Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Am J Physiol Gastrointest Liver Physiol 284: G853–G862, 2003.

First published January 15, 2003; 10.1152/ajpgi.00326.2002.

Novel MUC1 splice variants contribute to mucin


overexpression in CFTR-deficient mice
A. Marina Hinojosa-Kurtzberg,1 Malin E. V. Johansson,2 Cathy S. Madsen,1
Gunnar C. Hansson,2 and Sandra J. Gendler1
1
Mayo Clinic Scottsdale, Department of Biochemistry and Molecular Biology, Scottsdale, Arizona 85259;
and 2Göteborg University, Department of Medical Biochemistry, 413 90 Gothenburg, Sweden
Submitted 6 August 2002; accepted in final form 8 January 2003

Hinojosa-Kurtzberg, A. Marina, Malin E. V. Johans- forming membrane bound or secreted mucins MUC1,
son, Cathy S. Madsen, Gunnar C. Hansson, and Sandra -3, -4, -12, -13, and -17 (6, 13, 19, 39, 44, 50, 52). Some
J. Gendler. Novel MUC1 splice variants contribute to mucin biochemical and immunological analyses of mucus ob-
overexpressioninCFTR-deficientmice. AmJPhysiolGastroin- tained from CF patients suggest abnormalities in mu-

Downloaded from http://ajpgi.physiology.org/ by 10.220.33.3 on April 10, 2017


test Liver Physiol 284: G853–G862, 2003. First published
cus glycosylation, production, and secretion, but there
January 15, 2003; 10.1152/ajpgi.00326.2002.—A cystic fibro-
sis (CF) mouse expressing the human mucin MUC1 trans- is a lack of knowledge of specific alterations in mucin
gene (CFM) reverted the CF/Muc1⫺/⫺ phenotype (little mu- expression. It is unclear how the absence of a func-
cus accumulated in the intestine) to that of CF mice express- tional CFTR protein could modify the pattern of ex-
ing mouse Muc1, which exhibited increased mucus pression of some epithelial mucins. The defective ion
accumulation. Western blots and immunohistochemical anal- transport in CF may contribute to a reduction in the
ysis showed that the MUC1 protein was markedly increased volume of airway surface liquid (29) or alter the salt
in CFM mice in which it was both membrane bound and content of the airway surface liquid (43), and either of
secreted into the intestinal lumen. Studies to determine the these situations would change the milieu into which
reason for increased levels of the extracellular domain of the mucus is secreted. Other studies on CF mice have
MUC1 mucin identified mRNA and protein of two novel
suggested that the absence of CFTR in the lungs and
splice variants and the previously described secreted MUC1
lacking the cytoplasmic tail (MUC1/SEC). Novel MUC1 intestines perturbs normal differentiation of a secre-
splice variants, CT80 and CT58, were both transmembrane tory cell population (7, 12, 22). These situations would
proteins with cytoplasmic tails different from the normal potentially facilitate abnormal mucin/mucus produc-
MUC1. The MUC1-CT80 and MUC1/SEC forms are found tion.
expressed mainly in the CFM mice intestines. Thus MUC1 CFTR mutant mice (Cftrtm1UNC/Cftrtm1UNC), called
expression is increased, and it appears that alternate cyto- CF mice, exhibit intestinal goblet cell hyperplasia and
plasmic tails may change its role in signaling. MUC1 could be dilated crypts with excessive mucus accumulation and
an important contributor to the CF intestinal phenotype. obstruction mimicking human meconium ileus, but
cystic fibrosis; intestinal obstruction; small intestine; gene they do not show mucus accumulation in the lungs (45,
splicing 55). Interestingly, when CF mice lacking the mucin
Muc1 were analyzed, the intestinal mucus accumula-
tion was decreased compared with Muc1-expressing
CYSTIC FIBROSIS (CF) is caused by defects in the CF CF mice (34). The human mucin is designated as
transmembrane conductance regulator (CFTR) ion MUC1, whereas the mouse homologue is designated
channel. Some of the clinical manifestations of the Muc1. Despite the fact that Muc1 is expressed at low
disease are mucus accumulation in the respiratory and levels in the normal intestine and that it was originally
gastrointestinal tracts. Normally, a layer of mucus described as a transmembrane mucin, it appears to
covers the surface of the gastrointestinal tract, which play a role in the accumulation of intestinal mucus in
is indispensable to maintain the integrity of the intes- CF mice (34). MUC1 has a large extracellular domain
tinal mucosa. In CF, the mucus deposits (seemingly a (1,000–2,000 amino acids), a hydrophobic membrane-
mixture of several mucins) are one of the pleiotropic spanning domain, and a tyrosine-phosphorylated cyto-
manifestations of the disease. The excessive mucus plasmic domain of 72 amino acids (17, 18, 27). MUC1 is
results in severe intestinal obstruction at birth, known overexpressed in mammary tumors (18, 27), in colon
as meconium ileus. Intestinal obstruction can also oc- cancer (3, 10), and in other malignancies (56), and
cur later in life. Several mucins have been found to be soluble forms exist in tissue culture supernatants and
important in the normal or diseased intestine. These body fluids (1, 6). The MUC1 cytoplasmic tail (CT)
include the gel-forming secreted mucins MUC2, interacts with many proteins involved in signal trans-
MUC5B, and MUC5AC (4, 10, 11, 49) and the nongel- duction and cell adhesion, including ␤-catenin, p120ctr,

Address for reprint requests and other correspondence: S. J. Gen- The costs of publication of this article were defrayed in part by the
dler, Mayo Clinic Scottsdale, Dept. of Biochemistry and Molecular payment of page charges. The article must therefore be hereby
Biology, S. C. Johnson Medical Research Center, 13400 E. Shea marked ‘‘advertisement’’ in accordance with 18 U.S.C. Section 1734
Blvd., Scottsdale, AZ 85259 (E-mail: gendler.sandra@mayo.edu). solely to indicate this fact.

http://www.ajpgi.org 0193-1857/03 $5.00 Copyright © 2003 the American Physiological Society G853
G854 MUC1 SPLICE VARIANTS IN CF MUCUS

glycogen synthase kinase-3␤ (GSK-3␤), c-src, Grb2/ 2 EDTA, 2 sodium orthovanadate, and 10 sodium fluoride,
SOS, PKC␦, epidermal growth factor receptor (EGFR), with 50 ␮M ammonium molybdate and 1% Triton X-100, plus
erbB2, erbB3, and erbB4 (23–26, 33, 37, 38, 41, 53). complete inhibitor mixture (Sigma, St. Louis, MO). Protein
Our hypothesis is that Muc1 is participating in the lysates were centrifuged to separate cell debris. Protein con-
centration was determined using the bicinchoninic acid assay
intestinal mucus obstruction in the CF mice, it is
(Pierce) and samples were stored at ⫺80°C.
consequently increased in the CF intestines, and an Antibodies. We used three MAbs to recognize MUC1.
alternative shed or secreted MUC1 form is aberrantly HMFG-2 (kindly provided by Dr. Joyce Taylor-Papadimi-
produced under the altered environment generated by triou, Imperial Cancer Research Fund, London, UK) recog-
the absence of a functional CFTR. Here, we demon- nizes the epitope DTR (9) in the extracellular domain
strate the contribution of MUC1 to the CF intestinal (MUC1-EX). B27.29 (kindly provided by Biomira) recognizes
phenotype. We show 1) the differential expression of the epitope PDTRPAP (36) also in the MUC1-EX. CT2 rec-
MUC1 at the mRNA and protein level in CF MUC1 ognizes an epitope within the last 17 amino acids of the
transgenic (CFM) mice vs. the control MUC1 trans- MUC1 cytoplasmic domain (MUC1-CT) (41). A chicken poly-
genic (MUC1.Tg) mice with an intact CFTR, 2) histo- clonal antibody (PAb) directed to a synthetic peptide of a
unique (underlined) MUC1/SEC sequence (5⬘-CGGVSIGLS-
logical localization of MUC1 in the intestinal lumen of
FMPLP-3⬘) (44) and a rabbit PAb (CT80) directed to unique
CFM mice, 3) the presence of increased MUC1/SEC (underlined) COOH-terminal sequence of the splice variant
(message and protein) in the CFM mice, and 4) the

Downloaded from http://ajpgi.physiology.org/ by 10.220.33.3 on April 10, 2017


MUC1-CT80 (5⬘-CGKDSEGGTWKTQRAWKR-3⬘) were pro-
existence and characterization of a new MUC1 splice duced. The amino acids C or CGG were added to the amino
variant (MUC1-CT80) that shows elevated mRNA and terminal peptide sequences to facilitate their conjugation to
protein levels in the CFM mice. Additionally, the dif- keyhole limpet hemocyanin.
ferential expression of the MUC1 forms along the in- Western blots were performed using 25 ␮g of protein lysate
testinal tract consistently demonstrate that MUC1 is per sample detected with the antibodies HMFG-2, B27.29,
aberrantly expressed in the small intestine of CFM and MUC1/SEC, and 200 ␮g of protein lysate per sample for
mice in which expression resembles large intestine CT2 and CT80. Samples were separated by SDS-PAGE and
autochthonous MUC1 expression. These findings are transferred to polyvinylidene diflouride membrane (Immo-
bilon-P; Millipore) for Western blot analysis. Negative con-
consistent not only with a MUC1 role in the CF intes- trols included antibody-specific peptide blocking and Muc1
tinal phenotype but also suggest that CFTR has the knockout tissues. Dilutions of the antibodies were as follows:
ability to affect the nature and amount of mucus secre- CT2 (supernatant) 1:100, HMFG-2 (supernatant) 1:20,
tion of certain epithelial cells. Whether this is directly MUC1-SEC 1:1,000 (serum), and CT80 1:1,000 (serum).
related to CFTR function or indirectly due to secondary Mucin deglycosylation. Partial deglycosylation of MUC1
phenomena caused by the lack of CFTR is still to be was carried out by immersing the immobilized protein on
defined. Immobilon-P membrane in liquid trifluoromethanesulfonic
acid (TFMSA; Aldrich) for 6 h at 4°C (2). The membrane was
MATERIALS AND METHODS neutralized by washing in 50 mM Tris 䡠 HCl pH 8 for 30 s and
blocked before Western analysis as described above.
Animals. All studies were performed on inbred C57BL/6 Immunohistochemistry. Four longitudinal sections of 2 cm
mice containing either wild-type CFTR or mutant CFTR each and four transverse sections each of jejunum, ileum, and
(Cftrtm1UNC/Cftrtm1UNC) (45), the latter referring to the CF colon of CFM mice ranging in age from 2 wk to 11 mo were
mouse. Mice lacking Muc1 (46) were crossed with CF mice. analyzed. Control MUC1.Tg and CF/Muc1⫺/⫺ mice were an-
The resulting CF/Muc1⫺/⫺ mice were mated with MUC1.Tg alyzed the same way. These tissues were carefully handled to
mice (40) in a Muc1⫺/⫺ background, to produce the CFM prevent mechanical shearing of the intestinal mucus. Tissues
mouse (Cftrtm1UNC/Cftrtm1UNC, Muc1⫺/⫺, MUC1⫹). The con- were fixed in methacarn, embedded in paraffin, and 5-␮m
trol mice were MUC1.Tg in a Muc1⫺/⫺ background with a sections were cut from different areas of the small intestine.
functional CFTR (40). All mice were maintained in specific Before immunostaining with MAb CT2, tissues were treated
pathogen-free conditions in a barrier facility at Mayo Clinic with antigen retrieval reagent BD Retrievagen A, pH 6.5,
Scottsdale (Natalie Schafer Transgenic Animal Facility). according to the manufacturer’s instructions (Pharmingen,
Mice 3- to 4-wk-old were weaned onto a mouse liquid diet San Diego, CA), blocked for endogenous peroxidase activity,
(product no. F3017; BioServ, Frenchtown, NJ), and sterile blocked in normal goat serum, and incubated with primary
water was provided ad libitum. All experimental procedures antibodies overnight at 4°C. Slides were washed in enhanc-
were conducted according to Institutional Animal Care and ing wash buffer (Immunex), incubated with horseradish
Use Committee guidelines. Sixty-five animals were studied, peroxidase-conjugated secondary antibodies, washed in en-
ages 2 wk to 11 mo. CFM, MUC1.Tg, and CF/Muc1⫺/⫺ mice hancing wash buffer again, and developed with 3⬘,3⬘-diami-
were age matched within 1–2 days when younger than 8 wk, nobenzidine (Santa Cruz Biotechnology, Santa Cruz, CA) and
and older animals were matched within 1–2 wk. counterstained with Meyers hematoxylin (Sigma). Negative
Intestinal lysates. Intestinal lysates were prepared from controls included antibody-specific peptide blocking and
CFM mice (controls MUC1.Tg and CF/Muc1⫺/⫺). The small Muc1⫺/⫺ intestinal tissues. Antibodies were diluted as fol-
intestine (defined as the portion directly below the duodenum lows: CT2, 1:100 and MUC1/SEC, 1:300.
and above the cecum) was divided into two equal parts; the RNA extraction, RT-PCR, cloning, and DNA sequence
proximal part was taken as jejunum and the distal part as analysis. Total RNA was extracted from the jejunum, ileum,
the ileum. The colon was the portion between the cecum and and colon of CFM and MUC1.Tg mice, using the TRIzol
the anus. Intestinal segments were opened longitudinally reagent (Invitrogen, Carlsbad, CA). Samples of 20 ␮g of total
and cleaned with cold PBS. The lumenal mucus was scraped RNA were treated with DNase (DNA-free kit; Ambion, Aus-
away from the intestinal lumen and homogenized in Triton tin, TX) before cDNA synthesis. RT was done as previously
X-100 lysis buffer of (in mM): 20 HEPES (pH 8.0), 150 NaCl, described (34). The cDNA was aliquoted and stored at ⫺70°C.
AJP-Gastrointest Liver Physiol • VOL 284 • MAY 2003 • www.ajpgi.org
MUC1 SPLICE VARIANTS IN CF MUCUS G855

A single batch of cDNA was used for all the PCR results
shown here. Each experiment was repeated three times with
different batches of cDNA to confirm the consistency of the
results. The Expand High Fidelity PCR System (Roche Diag-
nostics, Indianapolis, IN) was used for all PCR reactions, as
recommended by the manufacturer for a 100-␮l volume. To
compare the presence or absence of transcripts compared
with a known RNA, we used a semiquantitative RT-PCR
approach featuring low-cycle number amplification (25 cy-
cles) and a mixture of actin primers/actin competimers (Am-
bion) as amplification reference. For this calibration control,
a 2:7.6 ratio of primers/competimers (0.5 ␮l of premixed actin
primers and 1.9 ␮l of actin competimers) was used for all the
RT-PCRs. Amplification conditions included an initial dena-
turing at 94°C for 5 min followed by 25 cycles of denaturing
94°C (30 s), annealing 57°C (1 min), and terminating 72°C (2
min) for MUC1, and MUC1 splice variants. For MUC1/SEC
the annealing temperature was 61°C (1 min). Routine con-

Downloaded from http://ajpgi.physiology.org/ by 10.220.33.3 on April 10, 2017


trols for all RT-PCRs included omission of the RT. Amplified
products were cloned and sequenced to confirm their identi-
ties.
The following primers were used for RT-PCR analysis: For
MUC1/SEC, primers were designed from the MUC1 (Gen-
Bank accession no. X52228) sequence: P2 (sense), 5⬘-GGTAC- Fig. 1. Replacement of mouse Muc1 with human MUC1 in cystic
CTCCTCTCACCTCCTCCAA-3⬘ (900–923 bp) (32), and fibrosis (CF) mice. Representative Alcian blue-stained intestinal
P7 (antisense), 5⬘-GGGGAAGGAAAGGCCGATACTCAC-3⬘ sections of the CF mice expressing the MUC1 gene (CFM) mice
(1089–1066 bp); a 190-bp product was amplified. For classic (Cftrtm1UNC/Cftrtm1UNC, MUC1⫹, Muc1⫺/⫺) (C and D) display the
MUC1 and MUC1 splice variants, primers were designed same intestinal phenotype of lumenal mucus accumulation as the CF
from the MUC1 genomic sequence (GenBank accession no. mouse with its native Muc1 (Cftrtm1UNC/Cftrtm1UNC) (A and B).
M61170 version gi:2055365). For classic MUC1, the primers
were Ex5 (sense), 5⬘-TGCTGGTCTGTGTTCTGGTTGC-3⬘
(4986–5007 bps) and Ex7 (antisense), 5⬘-TTGGCAGAAGTG- shown) (45, 55). Likewise, the life span, percentage of
GCTGCCACT-3⬘ (complementary to 6361–6341 bp), gener- survival, and general health of the CFM mice housed
ating a 265-bp fragment. To detect MUC1 splice variants, the under the same conditions as the CF mice have proven
primers were Ex5 (described above) and InVI (antisense), 5⬘- to be similar for both colonies over a period of 2.5 yr
CTTCTTCTTGCTTCTACCTGGG-3⬘ (5755–5734 bp). (data not shown).
RESULTS
MUC1-EX is substantially increased in the intestines
of the CFM mice. We examined the CFM mice vs.
Phenotype of the CFM mouse corresponds to the orig- MUC1.Tg mice for MUC1 protein expression along the
inal phenotype of the CF mouse. We (34) previously intestinal lumen. We performed Western blots from
described that Cftrtm1UNC/Cftrtm1UNC (CF) mice lack- intestinal mucus lysates with MUC1 MAbs directed
ing Muc1 exhibited a phenotype with decreased in- either to the MUC1-EX (B27.29 and HMFG-2) or to the
testinal mucus accumulation compared with the CF MUC1-CT (CT2). Immunoblotting with MAbs to the
mice. To further pursue the role of MUC1 in the mu- MUC1-EX revealed a noticeable increase in the levels
cus accumulation, we introduced the human MUC1 of MUC1 protein mainly in the jejunum and ileum of
gene into the Muc1⫺/⫺ mice by mating the CF/Muc1⫺/⫺ the CFM mice (Fig. 2, A and B). The detected MUC1
with the MUC1.Tg/Muc1⫺/⫺ mice to generate the protein appears heterogeneous in size, probably due to
Cftrtm1UNC/CftrtmUNC, Muc1⫺/⫺, MUC1⫹ (CFM) mice. variable O-linked glycosylation. The MAb B27.29,
This facilitated the detection of MUC1, because there which reacts with the heavily glycosylated MUC1-EX
is a lack of antibodies for the extracellular domain of (42), shows the most dramatic MUC1 increase in the
Muc1, and several well-characterized antibodies to hu- small intestine of the CFM mice (Fig. 2A). Further-
man MUC1 are available. The MUC1.Tg mouse con- more, an identical Western blot probed with MAb
tains a 10-kb piece of genomic DNA comprising the HMFG-2, which reacts with an epitope influenced by
entire MUC1 gene and regulatory sequences, and it the length of the oligosaccharide side chains (8),
expresses MUC1 at normal levels, conserving its tissue showed no reaction (data not shown). However, when a
specificity (40). Intestinal sections of CFM and the similarly prepared membrane blot was first partially
control MUC1.Tg mice were stained with Alcian blue deglycosylated with TFMSA and then immunoreacted
and compared with the sections previously obtained with MAb HMFG-2, MUC1 reactivity was found (Fig.
from CF and wild-type mice (34). Intestinal mucus 2B). This reaction with MAb HMFG-2 also showed that
accumulation was similar between CFM and CF mice MUC1 is augmented in the jejunum of the CFM mice
(Fig. 1). The CFM mouse also presents goblet cell and that the ileum and colon of both animals present
hyperplasia and dilation of the crypts filled with mucus very similar amounts of MUC1 (Fig. 2B). The immu-
as well as the physiological phenotype of early death of noblots with MAb CT2 (Fig. 2C) revealed that the
the pups as documented with the CF mouse (data not MUC1-CT is also increased in the CFM intestines. In
AJP-Gastrointest Liver Physiol • VOL 284 • MAY 2003 • www.ajpgi.org
G856 MUC1 SPLICE VARIANTS IN CF MUCUS

Fig. 2. Immunoblotting evidence of in-


creased MUC1 in the CFM intestines.
Intestinal mucosal lysates (25 ␮g) from
CFM and MUC1 transgenic (MUC1.Tg)
mice were immunoblotted with either
MUC1 extracellular domain (MUC1-
EX) MAb B27.29 (A) or (200 ␮g) with
MUC1 cytoplasmic tail (CT) MAb CT2
(C). A polyvinylidene diflouride mem-
brane containing the same lysates
shown on (A) was deglycosylated and
immunoblotted with the MUC1-EX
MAb HMFG2 (B), the binding of which
is influenced by the length of oligosac-
charide chains on MUC1.

Downloaded from http://ajpgi.physiology.org/ by 10.220.33.3 on April 10, 2017


this case, MUC1-CT was increased in jejunum, ileum, cycles) and actin for amplification reference, to search
and colon of the CFM mice. Furthermore, in CFM mice for transcripts of MUC1 alternate forms and to com-
MUC1-CT was more abundant in the colon, whereas pare their expression profiles in CFM and MUC1.Tg
MUC1-EX was more copious in the jejunum. In the mice. First, the normal transmembrane form of MUC1
jejunum of CFM mice, when MUC1-EX showed the was amplified by using primers unique to this sequence
highest accumulation (Fig. 2A), MUC1-CT was barely (Ex5-FOR, Ex7-REV). MUC1 levels appear to be in-
detectable (Fig. 2C). creased in both jejunum and ileum of the CFM mice,
MUC1-EX is localized abundantly in the crypts and but not in the colon.
in the mucus secreted into the intestinal lumen. To One of the known MUC1 splice variants is MUC1/
study the accumulation of MUC1 in the intestine, im- SEC, which lacks the cytoplasmic tail and has been
munostaining with several MUC1 MAbs was per- described in breast cancer cells (44) and in human
formed. Figure 3, A and E, shows a representative endometrial glands (1). The MUC1/SEC mucin is
Alcian blue stain of the mucus accumulated in the partly encoded by intron 2, which results in 11 unique
lumen of the small intestine in the CFM and CF/ amino acids (VSIGLSFPMLP) at its COOH terminus.
Muc1⫺/⫺ mice. The immunohistochemical analysis of We used the P2 and P7 primers to amplify MUC1/SEC,
intestinal sections of CFM mice with MAb B27.29 dis- which would not amplify the classic transmembrane
tinctly showed MUC1 protein, not only on the cell MUC1 (Fig. 5A). RT-PCR using 25 cycles revealed
membrane of the villi and crypts, but also in the se- MUC1/SEC mRNA in the jejunum, ileum, and colon of
creted mucus accumulated in the lumen of the small CFM mice, but only in the ileum and colon of the
intestine (Fig. 3B). That the reactivity was not due to MUC1.Tg mice (Fig. 4B). When the RT-PCR number of
nonspecific antibody-mucus interaction was shown by cycles was taken up to 40, the MUC1/SEC message was
the lack of staining in CF/Muc1⫺/⫺ mice containing also found in jejunum of MUC1.Tg mice (not shown).
some accumulated mucus but not MUC1 (Fig. 3F). The expression pattern of MUC1/SEC and MUC1 ap-
Immunostaining with CT2 also revealed MUC1 in the peared fairly similar along the small and large intes-
cell membrane of villi and crypt cells with very faint tines of the CFM mice (Fig. 4, A and B). This pattern
staining of the lumenal mucus (Fig. 3C). Also in this differed from the one obtained in the MUC1.Tg mice, in
case, the mucus lacking MUC1 did not react with MAb which MUC1 and MUC1/SEC messages were ex-
CT2 (Fig. 3G). These results are in agreement with the pressed in the colon, very little were expressed in the
results obtained in the Western blots of intestinal ileum, and no expression could be detected in the
mucus lysates with MAb CT2. In both, a strong reac- jejunum (Fig. 4, A and B).
tion in the small intestine with MUC1-EX was associ- Using the same RT-PCR approach, we further
ated with a weak reaction with MUC1-CT (Fig. 2, A screened the region between the MUC1 transmem-
and C). Comparable stains performed in colon sections brane domain in exon 5 and sequences in intron 6 (Fig.
of CFM and MUC1.Tg animals showed no clear differ- 5A). The classic MUC1-CT is comprised of 48 amino
ence (data not shown). acids in exon 6 and 24 amino acids in exon 7. We
Two novel MUC1 splice variants, MUC1-CT80 and detected two novel splice variants of the MUC1-CT, a
MUC1-CT58, were identified. Because the small intes- 689-bp transcript called MUC1-CT80 and a 280-bp
tine of CFM mice showed a strong reaction with transcript called MUC1-CT58 (Fig. 4C). The MUC1-
MUC1-EX accompanied with a weak reaction of its CT80 transcript was found in the jejunum, ileum, and
cytoplasmic domain (by immunoblotting and immuno- colon of the CFM mice but only in the colon of
histochemistry), we postulated the presence of a se- MUC1.Tg mice (Fig. 4C). RT-PCR reactions taken to 40
creted MUC1 form as one of the constituents of the cycles failed to produce the MUC1-CT80 transcript in
lumenal mucus. We used a semiquantitative RT-PCR small intestines of MUC1.Tg mice (data not shown).
approach, featuring low cycle number amplification (25 The shorter transcript, MUC1-CT58, was observed in
AJP-Gastrointest Liver Physiol • VOL 284 • MAY 2003 • www.ajpgi.org
MUC1 SPLICE VARIANTS IN CF MUCUS G857

Downloaded from http://ajpgi.physiology.org/ by 10.220.33.3 on April 10, 2017


Fig. 4. Semiquantitative RT-PCR of transcripts for MUC1 and 3 of
its isoforms differentially expressed in the CFM and MUC1.Tg in-
testines. Representative reactions of a semiquantitative RT-PCR
approach featuring low-cycle amplification (25 cycles) and actin as an
amplification reference are presented in A–C to show the expression
of different MUC1 transcripts in MUC1.Tg and CFM intestines. The
arrows point to the RT-PCR amplicons for 265-bp MUC1 (A), 190-bp
MUC1/SEC (B), and 689-bp MUC1-CT80 and 280-bp MUC1-CT58
(C). Increased expression of MUC1 (A) and MUC1/SEC (B) is ob-
served in the jejunum, ileum, and colon in the CFM mice, but only in
the ileum and colon of MUC1.Tg mice. MUC1-CT80 (C) is observed in
the CFM intestines, but only in the colon in the MUC1.Tg mice.
MUC1-CT58 is present along the intestines of CFM and MUC1.Tg
mice.

the jejunum, ileum, and colon of both types of mice


with higher expression in MUC1.Tg mice (Fig. 4C).
The predicted proteins for the splice variants would
have the following general characteristics (shown
in Fig. 5). MUC1-CT58 (GenBank accession no.
AF423031) and MUC1-CT80 (GenBank accession no.
AF423030) share identical coding sequences with
MUC1, from 1 to 5265 bp (GenBank accession no.
M61170.2 GI: 15321729). Thereafter, the clone MUC1-
CT58 has a new exon (exon 6b) located between MUC1
exon 6 and MUC1 exon 7 (Fig. 5A). In MUC1-CT58, the
Fig. 3. Immunohistochemical localization of MUC1-EX in the first 420 bp of MUC1 intron 6 are spliced out and the
CFM small intestine. Representative small intestine sections of new exon 6b starts at 5686 bps and continues at least
CFM (A–D), and CF/Muc1⫺/⫺(E–H) mice were stained with Alcian 60 more residues within the same MUC1 intron 6
blue (A and E) to detect the localization of lumenal mucus (L) or
were immunostained with the MUC1-EX MAb B27.29 (B and F), sequence. The clone MUC1-CT80 has a new exon in
MUC1-CT MAb CT2 (C and G), and MUC1/SEC PAb (D and H). place of MUC1 exon 6, named 6a (Fig. 5A). MUC1 exon
The CF/Muc1⫺/⫺ mice displayed no staining with either antibody 6 ends at 5265 bp but the new exon 6a continues to
(F, G, and H). Higher magnification of B shows abundant 5746 bp. The splice donor and acceptor sites conform to
MUC1-EX secreted into the intestinal lumen (red arrow) as well
as its localization on the cell membrane (blue arrow). The higher
known consensus sequences (see GenBank accession
magnification of C shows the intense staining of cell membrane- nos. AF423031 and AF423030 for specific sites). The
associated MUC1-CT (blue arrow), which appears scarce in the first 48 amino acids at the 5⬘ end of the cytoplasmic tail
lumen. Higher magnification of D indicates MUC1/SEC present in are the same in MUC1, MUC1-CT58, and MUC1-
the intestinal lumen (red arrows). CT80, because they are coded within exon 6 that is
unaltered in the splice variants (Fig. 5B). Exon 6b in
AJP-Gastrointest Liver Physiol • VOL 284 • MAY 2003 • www.ajpgi.org
G858 MUC1 SPLICE VARIANTS IN CF MUCUS

Downloaded from http://ajpgi.physiology.org/ by 10.220.33.3 on April 10, 2017


Fig. 5. Gene structure and nucleotide sequence for MUC1, MUC1/SEC, and the novel MUC1-CT splice variants
MUC1-CT58 and MUC1-CT80. A: Arabic numbers designate exons and the Roman numerals denote introns.
Primers used for RT-PCR are marked. UTR, 3⬘ untranslated region; VNTR, variable number of tandem repeats;
TM, transmembrane domain. B: comparison of MUC1-CT and the predicted sequences for the MUC1-CT splice
variants. Sites for characterized binding proteins are shown on the MUC1 sequences.

MUC1-CT58 generates a 3⬘ end of the cytoplasmic tail


with 10 unique amino acids substituting for the 3⬘ end
24 amino acids of the cytoplasmic tail (Fig. 5B). Exon
6a in MUC1-CT80 generates a cytoplasmic tail with 32
unique amino acids substituting for the 3⬘ end 24
amino acids of the cytoplasmic tail (Fig. 5B). The sites
for ␤-catenin and Grb2 interactions are therefore lack-
ing in both of the splice variants.
Sequencing the newly found transcripts revealed
several differences with the published sequence of
MUC1 intron 6 (GenBank accession no. M61170 ver-
sion GI: 2055365). We did extensive resequencing of
that region of the genomic DNA with several sense and
antisense primers and confirmed that the differences
were errors in the published intron 6 sequence. The
correct sequence has been submitted to GenBank and
the update can be viewed as accession no. M61170.2
GI: 15321729.
MUC1-CT80 is translated into a protein expressed
mainly in CFM intestines. To confirm that the MUC1-
CT80 transcript was translated into the corresponding
protein, a rabbit PAb (CT80) directed to a unique
sequence of MUC1-CT80 was used. This antiserum
was tested by immunoblotting and immunostainning
of CFM, MUC1.Tg, and CF/Muc1⫺/⫺ intestinal lysates
and fixed tissues, respectively. Intestinal lysates were
immunoblotted with the CT80 antiserum, revealing Fig. 6. Immunoblotting evidence of MUC1-CT80 and MUC1/SEC
several forms of the protein ranging between ⬃27 and differential expression in CFM and MUC1.Tg intestines. A: intesti-
45 kDa (Fig. 6A). Antiserum was proven to be specific nal lysates were immunoblotted (IB) either with polyclonal antibody
when its reactivity was partially or completely inhib- (PAb) CT80 or with nonimmune serum. Jejunum, ileum, and colon of
ited by preincubating the CT80 antiserum with the CFM mice are compared with MUC1.Tg mice. MUC1-CT80 bands
are designated with arrows and a bracket and bands that appear to
immunizing peptide and by the absence of reactivity in be nonspecific are designated NS. B: immunoblot with MUC1/SEC
CF/Muc1⫺/⫺ mice (not shown). Nonimmune rabbit se- PAb comparing CFM and MUC1.Tg intestines. The arrow indicates
rum lacked the MUC1-CT80 bands but showed nonspe- the MUC1/SEC protein.

AJP-Gastrointest Liver Physiol • VOL 284 • MAY 2003 • www.ajpgi.org


MUC1 SPLICE VARIANTS IN CF MUCUS G859

cific bands below 30 kDa (Fig. 6A). To further deter- the CFM mice. This increase was less prominent as one
mine the specificity of the CT80 antiserum, MUC1 was moved distally to the ileum and colon. Because there
immunoprecipitated with an antibody to MUC1-EX was no concordant increase in the cytoplasmic tail of
and blotted with CT80 antiserum and a similar band- MUC1, we investigated the expression of alternate
ing pattern was observed (data not shown). The differ- forms of MUC1 mRNA that could account for the ap-
ent MUC1-CT80 forms observed in the immunoblot parent inverse proportion of MUC1-EX and MUC1-CT
likely represent different levels of MUC1-CT80 glyco- in the small intestine. Both mRNAs and protein ex-
sylation, phosphorylation, or proteolytic processing. pression of novel and previously described MUC1 iso-
The phenomenon with multiple bands containing the forms matched those of increased MUC1-EX protein in
cytoplasmic tail is also observed for the normal MUC1 the small intestine of CFM mice.
by using the CT1 and CT2 antibodies (Fig. 2) (41). Alternate splicing is a common event among mucins
Multiple bands for MUC1-CT80 were expressed along and several splice variants have been described for
the whole CFM intestinal tract but only in the colon of MUC1 (5, 30, 32, 44). Therefore, one way to explain the
MUC1.Tg mice in which a single, diffuse band of ⬃30 large amount of MUC1-EX in the small intestine of the
kDa was observed (Fig. 6A). Even with increased CFM mice is by the presence of a MUC1 splice variant
amounts of protein, still no CT80 reactivity was found lacking the cytoplasmic tail, such as MUC1/SEC (44) or
in jejunum and ileum of MUC1.Tg mice. This observa- any other cytoplasmic tail variant either lacking some

Downloaded from http://ajpgi.physiology.org/ by 10.220.33.3 on April 10, 2017


tion was consistent with the pattern of MUC1-CT80 or all of the CT2 epitopes. We found roughly similar
mRNA seen in both types of mice. levels of MUC1/SEC and MUC1 mRNA and protein in
MUC1/SEC protein expression and histological lo- the CFM mice, whereas MUC1/SEC was very scarce in
calization patterns coincide with MUC1-EX. A chicken MUC1.Tg mice. Messages for the two novel splice vari-
PAb (MUC1/SEC) directed to a unique peptide se- ants (MUC1-CT58 and MUC1-CT80) lacking the CT2
quence in MUC1/SEC detected protein on immuno- epitope were found in CFM and MUC1.Tg mice; the
blots of intestinal lysates of CFM and MUC1.Tg mice message for MUC1-CT80 showed increased expression
(Fig. 6B). The amount of MUC1/SEC protein decreased in CFM small intestine vs. MUC1.Tg small intestine.
from jejunum to colon in CFM intestines. In MUC1.Tg Thus it appears that the large increase of MUC1-EX in
mice, no protein was seen in the small intestine but the small intestine of the CFM mouse may be due to
there is some reaction in the colon. Testing up to 100 increased transcription of classic MUC1 as well as to
␮g of protein did not give a positive reaction in the MUC1 isoforms expressing variant cytoplasmic tails
MUC1.Tg small intestine (not shown). CF/Muc1⫺/⫺ and at least one secreted MUC1 form lacking the cyto-
lysates tested with MUC1/SEC were always negative plasmic tail altogether. Immunoblots corroborated that
as expected (not shown). We investigated the histolog- MUC1-CT80 and MUC1/SEC proteins were expressed.
ical localization of MUC1/SEC in the CFM and CF/ The MUC1-CT80 and MUC1/SEC expression patterns
Muc1⫺/⫺ intestines. Figure 3D shows that MUC1/SEC matched that of the MUC1-EX pattern of expression in
was localized in the intestinal lumen similarly to the small intestine of CFM mice. The fact that MUC1/
MUC1-EX (Fig. 3B). The specificity of the MUC1/SEC SEC is overexpressed in CFM intestines, and it is
antibody was corroborated by the absence of staining present in the intestinal lumen, indicates that this
in the lumenal mucus present in the CF/Muc1⫺/⫺ in- secreted form is a contributor to the mucus accumu-
testines (Fig. 3H). lated in such mice. Thus it appears that the large
increase of MUC1-EX in the small intestine of the CFM
DISCUSSION mouse is mediated by increased transcription and
translation of classic transmembrane MUC1 and
In CF, the major pathological problems are the result MUC1/SEC as well as the novel cytoplasmic domain
of an accumulation of mucus within the respiratory variant MUC1-CT80.
and digestive systems. Despite progress in the identi- The functional significance of alternate splice vari-
fication and study of mucins and their obvious rele- ants in mucins has not been studied, but other trans-
vance as components of mucus, there is a lack of knowl- membrane proteins use this strategy with alternate
edge of specific alterations in mucin expression in CF. cytoplasmic domains to switch signaling pathways (14,
This is due, in part, to the fact that new mucin genes 31). MUC1-CT80 is lacking the region known to di-
are still being identified (MUC12, -13, -17) (19, 50, 51). rectly interact with ␤-catenin (53), which potentially
In addition, the rodent homologues of all the human could have a large impact on signaling events involving
mucins have not yet been obtained, which would facil- the MUC1-CT. Because MUC1-CT80 is overexpressed
itate the generation of animal models. We have used in the CFM small intestine in which the main
analyses of both RNA and protein to investigate the MUC1-EX and MUC1/SEC increase is noticed, it seems
participation of MUC1 in the intestinal mucus accu- possible that MUC1-CT80 is participating in some sig-
mulation in the CFM mouse compared with the naling leading to an atypical mucus secretion into the
MUC1.Tg mouse. Results showed that the extracellu- intestinal lumen. However, more studies are necessary
lar domain of MUC1 is an intrinsic part of the mucus to assess the function of the MUC1-CT splice variants.
accumulation observed in the intestines of the CFM It is known that the MUC1-CT can participate in a
mice. Western blots revealed an increase in the levels wide variety of signaling events, but until now, these
of the MUC1-EX, particularly evident in the jejunum of studies have mainly dealt with adhesion and transfor-
AJP-Gastrointest Liver Physiol • VOL 284 • MAY 2003 • www.ajpgi.org
G860 MUC1 SPLICE VARIANTS IN CF MUCUS

mation of cancer cells. One can suggest that the altered using quantitative immunoassays and immunohisto-
CFM intestine epithelium may trigger distinct signal- chemistry with a variety of anti-Muc3 antibodies, re-
ing events via alternative splicing of the MUC1 gene. vealed that the ectodomain, but not the cytoplasmic
The discordant MUC1-EX and MUC1-CT in the domain, of Muc3 is increased largely in the small
small intestine of CFM mice could also be due to intestine of CF mice. This pattern of expression for
shedding or proteolytic processing of the classic MUC1. Muc3 is similar to what we observed for MUC1. Given
Mature MUC1 results from the posttranscriptional the identification of message and protein for Muc4, -12,
processing of a large precursor that is cleaved in the and -13 in the intestines of control mice (39, 50, 52), it
endoplasmic reticulum, producing a noncovalently is likely that these mucins also participate in the mu-
bound heterodimer (28, 35). Although this cleavage cus accumulation in CF intestines.
does not directly lead to the release of the MUC1-EX, it The exact composition of the intestinal mucus accu-
may provide a mechanism for MUC1 shedding and a mulated as a result of a nonfunctional CFTR is cur-
concomitant degradation of the cytoplasmic tail. A sec- rently not known, but it is clear that such mucus
ond proteolytic cleavage could also liberate MUC1-EX contains a mixture of mucins, some of them tradition-
from the cell membrane. The feature of a proteolytic ally considered not important intestinal mucins, such
cleavage associated with posttranslational processing as MUC1. Considering our present data of increased
of membrane-associated mucins is shared by at least MUC1 mRNA and protein and that MUC1 expression

Downloaded from http://ajpgi.physiology.org/ by 10.220.33.3 on April 10, 2017


two other mucins of the same class, MUC4 and epigly- is regulated at the level of transcription (16, 21, 54), we
canin (47). Accordingly, the heterodimeric presentation establish that MUC1 is not only accumulated in the
of MUC1 and the other transmembrane mucins may CFM intestinal lumen due to an impaired mucus clear-
provide a mechanism for rapid shedding of the mucin ance but to its increased expression as well. We hy-
domain into the lumen where it appears to participate pothesize that MUC1 in the CF intestines is not only
in the accumulation of mucus detected in CF. contributing to the mucus accumulation through the
The major phenotype in the CF mouse occurs in the shedding and/or secretion of its extracellular domain
small intestine, an organ also affected in human CF but also via its proven role in signal transduction
patients (15). How the lack of a functional CFTR favors (23–26, 33, 37, 41, 53). MUC1 may signal to the cell
the increase of MUC1, in particular, and the increase interior an altered extracellular milieu, which would
of mucus secretion in general, remains unresolved. provoke a response intended to maintain the integrity
Considering our results of MUC1 message and protein of the intestinal mucosa under the stressful environ-
expression in the CF intestines, it appears that the ment generated by the absence of CFTR.
CFM small intestine takes on a resemblance to the
We thank Marvin Ruona for help with computer graphics and
normal colon, but it does not present histological signs Carol Williams for administrative assistance and manuscript sub-
of colonic differentiation. We have found that mucin mission.
fucosylation was also altered in the CF mouse small This work was supported by the Cystic Fibrosis Foundation, the
intestine and resembled fucosylation in the normal Swedish Foundation for International Cooperation in Research and
colon (48). This resemblance of the CFM small intes- Higher Education, The Swedish Research Council, and National
Institutes of Health Grant T32-H107897.
tine to the colon could be reflecting a subtle alteration
in the differentiation of some epithelial cells, leading to REFERENCES
atypical mucus secretion. Crypt enterocytes of the
small intestine are the cells most abundantly express- 1. Aplin JD, Hey NA, and Li TC. MUC1 as a cell surface and
secretory component of endometrial epithelium: reduced levels
ing CFTR, although it is expressed in the villus cells as in recurrent miscarriage. Am J Reprod Immunol 35: 261–266,
well. One would expect that the lack of a functional 1996.
CFTR could affect water secretion. It has been sug- 2. Axelsson MAB, Hansson EM, Sikut R, and Hansson GC.
gested that reduced airway-surface liquid in CF pa- Deglycosylation by gaseous hydrogen fluoride of mucus glyco-
proteins immobilized on nylon membranes and in microtiter
tients may create underhydrated mucus and impaired wells. Glycoconj J 15: 749–755, 1998.
mucociliary clearance. By inference, any reduced secre- 3. Baeckstrom D, Nilsson O, Price MR, Lindholm L, and
tion of water into the intestinal lumen in the CFM Hansson GC. Discrimination of MUC1 mucins from other sia-
mouse could result in impaired mucus clearance. How- lyl-Le(a)-carrying glycoproteins produced by colon carcinoma
ever, a dehydrated environment could, in principle, cells using a novel monoclonal antibody. Cancer Res 53: 755–761,
1993.
also generate a stress signal with the potential power 4. Bartman AE, Sanderson SJ, Ewing SL, Niehans GA, Wiehr
to activate a cascade of protective events to maintain CL, Evans MK, and Ho SB. Aberrant expression of MUC5AC
the integrity of the lumenal epithelium. Mucus produc- and MUC6 gastric mucin genes in colorectal polyps. Int J Cancer
tion may be increased as a primary line of defense 80: 210–218, 1999.
5. Baruch A, Hartmann M, Yoeli M, Adereth Y, Greenstein S,
within such a protective response. Our observations Stadler Y, Skornik Y, Zaretsky J, Smorodinsky NI, Keydar
and those of other researchers favor the idea that there I, and Wreschner DH. The breast cancer-associated MUC1
is increased expression of mucins and increased secre- gene generates both a receptor and its cognate binding protein.
tion of mucus contributing to the intestinal mucus Cancer Res 59: 1552–1561, 1999.
accumulation as well as an altered environment in 6. Boshell M, Lalani EN, Pemberton L, Burchell J, Gendler
SJ, and Taylor-Papadimitriou J. The product of the human
which such mucus is less readily cleared. Our results MUC1 gene when secreted by mouse cells transfected with the
clearly point out that MUC1 is part of the mucus full-length cDNA lacks the cytoplasmic tail. Biochem Biophys
accumulated. Recent findings of Khatri et al. (20), Res Commun 185: 1–8, 1992.

AJP-Gastrointest Liver Physiol • VOL 284 • MAY 2003 • www.ajpgi.org


MUC1 SPLICE VARIANTS IN CF MUCUS G861

7. Broackes-Carter FC, Mouchel N, Gill D, Hyde S, Bassett J, associated antigen and ␤–catenin. Mol Cell Biol 18: 7216–7224,
and Harris A. Temporal regulation of CFTR expression during 1998.
ovine lung development: implications for CF gene therapy. Hum 27. Ligtenberg MJ, Vos HL, Gennissen AM, and Hilkens J.
Mol Genet 11: 125–131, 2002. Episialin, a carcinoma-associated mucin, is generated by a poly-
8. Burchell J and Taylor-Papadimitriou J. Effect of modifica- morphic gene encoding splice variants with alternative amino
tion of carbohydrate side chains on the reactivity of antibodies termini. J Biol Chem 265: 5573–5578, 1990.
with core-protein epitopes of the MUC1 gene product. Epithelial 28. Ligtenberg MJL, Kruijshaar L, Buijs F, van Meijer M,
Cell Biol 2: 155–162, 1993. Litvinov SV, and Hilkens J. Cell-associated episialin is a
9. Burchell J, Taylor-Papadimitriou J, Boshell M, Gendler complex containing two proteins derived from a common precur-
SJ, and Duhig T. A short sequence within the amino acid sor. J Biol Chem 267: 6171–6177, 1992.
tandem repeat of a cancer-associated mucin contains immuno- 29. Matsui H, Grubb BR, Tarran R, Randell SH, Gatzy JT,
dominant epitopes. Int J Cancer 44: 691–696, 1989. Davis CW, and Boucher RC. Evidence for periciliary liquid
10. Carrato C, Balague C, de Bolos C, Gonzalez E, Gambus G, layer depletion, not abnormal ion composition, in the pathogen-
Planas J, Perini JM, Andreu D, and Real FX. Differential esis of cystic fibrosis airways disease. Cell 95: 1005–1015, 1998.
apomucin expression in normal and neoplastic human gastroin- 30. Obermair A, Schmid BC, Stimpfl M, Fasching B, Preyer O,
testinal tissues. Gastroenterology 107: 160–172, 1994. Leodolter S, Crandon AJ, and Zeillinger R. Novel muc1
11. Chang SK, Dohrman AF, Basbaum CB, Ho SB, Tsuda T, splice variants are expressed in cervical carcinoma. Gynecol
Toribara NW, Gum JR, and Kim YS. Localization of mucin
Oncol 83: 343–347, 2001.
(MUC2 and MUC3) messenger RNA and peptide expression in
31. Okabe S, Miwa A, and Okado H. Alternative splicing of the
human normal intestine and colon cancer. Gastroenterology 107:
C-terminal domain regulates cell surface expression of the

Downloaded from http://ajpgi.physiology.org/ by 10.220.33.3 on April 10, 2017


28–36, 1994.
NMDA receptor NR1 subunit. J Neurosci 19: 7781–7792, 1999.
12. Cohen JC, Morrow SL, Cork RJ, Delcarpio JB, and Larson
JE. Molecular pathophysiology of cystic fibrosis based on the 32. Oosterkamp HM, Scheiner L, Stefanova MC, Lloyd KO,
rescued knockout mouse model. Mol Genet Metab 64: 108–118, and Finstad CL. Comparison of MUC-1 mucin expression in
1998. epithelial and non-epithelial cancer cell lines and demonstration
13. Crawley SC, Gum JR Jr, Hicks JW, Pratt WS, Aubert JP, of a new short variant form (MUC-1/Z). Int J Cancer 72: 87–94,
Swallow DM and Kim YS. Genomic organization and structure 1997.
of the 3⬘ region of human MUC3: alternative splicing predicts 33. Pandey P, Kharbanda S, and Kufe D. Association of the
membrane-bound and soluble forms of the mucin. Biochem Bio- DF3/MUC1 breast cancer antigen with Grb2 and the Sos/Ras
phys Res Commun 263: 728–736, 1999. exchange protein. Cancer Res 55: 4000–4003, 1995.
14. De Melker AA and Sonnenberg A. Integrins: alternative 34. Parmley RR and Gendler SJ. Cystic fibrosis mice lacking
splicing as a mechanism to regulate ligand binding and integrin Muc1 have reduced amounts of intestinal mucus. J Clin Invest
signaling events. Bioessays 21: 499–509, 1999. 102: 1798–1806, 1998.
15. Eggermont E and De Boeck K. Small-intestinal abnormalities 35. Parry S, Silverman HS, McDermott K, Willis A, Holling-
in cystic fibrosis patients. Eur J Pediatr 150: 824–828, 1991. sworth MA, and Harris A. Identification of MUC1 proteolytic
16. Gaemers IC, Vos HL, Volders HH, van der Valk SW, and cleavage sites in vivo. Biochem Biophys Res Commun 283: 715–
Hilkens J. A STAT-responsive element in the promoter of the 720, 2001.
episialin/MUC1 gene is involved in its overexpression in carci- 36. Petrakou E, Murray A, and Price MR. Epitope mapping of
noma cells. J Biol Chem 276: 6191–6199, 2001. anti-MUC1 mucin protein core monoclonal antibodies. Tumour
17. Gendler SJ. MUC1, the renaissance molecule. J Mammary Biol 19, Suppl 1: 21–29, 1998.
Gland Biol Neoplasia 6: 339–353, 2001. 37. Quin RJ and McGuckin MA. Phosphorylation of the cytoplas-
18. Gendler SJ, Lancaster CA, Taylor-Papadimitriou J, Du- mic domain of the MUC1 mucin correlates with changes in
hig T, Peat N, Burchell J, Pemberton L, Lalani EN, and cell-cell adhesion. Int J Cancer 87: 499–506, 2000.
Wilson D. Molecular cloning and expression of human tumor- 38. Ren J, Li Y, and Kufe D. Protein kinase C delta regulates
associated polymorphic epithelial mucin. J Biol Chem 265: function of the DF3/MUC1 carcinoma antigen in ␤-catenin sig-
15286–15293, 1990. naling. J Biol Chem 277: 17616–17622, 2002.
19. Gum JR Jr, Crawley SC, Hicks JW, Szymkowski DE, and 39. Rossi EA, McNeer RR, Price-Schiavi SA, Van den Brande
Kim YS. MUC17, a novel membrane-tethered mucin. Biochem JM, Komatsu M, Thompson JF, Carraway CA, Fregien NL,
Biophys Res Commun 291: 466–475, 2002. and Carraway KL. Sialomucin complex, a heterodimeric gly-
20. Khatri IA, Ho C, Specian RD, and Forstner JF. Character- coprotein complex. Expression as a soluble, secretable form in
istics of rodent intestinal mucin Muc3 and alterations in a mouse lactating mammary gland and colon. J Biol Chem 271: 33476–
model of human cystic fibrosis. Am J Physiol Gastrointest Liver 33485, 1996.
Physiol 280: G1321–G1330, 2001. 40. Rowse GJ, Tempero RM, VanLith ML, Hollingsworth MA,
21. Kovarik A, Peat N, Wilson D, Gendler SJ, and Taylor-
and Gendler SJ. Tolerance and immunity to MUC1 in a human
Papadimitriou J. Analysis of the tissue specific promoter of the
MUC1 transgenic murine model. Cancer Res 58: 315–321, 1998.
MUC1 gene. J Biol Chem 268: 9917–9926, 1993.
41. Schroeder JA, Thompson MC, Gardner MM, and Gendler
22. Larson JE, Delcarpio JB, Farberman MM, Morrow SL,
SJ. Transgenic MUC1 interacts with EGFR and correlates with
and Cohen JC. CFTR modulates lung secretory cell prolifera-
tion and differentiation. Am J Physiol Lung Cell Mol Physiol MAP kinase activation in the mouse mammary gland. J Biol
279: L333–L341, 2000. Chem 276: 13057–13064, 2001.
23. Li Y and Kufe D. The human DF3/MUC1 carcinoma-associated 42. Sikut R, Sikut A, Zhang K, Baeckstrom D, and Hansson
antigen signals nuclear localization of the catenin p120(ctn). GC. Reactivity of antibodies with highly glycosylated muc1 mu-
Biochem Biophys Res Commun 281: 440–443, 2001. cins from colon carcinoma cells and bile. Tumor Biol 19, Suppl 1:
24. Li Y, Kuwahara H, Ren J, Wen G, and Kufe D. The c-Src 122–126, 1998.
tyrosine kinase regulates signaling of the human DF3/MUC1 43. Smith JJ, Travis SM, Greenberg EP, and Welsh MJ. Cystic
carcinoma-associated antigen with GSK3␤ and ␤-catenin. J Biol fibrosis airway epithelia fail to kill bacteria because of abnormal
Chem 276: 6061–6064, 2001. airway surface fluid. Cell 85: 229–236, 1996.
25. Li Y, Ren J, Yu Wh W, Li Q, Kuwahara H, Yin L, Carraway 44. Smorodinsky N, Weiss M, Hartmann ML, Baruch A, Har-
KL, 3rd, and Kufe D. The epidermal growth factor receptor ness E, Yaakobovitz M, Keydar I, and Wreschner DH.
regulates interaction of the human DF3/MUC1 carcinoma anti- Detection of a secreted MUC1/SEC protein by MUC1 isoform
gen with c-Src and ␤-catenin. J Biol Chem 276: 35239–35242, specific monoclonal antibodies. Biochem Biophys Res Commun
2001. 228: 115–121, 1996.
26. Li YQ, Bharti A, Chen DS, Gong JL, and Kufe D. Interaction 45. Snouwaert JN, Brigman KK, Latour AM, Malouf NN,
of glycogen synthase kinase-3␤ with the DF3/MUC1 carcinoma- Boucher RC, Smithies O, and Koller BH. An animal model

AJP-Gastrointest Liver Physiol • VOL 284 • MAY 2003 • www.ajpgi.org


G862 MUC1 SPLICE VARIANTS IN CF MUCUS

for cystic fibrosis made by gene targeting. Science 257: 1083– 51. Williams SJ, Munster DJ, Quin RJ, Gotley DC, and
1088, 1992. McGuckin MA. The MUC3 gene encodes a transmembrane
46. Spicer AP, Rowse GJ, Lidner TK, and Gendler SJ. Delayed mucin and is alternatively spliced. Biochem Biophys Res Com-
mammary tumor progression in Muc-1 null mice. J Biol Chem mun 261: 83–89, 1999.
270: 30093–30101, 1995. 52. Williams SJ, Wreschner DH, Tran M, Eyre HJ, Sutherland
47. Thingstad T, Vos HL, and Hilkens J. Biosynthesis and shed- GR, and McGuckin MA. Muc13, a novel human cell surface
ding of epiglycanin: a mucin-type glycoprotein of the mouse mucin expressed by epithelial and hemopoietic cells. J Biol
TA3Ha mammary carcinoma cell. Biochem J 353: 33–40, 2001. Chem 276: 18327–18336, 2001.
48. Thomsson KA, Hinojosa-Kurtzberg M, Axelsson KA, Dom- 53. Yamamoto M, Bharti A, Li Y, and Kufe D. Interaction of the
ino SE, Lowe JB, Gendler SJ, and Hansson GC. Intestinal DF3/MUC1 breast carcinoma-associated antigen and ␤-catenin
mucins from cystic fibrosis mice show increased fucosylation due in cell adhesion. J Biol Chem 272: 12492–12494, 1997.
to an induced Fuc␣1–2 glycosyltransferase. Biochem J 367: 54. Zaretsky JZ, Sarid R, Aylon Y, Mittelman LA, Wreschner
609–616, 2002. DH, and Keydar I. Analysis of the promoter of the MUC1 gene
49. Velcich A, Yang W, Heyer J, Fragale A, Nicholas C, Viani overexpressed in breast cancer. FEBS Lett 461: 189–195, 1999.
S, Kucherlapati R, Lipkin M, Yang K, and Augenlicht L. 55. Zhou L, Dey CR, Wert SE, DuVall MD, Frizzell RA, and
Colorectal cancer in mice genetically deficient in the mucin Whitsett JA. Correction of lethal intestinal defect in a mouse
Muc2. Science 295: 1726–1729, 2002. model of cystic fibrosis by human CFTR. Science 266: 1705–
50. Williams SJ, McGuckin MA, Gotley DC, Eyre HJ, Suther- 1708, 1994.
land GR, and Antalis TM. Two novel mucin genes down- 56. Zotter S, Hageman PC, Lossnitzer A, Mooi WJ, and Hilgers
regulated in colorectal cancer identified by differential display. J. Tissue and tumor distribution of human polymorphic epithe-

Downloaded from http://ajpgi.physiology.org/ by 10.220.33.3 on April 10, 2017


Cancer Res 59: 4083–4089, 1999. lial mucin. Cancer Reviews 11–12: 55–101, 1988.

AJP-Gastrointest Liver Physiol • VOL 284 • MAY 2003 • www.ajpgi.org

You might also like