• No results found

CD44 glycoproteins in colorectal cancer; expression, function and prognostic value - Thesis

N/A
N/A
Protected

Academic year: 2021

Share "CD44 glycoproteins in colorectal cancer; expression, function and prognostic value - Thesis"

Copied!
175
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

UvA-DARE (Digital Academic Repository)

CD44 glycoproteins in colorectal cancer; expression, function and prognostic

value

Wielenga, V.J.M.

Publication date 1999

Document Version Final published version

Link to publication

Citation for published version (APA):

Wielenga, V. J. M. (1999). CD44 glycoproteins in colorectal cancer; expression, function and prognostic value.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)

CD44 glycoproteins in

colorectal cancer

expression, function

and prognostic value

Y

o

n i

HIT

Il il II II INI II

Il II II II II II II INI II II II II II II

(3)
(4)

CD44 GLYCOPROTEINS IN COLORECTAL CANCER; EXPRESSION, FUNCTION AND PROGNOSTIC VALUE

(5)

This thesis was prepared at the Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands, and supported by grants from The Dutch Cancer Society and Het

Praeventiefonds.

(6)

CD44 GLYCOPROTEINS IN COLORECTAL CANCER; EXPRESSION, FUNCTION AND PROGNOSTIC VALUE

Academisch proefschrift

ter verkrijging van de graad van doctor aan de Universiteit van Amsterdam op gezag van de Rector Magnificus

prof.dr J.J.M. Franse

ten overstaande van een door het college voor promoties ingestelde commissie,

in het openbaar te verdedigen in de Aula der Universiteit op 29 Juni 1999, te 11.00 uur

door Vera Jacqueline Marita Wielenga geboren te Wettingen

(7)

Faculteit der Geneeskunde Promotores

Prof. dr S.T. Pals

Prof. dr G.J.A. Offerhaus Promotiecommissie

Prof, dr A. Berns Prof, dr H.C. Clevers Prof, dr R. Fodde Prof dr F.J.W. ten Kate Prof. dr C.J.F, van Noorden Prof. dr J.H.P. Wilson

(8)

Contents Abbreviations

Chapter 1 General introduction 7 Chapter 2 Expression of CD44 variant proteins in human colorectal

cancer is related to tumor progression.

Cancer Res 1993, 53:4754-4756 49 Chapter 3 Expression of mutant P53 protein and CD44 variant proteins

in colorectal tumorigenesis

Gut 1995, 36:76-80 53 Chapter 4 CD44 splice variants as prognostic markers in colorectal

cancer.

Scan J Gastroenterol 1998, 33:82-87 59 Chapter 5 Expression of CD44 in Ape and Tcf mutant mice implies

regulation by the Wnt- pathway.

Am J Pathol 1999, 154:515-524 67 Chapter 6 Heparan sulfate-modified CD44 promotes hepatocyte growth

factor/scatter factor-induced signal transduction through the receptor tyrosine kinase c-Met.

JBiolChem 1999,274:6499-6506 91 Chapter 7 Co-expression of c-Met and heparan sulfate proteoglycan

forms of CD44 in colorectal cancer 101 Chapter 8 CD44 glycoproteins in colorectal cancer; expression,

function and prognostic value.

Adv Cancer Res, In press 129

Summary/Samenvatting 159 Acknowledgements/Dankwoord 169

(9)

Abbreviations

APC Adenomatous polyposis coli

ACF Abberant crypt focus

CD44s CD44standard

CD44v CD44variants

ECM Extracellular matrix

ERM Ezrin, radixin, moesin

FAP Familial adenomatous polyposis

FGF-2 Fibroblast growth factor-2

GAG Glycosaminoglycans

HA Hyaluronic acid

HS Heparan sulfate

HGF/SF Hepatocyte growth factor/scatter factor

PG Proteoglycan

(10)

Chapter 1

(11)

INTRODUCTION General introduction.

Colorectal cancer is the second leading cause of cancer related death in the western world (American Cancer Society, 1994). Mortality due to colorectal cancer is almost always the result of tumor metastasis. Roughly half of the patients with apparent localized disease at the time of diagnosis, who are operated with the intention to cure, will nevertheless have an eventual recurrence of the disease, due to undetected micrometastasespresent at the time of surgery (Coleman et al, 1993; Greenson et al, 1994). In order to improve prognosis it is imperative that the molecular determinants of metastasis are identified. This would enable differentiation between patients with actual localized disease and patients in which the disease has spread already and who might benefit from aggressive adjuvant therapy. The aim of this thesis therefore, is to study the expression and regulation of CD44 and its isoforms, molecules which have been implicated in metastasis formation in animal models, and to explore their usefulness in predicting the natural history of colorectal cancer.

COLORECTAL CARCINOGENESIS

Cancer of the large bowel mucosa develops through a series of morphologically well described precursor lesions, known as the adenoma-carcinoma sequence. The mucosal changes are the result of progressive genetic alterations, that occur at specific time points during carcinogenesis, as summarized in "the Vogelgram"(Fig.l) (Muto et al, 1975; Vogelstein et al, 1988; Fearon et al, 1990). Four types of genes can be mutated, with different mutational routes. Stimulatory genes -proto-oncogenes- become hyperactive due to mutations with a dominant effect: only one of the gene copies need to undergo the change, and the altered gene is called an oncogene. Otherwise, inhibitory genes can be made inactive, and this type of mutation has a recessive effect: both gene copies must be inactivated or deleted to free the cell from inhibition, and the lost gene is called a tumor suppressor gene. A third

(12)

APC (5q) B-catenin (3p) K-ras (12p) SMAD2 (18q) SMAD4 (18q) p53 (17p) normal early polyp Tatï polyp

m

\i i/

mismatch repair

m

Figure 1. The adenoma-carcinoma sequence.

mechanism is the obstruction of the DNA mismatch proof reading system genes due to mutations in both gene copies of the mismatch-repair genes. Mismatch-repair genes repair DNA replication errors that occur during the S-phase. Disruption of the repair mechanism results in a hypermutable phenotype, leading to accumulation of other somatic genetic mutations at an accelerated rate. The last type of genes that can be mutated are the mitotic breakpoint genes, which are implicated in correcting failures during mitotic segregation of the chromosomes.

Although the majority of colorectal cancers occurs sporadically, well described inherited predispositions to colorectal cancer as, for example, familial adenomatous polyposis (FAP) and hereditary non polyposis colorectal cancer (HNPCC), have greatly increased the insight in the molecular genetics underlying sporadic colorectal cancer. Specific pathways are disrupted by molecular genetic changes of which the sequential order in the adenoma-carcinoma sequence results in progressive disturbance of cell growth and motility.

(13)

The ras oncogenes.

Three cellular transforming ras oncogenes genes have been described,

K-ras, U-ras and N-ras (Weinberg et al, 1994). The ras genes encode

GTP-binding proteins (Bourne et al, 1991). A point mutation at an appropriate site in a ras gene creates a Ras protein that fails to hydrolyze its bound GTP and thereby persists in its active state, resulting in a growth advantage during tumorigenesis. Point mutations of ras genes are acquired during adenoma progression, and are rarely present in adenomas smaller than 1 cm but are found in 50% of adenomas larger than 1 cm and in 50% of carcinomas (Vogelstein et al, 1988; Poylak et al, 1996). Ras mutations occur frequently in adenomas, but are not neoplastic per se, since they are also found in the majority of the self-limiting non-dysplastic aberrant crypt foci (ACF).

The GTP-ase activity of wild-type Ras is stimulated by GTP-ase activating protein (GAP), while mutant Ras does not respond. Recently, the gene responsible for neurofibromatosis type I (NF1) was found to have GAP activity. This is of interest since a colon tumor without a ras mutation was reported to have a mutation of NF1 (Xuetal, 1990; Li etal, 1992). Hence, in part of the ras mutation negative colon tumors, deregulation of the same pathway by mutation of a different component might play a role

The p53 tumor-suppressor gene.

The p53 gene is located on chromosome 17p and encodes a 53kD nuclear phosphoprotein identified as a tumor suppressor gene. The wild-type

p53 gene product was shown to function as a transcription factor causing arrest

of progression through the cell cycle in Gl and repair of genotoxic damage or otherwise pushing the cell into apoptosis (Kern et al, 1994). Targets regulated by p53 include the apoptosis-inducing BAX gene (Miyashita et al, 1994) and the growth-arrest genep21/WAFl/CIPI(El-Deiry WS et al, 1993; Harper et

al, 1993).

Loss of 17p was reported in 75% of the colorectal cancers, and in 30% of the late adenomas but rarely in early adenomas, suggesting a role during the transformation of in situ to invasive carcinoma during the adenoma-carcinoma sequence (Monpezat et al, 1988; Vogelstein et al, 1988; Baker et al, 1989,

(14)

1990). In concordance with the properties of tumor-suppressor genes, the normal function of p53 is disrupted in colorectal carcinomas mostly as a result of loss of one 17p allele, coupled with a mutation of the remaining copy of p53 (Baker et al, 1989, 1990). However, in some cases p53 mutant protein has a dominant-negative effect, due to formation of tetramers by mutant and wild-type molecules (Vogelstemen/., 1992). Disruption of the p53 pathway leads to increased cell proliferation with fixation of the genotoxic damage into the DNA and to clonal expansion of the tumor cells.

The tumor-suppressor genes on chromosome 18q.

Loss of 18q occurs in 73% of colorectal cancer and in 47% of late adenomas (Thiagalingam et al, 1996). Several (candidate) tumor-suppressor genes have been identified in this region and recently the importance of the

Smad-genes on 18q was discovered. Mutations in Smad2 (also known as

DPC4) and Smad4 (also known as JV18-1, MADR2, and hMAD-2) were reported in approximately 7% and 20% of the colon carcinomas, respectively (Riggins et al, 1996; Eppert et al, 1996). Inactivating mutations in the Smad genes are postulated to disrupt the growth inhibitory signal transduction pathway of the TGF-ßll receptors (Liu et al, 1996; Yingling et al, 1996). The TGF-ßll receptor gene has a poly-A-repeat and therefore, disruption of this pathway is frequently found in colorectal tumors with microsatellite instability. This observation supports the idea that the TGF-ßll receptor is a critical target gene of inactivation in mismatch-repair deficient tumors (Parsons etal, 1995).

Mismatch-repair genes

Errors in DNA replication during the S-phase result in single base pair mismatches or mispaired loops that occur at repetitive sequences such as microsatellites. These errors are repaired by mismatch-repairgenes. Disruption of the repair mechanism, due to mutations in these genes, result in accumulation of other somatic genetic mutations with an acceleration of the carcinogenic process. Patients with an inherited mutation of these mismatch-repair genes will develop HNPCC with formation of colorectal tumors distinct from the ones as initially described in the Vogelgram.

(15)

The APC tumor-suppressor gene.

The previously described genetic alterations accompany the adenoma-carcinoma sequence but are not a prerequisite for this process. Mutations involving components of the Wnt-Wingless signaling cascade i.e. APC and

ß-catenin (Fig. 1, Fig.2), however, appear to be a precondition for the initial

neoplastic transformation of colon epithelium. The APC gene, located on chromosome 5q, is mutated in 80% of sporadic intestinal tumors, whereas mutations of ß-catenin are found in most of the remaining tumors. The

APCIß-catenin mutations are already present in early adenomas. Familial adenomatous

polyposis (FAP) patients, individuals with a germline mutation in the APC gene, develop multiple colorectal adenomas (Powell et ai, 1992; Ishii et ai, 1992; Levy et ai, 1994; Vogelstein et ai, 1988), with virtually 100% chance of malignant transformation after a few decades. Like FAP patients, APC

163 8N and Min mice that are heterozygous for an A PC mutation at codon 163 8 and 850, respectively, will develop multiple adenomas of the intestine, following deletion of the remaining wild type allele of APC, thus confirming the role of APC as the gatekeeper in colorectal cancer (Smits et ai, 1997; Moser et ai, 1995; Luongo et ai, 1994; Levy et ai, 1994; Ishii et ai, 1992).

APC is part of the Wnt- pathway (Fig. 2) (Peifer et ai, 1996), the human homologue of the Wingless (WG)-pathway in Drosophila. One of the molecules that interact with APC in this pathway is ß-catenin (Rubinfeld et ai,

1993; Behrens et ai, 1996). ß-Cateninwas originally identified on the basis of its association with Cadherin adhesion molecules (Birchmeier et ai, 1994), but is now also recognized as an essential component of the Wnt-Wingless cascade. Under physiological circumstances APC and ß-catenin form a complex and the activity of ß-catenin as a signal transducer, is tightly regulated by this binding. During intestinal carcinogenesis,however, as the result of APC (or ß-catenin) mutations, this complex is no longer formed and large amounts of free ß-catenin are available. This ß-catenin will bind the transcription factor TCF-4 and the ß-catenin/TCF complex will enter the nucleus, resulting in uncontrolled transcription of TCF-4 target genes (Korinek et ai, 1997; Gumbiner et ai, 1990; Behrens et ai, 1996; Molenaar et ai, 1996). The mechanisms by which deregulation of the Wnt-pathway induces cancer, even

(16)

The Cell

Figure 2. The Wnt-pathway. In this cascade, ß-catenin functions as a transciptional

co-activator when complexée! with members of the Tcf family of DNA binding proteins. The amount of ß-catenin, however, is regulated by binding of APC and subsequent degradation. On the left, the Wnt-pathway is shown under physiological conditions. In the non-activatedpathway, ß-catenin is bound to APC and phosphorylation of APC by GSK3 ß results in degradation of ß-catenin. On the right side, the Wnt-pathway is constitiutively activated as in colorectal cancer. Due to mutations in either APC or in

ß-catenin, the APC/ß-catenin complex is not formed resulting in uncontrolled amounts

of nuclear complexes between co-activator ß-catenin and Tcf-4 with transcription of Tcf-4 target genes in intestinal epithelium.

as the target genes of the ß-catenin/Tcf-4 complex, are still largely unknown. However, transfection of mutated APC was shown to inhibit apoptosis of the transfected cells (Morin et al., 1996) and recent studies have identified c-MYC as potential target gene (He et al., 1998).

(17)

Dukes A

mucosa

submucosa

Figure 3. The Dukes' stages. Dukes A: the carcinoma is confined to the bowel wall. Dukes B: malignant cells penetrate through the muscularis propria. Dukes C: malignant cells metastasize to regional lymph nodes. Dukes D: malignant cells metastasize to

The morphological changes in colorectal cancer.

The result of these genetic alterations, is the evolution of the colorectal epithelium through a series of morphologically well described neoplastic lesions. The first morphologically detectable anomaly of the mucosa is the dysplastic aberrant crypt focus (ACF) (Nucci et al., 1997). Impaired maturation of the dysplastic epithelium leads to a disturbed architecture with atypical cells present throughout the crypt, up to the mucosal surface. When more crypts become involved, the mucosal surface will develop a polyploid aspect resulting in the formation of an adenomatous polyp. When the tumor cells penetrate the basement membrane, by definition^ carcinoma has developed. The subsequent extension of the carcinoma can be separated in distinctive stages as originally described by CE. Dukes in 1932 (Fig.3). Initially the carcinoma will be

(18)

confined to the bowel wall (stage Dukes A). When the carcinoma cells penetrate through the muscularis propria (stage Dukes B) they eventually metastasize, first to regional lymph nodes (stage Dukes C) and finally to distant organs (stage Dukes D).

THE

CD44

GENE AND ITS PROTEIN PRODUCTS.

The oncogenes and tumor suppressor genes, previously mentioned, are all involved in the regulation of tumor growth and/or apoptosis. The main cause of cancer related death, however, is not primary tumor growth, but formation of distant metastases. Although relatively little is known about molecular mechanisms underlying this complicated process, a large body of studies indicate an important role of CD44 in metastasis (Lesley et al, 1993; Herrlich et al, 1993; Naor étal, 1997).

CD44 was originally described as a homing receptor on lymphocytes, mediating lymphocyte interactions with high endothelial venules (HEV) (Jalkanen et al, 1986, 1987). Metastasizing tumor cells and recirculating (activated) lymphocytes share several properties including invasive behavior, migration involving reversible adhesive contacts, accumulation and expansion in draining lymph nodes, release into the circulation, and adhesion to vascular endothelium and extravasation. This analogy between lymphocyte recirculation and tumor dissemination prompted the hypothesis that malignant cells might "misuse" molecules like CD44 for metastasis formation (Herrlich et al., 1993). Support for this hypothesis has come from experimental studies in laboratory animals showing a causal role of specific CD44 isoforms in metastasis formation (Güntherte/ al, 1991), as well as from clinical studies documenting deregulated CD44 expression in human cancer, including colorectal cancer (Lesley et al, 1993; Herrlich et al, 1993; Koopman et al, 1993; Naor et al,

1997; Matsumura et al, 1992; Heider et al, 1993; Tanabe et al, 1993 Wielengae/a/., 1993; Finn et al, 1994; Kim et al, 1994; Mulder et al, 1994 Kaufmann et al, 1995; Orzechowski et al, 1995; Rodriguez et al, 1995 Imazeki et al, 1996; Yamaguchi et al, 1996).

(19)

The structure of CD44.

All CD44 family members are encoded by a single gene on chromosome 11 p 13 that consists of 19 exons (Fig.4) (Stamenkovic et al., 1989; Dougherty et al, 1991 ; Günthert et al, 1991 ; Screaton et al., 1992; Lesley et

al., 1993). They share the N-terminal cartilage link protein homology domain,

that binds hyaluronic acid (HA) (Aruffo et al., 1990; Stamenkovic et al., 1989; Peach et al, 1993), as well as the C-terminal (transmembrane, cytoplasmic) domains encoded by exon 1-5 and exon 15-19, respectively. However, they differ in the extracellular membrane proximal part as a result of extensive alternative splicing of exons 6-14 (also referred to as exons v2-v 10) (Dougherty

etal, 1991; Günthert et al, 1991; Screaton étal, 1992; Tölg étal, 1993). In

this way, more than a thousand CD44 variants (CD44v) can be potentially generated. Further diversity of CD44 results from post-translational modifications with N-and O-linked sugars and with glycosaminoglycan(GAG) side chains like chondroitin- and heparan-sulfate. The CD44 cytoplasmic domain associates with the actin cytoskeleton via ankyrin, as well as via the ERM-family proteins ezrin, radixin, and moesin (Bourguignon et al, 1993; Tsukita et al., 1994; Isacke et al, 1994). In T-lymphocytes, CD44 is physically

and functionally associated with the Src-family tyrosine kinase p56lck(Taher et

al, 1996).

The expression of CD44.

CD44 is a family of type I transmembrane glycoproteins that is widely expressed on a variety of cells including cells of epithelial, mesenchymal, and hematopoietic origin. The expression of CD44 isoforms is tissue specific, for example, the shortest isoform of CD44 (CD44s), which lacks v2-vl0, is themost common form on hematopoietic cells, while larger CD44 splice variants dominate on several normal and on neoplastic epithelia. In normal colorectal epithelium, for example, CD44 expression is low and confined to the base of the crypt, while during malignant transformation, the expression is greatly enhanced (Heider et al, 1993; Lesley et al, 1993; Wielenga et al,

1993; Fox et al, 1994). CD44 splice variants are also found on activated lymphocytes and malignant lymphomas (Arch et al, 1992; Koopman et al,

(20)

Cartilage link protein homology domain

Growth factors: HGF/SF FGF

P3 a a) 'JK% Plasma mantrane •

Figure 4. Schematical representation of the CD44 gene and its encoded proteins. The

extracellular domain and cytoplasmic tail of CD44 isoforms vary in size as the result of alternative splicing. The alternative spliced exons are indicated by open boxes. The human vl exon contains a stop codon. In the model of the potein, all putative glycosylation sites are indicated: 0-glycosylation(open circles);yV-glycosylation(closed circles); chondroitin sulfate (open squares); heparan sulfate chain (rod). As indicated, the heparan sulfate binding site in exon v3 has the ability to bind growth factors. In addition, the HA-binding sites (double lines); the disulfide bonds (S-S); the ankyrin binding site (...); the ezrin binding sites (—); the phosphorylation sites (P); and the putative interaction sites for SRC-family kinases, are indicated.

(21)

Factors that regulate expressionand alternative splicing of CD44 during physiological and non-physiological circumstances are still largely unknown. Activation of the AP-1 binding site in the CD44 promoter has been shown to induce CD44 expression (Lamb et al, 1997; Kogerman et al, 1996). CD44 overexpression was furthermore described in a embryonic intestinal cell line as result of ras and src transfections (Jamal et al, 1994). Finally, several growth factors were reported to induce upregulationof CD44v, as for example TGF-1 and PDGF with involvement of PI 3-kinase and PKC (Fichter et al,

1997) and HGF/SF (Hiscox et al, 1997). In epithelial cell lines, alternative splicing of CD44 is positively regulated by Trans- acting factors (König et al,

1996).

Functions of CD44.

CD44 is involved in a number of important biological processes, including embryogenesis (Wheatley et al, 1993 ; Ruiz et al. ,1995; Sherman et

al, 1998) hematopoiesis, angiogenesis (Trochon et al, 1996), lymphocyte

homing and activation (Arch et al, 1992; Koopman et al, 1990; Naujokas et

al., 1993), survival from apoptosis (Ayroldi et al, 1995), and tumor metastasis

(Lesley et al, 1993; Herrlich et al, 1993; Naor et al, 1997). Importantly, a number of studies have indicated that CD44 can function as a signal-transducing receptor. In lymphocytes, outside-in signaling through CD44 costimulates antigen-specific lymphocyte activation and proliferation. Furthermore, CD44 engagement leads to activation of integrins on the lymphocyte cell-surface (Miyakae/ al, 1990; Haynes etal, 1989; Shimizu et

al, 1989; Koopman et al, 1990).

A major obstacle towards a better insight into the functions of the heterogenous CD44 family is the thus far limited number of ligands that has been identified, the only extensively studied interaction being that between CD44 and hyaluronate (HA) (Lesley et al, 1993). CD44 acts as a major receptor for this glycosaminoclycan (GAG) that is abundant in the ECM of mesenchymal tissues and that is believed to play a regulating role in cell migration (Lesley et al, 1993; Knudson et al, 1993; Aruffo et al, 1990). Although the binding site for HA is located on the constant N-terminal part of CD44, present on all isoforms, HA-binding capacity is not a constant feature

(22)

of CD44, but is subject to complex regulation by mechanisms involving both alternative splicing, modulation of cytoskeletal interaction, and posttranslational modification of CD44 (Aruffo et al, 1990; Culty et al, 1990; Lesley et al, 1992; Bourguignon et al, 1993; Bennett et al, 1995a; Katoh et

al, 1995; Stamenkovic étal, 1991; Takahashi étal, 1996; Uffetal., 1995;

van der Voort et al, 1995; Liu et al, 1996; Sleeman et al, 1996; Pure et al, 1995) By altering cellular interactions with HA, CD44 may facilitate tumor metastasis at least at two distinct levels: It may promote cell migration through the ECM. Alternatively, it may facilitate rolling of tumor cells on HA expressed on the surface of vascular endothelium at inflammatory sites and thereby enhance extravasation (DeGrendele et al, 1996, 1997). Indeed, in studies by Bartolazzi et al (1994), HA binding of melanoma cell lines was reported to be essential for the metastasis promoting effect of CD44s. However, properties other than HA binding must also play a role, as the HA-binding capacity and metastatic potential of pancreatic carcinoma cell lines were not related (Sleeman et al, 1996). Other molecules that have been reported to interact with CD44 are collagen IV, fibronectin, serglycin, invariant chain, and osteopontin, (Jalkanen et al, 1992; Lesley et al, 1993; Toyama et

al, 1995; Weber et al, 1996). The role of CD44 in tumorigenesis.

Initial evidence for a role of CD44 in cancer comes from studies in human non-Hodgkin's lymphomas. In this disease, the expression of CD44 was found to be associated with tumor dissemination and unfavorable prognosis (Pals et al, 1989; Horst et al, 1990; Jalkanen et al, 1991; Stauder et al, 1995). In a study by Günthert et al. in 1991, specific CD44 splice variants containing exon v6 conferred metastatic potential to a non-metastatic rat pancreatic carcinoma cell line. This observation catalyzed a large number of studies of CD44 splice variant expression in a variety of epithelial malignancies, including colorectal cancer, breast cancer, cervical cancer and bladder cancer. In many epithelial cancers, increased levels of CD44 and/or different patterns of splice variants were found in tumors in comparison with their normal counterparts (Matsumura et al, 1992 ; Heider et al, 1993 ; Herrlich

(23)

Wielenga et al, 1993; Dali et al, 1994; Finn et al, 1994; Kim et al, 1994; Mulder ef a/., 1994; Coopère/al, 1995; Kaufmänner a/., 1995; Orzechowski

étal, 1995; Rodriguez et al, 1995; Stauder étal, 1995; Imazeki étal, 1996;

Yamaguchi et al., 1996). In colorectal cancer, as well as in breast cancer, enhanced CD44 expression was reported to be associated with unfavorable prognosis (Wielenga et al, 1993; Mulder et al, 1994; Kaufmann et al, 1995; Yamaguchi et al, 1996; Ropponen et al, 1998). It identifies patients with a high propensity to develop metastases, who may benefit from adjuvant therapy. The explanation for the promoting activity of CD44 on the process of tumor dissemination remains unclear but, as mentioned before, an altered tumor cell-ECM interaction could be responsible. Furthermore, recent studies have identified a novel function for CD44 that might be highly significant for tumorigenesis, i.e. binding and presentation of growth factors.

Interaction of CD44 with growth factors.

CD44 can be modified by both chondroitin- and heparan-sulfate side chains and is thus a "facultative" cell-surface proteoglycan (Jalkanen et al,

1988; Stamenkovic et al, 1989; Brown et al, 1991; Faassen et al, 1992; Screaton et al., 1992; Lesley et al., 1993). Recent studies have shown that these modifications are associated with CD44 isoforms containing the v3 alternative exon (Jackson et al, 1995), which encodes a consensus motif SGSG for glycosaminoclycan (Bourdon et al, 1987; Greenfield et al, 1999) addition, which is evolutionary conserved between rodents and man (Screaton et al,

1992; Tölg et al, 1993). Proteoglycans are believed to play an important regulatory role in cell growth and motility (Kjellén et al, 1991; Ruoslahti et

al, 1991; Schlessinger et al, 1995). They can bind growth factors via their

GAG side chains and target these factors to their high affinity signal transducing receptors. This process has been particularly well explored for fibroblast growth factor-2 (FGF-2). Binding of FGF-2 to the low affinity proteoglycan receptor on the cell-surface presumably allows more frequent encounters with the high affinity receptor and, furthermore, formation of a multivalent FGF-proteoglycancomplexmay be required for dimerizationof the high affinity signaling receptors (Ruoslahti et al, 1991; Schlessinger et al, 1995; Yayon et al, 1991). Interestingly, specific structural modifications of

(24)

heparan sulfate side chains appear to determine their affinity for a given heparin-binding growth factor. This creates a mechanism for cell or tissue selective growth factor binding (David et al, 1993; Lindahl et al, 1998; Tanaka et al, 1998). Altered composition and expression of cell-surface proteoglycans has been reported in tumors and has been implicated in tumor invasion and metastasis (Kjellén et al, 1991; David et al, 1993; van Muijen

étal, 1995).

As for CD44, Tanaka and colleagues have shown that heparan sulfate proteoglycan forms isolated from monocytes can present the chemokine macrophage inflammatory protein-lb to T-lymphocytes resulting in activation of T-cell integrins (Tanaka etal, 1993). Subsequent studies from Bennett and colleagues demonstrated that GAG-modified CD44 splice variants can bind FGF-2 (Bennett et al, 1995b), and presentation of FGF-2 by CD44v-HS expressed on the apical epidermal ridge appears to be involved in limb bud formation during embryogenesis (Sherman et al, 1998).

THE HGF/SF-C-MET-SIGNALING PATHWAY.

Among other molecules that have been implicated in tumor invasion and metastasis are c-Met and its ligand HGF/SF (Liu et al, 1992; Yamashita

et al, 1994; Boros et al, 1995; Di Renzo et al, 1995; Tuck et al, 1996).

HGF/SF induces epithelial cells to invade collagen matrices in vitro, and NIH-3T3 cells cotransfected with c-met and HGF/SF acquire an invasive and metastatic phenotype (Rong et al, 1992, 1994; Giordano et al, 1993). Furthermore, in HGF/SF-transgenicmice, tumorigenesis was reported in many different tissues including mammary glands, skeletal muscles, and melanocytes (Liang et al, 1996; Takayama et al, 1997). In human cancer, both HGF/SF and c-Met are often overexpressed, and germline mutations in the c-met gene have recently been reported in hereditary renal cancer (Schmidt et al, 1997). These mutations lead to an enhanced enzymatic activity of the c-Met tyrosine kinase (Jeffers et al, 1996a ).

The structure of c-Met and HGF/SF.

(25)

encodes a receptor tyrosine kinase of 190 kDa (Fig. 5) (Park et al, 1986). c-Met is synthesized as a 170 kDa precursor that is cleaved and glycosylated to give rise to a heterodimer (Giordano et al., 1989). This mature form of the receptor is composed of two disulfide-linked chains, i.e., an extracellular 50 kDa a-subunit and a transmembrane 145 kDa ß-a-subunit (Giordano et al, 1989) that is endowed with tyrosine kinase activity (Park et al, 1987, Giordano et al,

1988). Binding of the ligand HGF/SF results in activation of c-Met.

HGF/SF is a heparan-binding glycoprotein that consists of a 60 kDa a-chain and a 30 kDa ß-a-chain, linked by disulfide bonds (Fig.6) (Gherardi et al,

1989). HGF/SF belongs to the family of kringle proteins, characterized by triple disulfide loop structures (kringles) that mediate protein/protein interactions(Nakamuraet al, 1986). Like c-Met, HGF/SF is synthesized as a single chain precursor that is cleaved extracellularly by serine proteases including coagulation factor XII and HGF/SF activator (Miyazawa et al, 1993, 1996). In addition to the high affinity receptor c-Met, HGF/SF also binds to heparan sulfate (HS) proteoglycans on cell surfaces and in the extracelluar matrix (ECM). This binding, and possible sequestering and presentation of HGF/SF by HS- proteoglycans to c-Met, may enhance the activation of the HGF/SF - c-Met pathway (Zioncheck et al, 1995).

The expression of c-Met and HGF/SF.

Wereas c-Met is expressed on a variety of, predominantly epithelial, cells and is overexpressed on several carcinomas, HGF/SF is secreted by mesenchymal cells, like fibroblasts, vascular smooth muscle cells, and glial cells, as well as by macrophages and activated T-lymphocytes (Naidu et al,

1994, Rosen et al, 1990, 1994; Stoker et al, 1987). c-Met is expressed on normal intestinal epithelium and c-Met protein and mRNA are upregulated in colorectal tumorigenesis(Di Renzo et al, 1991,1995; Liu et al., 1992; Prat et

al, 1991). Inflammatory cytokines like II-la and II-Iß enhance HGF/SF

production by fibroblasts (Tamura et al, 1993), whereas TGF-ß inhibits the production (Gohda et al, 1992). Activation of the oncogenes ras and ret, may induce deregulated c-Met expression in carcinomas (Ivan et al, 1997).

(26)

Functions of the c-Met-HGF/SF pathway.

The HGF/SF-c-Met pathway induces growth and motility of target epithelial cells, endothelial cells, myoblasts, lymphocytes, and tumor cells (Stoker et al, 1987; Bottaro ef a/., 1991; Bussolino etal, 1992; Tajima et al,

1992; Weidner et al, 1990,1993; Donated al, 1994;Bladte/La/., 1995;Boros

et al., 1995 ; Brinkmann et al, 1995 ; Van der Voort et al, 1997). The pathway

is a key mediator of in mesenchymal-epithelial interactions that take place during embryogenesis (Wang et al, 1994; Sonnenberg et al, 1993), wound healing (Dignass et al, 1994), liver regeneration (Nakamura et al., 1986), and tumorigenesis (Tuck et al, 1996; Liuetal, 1992; Furukawa et al, 1995;Prat

et al, 1991; Di Renzo et al, 1991, 1995; Ebert et al, 1994). During these

processes, the c-Met pathway directs cells toward motility ("scattering"), by inducing the phosphorylation of ß-catenin and by downregulating cadherin-mediated adhesion (Shibamoto et al, 1994). Furthermore, it may mediate proliferation, and/or morphogenesis. These activities are not mutually exclusive: Studies using chimeric receptors containing the extracellular domains of other receptors fused to the cytoplasmic part of c-Met, demonstrate that c-Met can convey multiple biological functions (Weidner et al, 1993). Activation of the signaling pathways that propagate these different activities is determined at the receptor level by differential binding of cytoplasmic signaling proteins to phosphotyrosines in the cytoplasmic domain of c-Met (Koch et al, 1991; Ponzetto et al., 1994), or may be regulated more downstream.

HGF/SF induces intrinsic, tissue-specific morphogenic activities in a wide variety of epithelial cells that are grown in three-dimentional matrices. It induces colon cell lines to form crypt-like structures in (Brinkmann et al,

1995), and mammary epithelial cell lines (Brinkmann et al., 1995) and cultured mouse mammary glands (Yang et al, 1995) to form long branching ducts. Morphogenic activities of epithelial cells were reported to require ezrin, a cytoskeletal component that links the cell membrane to F-actin (Crepaldi et al,

1997). During mouse embryogenesis, c-Met mRNA is expressed in epithelial cells of various organs, while HGF/SF is present in surrounding mesenchymal tissue (Sonnenberg et al., 1993 ; Iyer et al., 1990; Kolatsi et al., 1997). HGF/SF as well as c-Met mutant mice show severe defects in liver development,

(27)

oc-chain

ß-chain

TK

DS

Figure 5. The structure of the receptor tyrosine kinase c-Met. The receptor is composed

of two disulfide- linked chains: an extracellular50 kDa a-subunit and a transmembrane 145 kDa ß-subunit. C-Met is synthesized as a 170 kDa precursor that is cleaved and posttranslationally glycosylated to give rise to the mature heterodimer with total

leading to death at E15, and in limb bud formation (Bladt et al., 1995; Schmidt

et al, 1995). The latter defect is caused by defective migration of myogenic

precursor cells.

The involvement of HGF/SF- c-Met in tumorigenesis was shown in several studies. For example, c-Met was isolated originally as the product of a human oncogene, Tpr-met, which encodes a consitutively dimerized/activated chimeric c-Met protein possessing transforming activity (Cooper et al, 1984; Rodrigues et ai, 1993). The generation of an autocrine loop as a result of

(28)

OC-chain

ß-chain

Figure 6. The structure of the HGF/SF heparin-binding growth factor. HGF/SF is a

glycoprotein that consists of a 60 kDa a-chain and a 30 kDa ß-chain, linked by disulfide bonds with triple disulfide loop structures (kringles) that mediate protein/protein and

coexpression of wild-type c-Met and HGF/SF molecules in the same cell is also oncogenic (Jeffers et al, 1996b). The tumorigenecity of both Tpr-Met and autocrine HGF/SF-Met signaling has been verified in transgenic mouse models, which develop tumors in many different tissues including mammary glands, skeletal muscles and melanocytes (Liang et al, 1996; Takayama et al. 1997). c-Met activationhas also been shown to promote the metastatic spread of cancer, a finding that most likely is due to its stimulatory effects on a variety of processes such as angiogenesis, cell motility, and protease secretion (Jeffers

et al, 1996a). Recently, missense mutations in c-Met were found to be

(29)

mutations deregulate the enzymatic activity of the receptor, thereby unleashing its oncogenic potential (Jeffers et al., 1997). Furthermore, the c-Met expression is upregulated in several carcinomas including those of the breast (Di Renzo

et ai, 1991; Tuck et al., 1996; Jin et al, 1997), kidney (Prat et al, 1991),

colorectum(DiRenzo^a/., 1991, 1995; Un et al, 1992; Prate/ al, 1991) and pancreas (Ebert et al, 1994; Furukawa et al, 1995). Congenital mutations in the tyrosine kinase domain of the c-Met gene, results in papillary renal carcinomas (Schmidt et al, 1997).

SCOPE OF THE THESIS

The aim of the work decribed in this thesis was to elucidate the role of CD44 in colorectal tumorigenesis and to assess the prognostic value of CD44 expression in patients with colorectal cancer.

CD44 splice variants play a causal role in the metastatic spread of cancer in animal models. In Chapter 2, we explored the expression of CD44 splice variants at the subsequent stages of colorectal tumor progression, i.e., in normal intestine, adenomas, and carcinomas. In Chapter 3, the relation between expression of the tumor suppressor gene p53 and CD44 splice variants containing v5 and v6 was studied.

We have previously provided evidence for an association of CD44v6 containing variants with tumor-related death in patients with colorectal cancer. In Chapter 4, we further investigated the prognostic significance of CD44 in relation to conventional prognosticators.

Overexpression of CD44 is an early event in the adenoma-carcinoma sequence. This suggests a link with disruption of APC tumor suppressor protein-mediated regulation of /?-catenin/Tcf-4 signaling, which is crucial in initiating tumorigenesis. In Chapter 5, we explore this hypothesis by analyzing CD44 expression in the intestinal mucosa of mice and humans with genetic defects in either APC or Tcf-4, leading to constitutive activation or blockade of the /î-catenin/Tcf-4 pathway, respectively.

The mechanism(s) by which CD44 promotes tumor growth and/or metastasis, is as yet poorly understood. Recent studies have shown that CD44 isoforms containing the alternatively spliced exon v3 carry heparan sulfate side

(30)

chains (HS) and are able to bind heparin-binding growth factors. In Chapter 6, we have explored the possibility of a physical and functional interaction between CD44 and hepatocyte growth factor/scatter factor (HGF/SF), the ligand of the receptor kinase c-Met, since this pathway plays a key role in invasion and metastasis. In Chapter 7, we studied the expression of c-Met, HGF/SF and CD44-HS, along the adenoma-carcinoma sequence.

In Chapter 8 the literature on CD44 in colorectal cancer is reviewed and a model for CD44 functioning in colorectal tumorigenesis is proposed.

(31)

REFERENCES

1. American Cancer Society. Cancer Facts & Figures-1994. Atlanta GA: American Cancer Society Inc., 1994

2. Arch R, Wirth K, Hofmann M, Ponta H, Matzku S, Herrlich P, Zöller M: Participation in normal immune responses of a metastasis-inducing splice variant of CD44. Science (Wash. DC) 257: 682-685, 1992 3. Aruffo A., StamenkovicI, Melnick M, Underhill CB, Seed B: CD44 is

the principal cell surface receptor for hyaluronate. Cell 61: 1303-13, 1990

4. Ayroldi E, Cannarile L, Migliorati G, Bartoli A, Nicoletti I, Riccardi C: CD44 (Pgp-1) inhibits CD3 and Dexamethasone-induced apoptosis. Blood 86: 2672-2678, 1995

5. Baker SJ, Fearon ER, Nigro JM, Hamilton SR, Preisinger AC, Jessup JM, van Tuinen P, Ledbetter DH, Barker DF, Nakamura Y, White R & Vogelstein B: Chromosome 17p deletions and p53 gene mutations in colorectal carcinomas. Science 244: 217-221, 1989

6. Baker SJ, Preisinger AC, Jessup M, Paraskeva C, Markowitzs S, Willson JKV, Hamilton SR & Vogelstein B: p53 gene mutations occur in combination with 17p allelic deletions as late events in colorectal tumorigenesis. Cancer Res 50: 7717-7722, 1990

7. Bartolazzi A, Peach R, Aruffo A, Stamenkovic I: Interaction between CD44 and hyaluronate is directly implicated in the regulation of tumor development. J Exp Med 180: 53-66, 1994

8. Behrens J, von Kries JP, Kühl M, Bruhn L, Wedlich D, Grosschedll R, Birchmeier W: Functional interaction of ß-catenin with the transcription factor LEF-1. Nature 382: 638-642, 1996

9. Bennett KL, Modrell B, Greenfield B, Bartolazzi A, Stamenkovic I, Peach R, Jackson D, Spring F, Aruffo A: Regulation of CD44 binding to hyaluronate by glycosylation of variable spliced exons. J Cell Biol 131: 1623-1933, 1995a

10. Bennett KL, Jackson DG, Simon JC, Tanczos E, Peach R, Modrell B, Stamenkovic I, Plowman G and Aruffo A: CD44 isoforms containing exon v3 are responsible for the presentation of heparin-binding growth

(32)

factor. J Cell Biol 128:687-698, 1995b

11. Birchmeier WKM, Semin Cancer Biol 4: 231-239, 1993

12. Birchmeier W, Behrens J: Cadherin expression in carcinomas ; role in the formation of cell junction and the prevention of invasiveness. Biochem Biophys Acta 1198: 11-26, 1994

13. Bladt F, Riethmacher D, Isenmann S, Aguzzi A, Birchmeier C: Essential role for the c-Met receptor in the migration of myogenic precursor cells into the limb bud. Nature 376: 768-771, 1995

14. Boros P and Miller CM: Hepatocyte growth factor: a multifunctional cytokine. Lancet 345: 293-295, 1995

15. Bottaro DP, Rubin JS, Faletto DL, Chan AML, Kmiecik TE, Woude GF van de, Aaronson SA: Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science 251 : 802-804

1991

16. Bourdon MA, Krusius T, Campbell S, Schwartz NB and Ruoslahti E: Identification and synthesis of a recognition signal for the attachment of glycosaminoglycansto proteins. Proc Natl Acad Sei USA 84- 3194-3198, 1987

17. Bourguignon LYW, Lokeshwar V, Chen X, Kerrick GL: Hyaluronic acid-induced lymphocyte signal transduction and HA receptor (GP85/CD44)-cytoskeleton interaction. J Immunol 151: 6634-6644 1993

18. Bourne HR, Sanders DA, McCormick F: The GTPase super family: Conversed structure and molecular mechanism. Nature 349-117-127 1991

19. Brinkmann V, Foroutan H, Sachs M, Weidner KM, Birchmeier W: Hepatocyte growth factor/scatter factor induces a variety of tissuespecific morphogenic programs in epithelial cells. J Cell Biol 131

-1573-1586, 1995

20. Brown T, Bouchard T, St. John T, Wayner E, Carter WG: Human keratinocytes express a new CD44 core protein (CD44E) as a heparan-sulfate intrinsic membrane proteoglycan with additional exons. J Cell Biol 113:207-221, 1991

(33)

L, Gaudino G, Tamagnone L, Coffer A, Comoglio PM: Hepatocyte growth factor is a potent angiogenic factor which stimulates endothelial cell motility and growth. J Cell Biol 119: 629-641, 1992

22. Coleman MP, Esteve J, Damiecki P, Arslan A, Renard H: Trends in cancer incidence and mortality. Lyon: IARC Scientific Publications No. 121, 1993

23. Cooper CS, Park M, Blair DG, Tainsky MA, Huebner K, Croce CM, Vande Woude GF: Molecular cloning of a new transforming gene from a chemically transformed human cell line. Nature 311:29-33, 1984 24. Cooper DL and Dougherty GJ: To mestastasize or not? Selection of

CD44 splice sites. Nature Med 1: 635-637, 1995

25. Crepaldi, T., A. Gautreau, P.M. Comoglio, D. Louvard, and M. Arpin: Ezrin is a effector of hepatocyte growth factor-mediated migration and morphogenesis in epithelial cells. J Cell Biol 138: 423-434, 1997 26. Culty M, Miyake K, Kincade PW, Sikorski E, Butcher EC, Underhill

CB: The hyaluronate receptor is a member of the CD44 (H-CAM) family of cell surface glycoproteins. J Cell Biol 111: 2765-2774, 1990 27. Dall P, Heider KH, Hekele A, von Minckwitz G, Kaufmann M, Ponta

H, Herrlich P: Surface protein expression and messenger RNA-splicing analysis of CD44 in uterine cervical cancer and normal cervix epithelium. Cancer Res 54: 3337-3341, 1994

28. Dignass AU, Lynch-Devaney K, Podolsky DK: Hepatocyte growth factor/scatter factor modulates intestinal epithelial cell proliferation and migration. Biochem Biophys Res Comm 202: 701-709, 1994

29. David G: Integral membrane heparan sulfate proteoglycans. Faseb J 7: 1023-1030, 1993

30. DeGrendele HC, Estess P, Picksr LJ, Siegelman MH: CD44 and its ligand hyaluronate mediate rolling under physiological flow: a novel lymphocyte-endotheliail cell primary adhesion pathway. J Exp Med

183: 1119-1130, 1996

31. DeGrendele HC, Estress P, Siegelman MH: Requirement for CD44 in activated T cell extravasation into an inflammatory site. Science 278: 672-675, 1997

(34)

E, Gaglia P, Zara P, Comoglio PM: Expression of the Met/HGF receptor in normal and neoplastic human tissues. Oncogene 6:

1997-2003, 1991

33. Di Renzo MF, Olivero M, Giacomini A, Porte H, Chastre E, Mirossay L, NordlingerB, Bretti S, Bottardi S, Giordano S, Plebani M, Gespach C and Comoglio PM: Overexpression and amplification of the

Met/HGF receptor gene during the progression of colorectal cancer.

Clin Cancer Res 1: 147-154, 1995

34. Donate LE, Gherardi E, Srinivasan N, Sowdhamini R, Aparicio S, Blundell TL: Molecular evolution and domain structure of plasminogen-related growth factors (HGF/SF and HGF1 /MSP). Protein Science 3: 2378-2394, 1994

35. Dougherty GJ, Lansdorp PM, Cooper DL, Humphries RK: Molecular cloning of CD44R1 and CD44R2, two novel isoforms of the human CD44 lymphcyte "homing" receptor expressed by hemopoietic cells. J Exp Med 174:1-5, 1991

36. Dukes CE: The classificationof cancer of the rectum. J Pathol Bacteriol 35:323-332, 1932

37. El-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM, Lin D, Mercer WE, Kinzler KW, Vogelstein B : WAF 1, a potential mediator of p53 tumor suppression. Cell 75: 817-825, 1993

38. Ebert M, YokoyamaM, Friess H, Buchler MW, Korc M: Coexpression of the c-met proto-oncogene and hepatocyte growth factor in human pancreatic cancer. Cancer Res 54: 5775-5778, 1994

39. Eppert K, Scherer SW, Ozcelik H, Pirone R, Hoodless P, Kim H, Tsui LC, Bapat B, Gallinger S, Andrulis IL, Thomsen GH, Wrana L, Attisano L: MADR2 maps to 18q21 and encodes a TGF-ß-regulated MAD-related protein that is mutated in colorectal carcinoma. Cell 86-543-552, 1996

Faassen AE, Schräger JA, Klein DJ, Oegema TR, Couchman JR and McCarthy JB: A cell surface chondroitin sulfate proteoglycan, immunologically related to CD44, is involved in type I collagen-mediated melanoma cell motility and invasion. J Cell Biol 116: 521-531,1992

(35)

41. Fearon ER, Vogelstein B: A genetic model for colorectal tumorigenesis. Cell 61:759-767, 1990

42. Fichter M, Hinrichs R, Eissner G, Scheffer B, Classen S, Ueffing M: Expression of CD44 isoforms in neuroblastoma cells is regulated by PI 3-kinase and protein kinase C. Oncogene 14: 2817-2824, 1997 43. Finn L, Dougherty G, Finley G, Meisler A, Becich M and Cooper DL:

Alternative splicing of CD44 pre-mRNA in human colorectal tumors. Biochem Biophys Res Comm 200: 1015-1022, 1994

44. Fox SB, Gatter KC, Jackson DG, Screaton GR, Bell MV, Bell JI, Harris AL, Simmons D, Fawcett J: CD44 and cancer screening. Lancet 342:548-549, 1993

45. Furukawa T, Duguid WP, Kobari M, Matsuno S, Tsao MS: Hepatocyte growth factor and Met receptor expression in human pancreatic carcinogenesis. Am J Pathol 147: 889-895, 1995

46. Gherardi E, Gray J, Stoker M, Perryman M, Furlong R: Purification of scatter factor, a fibroblast-derived basic protein that modulates epithelial interactions and movement. Proc Natl Acad Sei USA 86: 5844-5848, 1989

47. Giordano S, Di Renzo MF, Narsimhan RP, Tamagnone L, Gerbaudo EV, Chiado-Piat L, Comoglio PM: Evidence for autocrine activation of a tyrosine kinase in a human gastric carcinoma cell line. J Cell Biochem 38: 229-236, 1988

48. Giordano S, Ponzetto C, Di Renzo MF, Cooper CS, Comoglio PM: Tyrosine kinase receptor indistinguishable from the c-met protein. Nature 339: 155-156, 1989

49. Giordano S, Zhen Z, Medico E, Gaudino G, Galimi F, Comoglio PM: Transfer of motogenic and invasive response to scatter factor/hepatocyte growth factor by transfection of human MET protooncogene. Proc Natl Acad Sei USA 90: 649-653, 1993

50. Gohda E, Matsunaga T, Kataoka H, Yamamoto I: TGF-ß is a potent inhibitor of hepatocyte growth factor secretion by human fibroblasts. Cell Biol Int Rep 16: 917-926, 1992

51. Greenfield B, Wang WC, Marquardt H, Piepkorn M, Wolff E, Aruffo A, Bennet Kl: Characterization of the heparan sulfate and chondroitin

(36)

sulfate assembly sites in CD44. J Biol Chem 22; 2511-2517, 1999

52. Greenson JKA Isenhart CE, Rice R, Mojsisik C, Houchens D, Martin

EW: identification of occult micrometastases in pericolic lymphnodes of Dukes' B colorectal cancer patients using monoclonal antibodies against cytokeratin and CC49. Cancer 73: 563-569, 1994

53. Gumbiner B: Generation and maintenance of epithelial cell polarity. Curr Opin Cell Biol 2 (5):881-887, 1990

54. Günthert U, Hofmann M, Rudy W, Reber S, Zöller M, Haußmann I, Matzku S, Wenzel A, Ponta H, Herrlich P: A new variant of glycoprotein CD44 confers metastatic potential to rat carcinoma cell lines. Cell 65:13-24, 1991

55. Harper JW, Adami GR, Wei N, Keyomarsi K, Elledge S J. The p21 Cdk-interacting protein Cip 1 is a potent inhibitor of Gl cyclin-dependent kinases. Cell 75: 805-816, 1993

56. Haynes BF, Telen MJ, Hale LP, Denning SM: CD44-A molecule involved in leukocyte adherence an T-cell activation. Immunol Today

10:423-428, 1989

57. He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B, Kinzier KW: Identification of c-Myc as a target of the APC pathway. Science 281:1509-1512, 1998

58. Heider KH, Horst E, Berg van den F, Hofmann M, Ponta H, Herrlich P, Pals ST: A human homologue of the rat metastasis-associatedvariant of CD44 is expressed in colorectal carcinomas and adenomatous polyps. J Cell Biol 120: 227-233, 1993

59. Herrlich P, Zöller M, Pals ST, Ponta H: CD44 splice variants: metastases meet lymphocytes. Immunol Today 14: 395-399, 1993 60. Hiscox S, Jiang WG: Regulation of endothelial CD44 expression and

endothelium-tumour cell interactions by hepatocyte growth factor/scatter factor. Biochem Biophys Res Comm 233: 1-5, 1997 61. Horst E, Meijer CJLM, RadaskiewiczT, Ossekoppele GJ, van Krieken

JHJM, Pals ST: Adhesion molecules in prognosis of diffuse large-cell lymphoma: expression of a lymphoid homing receptor (CD44), LFA-1 (CDll/CD18),andICAM-l.Leukemia4: 595-599, 1990

(37)

M: Expression of variant CD44-messenger RNA in colorectal adenocarcinomas and adenomatous polyps in humans. Gastroenterol

110:362-368, 1996

63. Isacke C. The role of the cytoplasmic membrane in regulating CD44 function. J Cell Science 107: 2353-2359, 1994

64. Ishii S, HoriiA, NakatsuruS, FuruyamaJ, UtsunomiyaJ, NakamuraY. Inactivationof both APC alleles in an early stage of colon carcinomas in a patient with familial adenomatous polyposis (FAP). Hum Mol Genetics 1,387-390, 1992

65. Ivan M, Bond JA, Prat M, Comoglio PM. Wynford-Thomas D. Activated ras and ret oncogenes induce over-expression of c-met (hepatocyte growth factor receptor) in human thyroid epithelial cells. Oncogene 14: 2417-2423, 1997

66. Iyer A, Kmiecik TE, Park M, Daar I, Blair D, Dunn KJ, Sutrave P, Ihle JN, Bodescot M, Vande Woude GF: Structure, tissue-specific expression, and transforming activity of the mouse met protooncogene. Cell Growth Diff 1: 87-95, 1990

67. Jackson DG, Bell JI, Dickinson R, Timans J, Shields J and Whittle N: Proteoglycan forms of the lymphocyte homing receptor CD44 are alternatively spliced variants containing the v3 exon. J Cell Biol 128: 673-685, 1995

68. Jalkanen S, Bargatze RF, Herron LR, Butcher EC: A lymphoid cell surface glycoprotein involved in endothelial cell recognition and lymphocyte homing in man. Eur J Immunol 16: 1195-1202, 1986 69. Jalkanen S, Bargatze RF, de los Toyos J, and Butcher EC: Lymphocyte

recognition of high endothelium: antibodies to distinct epitopes of an 85-95-kD glycoprotein antigen differentially inhibit lymphocyte binding to lymph node, mucosal, or synovial endothelial cells. J Cell Biol 105: 983-990, 1987

70. Jalkanen S, Jalkanen M, Bargatze R, Tammi M, Butcher EC: Biochemical property of glycoproteins involved in lymphocyte recognition of high endothelial venules in man. J Immunol 141:

1615-1623, 1988

(38)

and clinical behaviour of non-Hodgkin's lymphoma. J Clin Invest 87: 1835-1840, 1991

72. Jalkanen S, Jalkanen M. Lymphocyte CD44 binds the COOH-terminal heparin binding domain of fibronectin. J. Cell Biol. 116: 817-825,1992 73. Jamal HH, Cano-Gauci DF, Buick RN, Filmus J. Activated ras and src induce CD44 overexpressionin rat intestinal epithelial cells. Oncogene 9: 417-423, 1994

74. Jeffers M, Rong S, Vande Woude GF: Enhanced tumorigenicity and invasion-metastasis by hepatocyte growth factor/scatter factor-met signalling in human cells concomitant with induction of the urokinase proteolysis network. Molec Cell Biol 16: 1115-1125, 1996a

75. Jeffers M, Rong S, Anver M, Vande Woude GF : Autocrine hepatocyte growth factor/scatter factor-Met signaling induces transformation and the invasive/metastastic phenotype in CI27 cells. Oncogene

13(4):853-856, 1996b

76. Jeffers M, Schmidt L, NakaigawaN, Webb CP, Weirich G, Kishida T, Zbar B, Vande Woude GF: Activating mutations for the met tyrosine kinase receptor in human cancer. Proc Natl Acad Sei USA 94:

11445-11450, 1997

77. Jin L, Fuchs A, Schnitt SJ, Yao Y, Joseph A, Lamszus K, Park M, Goldberg ID, Rosen EM: Expression of scatter factor and c-met receptor in benign and malignant breast tissue. Cancer 79: 749-760,

1997

78. Katoh S, Zheng Z, Oritani K, Shimozato T, Kincade PW: Glycosylation of CD44 negatively regulates its recognition of hyaluronan. J Exp Med 182: 419-429, 1995

79. Kaufmann M, Heider KH, Sinn HP, von Minckwitz G, Ponta H, Herrlich P: CD44 variant exon epitopes in primary breast cancer and lenght of survival. Lancet 345: 615-619, 1995

80. Kern SE: p53: Tumor suppression through control of the cell cycle. Gastroenterology 106: 1708-1711, 1994

81. Kim H, Yang XL, Rosada C, Hamilton SR and August JT: CD44 expression in colorectal adenomas is an early event occuring prior to K-ras and p53 gene mutation. Arch Biochem Biophys 310: 504-507,1994

(39)

82. Kjellén L and Lindahl U: Proteoglycans: structures and interactions. Ann Rev Biochem 60: 443-475, 1991

83. Knudson CH, Knudson W: Hyaluronan binding proteins in development, tissue homeostasis, and disease. Faseb J 7: 1233-1241,

1993

84. Koch CA, Anderson D, Moran MF, Ellis C, Pawson T. SH2 and SH3 domains: elements that control interactions of cytoplasmic signaling proteins. Science 252: 668-674, 1991

85. Kogerman P, Sy MS, Culp LA. CD44 protein levels and its biological activity are regulated in Balb/c 3T3 fibroblasts by serum factors and by transformation with the ras but not with the sis oncogene. J Cell Physiol 169:341-349, 1996

86. Kolatsi-Joannou M. Moore R. Winyard PJ. Woolf AS. Expression of hepatocyte growth factor/scatter factor and its receptor, MET, suggests roles in human embryonic organogenesis. Ped Res 41 : 657-665, 1997 87. König H, Moll J, Ponta Herlich P. Trans-acting factors regulate the

expression of CD44 splice variants. EMBO J 15: 4030-4039, 1996 88. Koopman G, van Kooyk Y, de Graaf M, Meijer CJLM, Figdor CG,

Pals ST: Triggering of the CD44 molecule induces T cell adhesion through activation of the LF A-1 pathway. J Immunol 145: 3589-3593,

1990

89. Koopman G, Heider KH, Horst E, Adolf GR, van den Berg F, Ponta H, Herrlich P, Pals ST: Activated human lymphocytes and aggresive non-Hodgkin's lymphomas express a homologue of the rat metastasis-associated variant of CD44. J Exp Med 177: 897-904, 1993

90. Korinek V, Barker N, Morin PJ, van Wichen D, de Weger R, Kinzier KW, Vogelstein B, and Clevers H: Constitutive transcriptional activation by a ß-catenin-Tcf complex in APC-/- colon carcinoma. Science 275: 1784-1787, 1997

91. Lamb RF, Hennigan RF, Turnbull K, Katsanakis KD, MacKenzie ED, Birnie GD, Ozanne BW. AP-1-mediated invasion requires increased expressionofthe hyaluronan receptor CD44.Molec Cell Biol 17: 963-976, 1997

(40)

Requirements for HA binding by CD44: a role for the cytoplasmic domain and activation by antibody. J Exp Med 175: 257-266, 1992 93. Lesley J, Hyman R, Kincade PW: CD44 and its interactions with the

extracellular matrix. Adv Immunol 4: 271-335, 1993

94. Levy DB, Smidt KJ, Beazer-Barclay Y, Hamilton SR, Vogelstein B, Kinzler KW. Inactivation of bpth APC alleles in human and mouse tumors. Cancer Res 54: 5953-5958, 1994

95. Li Y, Bollag G, Clark R, Stevens J, Conroy L, Fults D, Ward K, Friedman E, Samowitz W, Robertson M: Somatic mutations in the neurofibromatosis I gene in human tumors. Cell 69: 275-281, 1992 96. Liang TJ, Reid AE, Xavier R, Cardiff RD, Wang TC: Transgenic

expression of tpr-met oncogene leads to development of mammary hyperplasia and tumors. J Clin Invest 97: 2872-2877, 1996

97. Lindahl U, Lidholt K, Spillman D, Kjellen L: More to heparin than anticoagulation. Thrombosis Res 75: 1-32, 1994

98. Lindahl U, Kusche-Gullberg M, Kjellen L: Regulated diversity of heparan sulfate. J Biol Chem 273: 24979-24982, 1998

99. Liu C, Park M, Tsao MS: Overexpressionof c-met proto-oncogene but not epidermal growth factor receptor or c-erbB-2 in primary human colorectal carcinomas. Oncogene 7:181-185, 1992

100. Liu F, Hata A, Baker J, Doody J, Carcamo J, Harland R, Massague JA: human MAD protein acting as a BMP-regulated transcriptional activator. Nature 381: 620-623, 1996

101. Luongo C, Moser AR, Gledhill S, Dove WF. Loss of Apc+ in intestinal adenomas from Min mice. Cancer Res 54: 5947-5952, 1994

102. Matsumura Y, Tarin D: Significance of CD44 gene products for cancer diagnosis and disease evaluation. Lancet 340: 1053-1058, 1992 103. Miyaka K, Medina KL, Hayashi SI, Ono S, Hamaoka T, Kincade P:

Monoclonal antibodies to Pgp-1/CD44 block lympho-hemopoiesis in long-term bone marrow cultures. J Exp Med 171 : 477-483, 1990 104. Miyashita T, Krajewska M, Wang HG, Lin HK, Liebermann DA,

Hoffman B, Reed JC. Tumor suppressor p5 3 is a regulator of bcl-2 and bax gene expression in vitro and in vivo. Oncogene 9: 1799-1805,1994 105. Miyazawa K, Shimomura T, Kitamura A, Kondo J, Morimoto Y,

(41)

Kitamura N. Molecular cloning and sequence analysis of the cDNA for a human serine protease respomsible for activation of hepatocyte growth factor. Structural similarity of the protease precursor to blood coagulation factor XII. J Biol Chem 268; 10024-10028, 1993

106. Miyazawa K, Shimomura T, Kitamura N: Activation of hepatocyte growth factor in the injured tissues is mediated by hepatocyte growth factor activator. J Biol Chem 271: 3615-3618, 1996

107. Molenaar M, van de Wetering M, Oosterwegel H, Peterson-Maduro J, Godsave S, Korinek V, Roose J, Destree O, Clevers H: Xtcf-3 transcription factor mediates beta-catenin-induced axis formation in Xenopus embryos. Cell 86: 391-399, 1996

108. Monpezat JP, Delattre O, Bernard A, Grunwald D, Remvikos Y, Muleris M, Salmon RJ, Frelat G, Dutrillaux B, Thomas G. Loss of alleles on chromosome 18 and on the short arm of chromosome 17 in polypoid colorectal carcinomas. Int J Cancer 41 : 404-408, 1988 109. Morin PJ, Vogelstein B, Kinzler KW: Apoptosis and APC in colorectal

carcinogenesis. Proc Natl Acad Sei USA 93: 7950-7954, 1996 110. Moser AR, Luongo C, Gould KA, McNeley MK, Shoemaker AR, Dove

WF. ApcMin: a mouse model for intestinal and mammary tumorigenesis. Eur J Cancer 31 A: 1061-1064, 1995

111. Mulder JWR, Kruyt PM, Sewnath M, Oosting J, Seldenrijk CA, Weidema W, Offerhaus GJA, Pals ST: Colorectal cancer prognosis and expression of CD44 exon-v6-containing CD44 proteins. Lancet 344:

1470-1472, 1994

112. Muto T, Bussey HJR, Morson BC: The evolution of cancer of colon and rectum. Cancer 36: 2251-2270, 1975

113. Naidu YM, Rosen EM, Zitnick R, Goldberg I, Park M, Naujokas M, Polverini PJ, Nickoloff BJ. Role of scatter factor in the pathogenesis of AIDS-related Kaposi sarcoma. PNAS 91: 5281-5285, 1994

114. Nakamura T, Teramoto H, Ichihara A: Purification and characterization of a growth factor from rat platelets for mature parenchymal hepatocytes in primary cultures. Proc Natl Acad Sei USA 83:6489-6493, 1986

(42)

association with the malignant process. Adv Cancer Res 71 : 241-319, 1997

116. Naujokas MF, Morin M, Anderson MS, Peterson M, Miller Jim. The Chondroitin sulfate form of invariant chain can enhance stimulation of T cell responses through interaction with CD44. Cell 74:257-268,1993 117. Nucci MR, Robinson CR, Longo P, Campbell P, Hamilton SR:

Phenotypic and genotypic characteristics of aberrant crypt foci in human colorectal mucosa. Hum Pathol 28: 1396-1407, 1997

118. Orzechowski HD, Beckenbach C, Herbst H, Stölzel U, Riecken EO and Stallmach A: Expression of CD44v6 is associated with cellular dysplasia in colorectal epithelial cells. Eur J Cancer 31 A: 2073-2079, 1995

119. Pals ST, Horst E, Ossekoppele GJ, Figdor CG, Scheper RJ, Meijer CJLM: Expression of lymphocyte homing receptor (CD44) as a mechanism of dissemination in non-Hodgkin's lymphoma. Blood 73: 885-888, 1989

120. Park M, Dean M, Cooper CS, Schmidt M, O'Brien S J, Blair DG, Vande Woude GF. Mechanism of met oncogene activation. Cell 45: 895-904, 1986

121. Park M, Dean M, Kaul K, Braun M J, Gonda MA, Vande Woude G. Sequence of MET protooncogene cDNA has features characteristic of the tyrosine kinase family of growth-factor receptors. Proc Natl Acad Sei USA 84: 6379-6383, 1987

122. Parsons R, Myeroff LL, Liu B, Willson JKV, Markowitz SD, Kinzier K, Vogelstem B: Microsatellite instability and mutations of the transforming growth factor ß type II receptor gene in colorectal cancer. Cancer Res 55: 5548-5550, 1995

123. Peach RJ, Hollenbaugh D, Stamenkovic I, Aruffo A: Identification of hyaluronic acid binding sites in the extracellular domain of CD44. J Cell Biol 122:257-264, 1993

124. Peifer M: Regulating cell proliferation; as easy as APC. Science 272: 974-975, 1996

125. Ponzetto C, Bardelli A, Zhen Z, Maina F, dalla Zonca P, Giordano S, Graziani A, Panayotou G, Comoglio PM: A multifunctional docking

(43)

site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family. Cell 77:261- 271, 1994

126. Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, Vogelstein B, Kinzier KW: APC mutations occur early during colorectal carcinogenesis. Nature 359: 235-237, 1992

127. Poylak K, Hamilton SR, Vogelstein B, Kinzier KW: Early alteration of cell-cycle-regulated gene expression in colorectal neoplasia. Am J Pathol 149:381-387, 1996

128. Prat M, Narishman RP, Crepaldi T, Nicotra MR, Natali PG, Comoglio PM: The receptor encoded by the human c-met oncogene is expressed in hepatocytes, epithelial cells and solid tumors. Int J Cancer 49:323-328, 1991

129. Pure E, Camp RL, Peritt D, Panettieri RA, Lazaar AL, Nayak S. Defective phosphorylationand hyaluranate binding of CD44 with point mutations in the cytoplasmic domain. J Exp Med 181: 55-62, 1995 130. Rodrigues GA and Park M: Autophosphorylationmodulates the kinase

activity and oncogenic potential of the Met receptor tyrosine kinase. Oncogene 9: 2019-2027, 1994

131. Rodriguez C, Monges G, Rouanet P, Dutrillaux B, Lefrançois D and Theillet C: CD44 expression patterns in breast and colon tumors: a PCR-based study of splice variants. Int J Cancer 64: 347-354, 1995 132. Rong S, Bodescot M, Blair D, Dunn J, Nakamura T, Mizuno K, Park

M, Chan A, Aaronson S and Woude GF van de Tumorigenicity of the

met proto-oncogene and the gene for hepatocyte growth factor. Mol

Cell Biol 12:5152-5158, 1992

133. Rong S, Segal S, Anver M, Resau JH, Vande Woude GF: Invasiveness and metastasis of NIH 3T3 cells induced by M?/-hepatocyte growth factor/scatter factor autocrine stimulation. Proc Natl Acad Sei USA 91 : 4731-4735, 1994

134. Riggins GJ, Thiagalingam S, Rozenblum E, Weinstein CL, Kern SE, Hamilton SR, Wilson JK, Markowitz SD, Kinzler KW, Vogelstein B: Mad-related genes in the human. Nat Gen 13: 347-349, 1996

135. Ropponen KM, Eskelinen MJ, Lipponen PK, Alhava E, Kosma VM: Expression of CD44 and variant proteins in human colorectal cancer

(44)

and its relevance for prognosis. Scan J Gastroenterology 33: 303-309, 1998

136. Rosen EM, Meromsky L, Setter E, Vinter DW, Goldberg ID. Purified scatter factor stimulates epithelial and vascular endothelial cell migration. PSEBM 195: 34-43, 1990

137. Rosen EM, Nigam SK, Goldberg ID. Scatter factor and the c-met receptor: a paradigm for mesenchymal/epithelialinteraction. J Cell Biol

127:225-234, 1994

138. Rubinfeld B, Souza B, Albert I, Muller O, Chamberlain SH, Masiarz FR, Munemitsius S, Polakis P: Association of the APC gene product with ß-catenin. Science 262: 1731-1734, 1993

139. Ruiz P, Schwarzler C, Gunthert U: CD44 isoforms during differentiation and development. Bioessays 17: 17-24, 1995

140. Ruoslahti E, Yamaguchi Y: Proteoglycans as modulators of cell growth. Cell 64: 867-869, 1991

141. Schlessinger J, Lax I and Lemmon M: Regulation of growth factor activations by proteoglycans: what is the role of the low affinity receptors? Cell 83: 357-360, 1995

142. Schmidt C, BladtF, GoedeckeS, Brinkmann V, ZschlescheW, Sharpe M, Gherardi E, Birchmeier C. Scatter Factor/Hepatocyte growth factor is essential for liver development. Nature 373: 699-702, 1995

143. Schmidt L, Duh FM, Chen F, Kishida T, Glenn G, Choyke P, Scherer SW, Zhuang Z, Lubensky I, Dean M, Allikmets R, Chidambaram A, Bergerheim UR, Feltis JT, Casadevall C, Zamarron A, Bernues M, Richard S, Lips CJM, Walther MM, Tsui LC, Geil L, Orcutt ML, Stackhouse T, Lipan J, Slife L, Brauch H, Decker J, Niehans G, Hughson MD, Moch H, Storkel S, Lerman MI, Linehan WM, Zbar B: Germline and somatic mutations in the tyrosine kinase domain of the

METproto-oncogene in papillary renal carcinomas. Nature Genet 16:

68-73,1997

144. Screaton GR, Bell MV, Jackson DG, Cornells FB, Gerth U, Bell JI: Genomic structure of DNA encoding the lymphocyte homing receptor CD44 reveals at least 12 alternatively spliced exons. Proc Natl Acad Sei USA 89: 12160-12164, 1992

(45)

145. Sherman L, Wainwright D, Ponta P, Herrlich P: A splice variant of CD44 expressed on the apical ectodermal ridge presents fibroblast growth factors to limb mesenchyme and is required for limb outgrowth. Genes Dev 12: 1058-1071, 1998

146. Shibamoto S, Hayakawa M, Takeuchi K, Hori T, Oku N, Miyazawa K, Kitamura N, Takeichi M, Ito F: Tyrosine phosphorylation of beta-catenin and plakoglobin enhanced by hepatocyte growth factor and epidermal growth factor in human carcinoma cells. Cell Ad Comm 1 : 295-305, 1994

147. Shimizu Y, van Seventer GA, Siraganian R, Wahl L, Shaw S : Dual role of the CD44 molecule in T cell adhesion and activation. J Immunol

143: 2457-2463, 1989

148. Sleeman JP, Arming S, Moll JF, Hekele A, Rudy W, Sherman , Kreil G, Ponta H, Herrlich P: Hyaluronate-independantmetastatic behaviour of CD44 variant-expressingpancreatic carcinoma cells. Cancer Res 56: 3134-3141, 1996

149. Smits R, Kartheuser A, Jagmohan-Changur S, Leblanc V, Breukel C, de Vries A, van Kranen H, van Krieken JH, Williamson S, Edelmann W, Kucherlapati R, Khan PM, and Fodde R: Loss of Ape and the entire chromosome 18 but absence of mutations at the Ras and Tp53 genes in intestinal tumors from ^/?cl638N, a mouse model for Ape-driven carcinogenesis. Carcinogenesis 18: 321-327, 1997

150. Sonnenberg E, Meyer D, Weidner KM, Birchmeier C: Scatter factor/hepatocyte growth factor and its receptor, the c-met tyrosine kinase, can mediate a signal exchange between mesenchyme and epithelia during mouse development. J Cell Biol 123: 223-235, 1993 151. Stamenkovic I, Amiot M, Pesando JM, Seed B: A lymphocyte

molecule implicated in lymph node homing is a member of the cartilage link protein family. Cell 56: 1057-1062, 1989

152. Stamenkovic I, Aruffo A, Amiot M, Seed B: The hematopoietic and epithelial forms of CD44 are distinct polypeptides with different adhesion potentialsfor hyaluronatebearing cells. EMBO J 10: 343-348,

1991

Referenties

GERELATEERDE DOCUMENTEN

Hence, this thesis addresses the function of VISTA, microglia, and astrocytes in healthy developing and adult CNS, and in MS using post-mortem human tissue, bioinformatic

With the addition chain, the Itoh-Tsujii algorithm computes the inverse in less time using a recursive re-arrangement of finite field operations. In relation to speed, EEA

multiple risk factors for older adults who sustained fall-related injuries, compared to (a). those who did not fall, and (b) those who fell without sustaining

Further con- straints are provided by generating synthetic seismograms for a known model containing gas hydrate and free gas, and applying a waveform inversion modeling approach to

The width and mass of the Gaussian is fixed to the value from the fit to the signal Monte Carlo sample. All parameters are allowed to

The spectra we observe for our precise radial velocity program in­ clude a single stellar reference spectrum observed without the HF absorption cell in the telescope beam

Higher quality 6” x 9” black and white photographic prints are available for any photographs or illustrations appearing in this copy for an additional charge... By

If the number of surface species increases to three, for example CO(ads), OH(ads) and either free Pt sites or O(ads), two adsorption relaxations are needed, circuit 2L, in order