ArticlePDF Available

CD44 as a stem cell marker in head and neck squamous cell carcinoma

Authors:

Abstract and Figures

In the recent past, evidence is increasing indicating the existence of a subpopulation of resistant tumor cells in head and neck squamous cell carcinoma (HNSCC) that cannot be eradicated by established antineoplastic treatments. These cancer stem cells (CSCs) have features of somatic stem cells such as selfrenewal, proliferation and differentiation. CD44+ cells in tumors of the head and neck are referred to as CSCs of HNSCC. Expression profiling of CD44 in 29 HNSCC tumors was performed by fluorescence microscopy. ELISA analysis was performed to detect concentration of soluble CD44 in the peripheral blood of 29 HNSCC patients and 11 healthy controls. Expression of CD44 was determined in all HNSCC tissue samples (n=29). In all samples a surface staining pattern was found. The concentration of CD44 in the peripheral blood of HNSCC patients was significantly higher compared to a healthy control group (mHNSCC =13.5 ± 0.5 ng/ ml; mCont = 9.3 ± 0.6 ng/ml; P=0.6 x 10(-12)). The role of CD44 as a marker for CSCs in HNSCC remains to be ascertained. Further experiments might reveal its role as a diagnostic and prognostic factor, and possibly as a therapeutic target.
Content may be subject to copyright.
ONCOLOGY REPORTS 26: 321-326, 2011
Abstract. In the recent past, evidence is increasing indicating
the existence of a subpopulation of resistant tumor cells
in head and neck squamous cell carcinoma (HNSCC) that
cannot be eradicated by established antineoplastic treatments.
These cancer stem cells (CSCs) have features of somatic stem
cells such as selfrenewal, proliferation and differentiation.
CD44+ cells in tumors of the head and neck are referred to
as CSCs of HNSCC. Expression profiling of CD44 in 29
HNSCC tumors was performed by uorescence microscopy.
ELISA analysis was performed to detect concentration of
soluble CD44 in the peripheral blood of 29 HNSCC patients
and 11 healthy controls. Expression of CD44 was determined
in all HNSCC tissue samples (n=29). In all samples a surface
staining pattern was found. The concentration of CD44 in the
peripheral blood of HNSCC patients was signicantly higher
compared to a healthy control group (mHNSCC=13.5±0.5 ng/ ml;
mCont=9.3±0.6 ng/ml; P= 0.6x10-12). The role of CD44 as
a marker for CSCs in HNSCC remains to be ascertained.
Further experiments might reveal its role as a diagnostic and
prognostic factor, and possibly as a therapeutic target.
Introduction
Head and neck squamous cell carcinoma (HNSCC) is an
aggressive and recurrent malignancy, largely because it is
usually diagnosed at a late stage. Tobacco and alcohol exposure
are the main risk factors and account for ~85% of HNSCC (1).
De spit e adva nces in surgical a nd non -su rgica l th erapy mor tality
from this disease remains high because of the development of
distant metastases and the emergence of therapy-resistant local
and regional recurrences. Antineoplastic treatments such as
chemotherapy or radiation can efciently eradicate a majority
of proliferating malignant cells within malignant tumors.
However, there is increasing evidence that there is a subpopu-
lation of resistant tumor cells that can not be reached by
these regimens. These cancer stem cells (CSCs) have distinct
features of somatic stem cells such as selfrenewal, extensive
proliferation and differentiation. Therefore, these cells are
required and responsible for initiation, but also maintenance
and recurrence of disease. In recent years, the CSC hypothesis
has been coined for HNSCC as well (2,3). Prince at al showed
that CD44+ cancer cells, which typically comprise <10% of
the cells in an HNSCC tumor, but not the CD44- cancer cells,
gave rise to new tumors in vivo (2). Since then, CD44+ cells
in tumors of the head and neck are referred to as CSCs of
HNSCC.
CD44 is an integral cell membrane glycoprotein and it
comprises different isoforms that arise from alternative splicing
of a region of variable exons. They differ in primary amino
acid sequence as well as amount of N- and O-glycosylation
(4), thereby its apparent molecular mass ranges from 85 to
250 kD (5). At least 20 variants of CD44 have been reported
due to the alternative splicing of 10 exons that encode the
membrane's proximal portion of the extracellular domain
(6-8). Originally, it was described as a receptor on circulating
lymphocytes involved in homing, cell adhesion and migration
(9,10).
In 1991 Günthert et al showed that the expression of CD44
gave metastatic potential to a non-metastatic line of cells in a
rat carcinoma model (11,12). Since then, several analysis have
indicated that there is a correlation between the expression
of CD44 variants and progression, metastasis and prognosis
of malignant disease. This has also been shown in different
types of epithelial carcinoma in addition to HNSCC such
as colorectal carcinoma (13,14), breast carcinoma (15) and
certain types of gastric carcinoma (5,16).
The in-depth analysis of expression markers such as CD44
in tissue samples of HNSCC patients may reveal their role
as potential prognostic biomarkers or therapeutic targets,
e.g. for antigen directed immunotherapy. The analysis of
soluble CD44 in peripheral blood of HNSCC patients may
reveal important ndings concerning diagnosis and possible
pathways of metastasis in head and neck cancer.
CD44 as a stem cell marker in head and
neck squamous cell carcinoma
ANNE FABER, CHRISTINE BARTH, KARL HÖRMANN, STEFAN KASSNER, JOHANNES DAVID SCHULTZ,
ULRICH SOMMER, JENS STERN-STRAETER, CARSTEN THORN and ULRICH REINHART GOESSLER
Department of Otorhinolaryngology Head and Neck Surgery, University Medical Centre
Mannheim, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
Received February 14, 2011; Accepted March 30, 2011
DOI: 10.3892/or.2011.1322
Correspondence to: Dr Anne Faber, Department of Otorhino-
laryngology Head and Neck Surgery, University Medical Centre
Mannheim, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
E-mail: faberanne@gmx.de, anne.faber@umm.de
Key words: CD44, cancer stem cells, head and neck squamous cell
carcinoma
FABER et al: CD44 AS A STEM CELL MARKER IN HEAD AND NECK SCC
322
Materials and methods
Tissue and peripheral blood sample collection. A total of 29
HNSCC tissue samples from tumor patients were selected
out of a tissue data base collected from 1997 to 2010 at the
Department of Otorhinolaryngology, Head and Neck Surgery at
the University of Mannheim. Samples were xated immediately
after excision by freezing in liquid nitrogen. All samples were
conrmed by pathology after H&E staining. In addition to the
tissue collection, peripheral blood samples were taken from
HNSCC patients and healthy donors. Peripheral blood was
collected before, during, but never after tumor surgery. A group
of 11 healty blood donors served as a control. Blood samples
were centrifuged at 2500 rpm for 10 min. Afterwards serum
samples were harvested and xated by freezing at -80˚C. The
histological and clinical characteristics of all tumor samples are
summarized in Table I.
Immunof luorescence labeling. To detect the expression
of CD44 in HNSCC tissue samples, tumors underwent
xation by freezing in liquid nitrogen as mentioned above.
Specimens were prepared in slices of 5-8 µm, air-dried
and xed in acetone for 10 min. Afterwards, slices were
treated with 4% paraformaldehyde (PFA) for 10 min at room
temperature. After 3 washing steps with PBS tumor samples
were treated with 1% ser um (goat) for another 10 min.
Then slices were incubated with CD44 antibody (mouse
monoclonal, 1:100, Abcam, Cambridge, UK) for 1 h at 37˚C
followed by incubation with a second biotinylated antibody
(anti-mouse, 1:100) for 30 min. After further washing steps
with PBS, slices were treated with Streptavidin-Cy3 (1:1000)
for 30 min at room temperature. Subsequently, slices were
stained with DAPI after washing with PBS. Finally, slices
were covered in f luorsave and dried to be evaluated by
uorescence microscopy.
Enzyme-linked immunosorbent assay. Serum levels of CD44
were measured with a human CD44 ELISA Kit (Abcam). A
monoclonal antibody against soluble CD44 was adsorbed to
microwells in 96-well microtiter plates. Samples, including
standards of known CD44 concentrations and samples were
pipetted into these wells. During the rst incubation, the CD44
antigen was added to wells. After washing, a biotinylated
monoclonal antibody specic for CD44 was incubated and the
enzyme (streptavidin-peroxidase) was added. After incubation
and washing to remove a ll unbou nd enzyme, a subst rate solution
was added, which catalyzed a reaction on the bound enzyme
and so induced a coloured reaction product. The intensity of
this product is directly proportional to the concentration of
CD44 present in the samples.
Statistical analysis. All results were plotted as the mean ±
standard deviation. To estimate the probability of differences,
we employed the Student's t-test. P<0.05 denoted statistical
signica nce.
Results
Tissue and peripheral blood sample collection. A total of
29 HNSCC tissue samples and the according blood serum
samples were selected out of a data base collected from 1997
to 2010 at the Department of Otorhinolaryngology Head
and Neck Surgery at the Faculty of Medicine Mannheim,
University of Heidelberg, Germany. Samples entering the
study were derived from 5 female and 24 male patients aged
48-76 (mean age: 60) years. Locations of the primary tumor
were differentiated as larynx, oropharynx, hypopharynx and
oral cavity (10 larynx, 5 oropharynx, 7 hypopharynx, 7 oral
cavity). Patient characteristics are summarized in Table I.
A group of 11 healthy blood donors served as a control for
experiments concerning CD44 concentration in peripheral
blood of HNSCC patients. All studies were approved by the
Ethics Committee of the Faculty of Medicine Mannheim,
University of Heidelberg.
Expression of CD44 in HNSCC tissue samples. Immuno-
uorescence labeling of 29 tissue samples was performed to
detect the expression of CD44 in HNSCC. In all samples,
Table I. Tumor tissue and blood sample collection.
Patient Gender Age Tumor TNM Smoking/
(yrs.) location ETOH
1 M 54 Larynx T4N2xMx +/+
2 F 55 Larynx T1N0 +/-
3 M 62 Larynx T4N0 +/-
4 M 70 Larynx T4N0 1
5 M 68 Larynx T3N2b 1
6 M 68 Larynx 1 1
7 M 55 Larynx T4N2b 1
8 M 74 Larynx T4N0 1
9 M 59 Larynx 1 1
10 M 57 Larynx T4N1 +/+
11 M 55 Oral Cavity T4N1 +/+
12 M 69 Oral Cavity T1N2b 1
13 F 54 Oral Cavity T1N2b 1
14 M 48 Oral Cavity 1 1
15 M 49 Oral Cavity T3N1 1
16 M 51 Oral Cavity 1 1
17 M 66 Oral Cavity T2N0 +/+
18 F 50 Oropharynx T3N2c 1
19 M 64 Oropharynx 1 1
20 M 60 Oropharynx T4N2c 1
21 M 64 Oropharynx T3N2 1
22 F 55 Oropharynx T4N3 1
23 M 61 Hypophyarnx T4N2b +/+
24 M 66 Hypopharynx T2N0 1
25 M 76 Hypopharynx 1 1
26 M 59 Hypopharynx T4N2b 1
27 F 62 Hypopharynx T2N2b 1
28 M 51 Hypopharynx 1 1
29 M 57 Hypopharynx T3N2c +/-
1, information not available.
ONCOLOGY REPORTS 26: 321-326, 2011 323
pathology of HNSCC was conrmed via H&E staining by
pathology during routine diagnostics (Fig. 1A). CD44 was
visualised in red color by immunouorescence labeling via
Cy3. In all 29 HNSCC tissue samples an intense uorescence
signal could be detected (Fig. 1B and C). In HNSCC tissues,
CD44 was mainly expressed on cell surface in all samples
stained (Fig. 1C). Stromal cells did not show any uorescence
by staining of CD44 (Fig. 1B).
Concent ra tio n of CD44 in periph eral bloo d of HNSCC
patients. ELISA experiments were performed to measure
the concentration of soluble CD44 in the peripheral blood
of H NSCC patients compared to healthy cont rols. The
expression of soluble CD44 in serum of HNSCC and healthy
controls is shown in Fig. 2. There was a signicantly higher
concentration of CD44 in peripheral blood of HNSCC
patients compared to the healthy control group (mHNSCC =
13.5±0.5 ng/ml, mCont=9.3±0.6 ng/ml, P=0.6x10-12). Primary
tumor location ( Fig. 3A), T-stadium (B) and N-st adium
(C) did not significa ntly inf luence the concentration of
CD44 in peripheral blood of HNSCC patients. The values
determined and showed in Fig. 3 are: mlarynx=13. 0.4 ng/
ml, moroph=13.6±0.5 ng/ml, mhy poph=13.1±0.3 ng/ml, mor alcav
=13.5±0.3 ng/ ml (A); mT1=13.8±0.7 ng/ml, mT2=13. 0.3 ng/
ml, mT3=13.5±0.5 ng/ ml, mT4=13. 8 ± 0.5 ng /ml (B);
mN0=13.8±0.5 ng/ml, mN1=13.6±0.3 ng/ml, mN2=13.5±0.6 ng/
ml, mN3=14.1 (C); mCont 9.3±0.6 ng/ml (A-C).
Discussion
Expression of CD44 in HNSCC tissue samples. In this
study, we showed that CD44, which is referred to as a cancer
stem cell (CSC) marker of the head and neck (2), can be
detected in HNSCC tissue samples by immunouorescence
labeling. Others have used flow cytometry analysis (2,17),
immunohistochemistry (18-20), or microarray technology
(18) to verify CD44+ cells in HNSCC tissue samples. Results
consistently indicate the presence of CD44 in HNSCC tumors
on both protein and gene level. In our experiments CD44 was
abundantly expressed in all HNSCC samples tested (n=29)
and therefore conform to data collected by Han et al (18),
who performed immunohistochemical analysis of 16 HNSCC
tumor samples.
Figure 1. Expression of CD44 in HNSCC samples. Pathology of HNSCC
was established for each sample by H&E staining during routine diagnostics
(A). CD44 was visualised in red color by immunouorescence labeling via
Cy3 (B and C). Cell nuclei were stained in blue by DAPI. High uorescence
int en sity for CD44 was found in each of 29 HNSCC samples stained
(B and C). Each sample showed a surface staining patter n of CD44 (C).
Stromal cells did not show any uorescence by staining of CD44 via Cy3 (B).
A
B
C
Figu re 2. CD44 in per ipheral blood of HNSCC patients. Concentration
of soluble CD4 4 in p eripheral blood of HNSCC patients was evaluated
by ELISA and compared to healthy controls. There was a sig nificantly
higher concentration of CD44 in peripheral blood of HNSCC patients in
comparison to healthy controls (P=0.6x10-12).
FABER et al: CD44 AS A STEM CELL MARKER IN HEAD AND NECK SCC
324
The staining pattern shown in immunouorescence (Fig. 1B
and C) and immunohistochemistry (18) shows a monotonic
staining of almost all cells except stromal and vascular cells.
This stands in contrast to previous work e.g. by Prince et al,
who first postulated CD44+ cells as CSCs of the head and
neck (2,21). Prince et al reported, that CD44+ cancer cells
typically comprise <10% of the cells in an HNSCC tumor (2).
It is obvious that the percentage of cells of the tumor samples
stained as CD44+ in our experiments is much higher than 10%.
A possible reason might be the different experimental methods
of CD44 detection, e.g. ow cytometry versus immunouores-
cence labeling. It is even more reasonable that the percentage
of <10% considered by Prince et al comes off because of the
exclusion of Lin- cells. Lin- was dened as negative for the
surface markers CD2, CD3, CD10, CD18, CD31, CD64 and
CD140b (2). The ow cytometric analysis done by Pries et al
showed, that CD44 expression in HNSCC tumors differs inter-
individually. In their study, the percentage of cells in HNSCC
tumors varied from ~4 up to over 90% (17). It is possible that
this is correct because Pries et al had a look at the whole of the
tumor and not just at a small specimen cut out of the bulk of
an HNSCC. It has been shown in previous work, that CD44 is
a ubiquitary marker e.g. in the hematopoietic or mesenchymal
system (22).
Consistent with the work of others (18,19,23) CD44
showed a surface staining in immunouorescence labeling
in our study. The location of CD44 on the cell surface of
HNSCC might represent its role as an adhesion molecule
in tumor survival and progression. There is evidence that
CD44 proteins anchor the cells to the extracellular matrix
(ECM) and that adhesive properties of malignant cells have
to be altered to detach from the primary tumor in order to
build metastases (24). It has been shown that the extracellular
portion of CD44 serves as a substrate for proteolytic cleavage
by matrix metalloproteinases (MMPs) on several cancer cell
lines and in human tumors. This indicates that CD44 proteins
can serve as ‘platforms’ for enzymes such as MMPs, which
are required for their effective functions. The inhibition of
the reactions catalyzed by these MMPs can block tumor cell
migration (25,26). As tumor cells get in contact with the ECM
via several cell surface adhesion molecules such as CD44,
integrins and cadherins it is likely that these proteins are also
needed for metastatic processes (27). As CD44 obviously gets
in contact with the ECM it is imaginable that there is a CD44-
dependent assembly and organization of the ECM, which
might be useful to protect tumor cells from immune defence
(28,29). Pre-treatment of chondrocytes with an anti-CD44
antibody blocked their interaction with the ECM (30).
Concent ra tio n of CD44 in p eriph eral bloo d of HNSCC
patients. Soluble CD44 has been evaluated in blood serum
of patients with different primary cancers (5,21,31) as well as
in oral rinses of patients with HNSCC (4,32). Elevated CD44
serum levels have been found for several tumor types using
ELISA analysis (31,33,34).
In this study, we showed that the concentration of soluble
CD44 is signicantly higher in peripheral blood of HNSCC
patients compared to healthy controls. This is in contrast to
the ndings reported by Van Hal et al (21), who did not nd
signicant differences between the CD44v6 plasma levels of
HNSCC patients, healthy controls and non-cancer patients.
A reason might be the measurement of different isoforms of
CD44. Van Hal et al focussed on the detection of CD44v6, the
total quantity of CD44 including all isoforms was measured
A
B
C
Figure 3. CD44 in peripheral blood of HNSCC patients depending on
location of primary tumor, T-stage and N-stage. There are no sign ica nt
differenc es in the concentration of s oluble CD44 in HNSCC patient s
according to primary tumor location (A), T-stage (B) or N-stage (C). Note
the signica ntly higher concentr ation of CD44 in the periphe ra l blood
of HNSCC patients compared to healthy controls (A) P= 0.6x10-1 2, (B)
P=0,7x10-13, (C) P= 0.7x10-13.
ONCOLOGY REPORTS 26: 321-326, 2011 325
in our experiments. A n explanation for the findings of
Van Hal et al might be given by Herold-Mende et al (19).
They found that the variant exons of CD44 v5, v6, v7, v7-8
and v10 are expressed in epithelia of healthy donors and that
variant exons v7, v8 and v10 were signicantly downregulated
in primary squamous cell carcinoma and were not detected
at all in the majority of metastasis-derived specimens (19).
Expression of CD44v5 and CD44v6, on the other hand, was
mainly unaltered. However, this might give an explanation for
the ndings of Van Hal et al (21). In contrast to this Mack et al
found a slight increase of CD44s and CD44v6 levels in oral
leukoplakia and in moderately differentiated carcinomas (20).
For other types of cancer CD44v6 has already been shown to
be a potential marker of prognosis. Saito et al postulated that
the serum concentration of sCD44v6 and its expression in
tumors were associated signicantly with the depth of invasion
of the tumor, lymph node metastasis and clinical stage in
patients with diffuse type gastric carcinoma (5).
In our experiments, a highly signicant difference between
concentration of CD44 in serum of HNSCC patients was
found in comparison to healthy controls. In contrast to Saito et
al (5), there was no correlation to the stage of primary tumor
expansion, lymph node metastasis or distant metastasis in our
results. It is not the stage of disease, but rather the mass of the
tumor or the specic number of tumor cells that should be
seen in congruence with the concentration of CD44 in periph-
eral blood in HNSCC patients. In any case, the tissue samples
available for experimental analysis are small specimens out
of the bulk of a tumor. This is to guarantee benet of surgery
to patients and optimal terms and conditions of pathological
assessment to ensure the best possible treatment and follow-up
for the patient.
Acknowledgements
We gratefully thank Petra Prohaska for excellent technical
suppor t. Special thanks to Dr C. Barth, Dr U. Gössler,
Dr K. Götte, Dr J.T. Maurer, Dr H. Sadick, Dr A. Sauter,
Dr C. Schubotz-Mitgau, Professor B.A. Stuck and all the
colleagues in anaesthetics for their assistance in obtaining
tumor samples at the University Hospital of Mannheim,
Dep a rtment of Ot o r h i nola r yngology Hea d and Ne ck
S u rg e r y.
References
1. Vokes EE, Weichselbaum RR, Lippman SM and Hong W K:
Head and neck cancer. N Engl J Med 328: 184-194, 1993.
2. Prince ME, Sivanandan R, Kaczorowski A, et al: Identication
of a subpopulation of cells with cancer stem cell properties in
head and neck squamous cell carcinoma. Proc Natl Acad Sci
USA 104: 973-978, 2007.
3. Zhang P, Zuo H, Ozaki T, Nakagomi N and Kakudo K: Cancer
stem cell hypothesis in thyroid cancer. Pathol Int 56: 485-489,
2006.
4. Franzmann EJ, Reategui EP, Pedroso F, et al: Soluble CD44
is a potential marker for the early detection of head and neck
cancer. Cancer Epidemiol Biomarkers Prev 16: 1348-1355,
20 0 7.
5. Saito H, Tsujita ni S, Kat ano K, Ikeg uchi M, Maeta M and
Kaibara N: Serum concentration of CD44 variant 6 and its
relation to prognosis in patients with gastric carcinoma. Cancer
83: 1094-1101, 1998.
6. Ponta H, Wainwright D a nd Herrlich P: The CD44 protein
family. Int J Biochem Cell Biol 30: 299-305, 1998.
7. Screaton GR, Bell MV, Bell JI and Jackson DG: The identi-
cation of a new alternative exon with highly restricted tissue
expression in transcripts encoding the mouse Pgp-1 (CD44)
homing receptor. Comparison of all 10 variable exons between
mouse, human, and rat. J Biol Chem 268: 12235-12238, 1993.
8. Screaton GR, Bell MV, Jackson DG, Cornelis FB, Gerth U and
Bell JI: Genomic structure of DNA encoding the lymphocyte
homing receptor CD44 reveals at least 12 alternatively spliced
exons. Proc Natl Acad Sci USA 89: 12160-12164, 1992.
9. Picker LJ, Nakache M and Butcher EC: Monoclonal antibodies
to human lymphocyte homing receptors define a novel class
of adhesion molecules on diverse cell types. J Cell Biol 109:
927-937, 1989.
10. Stamenkovic I, Amiot M, Pesando JM and Seed B: A lympho-
cyte molecule implicated in lymph node homing is a member of
the cartilage link protein family. Cell 56: 1057-1062, 1989.
11. nthert U, Hofmann M, Rudy W, et al: A new va riant of
glycoprotein CD44 confers metastatic potential to rat carcinoma
cells. Cell 65: 13-24, 1991.
12. Hofmann M, Rudy W, Zoller M, et al: CD44 splice variants
confer metastatic behavior in rats: homologous sequences are
expressed in human tumor cell lines. Cancer Res 51: 5292-5297,
1991.
13. T henappan A, Li Y, Shet ty K, John son L, Reddy EP a nd
Mishra L: New Therapeutics Targeting Colon Ca ncer Stem
Cells. Curr Colorectal Cancer Rep 5: 209, 2009.
14. Zlobec I, Gunthert U, Tornillo L, et al: Systematic assessment
of the prognostic impact of membra nous CD44v6 prot ein
expression in colorectal cancer. Histopathology 55: 564-575,
20 09.
15. Park SY, Lee HE, Li H, Shipitsin M, Gelman R and Polyak K:
Heterogeneity for stem cell-related markers according to tumor
subtype and histologic stage in breast cancer. Clin Cancer Res
16: 876-887, 2010.
16. Okayama H, Kumamoto K, Saitou K, et al: CD44v6, MMP-7
and nuclear Cdx2 are signicant biomarkers for prediction of
lymph node metastasis in primary gastric cancer. Oncol Rep 22:
745-755, 2009.
17. Pries R, Wittkopf N, Hasselbach er K a nd Wollenberg B:
Constitutive expression of the potential stem cell marker CD44
in permanent HNSCC cell lines. HNO 56: 461-466, 2008 (In
German).
18. Han J, Kioi M, Chu WS, Kasperbauer JL, Strome SE a nd
Puri RK: Identication of potential therapeutic targets in human
head & neck squamous cell carcinoma. Head Neck Oncol 1: 27,
20 09.
19. Herold-Mende C, Seiter S, Born AI, et al: Expression of CD44
splice variants in squamous epithelia and squamous cell carci-
nomas of the head and neck. J Pathol 179: 66-73, 1996.
20. Mack B and Gires O: CD44s and CD44v6 expression in head
and neck epithelia. PLoS One 3: e3360, 2008.
21. Van Hal NL, Van Dongen GA, Ten Brink CB, et al: Evaluation
of soluble CD44v6 as a potential serum marker for head and
neck squamous cell carcinoma. Clin Cancer Res 5: 3534-3541,
1999.
22. Halfon S, Abra mov N, Grinblat B and Ginis I: Mar ker s
distinguishing mesenchymal stem cells from fibroblasts are
downregulated with passaging. Stem Cells Dev 20: 53-66, 2011.
23. van Hal NL, van Dongen GA, Stigter-van Walsum M, Snow GB
and Braken hoff RH: Characterization of CD44v6 isoforms
in head-and-neck squamous-cell carcinoma. Int J Cancer 82:
837-845, 1999.
24. Schirrmacher V: Cancer metastasis: experimental approaches,
theoretical concepts, and impacts for treatment strategies. Adv
Cancer Res 43: 1-73, 1985.
25. Okamoto I, Kawano Y, Tsuiki H, et al: CD44 cleavage induced
by a membrane-associated metalloprotease plays a critical role
in tumor cell migration. Oncogene 18: 1435-1446, 1999.
26. Okamoto I, Tsuiki H, Kenyon LC, et al: Proteolytic cleavage of
the CD44 adhesion molecule in multiple human tumors. Am J
Pathol 160: 441-447, 2002.
27. Liotta LA and Stetler-Stevenson WG: Tumor invasion and
metastasis: an imbalance of positive and negative regulation.
Cancer Res 51: S5054-S5059, 1991.
28. Naor D, Sionov RV and I sh- Sha lom D: CD44: struc ture,
function, and association with the ma lignant process. Adv
Cancer Res 71: 241-319, 1997.
29. Naor D, Wallach-Dayan SB, Zahalka MA and Sionov RV:
Involvement of CD44, a molecule with a thousand faces, in
cancer dissemination. Semin Cancer Biol 18: 260-267, 2008.
FABER et al: CD44 AS A STEM CELL MARKER IN HEAD AND NECK SCC
326
30. Knudson W: The role of CD44 as a cell surface hyaluronan
receptor during t umor invasion of connective tissue. Front
Biosci 3: D604-D615, 1998.
31. Harn HJ, Ho LI, Shyu RY, et al: Soluble CD44 isoforms in
serum as potential markers of metastatic gastric carcinoma. J
Clin Gastroenterol 22: 107-110, 1996.
32. Fra nzmann EJ, Reategui EP, Carraway KL, Ham ilton KL,
Weed DT and Goodwin WJ: Salivary soluble CD44: a potential
molecular marker for head and neck cancer. Cancer Epidemiol
Biomarkers Prev 14: 735-739, 2005.
33. De Rossi G, Marroni P, Paganuzzi M, et al: Increased serum
levels of soluble CD44 standard, but not of variant isoforms v5
and v6, in B cell chronic lymphocytic leukemia. Leukemia 11:
134-141, 1997.
34. Guo YJ, Liu G, Wang X, et al: Potential use of soluble CD44 in
serum as indicator of tumor burden and metastasis in patients
with gastric or colon cancer. Cancer Res 54: 422-426, 1994.
... Moreover, its initiation has been associated with multiple etiological factors, such as cigarette smoking, alcohol consumption, and human papillomavirus infection [1,4]. Furthermore, the pathogenesis of HNSCC has been proposed to be mediated by serial molecular events like CD44, CD133, and ALDH1 [5][6][7]. Although the recent clinical management of patients with HNSCC mainly depends on surgical procedures, chemoand radiotherapy, and immunotherapy, such as PD1, PDL1, and epidermal growth factor receptor (EGFR) inhibitors [1,8], in addition to the significant progress recorded in diagnosis and treatment during the past decades, the survival rates for HNSCC patients remains low. ...
... Levels of circRNA expression in HNSCC tissues were assessed using quantitative RT-PCR. N=12 (patient[3][4][5][6][7][8][9][10][11][12][13][14]. D RT-PCR-based validation of circRNA synthesis in HNSCC tissue samples (from patient 3). ...
Article
Full-text available
Objective Head and neck squamous cell carcinoma (HNSCC) is one severe malignancy driven by complex cellular and signaling mechanisms. However, the roles of circular RNAs (circRNAs) in HNSCC’s development remains poorly understood. Therefore, this study investigated the functions of differentially expressed circRNAs in regulating HNSCC cell functions. Methods Differentially expressed circRNAs were characterized through RNA sequencing in HNSCC tissues. CircRNA’s identity was then confirmed using RT-PCR and Sanger’s sequencing. Next, expression levels of circRNA and mRNA were detected by qRT-PCR, after which protein abundances were measured by Western blotting. Subsequently, the proliferation, migration, and invasion of HNSCC cells was assessed by MTS, wound healing, and Transwell system, respectively, followed by identification of circRNA-binding proteins in HNSCC cells by circRNA pull-down, coupled with mass spectrometry. Results Great alterations in circRNA profiles were detected in HNSCC tissues, including the elevated expression of circ_0000045. As observed, silencing of circ_0000045 effectively repressed the proliferation, migration, and invasion of HNSCC cell lines (FaDu and SCC-9). Contrarily, circ_0000045’s overexpression promoted the proliferation, migration, and invasion in FaDu and SCC-9 cells. Results also showed that circ_0000045 was associated with multiple RNA-binding proteins in HNSCC cells, such as HSP70. Moreover, circ_0000045 knockdown enhanced HSP70 expression and inhibited JNK2 and P38’s expression in HNSCC cells, which were oppositely regulated by circ_0000045’s overexpression. Conclusion The high expression of circ_0000045; therefore, promoted cell proliferation, migration, and invasion during HNSCC’s development through regulating HSP70 protein and mitogen-activated protein kinase signaling.
... HNSCC cell molecular markers CD44, CD133, and ALDH1 are of high prognostic importance. Hyaluronic acid and MMPs, critical for intercellular interaction and migration, are the ligands for cell surface receptor CD44, which increases the self-renewal property and metastasis of cells [107][108][109]. CD133, a transmembrane protein, is also upregulated [110]. ...
Article
Full-text available
Simple Summary According to WHO 2018 estimates, cancer is the second leading cause of mortality worldwide after cardiovascular disease, accounting for one in six fatalities. Mortality due to head and neck cancer is also significantly high, making it the seventh most prevalent cancer. Elucidating the mechanisms of action of key regulators, such as bioactive molecules secreted by the cancer cells in exosomes, their interaction with neighboring and distant cells, the extracellular matrix, and the tumor microenvironment, could be a valuable tool for future diagnostic and treatment approaches. Exosomes facilitate several critical functions in cancer cells, such as epithelial-to-mesenchymal transition, which makes head and neck cancer even worse by giving it a metastatic potential to evade the secondary site and spread cancer in the body. Exploring the molecules involved in this process could help targeting specific sites, for instance, by modifying the cell-specific proteins and exosome cargoes. Abstract Exosomes are nanosized vesicles that are produced in normal and cancer cells, promoting intracellular communication. In head and neck cancer (HNC), exosomes are involved in many undesirable events of cancer development and progression, including angiogenesis, tumor microenvironment (TME) remodeling, invasion, epithelial-to-mesenchymal transition (EMT), metastasis, extracellular matrix (ECM) degradation, and drug resistance. Exosomes are involved in altering the signaling pathways in recipient cells by the cargoes they carry. Proteins, lipids, and nucleic acids such as DNA fragments and RNAs (i.e., mRNAs, miRNAs, and long non-coding RNAs) are carried in the exosomes to promote cell communication. EMT is a critical cellular process in which epithelial cells are forced to become mesenchymal cells by the actions of SNAIL/SLUG, TWIST, and ZEB family transcription factors carried in exosomes that facilitate metastasis. In this critical review, we focused on exosome biogenesis, their cargoes, and their involvement in EMT induction and metastasis during HNC. Insights into exosome isolation and characterization, as well as their key role in ECM remodeling and degradation, are also presented and critically discussed. More importantly, this article addresses the role of exosomes in HNC and drug resistance induced in drug-sensitive cancer cells. In addition, exosomes have a great potential to be used as diagnostic and therapeutic tools. A better understanding on exosome biogenesis, composition, and functions in HNC will aid in developing novel therapeutic strategies to treat HNC, overcome therapy resistance, and avoid metastasis, which is a significant cause of cancer death.
... Previous reports demonstrate that exposure to tobacco, alcohol consumption, as well as human papillomavirus (HPV) and epsteinbarr virus (EBV) infection are the general risk factors for HNSC (Isayeva et al. 2012;Johnson et al. 2020). It has become increasingly clear that the epithelial-mesenchymal transition (EMT), altered expression of molecular biomarkers such as CD44, CD133 and matrix metalloproteinases (MMPs) that involved in cell adhesion and migration, along with disrupted TME have a key role in the formation and progression of HNSC (Faber et al. 2011;González-González et al. 2021;Yu and Cirillo 2020). In the present study, we discovered that ENY2 displayed significant correlation with HNSC. ...
Article
Full-text available
ENY2 (Enhancer of yellow 2 transcription factor) is a transcription nuclear protein and primarily participates in the course of mRNA export and histone deubiquitination to influence gene expression. Current studies have shown that the expression of ENY2 is significantly upregulated in multiple cancers. However, the exact association between ENY2 and pan-cancers has not been fully established. Here, we comprehensively analyzed ENY2 from the online public database and The Cancer Genome Atlas (TCGA) database, including gene expression level in pan-cancer, comparison of ENY2 expression in different molecular and immune subtypes of pan-cancer, targeted protein, biological functions, molecular signatures, diagnostic and prognostic value in pan-cancer. Moreover, we focused on head and neck squamous cell carcinoma (HNSC) and explored ENY2 from the perspective of the correlations with clinical characteristics, prognosis, co-expression genes, differentially expressed genes (DEGs) and immune Infiltration. Our findings showed that the expression of ENY2 differed enormously not only in most cancer types but also in different molecular and immune subtypes of cancers. High accuracy in predicting cancers and notable correlations with prognosis of certain cancers suggested that ENY2 might be a potential diagnostic and prognostic biomarker of cancers. In addition, ENY2 was identified to be significantly correlated with clinical stage, gender, histologic grade and lymphovascular invasion in HNSC. Overexpression of ENY2 could lead to a worse overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI) in HNSC, especially in different clinical subgroups of HNSC. Taken together, ENY2 showed strong correlation with the diagnosis and prognosis of pan-cancer, and was an independent prognostic risk factor of HNSC, which may serve as a potential target for cancer management.
... The HNSCC cells with high levels of CD44 glycoproteins are capable of self-renewal. CD44 levels in HNSCC tumours are associated with metastasis and a poor prognosis [23,24]. CD133 glycoproteins results in increased invasiveness and metastasis in HNSCC. ...
Chapter
Full-text available
Head and Neck cancer accounts for approximately 900,000 cases and over 400,000 deaths annually worldwide. The primary risk factors associated with Head and Neck cancer include usage of tobacco, alcohol consumption, Human Papillomavirus (HPV) infection and Epstein-Barr virus (EBV) infection. Few subsites of Head and Neck Squamous Cell Carcinoma (HNSCC) are associated with Human Papilloma Virus (HPV) while others remain non-associated. The anatomical, physiological, genetic, protein profile and epigenetic changes that occur in both HPV-positive and HPV-negative HNSCC has been discussed in this chapter. The mutational profile plays a crucial role in the treatment of the HNSCC patients as the HPV-positive HNSCC patients have a better prognosis compared to the HPV-negative HNSCC patients. This chapter mainly focusses on the mutational profile of both HPV-associated and non-HPV associated HNSCC tumours.
Article
Full-text available
In oral squamous cell carcinoma (OSCC), a highly aggressive and frequently lethal malignancy, the role and action mechanism of the microtubule regulatory protein CDK5RAP2 have not been fully understood. Here, we show that CDK5RAP2 is highly expressed in OSCC and its expression correlates with clinical stage and lymph node metastasis of the disease. The expression of CDK5RAP2 is regulated by the Wnt signaling pathway. Depletion of CDK5RAP2 inhibits the tumorigenesis and migration of OSCC cells and alters the OSCC cancer stem (-like) cell (CSC) signature. Notably, suppression of CDK5RAP2 expression disrupts spindle orientation during mitosis. Collectively, these results identify CDK5RAP2 as a potential CSC marker and reveal a mechanism that controls the CSC population in OSCC.
Thesis
Full-text available
Ce projet de recherche translationnelle sur les cancers des voies aérodigestives supérieures (VADS) a permis d’identifier la cavéoline-1 (Cav1) comme un biomarqueur pronostique de l’évolution d’une tumeur primitive des VADS. En effet, une surexpression de cette protéine favorise une rechute locorégionale alors qu’un déficit de Cav1 engage la tumeur vers un processus métastatique. De plus, nous avons mis en évidence l’implication de l’axe Cav1 / EREG / YAP dans la résistance au traitement (cétuximab et radiothérapie). Enfin, nous avons identifié l’épireguline (EREG) comme la protéine clé de la résistance au cétuximab. Ainsi, un déficit d’EREG sensibilise les cellules au cétuximab par activation de la ferroptose et l’association de cette thérapie ciblée à la molécule de RSL3 ou à la metformine restreint drastiquement la survie cellulaire en accentuant cette mort cellulaire programmée. Ces derniers résultats ont pu être confirmés grâce à un modèle 3D complexe récapitulant l’hétérogénéité intra- et inter-tumorale, à savoir le modèle tumoroïde établi à partir de pièces opératoires de patients atteints d’un cancer des VADS.
Article
Full-text available
Squamous cell carcinoma is the most common malignancy that arises in the head-and-neck district. Traditional treatment could be insufficient in case of recurrent and/or metastatic cancers; for this reason, more selective and enhanced treatments are in evaluation in preclinical and clinical trials to increase in situ concentration of chemotherapy drugs promoting a selectively antineoplastic activity. Among all cancer treatment types (i.e., surgery, chemotherapy, radiotherapy), electroporation (EP) has emerged as a safe, less invasive, and effective approach for cancer treatment. Reversible EP, using an intensive electric stimulus (i.e., 1000 V/cm) applied for a short time (i.e., 100 μs), determines a localized electric field that temporarily permealizes the tumor cell membranes while maintaining high cell viability, promoting cytoplasm cell uptake of antineoplastic agents such as bleomycin and cisplatin (electrochemotherapy), calcium (Ca2+ electroporation), siRNA and plasmid DNA (gene electroporation). The higher intracellular concentration of antineoplastic agents enhances the antineoplastic activity and promotes controlled tumor cell death (apoptosis). As secondary effects, localized EP (i) reduces the capillary blood flow in tumor tissue (“vascular lock”), lowering drug washout, and (ii) stimulates the immune system acting against cancer cells. After years of preclinical development, electrochemotherapy (ECT), in combination with bleomycin or cisplatin, is currently one of the most effective treatments used for cutaneous metastases and primary skin and mucosal cancers that are not amenable to surgery. To reach this clinical evidence, in vitro and in vivo models were preclinically developed for evaluating the efficacy and safety of ECT on different tumor cell lines and animal models to optimize dose and administration routes of drugs, duration, and intensity of the electric field. Improvements in reversible EP efficacy are under evaluation for HNSCC treatment, where the focus is on the development of a combination treatment between EP-enhanced nanotechnology and immunotherapy strategies.
Article
Recent years have seen rise in applications of differential co-expression analysis (DCE) for disease biomarker identification. This paper presents a centrality-based hub-gene centric method called Centrality Based Differential Co-Expression Method (CBDCEM), for crucial gene finding for critical diseases. A prominent task of DCE is the identification of hub-gene(s) for each differentially co-expressed module. We propose a consensus-based algorithm that employs seven centrality measures to detect hub-genes. Our method has been validated on three Esophageal Squamous Cell Carcinoma (ESCC) datasets and is found to perform well for denser modules. Through comparison of our proposed hub-gene finding algorithm with 4 other hub-gene finding methods, we have observed that the former was able to identify unique potential biomarkers that were undetected by the rest. CBDCEM is able to identify 15 genes as potential biomarkers for ESCC. Eleven other genes identified by CBDCEM have some evidence of being potential biomarkers for other Squamous Cell Carcinoma and can be considered candidates for ESCC, but require further in-depth validation.
Article
Full-text available
Background: Radiotherapy is the first-line treatment for patients nasopharyngeal carcinoma (NPC), but its therapeutic efficacy is poor in some patients due to radioresistance. Adoptive T cell-based immunotherapy has also shown promise to control NPC; however, its antitumor efficacy may be attenuated by an immunosuppressive tumor microenvironment. Exosomes derived from γδ-T cells (γδ-T-Exos) have potent antitumor potentials. However, it remains unknown whether γδ-T-Exos have synergistic effect with radiotherapy and preserve their antitumor activities against NPC in an immunosuppressive tumor microenvironment. Methods: γδ-T-Exos were stained with fluorescent membrane dye, and their interactions with NPC were determined both in vitro and in vivo. NPC cell deaths were detected after treatment with γδ-T-Exos and/or irradiation. Moreover, effects of γδ-T-Exos on radioresistant cancer stem-like cells (CSCs) were determined. The therapeutic efficacy of combination therapy using γδ-T-Exos and irradiation on NPC tumor progression was also monitored in vivo. Finally, the tumor-killing and T cell-promoting activities of γδ-T-Exos were determined under the culture in immunosuppressive NPC supernatant. Results: γδ-T-Exos effectively interacted with NPC tumor cells in vitro and in vivo. γδ-T-Exos not only killed NPC cells in vitro, which was mainly mediated by Fas/Fas ligand (FasL) and death receptor 5 (DR5)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathways, but also controlled NPC tumor growth and prolonged tumor-bearing mice survival in vivo. Furthermore, γδ-T-Exos selectively targeted the radioresistant CD44+/high CSCs and induced profound cell apoptosis. The combination of γδ-T-Exos with radiotherapy overcame the radioresistance of CD44+/high NPC cells and significantly improved its therapeutic efficacy against NPC in vitro and in vivo. In addition, γδ-T-Exos promoted T-cell migration into NPC tumors by upregulating CCR5 on T cells that were chemoattracted by CCR5 ligands in the NPC tumor microenvironment. Although NPC tumor cells secreted abundant tumor growth factor beta to suppress T-cell responses, γδ-T-Exos preserved their direct antitumor activities and overcame the immunosuppressive NPC microenvironment to amplify T-cell antitumor immunity. Conclusions: γδ-T-Exos synergized with radiotherapy to control NPC by overcoming the radioresistance of NPC CSCs. Moreover, γδ-T-Exos preserved their tumor-killing and T cell-promoting activities in the immunosuppressive NPC microenvironment. This study provides a proof of concept for a novel and potent strategy by combining γδ-T-Exos with radiotherapy in the control of NPC.
Article
Full-text available
The recent identification of tumor-initiating colorectal cancer (CRC) stem cells in the pathogenesis of CRC has provided a potential target for novel therapeutics. Many details about CRC stem cells, however, remain poorly understood. Several potential markers of CRC stem cells have been proposed, including CD133, CD44, and, recently, Lgr5. Attention also has been drawn to control of stem cell self-renewal, proliferation, and differentiation by the Wnt and transforming growth factor (TGF)-β pathways. Disruption of Wnt signaling, via loss of APC (adenomatous polyposis coli), is among the earliest events in the multistage progression of CRC and likely occurs in basal crypt stem cells, generating a neoplastic cell population that then expands upward to occupy the rest of the crypt. TGF-β signaling is a key tumor suppressor pathway, and mutations in the type II receptor and Smad4 are observed in CRC specimens and are associated with more aggressive disease in tumors with disrupted Wnt signaling. Loss of the TGF-β adaptor protein β(2)-spectrin is associated with loss of colonic cell polarity and architecture, and its expression parallels that of Smad4. This review suggests rational approaches to target CRC stem cells as a novel and effective way to treat advanced and difficult-to-treat CRC.
Article
Full-text available
To evaluate the expression of stem cell-related markers at the cellular level in human breast tumors of different subtypes and histologic stage. We performed immunohistochemical analyses of 12 proteins [CD44, CD24, ALDH1, vimentin, osteonectin, EPCR, caveolin 1, connexin 43, cytokeratin 18 (CK18), MUC1, claudin 7, and GATA3] selected based on their differential expression in breast cancer cells with more differentiated and stem cell-like characteristics in 47 cases of invasive ductal carcinoma (IDC) only, 135 cases of IDC with ductal carcinoma in situ (DCIS), 35 cases of DCIS with microinvasion, and 58 cases of pure DCIS. We also analyzed 73 IDCs with adjacent DCIS to determine the differences in the expression of markers by histology within individual tumors. CD44+/CD24- and CD24-/CD24+ cells were detected using double immunohistochemistry. CD44 and EPCR expression was different among the four histologic groups and was lower in invasive compared with in situ tumors, especially in luminal A subtype. The expression of vimentin, osteonectin, connexin 43, ALDH1, CK18, GATA3, and MUC1 differed by tumor subtype in some histologic groups. ALDH1-positive cells were more frequent in basal-like and HER2+ than in luminal tumors. CD44+/CD24- cells were detected in 69% of all tumors with 100% of the basal-like and 52% of HER2+ tumors having some of these cells. Our findings suggest that in breast cancer, the frequency of tumor cells positive for stem cell-like and more differentiated cell markers varies according to tumor subtype and histologic stage.
Article
Full-text available
The aim of this study was to examine the expression of CD44v6, CD54, Cdx2, CXCL5, Cyclin B1, MMP-7, nm23, RCAS1 and Survivin in primary gastric cancer and to investigate whether these molecules were useful in predicting the lymph node status. They were selected as candidates for indicators of lymph node metastasis from various kinds of cancer-associated genes reported previously. In 135 cases of radically resected primary gastric adenocarcinoma, we investigated the association between the expression of these molecules and clinocopathologic factors by immunohistochemistry. The results revealed that the expression of CD44v6 and MMP-7 were significantly associated with lymph node status. By contrast, nuclear Cdx2 expression was found to be inversely correlated with lymph node metastasis. Moreover, multivariate analysis demonstrated that CD44v6, MMP-7 and nuclear Cdx2 were independent predictors for lymph node status. In conclusion, our results suggest that positive expression of both CD44v6 and MMP-7, and negative expression of nuclear Cdx2 may serve as powerful predictors of lymph node metastasis in gastric cancer. Combined evaluation of these markers could be further useful to predict lymph node status clinically.
Article
Background Despite significant advances and the use of new diagnosic and therapy methods to treat head and neck squamous cell carcinoma (HNSCC), prognosis has improved only marginally in the last decades. Thus, there is an enormous need for novel immunotherapeutic approaches. It is becoming more and more obvious that stem cells play an important role in tumor development and progression. The identity of these cells and the underlying cellular and molecular mechanisms remain mostly unknown in HNSCC. Material and methods Solid tumors as well as cells from the peripheral blood of patients with HNSCC were analyzed by flow cytometry concerning the expression of different putative stem cell marker proteins. Results Distinct populations of CD44-positive (CD44+), lin-negative (lin-) potential stem cells could be identified in solid tumors of HNSCC patients with strong individual variations. Surprisingly, the potential stem cell marker CD44 was found to be constitutively expressed on the surface of all permanent HNSCC cell lines analyzed. Conclusion These data strongly suggest that CD44+ tumor stem cells may play a key role in establishment of permanent HNSCC cell lines, selecting especially robust cell entities that in real life might drive progression and metastasis of HNSCC. Individual analysis of tumor stem cell markers will be an important tool for innovative therapies and prognosis of patients with HNSCC. Additional stem cell markers will be investigated concerning their expression and cellular function.
Article
The designation CD44 describes a group of type I transmembrane proteins which share N-terminal and C-terminal sequences. These molecules differ in the central extracellular domain by the use of sequences encoded by ten variant exons which may be completely absent or included in various combinations and by cell type specific addition of glycosaminoglycan and carbohydrate moieties. Expression of variant proteins is observed in normal tissues such as on keratinocytes, dendritic cells and activated lymphocytes in the adult organism and on morphogenetically active epithelium such as the apical ectodermal ridge (AER) in the embryo. Certain CD44 proteins expressed on the AER can act as low affinity fibroblast growth factor receptors and are vital for epithelial–mesenchymal cell communication. CD44 variant proteins have also been implicated in tumour growth and metastasis and we speculate that CD44 mediated growth factor presentation may also be decisive in metastasis formation. Molecular strategies designed to block growth factor presentation by CD44 may aid in the therapy of metastatic cancer.
Article
Splice variants of the adhesion molecule CD44 have been described as essential for the lymphatic spread of rat tumour cells and are claimed to be involved in the metastatic spread of several human tumours. Immunohistochemistry has been used to analyse the expression pattern of CD44 standard (CD44s) and variant (CD44v) isoforms in normal and dysplastic squamous epithelia, as well as in primary and metastatic squamous cell carcinomas (SCCs), which spread predominantly by way of the lymphatic system. Frozen sections of squamous epithelia and of squamous cell carcinomas were stained with a panel of monoclonal antibodies recognizing epitopes of CD44s as well as of the variant exons v5, v6, v7, v7–v8, and v10. The stratum basale and stratum suprabasale of squamous epithelia stained with all antibodies; the stratum spinosum stained with anti-CD44v5, anti-CD44v6, anti-CD44v7–8 and anti-CD44v10; the lower layers of the stratum corneum stained with anti-CD44v5. This expression profile was seen in epithelia of the lip, the tongue, the gingiva, the hard palate, the floor of the mouth, the buccal mucosa, and the pharynx. The same pattern of expression was also noted in dysplastic epithelia, but expression of the variant exons v7, v8, and v10 was significantly downregulated in primary squamous cell carcinomas and was not detected at all in the majority of metastasis-derived specimens. Expression of CD44v5 and CD44v6, on the other hand, was mainly unaltered. Thus, epithelial cell layers representing different stages of differentiation express distinct sets of CD44 variant isoforms, where especially exons v8–v10 might be required for the maintenance of the structural integrity of squamous epithelium. Downregulation of these exons on tumour cells could indicate that they are irrelevant for tumour progression or may even hamper infiltration of surrounding tissue or of lymphatics.
Article
BACKGROUND The expression of variant isoforms of CD44 is correlated with the ability of tumor cells to metastasize in some clinical carcinomas. In this study, the serum concentration of soluble splice isoforms of CD44 that shared exon variant 6 (sCD44v6) were measured and the histologic expression of CD44v6 in tumors from patients with gastric carcinoma examined.METHODS Serum samples were obtained from 102 patients with primary gastric carcinoma before surgery and serum levels of sCD44v6 were determined with an enzyme-linked immunoadsorbent assay. The expression of CD44v6 in tumors was examined by immunohistochemical staining.RESULTSBoth the serum concentration of sCD44v6 and its expression in tumors were associated significantly with the depth of invasion of the tumor, lymph node metastasis, and clinical stage in patients with diffuse type gastric carcinoma. Among patients with gastric carcinoma, the serum level of sCD44v6 was higher in those with CD44v6 positive tumor cells than in those with CD44v6 negative tumor cells. The serum level of sCD44v6 was a prognostic indicator in patients with diffuse type gastric carcinoma, as was the histologic expression of CD44v6. However, neither CD44v6 nor sCD44v6 was a predictor of survival time in patients with intestinal type gastric carcinoma.CONCLUSIONS It appears that CD44v6 and sCD44v6 are related to the progression of diffuse type gastric carcinoma. An elevated serum level of sCD44v6 may be useful as a prognostic indicator in patients with diffuse type gastric carcinoma. Cancer 1998;83:1094-1101. © 1998 American Cancer Society.
Article
Monoclonal antibodies in the Hermes family recognize a lymphocyte structure that participates in lymphocyte adhesion to endothelium and has been suggested to be the human homolog of the murine Mel-14 lymph node homing receptor. Recently, antibodies against the Hermes antigen, the polymorphic glycoprotein Pgp-1 antigen, and the broadly expressed CDw44 antigen have been shown to recognize the same structure. In this work, cDNA clones encoding the CDw44 antigen were isolated and expressed in COS cells. Two forms were identified: a lymphoid form expressed in hematopoietic cells, and an epithelial form weakly expressed in normal epithelium but highly expressed in carcinomas. The extracellular domain of CDw44 bears homology to cartilage link proteins and a related segment of proteoglycan core protein. However, comparison with the recently identified sequence of the Mel-14 antigen shows that CDw44 and Mel-14 are unrelated.
Article
Expansion of plastic-adherent bone marrow-derived mesenchymal stem cells (MSCs) results in gradual loss of osteogenic potential after passage 5-6. One explanation is contamination of MSC cultures with mature cells including fibroblasts. Identification and elimination of fibroblasts from MSC cultures could improve MSC yield and differentiation potential and also prevent tumor formation after MSC transplantation. However, no specific markers currently exist that can reliably discriminate between MSCs and fibroblasts. Flow cytometry analysis demonstrated that markers currently used to define MSCs, such as CD105, CD166, CD90, CD44, CD29, CD73, and CD9, are also expressed on human skin or lung fibroblasts. However, the level of expression of CD166 was significantly higher and that of CD9 was significantly lower in MSCs than in fibroblasts. CD146 was expressed only in MSCs. Using small focused microarrays, new markers differentially expressed in MSCs and fibroblasts were identified. Real-time polymerase chain reaction confirmed that expression of CD106, integrin alpha 11, and insulin-like growth factor-2 in MSCs was at least 10-fold higher than in fibroblasts; whereas expression of matrix metalloproteinase 1 and matrix metalloproteinase 3 was almost 100-fold lower. Flow cytometry and immunostaining demonstrated that CD106 protein expression on cell surface could be upregulated in MSCs but not in fibroblasts by the treatment with tumor necrosis factor-alpha. Comparison of surface expression of commonly used and newly identified MSC markers in MSCs cultures of passage 2 and passage 6 demonstrated that CD106 (with and without tumor necrosis factor-alpha treatment), integrin alpha 11, and CD146 were downregulated in MSCs of passage 6, and CD9 was upregulated; whereas all other markers did not change. Newly identified markers that have robust differences of expression in MSCs and fibroblasts on gene and protein level could be used for quality control of MSC cultures after expansion, cryopreservation, gene transfection, and other manipulations.
Article
To assess systematically the membranous expression of CD44v6 in colorectal cancer by immunohistochemistry to determine its prognostic impact, the differential expression between primary and metastatic tumours and expression differences between the tumour centre and invasive front. Immunohistochemistry was performed for CD44v6 on two tissue microarrays. The first included 1279 colorectal tumours with full clinicopathological data. The second consisted of 50 matched primary and metastatic tumours sampled from the tumour centre and the invasive margin. A scoring system was tested by multiple observers. Receiver-operating characteristic curve analysis was used for cut-off point determination. Loss of membranous CD44v6 was associated with pT stage (P = 0.016; sensitivity 85.8%, specificity 20.1%), lymph node metastasis (P = 0.015; sensitivity 52.8%, specificity 55%), an infiltrating tumour margin (P < 0.001; sensitivity 71.4%, specificity 40%) and adverse prognosis (P = 0.011; hazard ratio 0.79, 95% confidence interval 0.7, 0.9), but was not an independent prognostic factor on multivariable analysis. Loss of expression occurred at the invasive front in both primary and metastatic lesions (P < 0.001). This study outlines an approach to help standardize the immunohistochemical evaluation of CD44v6 and similar markers in colorectal cancer and highlights a significant role for loss of membranous CD44v6 expression in colorectal cancer progression and prognosis.