD31-
`positive vascular elements and number of BrdUrd-positive cellsy
`field. The number of cells positive for BrdUrd staining ranged
`between 5% and 8% of total number of tumor cells. Data were
`collected from two independent observers without knowledge of
`which tumors were viewed. The average number of microvessels
`or cells positive for BrdUrd staining per digitized field was
`
`Wen et al.
`
`PNAS u April 10, 2001 u vol. 98 u no. 8 u 4623
`
`Breckenridge Exhibit 1096
`Wen
`Page 004
`
`

`

`U87 cells do not express PTEN and that after reconstitution of
`mutant or WT PTEN expression, U87 cells express amounts
`comparable to WT physiological
`levels. Expression of WT
`PTEN, to levels similar to those observed in a mouse brain lysate
`and primary human astrocytes, suppressed the activated state of
`AKT observed in PTEN-deficient U87 cells (Fig. 1 A, lanes 3 and
`5). After expression of the R130 M and G129E mutant forms of
`PTEN, the levels of phospho-AKT were similar to those ob-
`served in the parental U87 cells (Fig. 1 A, lanes 1, 2, and 4),
`suggesting that the lipid phosphatase activity of PTEN was
`essential for the effects on the PtdIns(3,4,5)P3-dependent acti-
`vation of AKT. Importantly, the growth of the different PTEN-
`expressing U87 cell lines in vitro was similar in 2%, 5%, and 10%
`FBS (data not shown and Fig. 1B). Therefore, we compared
`these cell lines further in our in vivo models.
`
`Effect of PTEN Reconstitution on inVivoGrowth and Proliferation. We
`implanted athymic nude mice s.c. and by intracranial injection.
`Production of s.c. tumors allowed us to monitor the size of the
`tumor and perform direct biochemical analysis of tumor tissue
`for PTEN expression and levels of AKT activation without
`significant contamination from other tissues. Tumor tissue
`blocks were processed for hematoxylinyeosin staining. Greater
`than 95% of tissue analyzed was tumor. We compared the levels
`of PTEN in tumor tissue and numerous normal tissues within the
`athymic nude mouse. Using anti-PTEN antisera, we detected the
`expression of PTEN in all tissues, with the exception of skeletal
`and heart muscle (data not shown), but no PTEN was detected
`in parental U87-derived tumor tissue (Fig. 2C, lane 4). These
`results indicate that the tumor tissue sampled represents pre-
`dominantly tumor cell-derived proteins. As observed in the cell
`lines grown in vitro, s.c. tumors, derived from U87 cells recon-
`stituted with mutant or WT PTEN, displayed similar levels of
`PTEN expression (Fig. 2C, lanes 1–3 and 5). Phospho-AKT
`activity was higher in PTEN-null U87 cells and U87 cells
`reconstituted with R130 M and to a lesser extent in U87 cells
`expressing the G129E mutant (Fig. 2C, lane 1) as compared with
`the WT PTEN-transduced cells (Fig. 2C, compare lanes 1, 2, and
`4 to lanes 3 and 5). The pattern of phosphorylated AKT was
`similar when the different U87 mutant expressing cell lines were
`assayed in vitro or in vivo (compare Fig. 1 A to 2C).
`Despite the similar in vitro growth rate, there was a dramatic
`difference in the growth of tumors derived from parental U87 cells
`compared with cells reconstituted with WT PTEN (Fig. 2 A and B).
`The average volume of U87-derived tumors on day 25 after
`implantation was 848 6 203 mm3, compared with 91 6 27 mm3 for
`tumors derived from WT PTEN-reconstituted cells (n 5 5, P ,
`0.0001). Interestingly, the reconstitution of U87 cells with catalyt-
`ically impaired PTEN (G129E or R130 M mutants) (Fig. 2A) shows
`an intermediate level of growth suppression, a result that suggests
`some residual function of these mutants in vivo.
`Despite dramatic differences in the size of these tumors, in vivo
`BrdUrd labeling of tumor cells revealed no significant difference
`in number of cells in S phase: 72 6 6 BrdUrd-positive cells per
`field in parental U87MG tumor mass versus 68.5 6 3 in WT
`PTEN reconstituted tumors (data not shown). The percentage
`of TUNEL-positive nuclei within the U87MG and WT PTEN
`reconstituted tumors was similar. These results suggested that
`something other than the proliferative or apoptotic rate of the
`U87MG versus U87MG reconstituted with WT PTEN ac-
`counted for the difference in growth potential. We observed that
`the loss of inositol phospholipid phosphatase activity results in
`deregulated tumor growth comparable to the total ablation of
`catalytic activity and that the despite differences in overall
`growth in vivo, the proliferative rate of these tumors is similar.
`These findings are consistent with a role for PTEN in the
`regulation of another aspect of tumor biology, such as angio-
`
`Effects of PTEN on growth of U87MG cells in vivo. (A) Cell growth in vivo.
`Fig. 2.
`To determine the rate of cell growth in vivo, equal amounts of cells (5 3 106) from
`each cell line were implanted at the right ventral flank by s.c. injection. The
`formation and growth of the s.c. tumor was monitored, and the volume of the
`tumor was determined by a three-dimensional measurement at the times indi-
`cated (day 0, the date of implantation, no tumor is detected). Data were analyzed
`by Student’s t test, and differences were significant comparing the PTEN deficient
`(U87MG, R130 M, G129E) to the WT PTEN (WT.E1, WT.C7), n 5 5, number of mice;
`P , 0.0001. (B) Stereophotography of s.c. tumor sites in mice implanted with the
`parental U87 tumor, PTEN minus (Left) versus WT PTEN reconstituted tumor cells
`(Right). These tumors represent 25 and 42 days after implantation for PTEN minus
`versus WT PTEN reconstituted tumors, respectively. (Magnification, 340.) (C)
`Immunoblot of cryostat tissue sections from s.c. tumor for the expression pattern
`of PTEN, AKT, and phosphorylated AKT. Frozen tissue sections were solubilized in
`Laemmli sample buffer, total protein was quantitated, and equal protein was
`loaded on SDSyPAGE. The data shown are representative of tissue analysis from
`five animals per experimental group.
`
`genesis. This led us to assess the effect of PTEN reconstitution
`on the induction of angiogenesis in this model.
`
`PTEN Suppresses Tumor-Induced Angiogenesis. To examine the
`effect of PTEN on angiogenesis, we compared parental U87 cells
`to cells reconstituted with WT or mutant PTEN. We stained
`cryostat sections from s.c. tumors for CD31 (PECAM). CD31 is
`an endothelial marker used to measure the MVD of these
`tumors. MVD was assessed from multiple digitized images of
`CD31-stained tumor tissue at 3100 magnification (three fields
`were evaluated per tumor) and counted blindly for the number
`of CD31-positive microvessels per unit surface area as described
`(32). Reconstitution of PTEN expression in U87 cells dramat-
`ically suppressed the angiogenic response in vivo (Fig. 3 A and
`B). Quantitation of MVD in tumors derived from parental U87
`cells (77 6 13) and U87 cells expressing WT PTEN (38 6 7)
`revealed an ’50% suppression of angiogenesis (Fig. 3C) (n 5 5,
`P , 0.001). The MVD of tumors derived from U87 cells
`reconstituted with catalytically impaired PTEN (R130 M, 84 6
`15 or G129E, 69 6 16) were not significantly different (P . 0.05)
`from the parental U87 cell line (Fig. 3C). The levels of phospho-
`AKT detected within the tumor mass in vivo provide a correla-
`tion between the loss of the inositol lipid phosphatase function
`of PTEN, the phosphorylation status AKT, and the angiogenic
`phenotype within the tumor.
`
`PTEN Induction of Thrombospondin. Recent in vitro data suggest a
`link between PTEN and downstream targets including AKT,
`
`4624 u www.pnas.orgycgiydoiy10.1073ypnas.081063798
`
`Wen et al.
`
`Breckenridge Exhibit 1096
`Wen
`Page 005
`
`

`

`A
`
`B
`
`C
`
`D
`
`E
`
`PTEN suppresses angiogenesis. Immunohistochemical analysis of staining with CD31 antibody to evaluate the angiogenesis response within the parental
`Fig. 3.
`U87MG tumor (A) and PTEN reconstituted tumors (B), implanted into the s.c. tissue. In PTEN minus and tumors expressing mutants of PTEN, there are more new
`vessels formed (angiogenesis) (Upper, arrow indicated) than in WT PTEN reconstituted tumor (Lower), indicating the PTEN has direct influence on angiogenesis
`during tumor growth. (C) MVD counts were performed on tumor tissue stained with anti-CD31 antibody as described (32) to determine the effect of expression
`of PTEN and specific PTEN mutants (G129E or R130 M) on tumor-induced angiogenesis. Bars represent SD, five animals per group. Statistical analysis by Student’s
`t test demonstrateed significant difference between MVD of PTEN null and PTEN catalytic mutants as compared with WT PTEN reconstituted tumors, n 5 5,
`number of mice; P , 0.001. (D) PTEN regulates the expression of TSP-1 in U87MG cells. RNase protection assay was used to measure levels of TSP-1 mRNA in WT
`PTEN expressing U87 cells or cells transduced with a mutant catalytically dead PTEN (G129R). U87MG cells were infected with retrovirus encoding WT PTEN (WT),
`the catalytically dead, G129R mutant (GR), or empty vector retrovirus (2) and selected for 10 days in puromycin. RNA was harvested and RNase protection assays
`were carried out by using probes for TSP-1 and glyceraldehyde-3-phosphate dehydrogenase (GAPDH). A probe for GAPDH was used as a normalization control.
`(E) Thrombospondin immunoblot analysis. U87MG transduced with WT PTEN (WT) or a catalytic mutant PTEN (G129R) in an ecdysone inducible expression system
`(36) were induced (48 h) with 0.5 mM muristirone or assayed without induction to determine the effect of PTEN expression on the induction of TSP-1 by Western
`blotting. Supernates from cells were prepared and proteins were resolved on SDSyPAGE and probed with anti-TSP-1 antibody. There is clear up-regulation of
`TSP-1 in WT PTEN-transduced U87 cultures compared with U87 cells expressing the lipid phosphatase-deficient G129R mutant PTEN.
`
`MEDICALSCIENCES
`
`HIF1a, and vascular endothelial growth factor in the potential
`control of angiogenesis (34, 35). We used the RNase protection
`assay to confirm an observation we initially made during a cDNA
`microarray analysis comparing the effect of PTEN reconstitu-
`tion on U87MG TSP-1 expression. RNase protection assay was
`performed with a TSP-1 specific probe in the same U87MG cells
`constitutively expressing WT PTEN or G129R PTEN (Fig. 3D).
`The data demonstrate that WT but not mutant PTEN expression
`induces TSP-1 in U87 cells. To confirm these results, we per-
`formed Western blot analysis for TSP-1 expression with a
`retroviral-based ecdysone-inducible PTEN expression system
`(36). Inducible and dose-dependent expression of PTEN was
`confirmed in U87 cells, and we noted that the induced expression
`of WT PTEN and not G129R PTEN (not shown) resulted in
`augmented TSP-1 expression (Fig. 3E), and WT PTEN sup-
`pressed the activation of phospho-AKT without affecting total
`AKT (data not shown). The induced expression of mutant
`G129R had no effect on phospho-AKT. Therefore, our data
`demonstrate that PTEN positively modulates the expression of
`TSP-1, a negative regulator of angiogenesis (Fig. 3 D and E) (37,
`38). These data identify one potential mechanism by which
`PTEN may regulate angiogenesis through the control of TSP-1.
`
`PTEN Is a Determinant of Survival in Orthotopic Brain Tumor Model.
`Brain tumor-induced angiogenic responses occur in the context
`of brain-specific stromal and extracellular matrix interactions.
`This fact led us to determine whether the expression of the
`PTEN affects the survival of mice in an orthotopic brain tumor
`model. For these experiments, U87 cells expressing either WT or
`mutant forms of PTEN were implanted under stereotactic
`control into the right frontal lobe of nude mice (Fig. 4B, see
`
`arrow for site of implantation). The results indicate that recon-
`stitution of WT PTEN in U87 cells suppressed the malignant
`potential of these cells in an orthotopic animal model. Thus,
`there was 90% survival at 40 days in animals implanted with the
`WT PTEN-reconstituted U87 cells compared with 100% mor-
`tality of mice implanted with the parental cells at 27 days (Fig.
`4C) (n 5 15, P , 0.0001). PTEN reconstituted tumor cells grew
`more slowly when implanted in the frontal lobe (Fig. 4, compare
`A and B) and remained circumscribed to that area of brain (data
`not shown). U87 cells reconstituted with mutants of PTEN,
`either ablated in inositol lipid phosphatase activity (G129E) or
`catalytically inactive (R130 M), displayed a phenotype similar to
`the PTEN-negative, parental U87 cells (Fig. 4C). Animals with
`tumors derived from U87 cells rec

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket