search
for
 About Bioline  All Journals  Testimonials  Membership  News


Indian Journal of Cancer
Medknow Publications on behalf of Indian Cancer Society
ISSN: 0019-509X EISSN: 1998-4774
Vol. 48, Num. 3, 2011, pp. 351-360

Indian Journal of Cancer, Vol. 48, No. 3, July-September, 2011, pp. 351-360

Review Article

The Smad family and its role in pancreatic cancer

1 Cancer Genetics Unit, Hormones and Cancer Group, Kolling Institute of Medical Research, NSW, 2006, Australia
2 Department of General Surgery, Government Medical College and Hospital, Sector-32, Chandigarh, India
3 Department of Cytology and Gynaecological Pathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
Correspondence Address: P Singh, Cancer Genetics Unit, Hormones and Cancer Group, Kolling Institute of Medical Research, NSW, 2006 Australia, puneetsingh_bio@y7mail.com

Code Number: cn11091

PMID: 21921337

DOI: 10.4103/0019-509X.84939

Abstract

One of the major signaling pathways that determine the tumor aggression and patient outcome in pancreatic cancer is the transforming growth factor-beta (TGF-ß) pathway. It is inactivated at various levels in pancreatic cancer and plays a dual role in tumor initiation and progression. The Smad family of proteins transduce signals from the TGF-ß superfamily ligands that regulate cell proliferation, differentiation and death through activation of receptor serine/threonine kinases. This review discusses the structure, function and regulation of various participating Smad family members, and their individual roles in determining the progression and outcome of pancreatic cancer patients, with a special emphasis on Smad4.

Keywords: Pancreatic cancer, smad family, Smad4, Smad6, Smad7, transforming growth factor-beta

Introduction

The members of the transforming growth factor-β (TGF-β) family control growth, differentiation and apoptosis of cells and have important functions during embryonic development. The TGF-β family ligands initiate downstream signaling events by activation of transmembrane serine-threonine kinase receptors, namely, TGF-β type I receptor (TbRI) and TGF-β type II receptor (TβRI) [Figure - 1]. On ligand binding, TβRII becomes constitutively active, heterodimerizes with TβRI and transphosphorylates its GS domain, resulting in its activation. [1],[2] Once activated, TβRI phosphorylates a class of molecules known as receptor-regulated Smads (R-Smads) at an SSXS motif at their C-terminal end. [3] Active, phosphorylated Smad2 or Smad3 heterodimerize with Smad4, translocate to the nucleus and regulate transcription. [4],[5] Smad6 and Smad7 are the negative regulators of TGF-β pathway and act by blocking R-Smads' interaction with TβRI, phosphorylation by TβRI or heterodimerization with Smad4. [6],[7]

Although Smad4 is inactivated in many cancers, its specificity for pancreatic cancer makes it one of the most extensively studied markers for pancreatic cancer. It is lost in 55% of the cases, and its loss often predicts a poorer prognosis. Besides Smad7, which is emerging as a new molecule of consequence in this disease, other components of the Smad family do not have a significant role to play. TGF-β itself is one of the important growth factors in pancreatic cancer, which, in the event of Smad4 loss, through alternative pathways, supports tumor growth and progression.

Discovery and Nomenclature of Smads

Proteins belonging to the Smad family were first identified in the fruit fly Drosophila melanogaster in the mid 1990s, where an intracellular protein named Mad mediates the signaling of decapentaplegic (dpp), a member of the TGF-β superfamily corresponding to mammalian bone morphogenetic protein 2 or 4 (BMP-2/4). [8] The discovery of orthologous proteins in Caenorhabditis elegans as well as in vertebrates soon followed, which paved the way to the discovery of a whole new family of proteins, the Smad family, which functions as important mediator of the TGF-β signaling pathway. The term "Smad" is derived from a combination of the gene name from two orthologous proteins sma from Caenorhabditis elegans, and Mad from Drosophila melanogaster. [9] The first Smad to be discovered was Smad4, or DPC4 (deleted in pancreatic cancer 4). [10]

General Structure of Smads

Smads consist of two conserved domains: The N-terminal Mad Homology domain1 (MH1 domain) and the C-terminal Mad Homology domain 2 (MH2 domain), joined together by a linker region, which is full of regulatory sites [Figure - 2]. MH1 domain has DNA-binding activity (except in the major splice form of Smad2, which contains an insert that prevents DNA binding), whereas the MH2 has transcriptional activity. [11] Receptor-mediated phosphorylation appears to relieve these two domains from a mutually inhibitory interaction. The L3 loop and the α-helix-1 (αH-1) in the MH2 domain of a R-Smad and the L45 loop in the kinase domain of a type I receptor specify the Smad-receptor interaction. [12] The αH-2 domain of Smads specifies interactions with certain DNA-binding cofactors. [13],[14],[15] A highly basic surface patch conserved around the L3 loop of all R-Smads and a complementary surface pattern on TβR-I kinase domain may facilitate receptor-Smad recognition. [16] This L3 loop is absent in Smad4. [17]

The linker region has a regulatory function and contains phosphorylation sites for Erk and MAPKs, consensus sites for calcium-regulated kinases and one prolin-tyrosine (PY) motif for recognition by WW domains for ubiquitination or sumoylation of the Smads.

Members of the Smad Family

There are eight different Smads (Smad 1-8) in mammalian cell populations, which are classified into three categories: (i) receptor-regulated or regulatory Smads (R-Smads), which include Smads 1, 2, 3, 5 and 8, (ii) common Smad (Co-Smad), i.e. Smad 4 and (iii) inhibitory or anti-Smads (I-Smads), which are Smad 6 and Smad 7.

The general structure of Smads is more or less the same, with a few differences among different categories of Smads, which influence their respective functions [Figure - 3].

Receptor-Regulated Smads or R-Smads

R-Smads directly interact with activated TβRI in the receptor complex and, as a result, are phosphorylated at two of the three serine residues in a C-terminal SSXS motif [18] [Figure - 4]. Of the five R-Smads, Smad2 and Smad3 mediate activin and TGF-β signal transduction, while Smad1, Smad5 and Smad8 mediate BMP signals. [19],[20]

Following phosphorylation of their MH1 domain, R-Smads can form oligomers at different stochiometries. Early experiments indicated that oligomeric Smads are trimers. [21],[22] Equilibrium centrifugation and crystallographic studies have confirmed this in the case of Smad3, [23],[24] but a dimeric configuration for the Smad2-Smad4 complex has also been proposed. [25] Therefore, different R-Smad-Co-Smad oligomers with distinct stoichiometries are possible.

Once activated, Smad heterooligomers translocate to the nucleus, where they control target gene expression in a cell type-specific manner through interactions with other transcription factors, corepressors and coactivators. Studies with Smad4-null cell lines have shown that Smad2 and Smad3 are able to translocate to the nucleus on their own, but are unable to activate reporter genes on their own, suggesting that the whole R-Smad/Co-Smad complex is essential for transcriptional regulation. [26]

Common Smad or Co-Smad

There is only one Co-Smad, i.e. Smad4. The only other Co-Smad is Smad4β, found in Xenopus laevis. [27] Smad4 is recognized as a key tumor suppressor as it is defective in a number of cancers. It interacts with Smads 1, 2, 3 and 5 and hence participates in the intracellular signaling pathways of all three classes of TGF-β ligands. [19] This indicates that wild-type Smad4 in the TGF-β signal pathway may be critical in maintaining an environment that inhibits tumorigenesis. The SMAD4 gene is located on the long (q) arm of chromosome 18 at position 21.1, [28] and covers the region from 46,810,610 to 46,860,144 base pairs. The size of the whole gene is 49,539 bases, and it lies on the plus stand of DNA and transcribes 3220 nucleotides mRNA. The gene translates into a protein of molecular weight of 60,439 Daltons and contains 552 amino acids in its structure. Earlier, it was known to contain one to 11 exons, but recent mutational studies have led to the discovery of another exon that lies upstream of exon 1 and is named exon 0. [29] It belongs to the Dwarfin family of proteins, which harbor two conserved amino- and carboxyl-terminal domains known as MH1 and MH2, respectively.

Smad4 in the basal state is found mostly as a homooligomer, most likely a trimer. On activation of R-Smads, it forms a heterooligomer with them, translocates into the nucleus and acts as a transcription factor.

Anti-Smads or Inhibitory Smads or I-Smads

The inhibitory Smads, Smad6 and Smad7, act antagonistically, abrogating TGF-β signal transduction. They contain an MH2 domain but have very little similarity to the cannonical MH1 domain of other Smads [Figure - 4].

Smad7 inhibits Smad phosphorylation by occupying type I receptors for TGF-β, Activin and BMP. [11],[30] Mouse Smad7 preferentially inhibits Activin and TGF-β signaling over BMP signaling, while the reverse is true of a Xenopus Smad7 homolog. [31] Smad7 appears to reside predominantly in the nucleus at basal state and translocates to the cytoplasm upon TGF-β stimulation. [32]

Smad6 preferentially inhibits BMP signaling by a mechanism different from that of Smad7. [33],[34] When expressed at levels that are sufficient for inhibition of BMP signaling but not TGF-β signaling, Smad6 does not interfere with receptor function but competes with Smad4 for binding to receptor-activated Smad1 and yields inactive Smad1-Smad6 complexes. Overexpression of Smad4 can outcompete Smad6 and rescue BMP signaling. [33] At higher expression levels, Smad6 can mimic Smad7 and inhibit signaling by BMP and TGF-β receptors. [7]

The expression of both Smad6 and Smad7 is increased in response to BMP, Activin and TGF-β, suggesting their roles in negative feedback of these pathways. [34],[35] The expression of Smad7 can also be increased by pathways that negatively regulate TGF-β signaling, e.g. interferon-γ (IFN-γ), acting via the Jak1 tyrosine kinase and the Stat1 transcription factor, providing a basis for the known antagonism between TGF-β and IFN-γ in the regulation of immune cell functions. [36] Similarly, in response to the proinflammatory cytokines, tumor necrosis factor-α and interleukin-1β activate Smad7 expression via the NF-κ B/RelA transcription factor. [37]

Smad Activation

Inactive, cytoplasmic Smads are intrinsically autoinhibited through an intramolecular interaction between the MH1 and MH2 domains. [38] Receptor-mediated phosphorylation induces conformational changes that relieve the autoinhibition and expose buried epitopes on to the surface, enabling Smads to interact with other components important for nuclear import, transcriptional regulation or degradation.

Smad2 and Smad3, in their unphosphorylated and inactive form, reside predominantly in the cytoplasm bound to microtubule filaments [39] and filamins. [40] TGF-β signaling induces their dissociation by phosphorylating their C-terminal domains by TβRI receptor. TβRI receptor is associated with a FYVE domain containing scaffolding protein named SARA (Smad anchor for receptor activation). [41] Another cytoplasmic protein named PML (promyelocytic leukemia tumor suppressor) physically interacts with Smad2/Smad3 and SARA and is required for the association of Smad2/Smad3 with SARA. [42] SARA interacts with the MH2 domain of inactive Smad2 and Smad3, targeting them to early endosomes and aiding in their recruitment to the receptors, thus promoting Smad phosphorylation and TGF-β signaling. [43]

On ligand binding, the activated type I and II receptors may be internalized via clathrin-coated pits into early endosomes that contain SARA along with Smad2/3. PML expression is induced by TGF-β and it is required for the accumulation of SARA and the TGF-β receptors in the early endosomes. Several other accessory/scaffolding proteins like SARA have been discovered for the TGF-β pathway R-Smads, e.g. axin and disabled2 (Dab2), [44],[45] but no accessory proteins have yet been discovered for the BMP-pathway Smads. These regions, where two types of receptors are found in combination with SARA, are called "Smad signaling centers" [Figure - 5].

Regulation of the Activation Process

TGF-β signaling is modulated at multiple levels. Extracellular ligand trapping molecules or antagonists, including gremlin, noggin, chordin (all members of DAN/Cerberus protein family) and follistatin, can block ligand binding to the receptors. A pseudoreceptor BAMBI (BMP and Activin membrane bound inhibitor) extracellularly resembles a type I receptor but lacks the cytosolic kinase domain, traps and thus blocks activation by TGF-β ligands. [46] Posttranslational regulation of Smads, through a ubiquitin-mediated proteasomal degradation pathway, can profoundly affect signaling. [47],[48]

Smad6 and Smad7 inhibit the Smad-mediated signaling; Smad7 by interacting with the type I receptor while Smad6 by competing with the activated R-Smads for heteromeric complex formation with co-Smad4. [49] c-Ski and SnoN, members of the Ski family of protooncoproteins, can antagonize TGF-β signaling through direct interactions with Smad4 and R-Smads. [50] Phosphorylation of Smads in the MAP kinase sites at the linker region attenuates ligand-induced nuclear translocation. Dephosphorylation of the Smads by as yet unidentified phosphatases may be another mechanism for the termination of Smad signaling.

Phosphorylation of R-Smads by Other Signaling Pathways: A Cross-Talk between Different Pathways

The linker region connecting the MH1 and MH2 domains is less conserved between different Smads, but contains several important regulatory peptide motifs, including potential phosphorylation sites for mitogen-activated protein kinases (MAPKs), Erk-family MAP kinases, [51] the Ca 2+ /calmodulin-dependent protein kinase II (CamKII) [52] and protein kinase C (PKC). [53] A proline-tyrosine (PY) motif present in the linker region of most R-Smads and I-Smads enables Smad interaction with the WW domains of the E3 ubiquitin ligases Smurf1 and Smurf2 (Smad ubiquitination-related factors) and SCF/Roc1 enabling ubiquitin-mediated proteasomal degradation. [54],[55] Smad4 linker region also includes a nuclear export signal (NES) [56] and a Smad4 activation domain that is required in transcriptional complexes, mediating the activation of Smad-dependent target genes. [4] Thus, phosphorylation of the Smads not only causes their activation but also modulates their activity and provides a mechanism for integration of the Smad pathway with other signaling pathways that can modulate TGF-β signaling.

Mechanism of Smad Nucleo-Cytoplasmic Shuttling

Basal state

At the basal level, all R-Smads, mammalian Smad4 and Xenopus Smad4α reside in the cytoplasm. In contrast, Xenopus Smad4β and I-Smads localize to the cell nucleus. [27],[32],[49],[57] Smad4 constitutively shuttles between the cytoplasm and the nucleus, and its predominant cytoplasmic localization in unstimulated cells is due to active nuclear export mediated by a unique leucine-rich NES located in its linker region. Smad4 is continuously exported out of the nucleus by chromosome region maintenance 1 (CRM-1). Although the nuclear entry of Smad4 is a spontaneous event not requiring TGF-β signaling, CRM-1-mediated nuclear export must be suppressed by TGF-β in order to retain Smad4 in the nucleus. Smad4 therefore continuously shuttles in and out of the nucleus, and its oligomerization with R-Smads might occur en route to the nucleus. [56],[59],[60]

C-terminal phosphorylation of Smad3 results in a conformational change that exposes the NLS for importinα1-dependent Ran-mediated nuclear import. [59],[61] Smad2, however, translocates into the nucleus by a cytosolic factor-independent import activity. This difference in the pathway taken up by two R-Smads is due to the presence of the unique exon 3 in the MH1 domain of Smad2. [59]

Many of the previous studies have even suggested an importin-independent nuclear import mechanism for unphosphorylated Smad2, Smad3 and Smad4. Such a mechanism relies on the ability of Smad2, Smad3 and Smad4 to directly interact with phenylalanineglycine (FG) repeat-containing nucleoporins, including CAN/Nup214 and Nup153. [62],[63]

Activated state

In the activated state, counteracting nuclear import and export forces control the subcellular localization of Smad4. Because TGF-β stimulation does not noticeably accelerate nuclear import of Smad4, it most likely acts upon the CRM-1-mediated export to promote Smad4 accumulation in the nucleus. DNA binding cannot block, but instead promotes, CRM-1 association with Smad4. [64] Therefore, it is heterotrimerization that may cause physical occlusion of the CRM-1-binding site or may induce long-range conformational changes in Smad4, making it incapable of recognizing CRM-1. This Smad4 heterotrimeric complex stays in the nucleus until its dissociation is triggered by dephosphorylation of Smad2 and Smad3, resulting in its nuclear export by CRM-1. [65]

Transcriptional Regulation of Target Genes by Smads

Smad binding elements

The Smad4 heterodimeric complex recognizes and binds to Smad Binding Elements (SBEs) present in the promoter sequences of various cell cycle regulatory genes. [30] SBE comprises of an 8 bp pallindromic sequence GTCTAGAC [66] or multiple copies of GTCT or AGAC. [67]

While, full-length Smad4 protein can bind as such to SBE, binding by Smad3 to DNA requires either C-terminal phosphorylation or C-terminal truncation of the MH2 domain, both of which would release the inhibition of its MH1 domain. [68]

The crystal structure analysis revealed a b-hairpin loop in Smad3 responsible for its direct DNA contact. The β-hairpin loop is conserved among R-Smads and Co-Smad. [69] Smad3 and Smad4 have also been reported to bind to GC-rich sequences. [70] Smad2, however, lacks the ability to bind DNA because of the presence of a small exon upstream of the b-hairpin, which when removed, confers DNA-binding ability to Smad2. [71],[72]

The SBE AGAC sequence is calculated to be present on average once every 1024 bp in the genome, or about once in the regulatory region of any average size gene. If binding to the SBE were sufficient for Smad-dependent transcriptional activation, an activated Smad protein would lead to the nonselective activation of massive numbers of genes. However, activation of target genes solely via a SBE is not feasible for two reasons. Firstly, the affinity of a Smad MH1 domain for the SBE is in the 10 -7 M range, [69] which is too weak for effective binding in vivo without the involvement of additional transcriptional cofactors. This is supported by the fact that it takes many SBEs to achieve Smad activation of a reporter gene in artificial concatemers. [66] Secondly, Smad binding to the SBE lacks selectivity, as Smads 1, 3 and 4 have a similar affinity for the SBE. This is not surprising because the β-hairpin sequence is identical in all R-Smads and is highly conserved in Smad4. Therefore, additional transcriptional cofactors appear necessary for specific, high-affinity binding of a Smad complex to a target gene.

TGF-β/Smad4 Signaling in Pancreatic Cancer

Transforming growth factor-beta

All three mammalian Transforming growth factor-beta (TGF-β) isoforms are overexpressed in pancreatic ductal adenocarcinoma (PDAC), and overexpression has been found to corDPC4relate with decreased patient survival. [73],[74] This is surprising, considering the negative regulatory effect of TGF-β on epithelial cell growth. Cancer cells probably lose the ability to respond to the growth inhibitory signals of TGF-β.

The inactivating mutations in the Smad4 gene and upregulation of the inhibitory Smad6 and Smad7 genes have been found in many pancreatic cancers in which they inhibit TGF-β signaling. [10],[76]

This was shown in pancreatic cancer cells that were resistant to TGF-β signaling despite high levels of TGF-β receptors, TβRI and TβRII. In pancreatic cancers, TGF-β deregulation, besides losing its ability to bring about growth suppression, promotes metastasis and brings about Epithelial Mesenchymal Transition (EMT). Smad4 downregulation counteracted TGF-β-induced cell cycle arrest and migration but not EMT, which results in increased motility and invasion. [77] There are some alternative TGF-β pathways, independent of Smad4, which contribute towards its metastatic role in transformed cells. Oncogenic K-ras has been shown to facilitate TGF-β-induced transcriptional downregulation of the tumor suppressor PTEN in a SMAD4-independent manner, and could constitute a signaling switch mechanism from growth suppression to growth promotion in pancreatic cancers. [78] The contribution of TGF-β toward enhanced cancer progression can also be through its positive regulation of certain stromal elements, like membrane type 1-matrix metalloproteinase (MT1-MMP). [79] TGF-β suppresses PTEN in pancreatic cancer cells through NF-kappa B activation and enhances cell motility and invasiveness in a Smad4-independent manner that can be counteracted when TGF-β-Smad4 signaling is restored. The TGF-β/NF-kappaB/PTEN may be a critical pathway for pancreatic cancer cells to proliferate and metastasize. [80] Loss of Smad4, leading to aberrant activation of Stat3, contributes to the switch of TGF-βeta from a tumor-suppressive to a tumor-promoting pathway in pancreatic cancer. [81] The TGF-β/PKC/α -PTEN is yet another pathway implicating the change in the role of TGF-β signaling to a metastatic one. [82]

In case where TGF-β is overexpressed, its inhibition by TbRI inhibitor reduced primary tumor growth and decreased the incidence of metastasis. [83],[84]

A recent role identified for defective TGF-β signaling and can be utilized for future therapies is that it selectively sensitizes the pancreatic as well as colon cancers to rapamycin or other drugs targeting mTOR. [85]

TGF-beta Receptors

Reduced expression or inactivation of TGF-β receptors often correlates with malignant progression and loss of sensitivity to the antiproliferative effects of TGF-β. [86],[87] There are only sporadic reports of mutations or deletions in TβRI in malignancy. [88] A study showed that the individuals with constitutively decreased TβR1 expression may have a decreased risk of pancreatic cancer. [88] TβRII is a frequent locus of inactivating mutations, which may be because of a deficient DNA mismatch repair system. In one such case, frameshift mutations in a 10-basepair polyadenine tract (big polyadenine tract [BAT]) in exon 3 of the TβRII gene (BAT-RII) resulted in the production of truncated receptors where the serine-threonine kinase domain was absent. [86],[89] Repression of TGF-β receptor expression is a common mechanism that enables tumor cells to escape from negative regulation of growth by TGF-β. [90]

Smad4

Although lost in many cancers, loss of Smad4 is more sensitive and specific to pancreatic cancer. [91] Studies have shown SMAD4 to be inactivated in 55% of pancreatic cancers. [10],[92],[93] The inactivation of SMAD4 gene occurs either by deletion of both alleles (35%) or by intragenic mutation in one allele coupled with the loss of the other allele (20%). [94]

The mutations are often found in the C-terminal MH2 domain, which harbors a mutational hotspot corresponding to codons 330-370, termed as mutation cluster region (MCR). [95] The mutant protein cannot be recruited to DNA by transcription factors and hence cannot form transcriptionally active DNA-binding complexes. C-terminal truncation leads to decreased stability and prevents Smad4 homomeric complex formation and heteromeric complex formation with activated Smad2. Molecular modeling indicates that the truncation removes residues critical for homomeric and heteromeric Smad complex formation. [96] Another alternative route that the mutated Smad4 proteins may take is ubiquitin-proteosome degradation. [97]

Deletion of Smad4 occurs at a later stage of Pan IN. [98] Loss of Smad4 protein expression highly correlated with the presence of widespread metastasis but not with locally destructive tumors. [99] Involvement of K-ras/ERK pathway, PTEN and RON, respectively, are implicated in the gain of metastatic potential of pancreatic cancers in the absence of Smad4. [81],[82],[100] Smad4 is thought to be dispensable for normal pancreas but critical for the progression of tumors with mutated K-ras gene. [101] Thus, oncogenic K-RAS/ERK in pancreatic adenocarcinoma facilitates TGF-β-induced transcriptional downregulation of the tumor suppressor PTEN in a Smad4-independent manner and could constitute a signaling switch mechanism from growth suppression to growth promotion in pancreatic cancers.

SMAD4 gene inactivation is associated with poorer prognosis in patients with surgically resected adenocarcinoma of the pancreas. [102],[103] Determination of Smad4 status at initial diagnosis may be of value in determining the stage and metastatic status of disease and will help in stratifying patients into treatment regimens accordingly. [104],[105]

Success with SMAD4 gene transfection of Smad4-deficient cell lines has been variable. Introduction of Smad4 in BxPC3 cells with Smad4 homozygous deletion inhibited cell proliferation, but this effect was transient, indicating that Smad4 growth inhibitory actions were circumvented in the later stages of pancreatic tumorigenicity. [105] However, a recent gene transfection with SMAD4, using retroviral vector pLXSN, inhibited prolifertion in pancreatic cancer cell line. [106] Utilizing the loss of Smad4, some novel molecules (UA62001, UA62784) that specifically target and inhibit Smad4-deficient cancer cells have been discovered. [107],[108]

Inhibitory Smads

Previous studies have shown Smad7 overexpression in pancreatic cancer cell lines. [109],[110],[111] A recent study in patient samples, however, contradicts these results and shows a low expression of Smad7 in pancreatic cancer patients. The authors have demonstrated a significant correlation between low-Smad7 expression with lymph node metastasis and poor prognosis. [112] Different studies have isolated different molecules, like KLF11, retinoblastoma and thioredoxin, which are involved in Smad7-dependent aggressiveness of pancreatic cancer. [111],[113],[114] There are not many reports on Smad6 expression in pancreatic cancer. Smad6, along with Smad7 expression, was found to be elevated in a pancreatic cancer cell line. [109] One report on pancreatic cancer patient samples, however, contradicts this and demonstrates that the increased expression of Smad6 and Smad7 are infrequent in tumor compared with normal tissues. [115] The variation in the observed expression of two inhibitory Smads in cell lines as compared with the tissue samples can be because of a possible reversal of phenotype in artificial tissue culture systems.

R-Smads

There are not many reports regarding the role of R-Smads in pancreatic cancer. Smad2 mRNA levels were significantly increased in pancreatic cancer samples in comparison with normal pancreatic tissues. Authors suggest that this might lead to excessive activation of specific components of the TGF-beta-signaling pathway. [109]

A missense mutation in an arginine residue of the MH1 domain of Smad2 was identified in pancreatic tissue samples from multiple patients. Although this alteration did not interfere with the majority of the Smad's cellular functions, it directed the Smad protein for degradation via the UbcH5 family of E2 ubiquitin ligases. Similar results were also found with mutations in the MH2 domain. This resulted in the disruption of the steady state kinetics of R-Smads due to amino acid substitutions, making cells resistant to the suppressive effects of TGF-β. [97]

References

1.Wrana JL, Attisano L, Wieser R, Ventura F, Massagué J. Mechanism of activation of the TGF-beta receptor. Nature 1994;370:341-7.  Back to cited text no. 1    
2.Massagué J. TGF-beta signal transduction. Annu Rev Biochem 1998;67:753-91.  Back to cited text no. 2    
3.Souchelnytskyi S, Tamaki K, Engström U, Wernstedt C, ten Dijke P, Heldin CH. Phosphorylation of Ser465 and Ser467 in the C terminus of Smad2 mediates interaction with Smad4 and is required for transforming growth factor-beta signaling. J Biol Chem 1997;272:28107-15.  Back to cited text no. 3    
4.de Caestecker MP, Yahata T, Wang D, Parks WT, Huang S, Hill CS, et al. The Smad4 activation domain is a proline-rich, p300-dependent transcriptional activation domain. J Biol Chem 2000;275:2115-22.  Back to cited text no. 4    
5.de Winter JP, Roelen BA, ten Dijke P, van der Burg B, van den Eijnden-van Raaij AJ. DPC4 (SMAD4) mediates transforming growth factor-beta1 (TGF-beta1) induced growth inhibition and transcriptional response in breast tumour cells. Oncogene 1997;14:1891-9.  Back to cited text no. 5    
6.Hayashi H, Abdollah S, Qiu Y, Cai J, Xu YY, Grinnell BW, et al. The MAD-related protein Smad7 associates with the TGF-beta receptor and functions as an antagonist of TGF-beta signaling. Cell 1997;89:1165-73.  Back to cited text no. 6    
7.Imamura T, Takase M, Nishihara A, Oeda E, Hanai J, Kawabata M, et al. Smad6 inhibits signalling by the TGF-beta superfamily. Nature 1997;389:622-6.  Back to cited text no. 7    
8.Sekelsky JJ, Newfeld SJ, Raftery LA, Chartoff EH, Gelbart WM. Genetic characterization and cloning of mothers against dpp, a gene required for decapentaplegic function in Drosophila melanogaster. Genetics 1995;139:1347-58.  Back to cited text no. 8    
9.Derynck R, Gelbart WM, Harland RM, Heldin CH, Kern SE, Massague J, et al. Nomenclature: Vertebrate mediators of TGF-beta family signals. Cell 1996;87:173.  Back to cited text no. 9    
10.Hahn SA, Schutte M, Hoque AT, Moskaluk CA, da Costa LT, Rozenblum E, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science 1996;271:350-3.  Back to cited text no. 10    
11.Shi Y, Hata A, Massague J, Pavletich NP. A structural basis for mutational inactivation of the tumour suppressor Smad4. Nature 1997;388:87-93.  Back to cited text no. 11    
12.Massague J, Wotton D. Transcriptional control by the TGF-b/Smad signaling system. EMBO J 2000;19:1745-54.  Back to cited text no. 12    
13.Massague J, Chen YG. Controlling TGF signaling. Genes Dev 2000;14:627-44.  Back to cited text no. 13    
14.Feng XH, Derynck R. A kinase subdomain of transforming growth factor-b (TGF-b) type I receptor determines the TGF-b intracellular signaling activity. EMBO J 1997;16:3912-22.  Back to cited text no. 14    
15.Chen YG, Hata A, Lo RS, Wotton D, Shi Y, Pavletich N, et al. Determinants of specificity in TGF-beta signal transduction. Genes Dev 1998;12:2144-52.  Back to cited text no. 15    
16.Huse M, Chen YG, Massagué J, Kuriyan J. Crystal structure of the cytoplasmic domain of the type I TGF beta receptor in complex with FKBP12. Cell 1999;96:425-36.  Back to cited text no. 16    
17.Wu G, Chen YG, Ozdamar B, Gyuricza CA, Chong PA, Wrana JL, et al. Structural basis of Smad2 recognition by the Smad anchor for receptor activation. Science 2000;287:92-7.  Back to cited text no. 17    
18.Moustakas A, Souchelnytskyi S, Heldin CH. Smad regulation in TGF-beta signal transduction. J Cell Sci 2001;114:4359-69.  Back to cited text no. 18    
19.Cook T, Urrutia R. TIEG proteins join the Smads as TGF-beta-regulated transcription factors that control pancreatic cell growth. Am J Physiol Gastrointest Liver Physiol 2000;278:G513-21.  Back to cited text no. 19    
20.Shi Y, Massague J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 2003;113:685-700.  Back to cited text no. 20    
21.Kawabata M, Inoue H, Hanyu A, Imamura T, Miyazono K. Smad proteins exist as monomers in vivo and undergo homo- and hetero-oligomerization upon activation by serine/threonine kinase receptors. EMBO J 1998;17:4056-65.  Back to cited text no. 21    
22.Shi Y. Structural insights on Smad function in TGF-beta signaling. Bioessays 2001;23:223-32.  Back to cited text no. 22    
23.Chacko BM, Qin B, Correia JJ, Lam SS, de Caestecker MP, Lin K. The L3 loop and C-terminal phosphorylation jointly define Smad protein trimerization. Nat Struct Biol 2001;8:248-53.  Back to cited text no. 23    
24.Correia JJ, Chacko BM, Lam SS, Lin K. Sedimentation studies reveal a direct role of phosphorylation in Smad3:Smad4 homo- and hetero-trimerization. Biochemistry 2001;40:1473-82.  Back to cited text no. 24    
25.Wu RY, Zhang Y, Feng XH, Derynck R. Heteromeric and homomeric interactions correlate with signaling activity and functional cooperativity of Smad3 and Smad4/DPC4. Mol Cell Biol 1997;17:2521-8.  Back to cited text no. 25    
26.Fink SP, Mikkola D, Willson JK, Markowitz S. TGF-beta-induced nuclear localization of Smad2 and Smad3 in Smad4 null cancer cell lines. Oncogene 2003;22:1317-23.  Back to cited text no. 26    
27.Masuyama N, Hanafusa H, Kusakabe M, Shibuya H, Nishida E. Identification of two Smad4 proteins in Xenopus. Their common and distinct properties. J Biol Chem 1999;274:12163-70.  Back to cited text no. 27    
28.Thiagalingam S, Lengauer C, Leach FS, Schutte M, Hahn SA, Overhauser J, et al. Evaluation of candidate tumour suppressor genes on chromosome 18 in colorectal cancers. Nat Genet 1996;13:343-6.  Back to cited text no. 28    
29.Mamot C, Mild G, Reuter J, Laffer U, Metzger U, Terracciano L, et al. Infrequent mutation of the tumour-suppressor gene Smad4 in early-stage colorectal cancer. Br J Cancer 2003;88:420-3.  Back to cited text no. 29    
30.Heldin CH, Miyazono K, ten Dijke P. TGF-beta signalling from cell membrane to nucleus through SMAD proteins. Nature 1997;390:465-71.  Back to cited text no. 30    
31.Souchelnytskyi S, Nakayama T, Nakao A, Morén A, Heldin CH, Christian JL, et al. Physical and functional interaction of murine and Xenopus Smad7 with bone morphogenetic protein receptors and transforming growth factor-beta receptors. J Biol Chem 1998;273:25364-70.  Back to cited text no. 31    
32.Itoh S, Landstrom M, Hermansson A, Itoh F, Heldin CH, Heldin NE, et al. Transforming growth factor beta1 induces nuclear export of inhibitory Smad7. J Biol Chem 1998;273:29195-201.  Back to cited text no. 32    
33.Hata A, Lagna G, Massague J, Hemmati-Brivanlou A. Smad6 inhibits BMP/Smad1 signaling by specifically competing with the Smad4 tumor suppressor. Genes Dev 1998;12:186-97.  Back to cited text no. 33    
34.Ishisaki A, Yamato K, Hashimoto S, Nakao A, Tamaki K, Nonaka K, et al. Differential inhibition of Smad6 and Smad7 on bone morphogenetic protein- and Activin-mediated growth arrest and apoptosis in B cells. J Biol Chem 1999;274:13637-42.  Back to cited text no. 34    
35.Nakao A, Afrakhte M, Morén A, Nakayama T, Christian JL, Heuchel R, et al. Identification of Smad7, a TGFbeta-inducible antagonist of TGF-beta signalling. Nature 1997;389:631-5.  Back to cited text no. 35    
36.Ulloa L, Doody J, Massagué J. Inhibition of transforming growth factor-beta/SMAD signalling by the interferon-gamma/STAT pathway. Nature 1999;397:710-3.  Back to cited text no. 36    
37.Bitzer M, von Gersdorff G, Liang D, Dominguez-Rosales A, Beg AA, Rojkind M, et al. A mechanism of suppression of TGF-beta/SMAD signaling by NF-kappa B/RelA. Genes Dev 2000;14:187-97.  Back to cited text no. 37    
38.Hata A, Lo RS, Wotton D, Lagna G, Massagué J. Mutations increasing autoinhibition inactivate tumour suppressors Smad2 and Smad4. Nature 1997;388:82-7.  Back to cited text no. 38    
39.Dong C, Li Z, Alvarez R, Feng XH, Goldschmidt-Clermont PJ. Microtubule binding to Smads may regulate TGF beta activity. Mol Cell 2000;5:27-34.  Back to cited text no. 39    
40.Sasaki A, Masuda Y, Ohta Y, Ikeda K, Watanabe K. Filamin associates with Smads and regulates transforming growth factor-beta signaling. J Biol Chem 2001;276:17871-7.  Back to cited text no. 40    
41.Di Guglielmo GM, Le Roy C, Goodfellow AF, Wrana JL. Distinct endocytic pathways regulate TGF-beta receptor signalling and turnover. Nat Cell Biol 2003;5:410-21.  Back to cited text no. 41    
42.Lin HK, Bergmann S, Pandolfi PP. Cytoplasmic PML function in TGF-beta signalling. Nature 2004;431:205-11.  Back to cited text no. 42    
43.Tsukazaki T, Chiang TA, Davison AF, Attisano L, Wrana JL. SARA, a FYVE domain protein that recruits Smad2 to the TGFbeta receptor. Cell 1998;95:779-91.  Back to cited text no. 43    
44.Furuhashi M, Yagi K, Yamamoto H, Furukawa Y, Shimada S, Nakamura Y, et al. Axin facilitates Smad3 activation in the transforming growth factor beta signaling pathway. Mol Cell Biol 2001;21:5132-41.  Back to cited text no. 44    
45.Hocevar BA, Smine A, Xu XX, Howe PH. The adaptor molecule Disabled-2 links the transforming growth factor beta receptors to the Smad pathway. EMBO J 2001;20:2789-801.  Back to cited text no. 45    
46.Onichtchouk D, Chen YG, Dosch R, Gawantka V, Delius H, Massague J, et al. Silencing of TGF-beta signalling by the pseudoreceptor BAMBI. Nature 1999;401:480-5.  Back to cited text no. 46    
47.Zhu H, Kavsak P, Abdollah S, Wrana JL, Thomsen GH. A SMAD ubiquitin ligase targets the BMP pathway and affects embryonic pattern formation. Nature 1999;400:687-93.  Back to cited text no. 47    
48.Lin X, Liang M, Feng XH. Smurf2 is a ubiquitin E3 ligase mediating proteasome-dependent degradation of Smad2 in transforming growth factor-beta signaling. J Biol Chem 2000;275:36818-22.  Back to cited text no. 48    
49.Itoh F, Asao H. Sugamura K, Heldin CH, ten Dijke P, Itoh S. Promoting bone morphogenetic protein signaling through negative regulation of inhibitory Smads. EMBO J 2001;20:4132-42.  Back to cited text no. 49    
50.Liu X, Sun Y, Weinberg RA, Lodish HF. Ski/Sno and TGF-beta signaling. Cytokine Growth Factor Rev 2001;12:1-8.  Back to cited text no. 50    
51.Kretzschmar M, Doody J, Massague J. Opposing BMP and EGF signaling pathways converge on the TGF-beta family mediator Smad1. Nature 1997;389:618-22.  Back to cited text no. 51    
52.Wicks SJ, Lui S, Abdel-Wahab N, Mason RM, Chantry A. Inactivation of smad-transforming growth factor beta signaling by Ca(2+)-calmodulin-dependent protein kinase II. Mol Cell Biol 2000;20:8103-11.  Back to cited text no. 52    
53.Yakymovych I, Ten Dijke P, Heldin CH, Souchelnytskyi S. Regulation of Smad signaling by protein kinase C. FASEB J 2001;15:553-5.  Back to cited text no. 53    
54.Fukuchi M, Imamura T, Chiba T, Ebisawa T, Kawabata M, Tanaka K, et al. Ligand-dependent degradation of Smad3 by a ubiquitin ligase complex of ROC1 and associated proteins. Mol Biol Cell 2001;12:1431-43.  Back to cited text no. 54    
55.Izzi L, Attisano L. Regulation of the TGF-beta signalling pathway by ubiquitin-mediated degradation. Oncogene 2004;23:2071-8.  Back to cited text no. 55    
56.Watanabe M, Masuyama N, Fukuda M, Nishida E. Regulation of intracellular dynamics of Smad4 by its leucine-rich nuclear export signal. EMBO Rep 2000;1:176-82.  Back to cited text no. 56    
57.Howell M, Itoh F, Pierreux CE, Valgeirsdottir S, Itoh S, ten Dijke P, et al. Xenopus Smad4beta is the co-Smad component of developmentally regulated transcription factor complexes responsible for induction of early mesodermal genes. Dev Biol 1999;214:354-69.  Back to cited text no. 57    
58.Itoh S, Landstrom M, Hermansson A, Itoh F, Heldin CH, Heldin NE, et al. Transforming growth factor beta1 induces nuclear export of inhibitory Smad7. J Biol Chem 1998;273:29195-201.  Back to cited text no. 58    
59.Kurisaki A, Kose S, Yoneda Y, Heldin CH, Moustakas A. Transforming growth factor-beta induces nuclear import of Smad3 in an importin-beta1 and Ran-dependent manner. Mol Biol Cell 2001;12:1079-91.  Back to cited text no. 59    
60.Pierreux CE, Nicolás FJ, Hill CS. Transforming growth factor beta-independent shuttling of Smad4 between the cytoplasm and nucleus. Mol Cell Biol 2000;20:9041-54.  Back to cited text no. 60    
61.Xiao Z, Liu X, Henis YI, Lodish HF. A distinct nuclear localization signal in the N terminus of Smad 3 determines its ligand induced nuclear translocation. Proc Natl Acad Sci USA 2000;97:7853-8.  Back to cited text no. 61    
62.Xu L, Kang Y, Cöl S, Massagué J. Smad2 nucleocytoplasmic shuttling by nucleoporins CAN/Nup214 and Nup153 feeds TGF-beta signaling complexes in the cytoplasm and nucleus. Mol Cell 2002;10:271-82.  Back to cited text no. 62    
63.Xu L, Alarcón C, Cöl S, Massagué J. Distinct domain utilization by Smad3 and Smad4 for nucleoporin interaction and nuclear import. J Biol Chem 2003;278:42569-77.  Back to cited text no. 63    
64.Chen HB, Rud JG, Lin K, Xu L. Nuclear Targeting of Transforming Growth Factor-b-activated Smad Complexes. J Biol Chem 2005;280:21329-36.  Back to cited text no. 64    
65.Inman GJ, Nicolas FJ, Hill CS. Nucleocytoplasmic shuttling of Smads 2, 3, and 4 permits sensing of TGF-beta receptor activity. Mol Cell 2002;10:283-94.  Back to cited text no. 65    
66.Zawel L, Dai JL, Buckhaults P, Zhou S, Kinzler KW, Vogelstein B, et al. Human Smad3 and Smad4 are sequence-specific transcription activators. Mol Cell 1998;1:611-7.  Back to cited text no. 66    
67.Denissova NG, Pouponnot C, Long J, He D, Liu F. Transforming growth factor beta -inducible independent binding of SMAD to the Smad7 promoter. Proc Natl Acad Sci USA 2000;97:6397-402.  Back to cited text no. 67    
68.Dennler S, Itoh S, Vivien D, ten Dijke P, Huet S, Gauthier JM. Direct binding of Smad3 and Smad4 to critical TGF beta-inducible elements in the promoter of human plasminogen activator inhibitor-type 1 gene. EMBO J 1998;17:3091-100.  Back to cited text no. 68    
69.Shi Y, Wang YF, Jayaraman L, Yang H, Massagué J, Pavletich NP. Crystal structure of a Smad MH1 domain bound to DNA: Insights on DNA binding in TGF-beta signaling. Cell 1998;94:585-94.  Back to cited text no. 69    
70.Labbé E, Silvestri C, Hoodless PA, Wrana JL, Attisano L. Smad2 and Smad3 positively and negatively regulate TGF beta-dependent transcription through the forkhead DNA-binding protein FAST2. Mol Cell 1998;2:109-20.  Back to cited text no. 70    
71.Dennler S, Huet S, Gauthier JM. A short amino-acid sequence in MH1 domain is responsible for functional differences between Smad2 and Smad3. Oncogene 1999;18:1643-8.  Back to cited text no. 71    
72.Yagi K, Goto D, Hamamoto T, Takenoshita S, Kato M, Miyazono K. Alternatively-spliced variant of Smad2 lacking exon 3: Comparison with wild-type Smad2 and Smad3. J Biol Chem 1999;274:703-9.  Back to cited text no. 72    
73.Friess H, Yamanaka Y, Büchler M, Ebert M, Beger HG, Gold LI, et al. Enhanced expression of transforming growth factor-beta isoforms in pancreatic cancer correlates with decreased survival. Gastroenterology 1993;105:1846-56.  Back to cited text no. 73    
74.Lu Z, Friess H, Graber HU, Guo X, Schilling M, Zimmermann A, et al. Presence of two signaling TGF- beta receptors in human pancreatic cancer correlates with advanced tumor stage. Dig Dis Sci 1997;42:2054-63.  Back to cited text no. 74    
75.Hahn SA, Schutte M, Hoque AT, Moskaluk CA, da Costa LT, Rozenblum E, et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science 1996;271:350-3.  Back to cited text no. 75    
76.Kleeff J, Maruyama H, Ishiwata T, Sawhney H, Friess H, Büchler MW, et al. Bone morphogenetic protein 2 exerts diverse effects on cell growth in vitro and is expressed in human pancreatic cancer in vivo. Gastroenterology 1999;116:1202-16.  Back to cited text no. 76    
77.Levy L, Hill CS. Smad4 dependency defines two classes of transforming growth factor {beta} (TGF-{beta}) target genes and distinguishes TGF-{beta}-induced epithelial-mesenchymal transition from its antiproliferative and migratory responses. Mol Cell Biol 2005;25:8108-25.  Back to cited text no. 77    
78.Chow JY, Quach KT, Cabrera BL, Cabral JA, Beck SE, Carethers JM. RAS/ERK modulates TGFbeta-regulated PTEN expression in human pancreatic adenocarcinoma cells. Carcinogenesis 2007;28:2321-7.  Back to cited text no. 78    
79.Ottaviano AJ, Sun L, Ananthanarayanan V, Munshi HG. Extracellular matrix-mediated membrane-type 1 matrix metalloproteinase expression in pancreatic ductal cells is regulated by transforming growth factor-beta1. Cancer Res 2006;66:7032-40.  Back to cited text no. 79    
80.Chow JY, Ban M, Wu HL, Nguyen F, Huang M, Chung H, et al. TGF-beta downregulates PTEN via activation of NF-kappaB in pancreatic cancer cells. Am J Physiol Gastrointest Liver Physiol 2010;298:G275-82.  Back to cited text no. 80    
81.Zhao S, Venkatasubbarao K, Lazor JW, Sperry J, Jin C, Cao L, et al. Inhibition of STAT3 Tyr705 phosphorylation by Smad4 suppresses transforming growth factor beta-mediated invasion and metastasis in pancreatic cancer cells. Cancer Res 2008;68:4221-8.  Back to cited text no. 81    
82.Chow JY, Dong H, Quach KT, Van Nguyen PN, Chen K, Carethers JM. TGF-beta mediates PTEN suppression and cell motility through calcium-dependent PKC-alpha activation in pancreatic cancer cells. Am J Physiol Gastrointest Liver Physiol 2008;294:G899-905.  Back to cited text no. 82    
83.Subramanian G, Schwarz RE, Higgins L, McEnroe G, Chakravarty S, Dugar S, et al. Targeting endogenous transforming growth factor beta receptor signaling in SMAD4-deficient human pancreatic carcinoma cells inhibits their invasive phenotype1. Cancer Res 2004;64:5200-11.  Back to cited text no. 83    
84.Gaspar NJ, Li L, Kapoun AM, Medicherla S, Reddy M, Li G, et al. Inhibition of transforming growth factor beta signaling reduces pancreatic adenocarcinoma growth and invasiveness. Mol Pharmacol 2007;72:152-61.  Back to cited text no. 84    
85.Gadir N, Jackson DN, Lee E, Foster DA. Defective TGF-beta signaling sensitizes human cancer cells to rapamycin. Oncogene 2008;27:1055-62.  Back to cited text no. 85    
86.Markowitz S, Wang J, Myeroff L, Parsons R, Sun L, Lutterbaugh J, et al. Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science 1995;268:1336-8.  Back to cited text no. 86    
87.Kim SJ, Im YH, Markowitz SD, Bang YJ. Molecular mechanisms of inactivation of TGF-beta receptors during carcinogenesis. Cytokine Growth Factor Rev 2000;11:159-68.  Back to cited text no. 87    
88.Adrian K, Strouch MJ, Zeng Q, Barron MR, Cheon EC, Honasoge A, et al. Tgfbr1 haploinsufficiency inhibits the development of murine mutant Kras-induced pancreatic precancer. Cancer Res 2009;69:9169-74.  Back to cited text no. 88    
89.Grady WM, Rajput A, Myeroff L, Liu DF, Kwon KH, Willis J, et al. Mutation of the Type II Transforming Growth Factor-ß Receptor Is Coincident with the Transformation of Human Colon Adenomas to Malignant Carcinomas. Cancer Res 1998;58:3101-4.  Back to cited text no. 89    
90.Kano MR, Bae Y, Iwata C, Morishita Y, Yashiro M, Oka M, et al. Improvement of cancer-targeting therapy, using nanocarriers for intractable solid tumors by inhibition of TGF- ß signaling. Proc Natl Acad Sci USA 2007;104:3460-5.  Back to cited text no. 90    
91.van Heek T, Rader AE, Offerhaus GJ, McCarthy DM, Goggins M, Hruban RH, et al. K-ras, p53, and DPC4 (MAD4) alterations in fine-needle aspirates of the pancreas: A molecular panel correlates with and supplements cytologic diagnosis. Am J Clin Pathol 2002;117:755-65.  Back to cited text no. 91    
92.Schutte M, Hruban RH, Hedrick L, Cho KR, Nadasdy GM, Weinstein CL, et al. DPC4 gene in various tumor types. Cancer Res 1996;56:2527-30.  Back to cited text no. 92    
93.Hruban RH, Wilentz RE, Goggins M, Offerhaus GJ, Yeo CJ, Kern SE. Pathology of incipient pancreatic cancer. Ann Oncol 1999;10:9-11.  Back to cited text no. 93    
94.Hruban RH, Offerhaus GJ, Kern SE, Goggins M, Wilentz RE, Yeo CJ. Tumor-suppressor genes in pancreatic cancer. J Hepatobiliary Pancreat Surg 1998;5:383-91.  Back to cited text no. 94    
95.Iacobuzio-Donahue CA, Song J, Parmiagiani G, Yeo CJ, Hruban RH, Kern SE. Missense mutations of MADH4: Characterization of the mutational hot spot and functional consequences in human tumors. Clin Cancer Res 2004;10:1597-604.  Back to cited text no. 95    
96.Maurice D, Pierreux CE, Howell M, Wilentz RE, Owen MJ, Hill CS. Loss of Smad4 function in pancreatic tumors: C-terminal truncation leads to decreased stability. J Biol Chem 2001;276:43175-81.  Back to cited text no. 96    
97.Xu J, Attisano L. Mutations in the tumor suppressors Smad2 and Smad4 inactivate transforming growth factor-beta signaling by targeting Smads to the ubiquitin-proteasome pathway. Proc Natl Acad Sci USA 2000;97:4820-5.  Back to cited text no. 97    
98.Hruban RH, Goggins M, Parsons J, Kern SE. Progression model for pancreatic cancer. Clin Cancer Res 2000;6:2969-72.  Back to cited text no. 98    
99.Iacobuzio-Donahue CA, Fu B, Yachida S, Luo M, Abe H, Henderson CM, et al. DPC4 gene status of the primary carcinoma correlates with patterns of failure in patients with pancreatic cancer. J Clin Oncol 2009;27:1806-13.  Back to cited text no. 99    
100.Xu X, Ehdaie B, Ohara N, Yoshino T, Deng CX. Synergistic action of Smad4 and Pten in supressing pancreatic ductal adenocarcinoma formation in mice. Oncogene 2010;29:674-86.  Back to cited text no. 100    
101.Bardeesy N, Cheng KH, Berger JH, Chu GC, Pahler J, Olson P, et al. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes Dev 2006;20:3130-46.  Back to cited text no. 101    
102.Hua Z, Zhang YC, Hu XM, Jia ZG. Loss of DPC4 expression and its correlation with clinicopathological parameters in pancreatic carcinoma. World J Gastroenterol 2003;9:2764-7.  Back to cited text no. 102    
103.Blackford A, Serrano OK, Wolfgang CL, Parmigiani G, Jones S, Zhang X, et al. SMAD4 gene mutations are associated with poor prognosis in pancreatic cancer. Clin Cancer Res 2009;15:4674-9.  Back to cited text no. 103    
104.Ali S, Cohen C, Little JV, Sequeira, JH, Mosunjac MB, Siddiqui MT. The utility of SMAD4 as a diagnostic immunohistochemical marker for pancreatic adenocarcinoma, and its expression in other solid tumors. Diagn Cytopathol 2007;35:644-8.  Back to cited text no. 104    
105.Yasutome M, Gunn J, Korc M. Restoration of Smad4 in BxPC3 pancreatic cancer cells attenuates proliferation without altering angiogenesis. Clin Exp Metastasis 2005;22:461-73.  Back to cited text no. 105    
106.Shen W, Tao GQ, Li DC, Zhu XG, Bai X, Cai B. Inhibition of pancreatic carcinoma cell growth in vitro by DPC4 gene transfection. World J Gastroenterol 2008;14:6254-60.  Back to cited text no. 106    
107.Wang H, Han H, Von Hoff DD. Identification of an agent selectively targeting DPC4 (deleted in pancreatic cancer locus 4)-deficient pancreatic cancer cells. Cancer Res 2006;66:9722-30.  Back to cited text no. 107    
108.Wang H, Stephens B, Von Hoff DD, Han H. Identification and characterization of a novel anticancer agent with selectivity against deleted in pancreatic cancer locus 4 (DPC4)-deficient pancreatic and colon cancer cells. Pancreas 2009;38:551-7.  Back to cited text no. 108    
109.Kleeff J, Ishiwata T, Maruyama H, Friess H, Truong P, Buchler MW, et al. The TGF-beta signaling inhibitor Smad7 enhances tumorigenicity in pancreatic cancer. Oncogene 1999;18:5363-72.  Back to cited text no. 109    
110.Guo J, Kleeff J, Zhao Y, Li J, Giese T, Esposito I, et al. Yes-associated protein (YAP65) in relation to Smad7 expression in human pancreatic ductal adenocarcinoma. Int J Mol Med 2006;17:761-7.  Back to cited text no. 110    
111.Arnold NB, Ketterer K, Kleef J, Friess H, Buchler MW, Korc M. Thioredoxin is downstream of Smad7 in a pathway that promotes growth and suppresses cisplatin-induced apoptosis in pancreatic cancer. Cancer Res 2004;64:3599-606.  Back to cited text no. 111    
112.Wang P, Fan J, Chen Z, Meng ZQ, Luo JM, Lin JH, et al. Low-level expression of Smad7 correlates with lymph node metastasis and poor prognosis in patients with pancreatic cancer. Ann Surg Oncol 2009;16:826-35.  Back to cited text no. 112    
113.Ellenrieder V, Buck A, Harth A, Jungert K, Buchholz M, Adler G, et al. KLF11 mediates a critical mechanism in TGF-beta signaling that is inactivated by Erk-MAPK in pancreatic cancer cells. Gastroenterology 2004;127:607-20.  Back to cited text no. 113    
114.Arnold NB, Korc M. Smad7 abrogates transforming growth factor-beta1-mediated growth inhibition in COLO-357 cells through functional inactivation of the retinoblastoma protein. J Biol Chem 2005;280:21858-66.  Back to cited text no. 114    
115.Jonson T, Gorunova L, Dawiskiba S, Andren-Sandberg A, Stenman G, ten Dijke P, et al. Molecular analyses of the 15q and 18q SMAD genes in pancreatic cancer. Genes Chromosomes Cancer 1999;24:62-71.  Back to cited text no. 115    

Copyright 2011 - Indian Journal of Cancer


The following images related to this document are available:

Photo images

[cn11091f4.jpg] [cn11091f5.jpg] [cn11091f3.jpg] [cn11091f2.jpg] [cn11091f1.jpg]
Home Faq Resources Email Bioline
© Bioline International, 1989 - 2024, Site last up-dated on 01-Sep-2022.
Site created and maintained by the Reference Center on Environmental Information, CRIA, Brazil
System hosted by the Google Cloud Platform, GCP, Brazil