throbber
on June 6, 2016
`
`http://science.sciencemag.org/
`
`Downloaded from
`
`not related to an inability of deoxycholate
`to release MTP from the microsomes, we
`sonicated the microsomes from one abeta-
`lipoproteinemic subject for 5 min after de-
`tergent treatment. Sonication releases TG
`transfer activity with an efficiency compa-
`rable to that of detergent treatment. Even
`after sonication, no TG transfer activity
`was detected.
`To demonstrate that the lack of detect-
`able TG transfer activity in the abetali-
`poproteinemic individuals was not related
`to an inability to detect activity in cells that
`contain large intracellular fat droplets, as
`occur in abetalipoproteinemia, we mea-
`sured TG transfer activity in biopsy samples
`from a subject with Anderson's disease (also
`referred to as chylomicron retention dis-
`ease) (11) and a subject with homozygous
`hypobetalipoproteinemia (12). In these two
`genetic diseases, defects occur in the assem-
`bly or secretion of chylomicrons, and affect-
`ed individuals have large fat droplets in
`their enterocytes, analogous to individuals
`with abetalipoproteinemia.
`In addition,
`TG transfer activity was measured in an
`intestinal biopsy sample taken from a nor-
`mal subject who had not fasted before the
`biopsy. In all three individuals, TG transfer
`activity comparable to that of the control
`subjects was measured (Table 1), confirm-
`
`25
`
`20
`
`15
`
`10
`
`ag 5
`
`= O
`Ch
`S%_ 25
`
`o 20
`I-
`
`.B
`
`15 I
`
`10 h
`
`5
`
`80
`
`O I F
`0°
`
`20
`
`ga-g--
`60
`
`80
`
`40
`Protein (rp)
`Fig. 1. TG transfer activity in (A) five normal and
`(B) four abetalipoproteinemic individuals. TG
`transfer activity was measured in homogenized
`intestinal biopsy samples. Results are ex-
`pressed as the percentage of the total [14C]TG
`transferred from donor to acceptor membranes
`in a 1-hour assay as a function of the amount of
`intestinal protein that was treated with deoxy-
`cholate to release TG transfer activity.
`
`9119219~
`
`ig..9999RRI
`
`Absence of Microsomal Triglyceride Transfer
`Protein in Individuals with Abetalipoproteinemia
`
`John R. Wetterau,* Lawrence P. Aggerbeck,
`Marie-Elisabeth Bouma, Claude Eisenberg, Anne Munck,
`Michel Hermier, Jacques Schmitz, Gerard Gay, Daniel J. Rader,
`Richard E. Gregg
`Abetalipoproteinemia is a human genetic disease that is characterized by a defect in the
`assembly or secretion of plasma very low density lipoproteins and chylomicrons. The
`microsomal triglyceride transfer protein (MTP), which is located in the lumen of microsomes
`isolated from the liver and intestine, has been proposed to function in lipoprotein assembly.
`MTP activity and the 88-kilodalton component of MTP were present in intestinal biopsy
`samples from eight control individuals but were absent in four abetalipoproteinemic sub-
`jects. This finding suggests that a defect in MTP is the basis for abetalipoproteinemia and
`that MTP is indeed required for lipoprotein assembly.
`
`Abetalipoproteinemia is an autosomal-re-
`cessive disease that is characterized by a
`virtual absence of plasma lipoproteins that
`contain apolipoprotein B (apoB) and by
`low plasma concentrations of triglyceride
`(TG) and cholesterol (1). These abnormal-
`ities are the result of a genetic defect in the
`assembly or secretion of very low density
`lipoproteins (VLDLs) in the liver and of
`chylomicrons in the intestine, resulting in
`retinitis pigmentosa, spinocerebellar degen-
`eration with ataxia, and a bleeding diathe-
`sis secondary to malabsorption of fat-soluble
`vitamins. The molecular basis for the pri-
`mary defect in abetalipoproteinemia has
`not been determined. TG, phospholipid,
`and cholesterol synthesis are not impaired
`(1), and linkage between the apoB gene
`and abetalipoproteinemia has been exclud-
`ed by restriction fragment length polymor-
`phism (RFLP) analysis in several families
`(2, 3).
`We investigated the possibility that a
`defect in the microsomal TG transfer pro-
`tein (MTP) may be the proximal cause of
`abetalipoproteinemia. MTP is a soluble
`protein present in the lumen of microsomes
`J. R. Wetterau and R. E. Gregg, Department of Meta-
`bolic Diseases, Bristol-Myers Squibb, Princeton, NJ
`08543-4000.
`L. P. Aggerbeck, Centre de Genetique Mol6culaire,
`Centre National de Ia Recherche Scientifique, 91198
`Gif-sur-Yvette, France.
`M.-E. Bouma and C. Eisenberg, FacultM de M6decine
`Xavier Bichat, Unite 327 Institut National de Ia Sante et
`de Ia Recherche M6dicale, 75018 Paris, France.
`A. Munck, Service Gastroent6rologie Nutrition Podia-
`triques, H6pital Robert Debr6, 75019 Paris, France.
`M. Hermier, Clinique Modicale Infantile B, Hepatogas-
`troenterologie et Nutrition Infantiles, H6pital Edouard
`Herriot, 69437 Lyon, France.
`J. Schmitz, Departement de Podiatrie, H6pital des
`Enfants Malades, 75015 Paris, France.
`G. Gay, Service de M6decine Interne, H6pital Saint
`Nicolas, 55107 Verdun, France.
`D. J. Rader, Molecular Disease Branch, National
`Heart, Lung and Blood Institute, National Institutes of
`Health, Bethesda, MD 20892.
`*To whom correspondence should be addressed.
`
`isolated from liver and intestine (4). It
`mediates the transport of TG, cholesteryl
`ester, and phosphatidylcholine (PC) be-
`tween membranes (5). The ability of MTP
`to transport TG between membranes, to-
`gether with its tissue distribution and sub-
`cellular location, has led to the suggestion
`that MTP functions in the assembly of
`plasma lipoproteins (4).
`MTP has been purified from bovine liver
`and characterized (5). It is a heterodimer of
`58- and 88-kD peptides (6). Characteriza-
`tion of the 58-kD component indicated
`that it is the previously described multi-
`disulfide
`functional
`protein,
`protein
`isomerase (PDI) (7). The role of PDI in the
`transfer protein complex is not known. At a
`minimum, PDI appears to be necessary to
`maintain the structural integrity of the
`transfer protein (8), but a larger role cannot
`be excluded. Because PDI by itself does not
`have lipid transfer activity, the 88-kD sub-
`unit is either the active component or it
`confers transfer activity to the protein com-
`plex.
`MTP activities in duodenal or duodenal-
`jejunal junction biopsy samples obtained
`from abetalipoproteinemic (9) and normal
`control subjects after an overnight fast were
`compared. Intestinal biopsy tissue was ho-
`mogenized and treated with deoxycholate
`to release TG transfer activity from the
`microsomal fraction (10). The membrane
`fractions were removed by centrifugation,
`and TG transfer activity was measured in
`the supermatants. In biopsy samples from all
`five normal control subjects tested (Fig.
`1A), TG transfer activity (10) was readily
`detectable. TG transfer activity was not
`detected ('5% of the mean of the normal
`subjects) in the biopsy tissue from any of
`the four abetalipoproteinemic subjects (Fig.
`lB and Table 1). To demonstrate that the
`lack of detectable TG transfer activity in
`individuals with abetalipoproteinemia was
`
`SCIENCE * VOL. 258
`
`*
`
`6 NOVEMBER 1992
`
`999
`
`1 of 4
`
`PENN EX. 2056
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`up
`
`:. a
`
`g
`
` on June 6, 2016
`
`http://science.sciencemag.org/
`
`Downloaded from
`
`that lipoproteins isolated from a rat liver
`Golgi fraction were consistently larger than
`those isolated from an ER fraction, which
`suggests the addition or transfer of lipid
`molecules to the nascent particles. The
`progressive addition of lipid to a developing
`lipoprotein particle was also demonstrated
`in the pulse-chase studies of Janero and
`Lane (18) and Bostrom et al. (19).
`The absence of the 88-kD component of
`MTP in individuals with abetalipoproteine-
`mia could be attributable either to its down-
`regulation to a nondetectable level or to a
`genetic defect in MTP or a factor that
`controls MTP concentration. Although
`MTP could be down-regulated in response
`to the cells not secreting lipoproteins, this
`explanation is unlikely because MTP con-
`centrations were normal in the subjects
`with Anderson's disease or homozygous hy-
`pobetalipoproteinemia, diseases in which
`enterocytes do not secrete lipoproteins.
`Given that all other known aspects of
`lipoprotein synthesis and assembly-the ex-
`pression of a normal apoB gene (2, 3, 20),
`as well as TG, phospholipid, and cholester-
`ol synthesis-are not impaired in abetali-
`poproteinemic subjects, it is likely that the
`proximal cause of abetalipoproteinemia is a
`genetic defect in the 88-kD component of
`MTP or in the regulation of its synthesis or
`degradation.
`
`REFERENCES AND NOTES
`1. J. P. Kane and R. J. Havel, in The Metabolic Basis
`of Inherited Disease, C. R. Scriver, A. L. Beaudet,
`W. S. Sly, D. Valle, Eds. (McGraw-Hill, New York,
`ed. 6, 1989), pp. 1139-1164.
`2. P. J. Talmud et al., J. Clin. Invest. 82, 1803 (1988).
`3. L. S. Huang et al., Am. J. Hum. Genet. 46, 1141
`(1 990).
`4. J. R. Wetterau and D. B. Zilversmit, Biochim.
`Biophys. Acta 875, 610 (1986).
`_ , Chem. Phys. Lipids 38, 205 (1985).
`5.
`6. J. R. Wetterau, L. P. Aggerbeck, P. M. Laplaud, L.
`R. Mclean, Biochemistry 30, 4406 (1991).
`7. J. R. Wetterau, K. A. Combs, S. N. Spinner, B. J.
`Joiner, J. Biol. Chem. 265, 9800 (1990).
`8. J. R. Wetterau, K. A. Combs, L. R. McLean, S. N.
`Spinner, L. P. Aggerbeck, Biochemistry 30, 9728
`(1991).
`9. Of the four abetalipoproteinemic subjects, two
`were female (ages 14 and 39 years) and two were
`male (ages 3 and 49 years). All had healthy,
`normolipidemic parents. The subjects had plas-
`ma cholesterol concentrations of between 24 and
`58 mg/dl and no detectable plasma apoB. Their
`current therapy includes low-fat diets and lipid-
`soluble vitamin supplements. The two adult pa-
`tients were the subject of a previously described
`study (20).
`10. TG transfer activity was measured by a modifica-
`tion of a previously described method (4). Before
`performing intestinal biopsies, we explained the
`nature of the study and its possible consequenc-
`es to the subjects or their guardians and obtained
`informed consent. In some instances biopsy sam-
`ples used in this study were part of material
`obtained for diagnostic purposes. Intestinal biop-
`sy samples were frozen and stored at -70°C until
`analyzed. Biopsy tissue was homogenized in a
`polytron homogenizer (Polytron PT3000, Brink-
`mann) at half-maximal setting. Typically, one sam-
`ple was homogenized in 0.25 ml of homogeniza-
`tion buffer [50 mM tris (pH 7.4), 50 mM KCI, 5mM
`
`ing that the presence of intracellular lipid
`droplets does not preclude the measurement
`of TG transfer activity.
`Soluble proteins obtained after deter-
`gent treatment of the intestinal biopsy tis-
`sue homogenates were analyzed by protein
`immunoblotting (13) with antibodies to the
`88-kD component of bovine MTP (14).
`The initial immunoblot analysis of two
`control subjects was performed with anti-
`bodies that had been affinity purified with
`bovine MTP. In both control subjects, a
`single band corresponding to the 88-kD
`component of bovine MTP was identified
`(Fig. 2A). To increase the probability of
`detecting small amounts of the 88-kD com-
`ponent of MTP (15), we used unfraction-
`ated antiserum in subsequent analyses, even
`though some cross-reactivity with other
`
`Table 1. TG transfer activity in intestinal biopsy
`samples.
`
`Subjects
`
`Normalized TG
`transfer activity*
`
`0.33 ± 0.16
`0.011 ± 0.004
`0.28
`0.18
`
`Normal controls (n = 5)
`Abetalipoproteinemia (n = 4)
`Anderson's disease (n = 1)
`Homozygous hypobeta-
`lipoproteinemia (n = 1)
`Nonfasted control (n = 1)
`0.36
`*The activity of a bovine MTP standard was measured
`each time an assay was performed. The TG transfer
`activity from each intestinal biopsy sample was divid-
`ed by that of the standard MTP to normalize the
`activities between experiments. For the first two groups
`of subjects, TG transfer activity is the mean ± SD.
`
`proteins was apparent. Bands comparable to
`that of the 88-kD component of bovine
`MTP were observed in all six control sub-
`jects examined (Fig. 2, B and C). In con-
`trast, no protein corresponding to the 88-
`kD component was detected in the four
`abetalipoproteinemic subjects (Fig. 2D). A
`similar analysis was performed with the
`unfractionated intestinal biopsy tissue ho-
`mogenates from two of the abetalipopro-
`teinemic subjects. Again, no band corre-
`sponding to the 88-kD component of MTP
`was apparent. Two bands with mobilities
`intermediate between the 58- and 88-kD
`components of MTP were present in all six
`control and four abetalipoproteinemic sub-
`jects examined with unfractionated antise-
`rum. Because these bands were not ob-
`served with affinity-purified antibodies (Fig.
`2A), they have been attributed to contam-
`inating antibodies that are specific for pro-
`teins other than MTP. As a control, immu-
`noblot analysis with antibodies to PDI
`demonstrated the presence of the 58-kD
`component of MTP (PDI) in the two abe-
`talipoproteinemic subjects tested (16).
`Our study suggests that MTP plays an
`obligatory role in the assembly of VLDL in
`the liver and chylomicrons in the intestine,
`probably by mediating the transport of lipid
`molecules from their site of synthesis in the
`endoplasmic reticulum (ER) membrane to
`nascent lipoprotein particles within the ER
`as they are assembled. This model for lipo-
`protein assembly is consistent with previous
`studies: Higgins and Hutson (17) showed
`
`A
`
`1
`
`2
`
`3
`
`B
`
`1
`
`2
`
`3
`
`4
`
`88-
`
`88-
`
`58-
`
`Fig. 2. Immunoblot analysis of MTP. Aliquots of
`purified bovine MTP (lane 1 of all four panels) or
`the 103,000g supernatant after treatment of
`homogenized intestinal biopsy samples with
`deoxycholate were subjected to SDS-poly-
`acrylamide gel electrophoresis and immuno-
`blotting with either antiserum or affinity-purified
`antibodies to the 88-kD subunit of MTP. (A)
`Lanes 2 and 3, soluble protein corresponding
`to 34 and 25 ±g
`of homogenate protein, re-
`spectively, from two normal subjects, probed
`with affinity-purified antibodies. (B) Lanes 2 to
`4, soluble protein corresponding to 23 >g of
`homogenate protein from three additional nor-
`mal subjects, probed with unfractionated anti-
`serum. (C) Lane 2, soluble protein correspond-
`ing to 15 pg
`of homogenate protein from a
`subject with Anderson's disease; lane 3, solu-
`ble protein corresponding to 25 ,ug of homog-
`enate protein from an individual with homozy-
`gous hypobetalipoproteinemia; lane 4, soluble
`protein corresponding to 25 pgg of homogenate
`protein from a nonfasted normal individual. Sam-
`ples were probed with unfractionated antiserum.
`(D) Lanes 2 to 5, soluble protein corresponding
`to 18 ,g (lane 2) and 23 ig (lanes 3 to 5) of
`homogenate protein from four abetalipoproteinemic subjects, probed with unfractionated antiserum.
`Lanes 6 and 7, 100 1g of unfractionated intestinal homogenate protein (from abetalipoproteinemic
`subjects corresponding to lanes 4 and 5) were subjected to electrophoresis and immunoblotting with
`unfractionated antiserum. The mobilities of the 58- and 88-kD components of bovine MTP are
`indicated. The figure represents a composite from several independent immunoblots.
`6 NOVEMBER 1992
`SCIENCE * VOL. 258
`*
`
`c
`
`1
`
`2
`
`3
`
`4
`
`D
`
`1
`
`2
`
`3
`
`4 5
`
`6 7
`
`88-
`
`58
`
`88-
`
`58-
`
`-
`
`1 000
`
`2 of 4
`
`PENN EX. 2056
`CFAD V. UPENN
`IPR2015-01836
`
`

`
` on June 6, 2016
`
`http://science.sciencemag.org/
`
`Downloaded from
`
`.,1,.,,,.,,..mp
`
`EDTA, leupeptin (5 jig/ml), and 2 mM phenyl-
`methylsulfonyl fluoride]. A portion of the homoge-
`nate was diluted to 0.6 ml and adjusted to 1.4%
`SDS before measurement of the protein concen-
`tration [0. H. Lowry, N. J. Rosebrough, A. L. Farr,
`R. J. Randall, J. Biol. Chem. 193,265(1951)]. The
`homogenate was then diluted with homogeniza-
`tion buffer to a protein concentration of -1.75
`mg/mI. One part deoxycholate solution (0.56%,
`pH 7.5) was added to 10 parts diluted homoge-
`nate while mixing. Each sample was incubated at
`4,C for 30 min and then centrifuged at 103,000g
`for 60 min. The supernatant was removed, diluted
`1:1 with 15/40 buffer [15 mm tris (pH 7.4), 40 mM
`NaCI, 1 mM EDTA, and 0.02% NaN3], and then
`dialyzed overnight against 15/40 buffer at 4CC.
`Portions of the dialyzed supematant were as-
`sayed for TG transfer activity, and immunoblot
`analysis was performed to detect the 88-kD MTP
`subunit. TG transfer activity was measured as the
`protein-stimulated rate of TG transfer from donor
`small unilamellar vesicles (SUVs) to acceptor
`SUVs. Vesicles of the desired composition were
`prepared by bath sonication in 15/40 buffer as
`described previously (5). The donor and accep-
`tor PC was labeled by adding traces of [3H]di-
`L-alpha-di-
`palmitoyl PC {phosphatidylcholine
`palmitoyl-[2-palmitoyl-9,10,3H(N)]; 33 Ci/mmol;
`DuPont Biotechnology Systems} to a specific ac-
`tivity of -100 cpm/nmol. Donor vesicles contain-
`ing 40 nmol of egg PC, 0.2 mole percent [14C]tri-
`olein {triolein [carboxyl-14C]; -110 Ci/mol; Du-
`Pont Biotechnology Systems}, and 7.3 mole per-
`cent bovine heart cardiolipin (Sigma) were mixed
`with acceptor vesicles containing 240 nmol of egg
`PC and 0.2 mol percent unlabeled TG, 5 mg of
`fatty acid-free bovine serum albumin, and a por-
`tion of the MTP samples in 0.9 ml of 15/40 buffer,
`and the mixture was incubated for 1 hour at 37°C.
`The transfer reaction was terminated by the addi-
`tion of 0.5 ml of a DEAE-cellulose suspension (5)
`and low-speed centrifugation to sediment selec-
`tively the donor vesicles containing the negatively
`charged cardiolipin. The measured amounts of
`[14C]TG (transferred from donor to acceptor
`SUVs) and [3H]PC (marker of acceptor SUV re-
`covery) were used to calculate the percentage TG
`transfer from donor to acceptor SUVs. First-order
`kinetics were used to calculate the total TG trans-
`fer (5). To calculate the protein-stimulated rate of
`TG transfer, the rate of TG transfer in the absence
`of transfer protein was subtracted from that in the
`presence of MTP. To confirm that TG hydrolysis
`was not interfering with our ability to measure lipid
`transfer, after the assay of two subjects we ex-
`tracted the acceptor vesicle lipid (which con-
`tained the transported lipid) and confirmed the
`identity of the TG by thin-layer chromatography.
`All the 14C had a mobility identical to that of
`authentic TG, confirming that intact TG was trans-
`ported in the assays. In addition, the human MTP
`was characterized for its heat stability. MTP was
`inactivated when heated to 60°C for 5 min. The
`loss of activity demonstrates that transfer activity
`attributable to an intracellular form of the cho-
`lesteryl ester transfer protein, which is stable at
`600C [J. lhm, J. L. Ellsworth, B. Chataing, J. A. K.
`Harmony, J. Biol. Chem. 257, 4818 (1982)], was
`not being measured.
`11. The clinical description and other relevant infor-
`mation for patient M.K. with Anderson's disease
`are presented elsewhere [F. Lacaille et al., Arch.
`Fr. Pediatr. 46, 491 (1989); M.-E. Bouma, I. Beu-
`cler, L. P. Aggerbeck, R. Infante, J. Schmitz, J.
`Clin. Invest. 78, 398 (1986)].
`12. The clinical description and relevant data for
`patient C.D. with homozygous hypobetalipopro-
`teinemia are presented elsewhere [G. Gay et al.,
`Rev. Med. Interne 11, 273 (1990); J.-Y. Scoazec
`et al., Gut 33, 414 (1992)].
`13. To identify the 88-kD component of MTP in tissue
`homogenates, we fractionated aliquots of proteins
`to be tested by SDS-polyacrylamide gel electro-
`phoresis and then transferred the separated pro-
`teins to nitrocellulose with a Bio-Rad Trans-blot
`cell. After incubation with a nonfat milk solution, the
`
`W-
`
`&,.9.EKWA
`
`1...
`
`-'I', 'M
`
`'10. -
`
`.... -
`
`'ll, 1,1'~
`
`W, "'
`&..
`1.
`
`1-11m
`
`nitrocellulose filter was incubated ovemight at
`room temperature with an aliquot of antiserum to
`the 88-kD protein (1:300 dilution) or affinity-purified
`antibodies (1:25 dilution). Immunoreactive pro-
`teins were visualized with horseradish peroxidase-
`coupled goat antibodies to rabbit immunoglobulin
`G (Bio-Rad) and a standard developing solution.
`14. The production and characterization of the anti-
`serum to the 88-kD protein have been previously
`described (7). The antiserum immunoprecipitates
`MTP protein and activity, but direct inhibition of
`MTP activity has not been demonstrated. Affinity-
`purified antibodies were prepared as follows:
`Purified MTP (8 to 10 mg) was coupled to 4 ml of
`Bio-Rad Affigel 15. Antibodies were partially puri-
`fied from the antiserum by (NH4)2S04 precipita-
`tion [226 mg of (NH4)2S04 per milliliter of serum].
`After centrifugation, the pellet was suspended
`and applied to the MTP-affigel. The column was
`of 10 mM tris (pH 7.5),
`washed with 100 ml
`followed by 100 ml of 10mM tris (pH 7.5) contain-
`ing 500 mM NaCI. Antibodies were eluted with 50
`ml of 100 mM glycine (pH 2.5) into 5 ml of 1 M tris
`(pH 8.0).
`15. With unfractionated antiserum, the 88-kD band of
`MTP was detectable in the soluble proteins re-
`leased from <3>g of intestinal homogenate pro-
`tein of three of four control subjects investigated.
`
`In the fourth subject, who had the lowest level of
`TG transfer activity of the controls, the 88-kD
`component of MTP was detectable in the soluble
`ig of intestinal homogenate
`proteins from 10
`protein.
`16. J. R. Wetterau et at., unpublished data.
`17. J. A. Higgins and J. L. Hutson, J. Lipid Res. 25,
`1295 (1984).
`18. D. R. Janero and M. D. Lane, J. Biol. Chem. 258,
`14496 (1983).
`19. K. Bostrom et al., ibid. 263, 4434 (1988).
`Clin. Invest. 78, 1707
`20. K. J. Lackner et al.,
`J.
`(1986).
`21. We thank F. Maurer Chagrin (H6pital Bichat, Par-
`is, France) for providing biopsy samples from
`normal subjects, N. Verthier for technical assis-
`tance, and the University of Cincinnati for the
`antiserum to the 88-kD component of MTP. Sup-
`ported by the Bristol-Myers Squibb Pharmaceuti-
`cal Research Institute, the Centre National de la
`Recherche Scientifique, the Institut National de la
`Sant6 et Recherche Medicale (grant CRE 910301
`and a France-U.S. cooperation grant), A.R.C.O.L.
`(the Committee for the Coordination of Research
`on Cholesterol in France), and NIH grant HL1 8577
`(to L.P.A.).
`
`26 May 1992; accepted 25 August 1992
`
`Tyrosine Phosphorylation of CD22
`During B Cell Activation
`Roberta J. Schulte, Mary-Ann Campbell, Wolfgang H. Fischer,
`Bartholomew M. Sefton*
`Ugation of the antigen receptor on B cells induces the rapid phosphorylation of tyrosine
`on a number of cellular proteins. A monoclonal antibody that recognized a tyrosine-
`phosphorylated cell surface protein that was present in activated B cells was generated.
`Amino acid sequence analysis showed that this 140-kilodalton protein was CD22, a B
`cell-specific cell surface glycoprotein and putative extracellular ligand of the protein ty-
`rosine phosphatase CD45. Tyrosine phosphorylation of CD22 may be important in B cell
`signal transduction, possibly through regulation of the adhesiveness of activated B cells.
`
`The B lymphocyte antigen receptor com-
`plex consists of membrane immunoglobulin
`(Ig), at least two accessory molecules (Ig-a
`and Ig-0) (1), several members of the Src
`family of protein tyrosine kinases (2, 3),
`and a 72-kD protein tyrosine kinase that
`may be encoded by the syk gene (4, 5).
`Cross-linking of surface Ig induces rapid
`increases in both tyrosine protein phos-
`phorylation (6-8) and inositol phospholip-
`id hydrolysis (9). Evidence suggests that the
`increased inositol phospholipid hydrolysis is
`induced, at least in part, by tyrosine phos-
`phorylation. (i) Phospholipase C-y, which
`is regulated by tyrosine phosphorylation in
`fibroblasts (10), is phosphorylated on tyro-
`sine during B cell activation (1 1). (ii) The
`increase in free intracellular Ca2+ that re-
`sults from inositol phospholipid hydrolysis
`
`R. J. Schulte, M.-A. Campbell, B. M. Sefton, Molecular
`Biology and Virology Laboratory, The Salk Institute,
`San Diego, CA 92186.
`W. H. Fischer, Peptide Biology Laboratory, The Salk
`Institute, San Diego, CA 92186.
`*To whom correspondence should be addressed.
`
`SCIENCE * VOL. 258
`
`*
`
`6 NOVEMBER 1992
`
`is prevented by treatment of B cells with
`herbimycin, an inhibitor of tyrosine protein
`phosphorylation (12). (iii) Expression of
`the protein tyrosine phosphatase CD45 is
`required for the stimulation of phosphati-
`dylinositol hydrolysis in a murine plasmacy-
`toma (13).
`Protein tyrosine phosphorylation may in
`fact represent the trigger or initial intracel-
`lular biochemical signaling event induced
`by the ligation of surface lg. It is not clear
`how ligation of this receptor complex in-
`duces increased substrate phosphorylation,
`but it is likely that the Src-family kinases or
`the 72-kD kinase plays a role.
`Phospholipase C-y is not the only pro-
`tein to undergo rapid tyrosine phosphoryla-
`tion after cross-linking of surface Ig with
`antibody. Approximately ten newly phos-
`phorylated proteins can be detected by im-
`munoblotting of total cell lysates with an-
`tibodies to phosphotyrosine (6-8), includ-
`ing the vav proto-oncogene product (14),
`the 72-kD cytosolic protein tyrosine kinase
`(4, 6), the 42-kD mitogen-activated (MAP)/
`1001
`
`3 of 4
`
`PENN EX. 2056
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`Absence of microsomal triglyceride transfer protein in individuals
`with abetalipoproteinemia
`Wetterau JR, LP Aggerbeck, ME Bouma, C Eisenberg, A Munck, M
`Hermier, J Schmitz, G Gay, DJ Rader and RE Gregg (November 6,
`1992)
`Science(cid:160)
`
` (5084), 999-1001. [doi: 10.1126/science.1439810]
`
`258
`
` on June 6, 2016
`
`http://science.sciencemag.org/
`
`Downloaded from
`
`(cid:160)E
`
`ditor's Summary
`
`This copy is for your personal, non-commercial use only.
`
`Article Tools
`
`Visit the online version of this article to access the personalization and article
`tools:
`http://science.sciencemag.org/content/258/5084/999
`
`Permissions
`
`Obtain information about reproducing this article:
`http://www.sciencemag.org/about/permissions.dtl
`
`(print ISSN 0036-8075; online ISSN 1095-9203) is published weekly, except the last week in
`Science
`December, by the American Association for the Advancement of Science, 1200 New York Avenue NW,
`Washington, DC 20005. Copyright 2016 by the American Association for the Advancement of Science;
` is a registered trademark of AAAS.
`all rights reserved. The title
`Science
`
`4 of 4
`
`PENN EX. 2056
`CFAD V. UPENN
`IPR2015-01836
`
`(cid:160)(cid:160)(cid:160)

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