Next Article in Journal
Effect of Exercise on Sarcopenia among Cancer Survivors: A Systematic Review
Next Article in Special Issue
Targeting Mitochondrial COX-2 Enhances Chemosensitivity via Drp1-Dependent Remodeling of Mitochondrial Dynamics in Hepatocellular Carcinoma
Previous Article in Journal
Association between Sarcopenia and Immediate Complications and Mortality in Patients with Oral Cavity Squamous Cell Carcinoma Undergoing Surgery
Previous Article in Special Issue
High Dose Thoracic Re-Irradiation and Chemo-Immunotherapy for Centrally Recurrent NSCLC
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Shifting the Focus of Signaling Abnormalities in Colon Cancer

Genetics Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(3), 784; https://doi.org/10.3390/cancers14030784
Submission received: 11 January 2022 / Revised: 26 January 2022 / Accepted: 30 January 2022 / Published: 3 February 2022
(This article belongs to the Special Issue Feature Paper from Journal Reviewers)

Abstract

:

Simple Summary

The major signaling pathways in colon cancer are WNT, RAS, and TGF-β. Components of these pathways are mutated in the majority of colon cancers, resulting in aberrantly high or low activity of the pathway. The functional consequences of the mutations reflect the behavior of these signaling pathways in intestinal stem cells. To better understand the roles of each pathway, we cover the basic function as well as points of intersection between the different pathways, to describe how they function individually, as well as together, to regulate cell proliferation.

Abstract

Colon cancer tumorigenesis occurs incrementally. The process involves the acquisition of mutations which typically follow an established pattern: activation of WNT signaling, activation of RAS signaling, and inhibition of TGF-β signaling. This arrangement recapitulates, to some degree, the stem cell niche of the intestinal epithelium, which maintains WNT and EGF activity while suppressing TGF-β. The resemblance between the intestinal stem cell environment and colon cancer suggests that the concerted activity of these pathways generates and maintains a potent growth-inducing stimulus. However, each pathway has a myriad of downstream targets, making it difficult to identify which aspects of these pathways are drivers. To address this, we utilize the cell cycle, the ultimate regulator of cell proliferation, as a foundation for cross-pathway integration. We attempt to generate an overview of colon cancer signaling patterns by integrating the major colon cancer signaling pathways in the context of cell replication, specifically, the entrance from G1 into S-phase.

1. Introduction

Cancer is a disease in which cells of the body reject the commands of their tissue. This disobedience is a result of the disruption of the signaling pathways, which are no longer able to balance the activities of the cell nor effectively communicate with the extracellular environment. Signaling pathways are disrupted through a myriad of causes, though the most common is the mutation of a gene which produces a critical regulatory protein in the signaling cascade. The mutation severs the connection between the stimulus and its downstream response, because one of the proteins required to relay the appropriate signal is unable to do so. Thereby, the cell loses control over a specific aspect of its behavior and the output of the pathway becomes relatively constant, as opposed to dynamic. This influences the resting state of the cell as well as any dynamic cellular responses which require input of this (now defunct) pathway. As the mutant cell multiplies, more mutations can occur, providing the basis for selection, which results in disease progression as the cancer cell further deviates from normal cell function. Eventually, the cancer cell accumulates a sufficient number of driver mutations, approximately 5–7 in colon cancer, in various pathways, which render it ignorant of its surrounding environment. This forms the basis for metastasis, as the cancer cell is able to traverse the surrounding tissues, withstand secreted growth inhibitory signals, as well as grow in a remote site of the body, independent of the signaling pathway requirements of its tissue of origin.

1.1. Signaling in the Healthy Intestines

In the healthy intestinal epithelium, the WNT, EGF, Notch, and BMP pathways maintain homeostasis [1]. The intestinal stem cells are maintained by high levels of WNT, EGF, and Notch, as well as low levels of BMP signaling [2,3,4,5]. The activity of these pathways is maintained by signals from the mesenchyme surrounding the stem cell compartment, as well as other cells (such as Paneth or cKit+/Reg4+) which are intermingled with the stem cells [6,7,8]. As the stem cell progeny leave the compartment and ascend the crypt, the WNT, EGF, and Notch levels decrease, while the activity of BMP increases [9,10,11]. The changing levels of these signaling pathways results in the differentiation of the stem cells, now transit amplifying cells, into the various cell types necessary to perform the intestinal functions. Cell type is, therefore, an emergent property determined by the output of several key signaling pathways (given the context of the cell type and its epigenetic status).

1.2. Signaling in Colon Cancer

In colon cancer, the activities of the previously mentioned pathways are altered by mutation and closely mirror the signaling pathway arrangement found within the intestinal stem cell niche, i.e., high WNT, high EGF, and low BMP. However, Notch signaling does not appear to be heavily targeted by mutation in colon cancer [12]. Given the stochastic nature of mutation, higher mutation prevalence for specific genes is indicative of increased cellular fitness of the mutation-harboring cell compared to the surrounding normal intestinal cells, i.e., selection. Recapitulation of the stem cell state by the cancer cell indicates that the cooperativity of these pathways results in an intestinal cell with indefinite replicative potential. However, even the increased activity of a single pathway (WNT or EGF) through mutation is sufficient to result in aberrant growth and adenoma formation [13,14,15], though more faithful recapitulation of the stem cell state, i.e., increased WNT and EGF, and low BMP signaling, results in a cancer cell with malignant potential [13,16,17]. It is, therefore, the combined influence of these signaling pathways, rather than the actions of any one pathway in particular, that is necessary for the transition to malignant disease. However, cooperation between pathways does not imply synergism.

2. WNT Signaling

The WNT signaling pathway is an ancient mechanism necessary for guiding development and maintaining tissue homeostasis throughout multicellular life [18]. In the intestinal epithelium, canonical WNT signaling is crucial for stem cell maintenance, and thereby, homeostasis, while WNT signaling loss abrogates the intestinal proliferative compartment [2,19]. It is no coincidence that the signaling pathway most critical for maintaining the intestinal stem cell state is also the most frequently mutated in colon cancer.

2.1. WNT Production

WNT signaling begins with the production of a small (~40 kDa), cysteine-rich protein, referred to as a WNT [20,21]. Humans produce 19 different WNTs, which perform various developmental functions with limited functional redundancy [22,23,24,25,26,27]. As part of their maturation, WNTs are lipid-modified by the addition of palmitoleic acid, which is appended by the O-acyltransferase enzyme, Porcupine, within the endoplasmic reticulum (ER) [28,29,30,31,32,33,34]. WNT acylation is required for binding to WLS (Wntless/Evi), an essential component of the WNT secretory pathway, while inhibition of WNT acylation results in WNT accumulation within the ER [34,35,36,37,38,39,40]. Following the binding of WNT to WLS, the WNT is transported from the Golgi to the plasma membrane for secretion and transfer to its target cells via WNT-binding proteins, exosomes, or cytonemes [41,42,43,44,45]. WNT acylation renders it hydrophobic and limits diffusion through the extracellular space [30].

2.2. WNT Binding

Once at the target cell membrane, the WNT ligand binds to a Frizzled (FZD) receptor as well as a Low-Density Lipoprotein Receptor-Related Protein (LRP) to form a FZD–LRP coreceptor, thus initiating canonical WNT signaling [46,47,48,49,50,51] (Figure 1, left). Humans have 10 FZDs, to which the 19 WNTs bind promiscuously [52,53,54]. The LRPs associated with WNT signaling are LRP5 and LRP6 [47,48,49]. Importantly, the palmitoleic acid appended to the WNT is also required for FZD and LRP binding [50,51,55]. Lipid modification is, therefore, necessary for WNT secretion, as well as receptor binding (which leads to active WNT signaling). Taken together, the hydrophobicity of the acylated WNT severely limits its diffusion in the extracellular space, enforcing strict control over which cells receive a productive WNT ligand [30,34,56]. However, additional regulatory mechanisms exist to modulate WNT–FZD–LRP binding. These include NOTUM, which functions by WNT deacylation; DKK, which functions by binding LRP and blocking WNT binding; or sFRP, which sequesters the WNT itself [55,57,58,59]. Additionally, FZD may be ubiquitinated and internalized by the transmembrane E3 ubiquitin ligases, RNF43 and ZNRF3, which block WNT signaling activity [60,61]. However, RNF43 and ZNRF3 activity can be inhibited by the intestinal stem cell marker, LGR5, in the presence of its ligand, RSPO [19,60,61,62]. RSPO and LGR5, therefore, permit WNT signal transduction by maintaining FZD at the plasma membrane for WNT binding through inhibition of RNF43 and ZNRF3.

2.3. WNT Signal Transduction

Upon successful WNT–FZD–LRP binding, the cytoplasmic domains of FZD and LRP become associated, which results in the recruitment of DVL and AXIN and the phosphorylation of LRP by CKIγ and GSK3, in an incompletely understood mechanism [63,64,65,66,67]. AXIN is a core component of the Destruction Complex (DC), and thus, sequestration of AXIN at FZD–LRP results in sequestration of the DC [68]. The DC is comprised of AXIN, APC, CKIα, GSK3, β-catenin, and β-TrCP [69,70,71,72,73,74]. DC sequestration inhibits β-catenin ubiquitination, but not phosphorylation, halting β-catenin turnover and saturating the DCs [68]. Since β-catenin is consistently produced, DC saturation results in β-catenin accumulation in the cytoplasm, which then migrates to the nucleus [68]. Nuclear migration of β-catenin may be facilitated by RAPGEF5 [75]. Once within the nucleus, β-catenin competes with the transcriptionally repressive TLE factors to bind with the TCF/LEF family of transcription factors, which are the canonical WNT transcription factors [76,77,78,79]. Association of the potent transcriptional activation domain of β-catenin with the TCF/LEF transcription factors results in the activation of WNT target gene expression [78,79] (Figure 1, left). The role of each TCF/LEF factor is largely distinct, though there remains limited functional redundancy, such as between TCF1 and LEF1 in the lymphoid lineage [80,81,82,83,84,85,86,87,88]. In the colon, TCF4, encoded by TCF7L2, is necessary for intestinal development in mouse embryos as well as intestinal maintenance in adult mice [2,83,84]. While it is difficult to comprehensively identify all crucial downstream TCF4 targets in the intestine; MYC, CCND1, and AXIN2 are generally considered to be key transcriptional targets of an active WNT signal and serve as WNT reporter genes [89,90,91].

2.4. Inactive WNT Signaling

In the WNT OFF state, the FZD and LRP receptors remain unbound by WNT, and therefore, remain unassociated in the cytoplasm. FZD receptors are internalized by ZNRF3 and RNF43, blocking accessibility to WNT [60,61]. The DC remains, unrestricted, in the cytoplasm. APC facilitates β-catenin retrieval from the nucleus, as APC contains nuclear import and export domains, as well as incorporation of β-catenin into the DC via the 15- and 20-amino acid domains (β-catenin binding), and SAMP domains (AXIN binding) of APC [69,70,92,93,94,95,96]. Once within the DC, CKIα phosphorylates β-catenin at S45, which primes it for subsequent phosphorylation by GSK3 at T41, S37, and S33 [71,97]. The phosphorylated residues form a motif recognized by β-TrCP, which ubiquitinates β-catenin, targeting it for destruction by the proteasomal machinery [71,72,98]. β-catenin is rapidly degraded and does not accumulate in the cytoplasm, nor does it migrate to the nucleus. The TCF/LEF factors, therefore, remain bound by the TLE family of transcriptional repressors and inhibit WNT target gene expression [99,100] (Figure 1, center).

2.5. WNT Signaling in Colon Cancer

The majority (93%) of colon cancers harbor mutations in the WNT signaling pathway, which result in altered WNT signaling activity [12]. The majority of these mutations arise in APC, resulting in a truncated APC protein which can not efficiently facilitate the degradation of β-catenin, resulting in constitutively active WNT signaling [101,102,103,104] (Figure 1, right). APC appears to follow Knudsen’s two-hit theory, as both alleles have been found to be mutated in the majority of adenomas from patients with familial adenomatous polyposis (FAP) [105,106,107,108,109]. Stabilizing mutations in β-catenin also occur, rendering it refractory to phosphorylation, and therefore degradation, resulting in constitutive WNT signaling [103]. Together, APC and CTNNB1 account for the majority of WNT mutations in colon cancer [12]. These mutations sever the dependence of the cell upon WNT ligands for activated WNT signaling activity, and mutant cells no longer require signals from the mesenchyme or Paneth cells to maintain active WNT. These mutations, therefore, result in the protracted maintenance of a WNT signaling phenotype outside of the intestinal stem cell niche. The APC and CTNNB1 mutations are mutually exclusive, indicating that they serve equivalent roles in WNT signaling activation [110]. Unfortunately, β-catenin accesses a wide array of target genes through its association with the TCF/LEF factors, complicating our understanding of the downstream consequences of high levels of nuclear β-catenin. However, MYC appears to be the single most important target, as MYC deletion rescues the consequences of APC loss, despite high levels of β-catenin [111].

3. RAS Signaling

RAS signaling is crucial for mediating cellular growth, differentiation, and survival [112]. As a result of their growth and anti-apoptotic functions, members of the RAS signaling pathway, most notably K-RAS, are frequently mutated in cancer [113]. Approximately 20% of all cancers and 50% of all colon cancers harbor an activating mutation in RAS [112,113]. In colon cancer these mutations typically occur following APC mutation during tumorigenesis, though this order is not invariant [113].

3.1. RAS Signaling Receptors and Ligands

RAS signaling relies on the actions of the Epidermal Growth Factor Receptors (EGFR), which are Receptor Tyrosine Kinases (RTKs), for signal transduction through the plasma membrane. The EGFR family consists of four members: ERBB1 (EGFR), ERBB2 (HER2), ERBB3 (HER3), and ERBB4 (HER4) [114,115,116,117]. EGFR is capable of binding a variety of ligands, including Epidermal Growth Factor (EGF), Transforming Growth Factor-α (TGF-α), and heparin-binding EGF-like growth factor (HB-EGF), as well as several others [114,118,119,120,121,122]. ERBB2 has no known ligands and ERBB3 and ERBB4 bind to the neuregulins (NRG) [123,124]. The major RAS ligands in the intestinal epithelium are EGF and TGF-α [6]. EGF–EGFR binding results in EGFR dimerization into homo or heterodimers with other members of the EGFR family, such as ERBB2 [125,126,127] (Figure 2, left). The ability to form receptor heterodimers allows for a wider variety of ligand binding and diversifies the intracellular responses based on the participating receptors [128].

3.2. RAS Signal Transduction

Oligomerization of the EGFR family of receptors results in transphosphorylation of the dimerized, cytoplasmic receptor domains [129,130,131,132]. The phosphorylated residue Y1068 on EGFR forms a docking site for GRB2, via its SH2 domain, which recruits its binding partner, SOS, a RAS-specific Guanine nucleotide Exchange Factor (GEF) [133,134,135,136,137]. SOS activates the small guanosine triphosphatase (GTPase) RAS by inducing the exchange of GDP for GTP [138,139,140]. When activated (bound by GTP), RAS activates a variety of downstream processes involving PI3K, RALGDS, and PLCε, as well as the protein serine/threonine kinase RAF [112,141,142,143]. This review discusses the major downstream process, the MAPK cascade, which is initiated by RAF. Once activated by RAS, RAF phosphorylates, and by doing so activates, the mitogen-activated protein kinase kinase, MEK [144,145,146,147]. Subsequently, MEK phosphorylates ERK on its activation loop [146,147,148,149]. A fraction of ERK is transported to the nucleus following activation, resulting in the phosphorylation of both cytosolic and nuclear substrates [150,151,152,153]. Within the nucleus, ERK phosphorylates a variety of transcription factors, including MYC and members of the ETS family, such as ELK1 [154,155,156]. Phosphorylation of ELK1 allows it to form a ternary complex with serum response factor (SRF) at serum response elements (SREs) [155,156]. A notable transcriptional target of ELK1 is FOS, a member of the AP-1 transcription factor [155,156]. Additionally, SREs are present within many immediate early response genes [157]. RAS signaling therefore results in the expression of key cell-cycle regulatory proteins which promote proliferation and survival.

3.3. Inactive RAS Signaling

In the absence of EGF or an EGF-like ligand, EGFR does not dimerize, since the dimerization interface remains sequestered until EGF binding, which induces a conformational change in the receptor [132,158]. EGFR dimerization alters the conformation of the kinase domains which permits phosphorylation [159,160]. Following EGFR autophosphorylation, the EGF–EGFR complex is rapidly internalized and subsequently degraded, limiting the amount of RAS activation [161,162,163]. RAS proteins contain intrinsic GTPase activity and gradually exchange GTP for GDP, leading to their own inactivation, a process greatly facilitated by the GTPase-activating proteins (GAPs) [164,165,166] (Figure 2, center).

3.4. RAS Signaling in Colon Cancer

In colon cancer, K-RAS mutations, which occur almost exclusively in codons 12, 13, or 61, are found in approximately 50% of tumors [12,113]. These mutations alter the residues surrounding the GTP–GDP exchange site on RAS, inhibiting the ability of rasGAPs to hydrolyze GTP and return K-RAS to the GDP-bound state [167,168] (Figure 2, right). RAS signaling, therefore, remains persistently active in K-RAS mutant cells and significantly reduces the reliance of the cell upon EGF and EGFR for an active RAS growth signal. Mutations in K-RAS are mutually exclusive with mutations in N-RAS or B-RAF, implying similar regulatory functions [12,169]. Consistent activation of RAS signaling results in the phosphorylation of ERK targets, such as the ETS transcription factors, which are necessary for the transforming effects of RAS [155,156]. ELK1 phosphorylation leads to ternary complex formation and the expression of FOS [155,156]. The other major component of the AP-1 transcription factor, JUN is also mediated by RAS signaling; however, it relies upon the JNK cascade [170,171,172]. Tumors containing an activated RAS pathway display increased proliferation through the actions of the ETS factors and AP-1 [112].

4. TGF-β Signaling

TGF-β signaling influences a diverse array of cellular processes throughout the lifespan of the organism, including proliferation, differentiation, morphogenesis, and regeneration [173]. In the colonic epithelium, finely regulated gradients of TGF-β, alongside WNT, mediate differentiation of the intestinal stem cells and transit-amplifying cells [1]. In colon cancer, mutations which influence TGF-β are typically the last perturbation preceding malignant transformation [174].

4.1. TGF-β Ligands and Receptors

The TGF-β family of ligands constitutes the largest family of secreted morphogens (33 members) and can be divided into the TGFβ–Activin–Nodal and BMP subfamilies [175]. These ligands are produced by cleavage of a pro-domain, which releases the mature domain of the ligand [176,177]. The mature ligand dimerizes, which is essential for receptor activation [178] (Figure 3, left). These dimeric ligands bind to the serine/threonine kinase family of receptors [179,180,181]. Humans utilize seven Type I and five Type II receptors, which are dedicated to TGF-β signaling [182,183]. In the intestines, the major TGF-β ligands are BMP2 and BMP4, which bind the Type I receptors BMPRIA and BMPRIB as well as the Type II receptor, BMPRII; however, the TGF-β subfamily is active as well [184,185].

4.2. TGF-β Signal Transduction

The intracellular workhorses of the TGF-β signaling pathway are the SMAD proteins. There are three functional classes of SMADs: receptor-regulated (R-SMAD), co-mediator (Co-SMAD), and inhibitory (I-SMAD) [186]. The R-SMADs include SMAD1, 2, 3, 5, and 8, and the Co-SMAD is SMAD4, while the I-SMADs are SMAD6 and 7 [182,186]. SMAD2 and 3 transduce the TGF-β ligand message, while SMAD1, 5, and 8 transmit the BMP message. BMP ligand binding cooperatively brings together two Type I and Type II BMP receptors, forming a hetero-tetramer [186]. Complex formation allows the Type II receptor to phosphorylate the Type I receptor’s GS domain, facilitating the exchange of the inhibitory FKBP12 for an R-SMAD(1, 5, 8) [187,188,189,190]. R-SMAD localization to the receptor is facilitated by the protein SARA [191]. Upon R-SMAD binding, the Type I kinase domain phosphorylates the R-SMAD, after which it dissociates from the receptor [187,192,193]. Phosphorylated R-SMADs then form a hetero-trimeric complex with SMAD4, the Co-SMAD [194,195]. The SMAD complex then translocates to the nucleus, where it regulates target gene transcription alongside various nuclear cofactors [187,193,196]. These nuclear co-factors bind the R-SMAD, ensuring the specificity of the transcriptional response [182]. TGF-β target genes include p15Ink4b and p21Cip1, the cyclin-dependent kinase inhibitors which mediate the growth-repressive actions of TGF-β [197,198,199].

4.3. Inactive TGF-β Signaling

TGF-β signaling is regulated at several levels. At the ligand level, Noggin and Chordin, are both capable of binding to the BMP ligands, which prevents their association with the BMP receptors [200,201,202] (Figure 3, center). When the receptors remain unassociated, either through a lack of BMP ligand or the presence of Noggin or Chordin, FKBP12 binds to, and inhibits, the phosphorylation of the Type I receptors [203]. The R-SMAD is unable to bind to the Type I receptor and the Co-SMAD::R-SMAD complex is not formed. The I-SMADs also negatively regulate TGF-β signaling by competing with R-SMADs for receptor binding or with SMAD4 complex formation [204,205]. SMAD6 inhibits BMP signaling, while SMAD7 can inhibit either TGF-β or BMP signaling.

4.4. TGF-β Signaling in Colon Cancer

TGF-β signaling negatively regulates cell proliferation in the colon and is often mutated in colon cancer [13,206]. These mutations, which occur in TGFBRII, SMAD2, or SMAD4 inactivate the protein and abrogate the ability of TGF-β to regulate proliferation [206,207,208,209] (Figure 3, right). Mutations in SMAD4 (10%) are more common than mutations in SMAD2 (6%); however, in hypermutated tumors, mutations in TGFBRII were observed in 51% of tumors [12]. The SMAD4 mutations may occur within the MH2 domain, which inhibits the ability of SMAD4 to bind the R-SMAD and transfer to the nucleus, or may occur within the MH1 domain, which results in autoinhibition of the SMAD, which also inhibits SMAD complex formation [210,211]. Ultimately, biallelic mutation of the TGF-β signaling pathway components results in decreased regulation of cell growth and is typically the last mutation to occur prior to the transition to malignancy [212].

5. The Cell Cycle in Cancer

The cell cycle is initiated by progression of the cells past the restriction point in G1 [213,214]. Once this point has been passed, no additional external stimuli are required for progression through the cell cycle [213,214]. The restriction point represents the shift from a reliance on cyclin D-CDK4/6 for progression to S-phase, to a reliance on cyclin E-CDK2 [214,215,216]. The goal of the cancer cell is, therefore, to progress through the restriction point by activating cyclin E, which depends upon cyclin D.

6. WNT, RAS and TGF-β

The WNT, RAS, and TGF-β signaling pathways are frequently mutated in sporadic colon cancer [12,13]. WNT signaling is necessary for intestinal homeostasis throughout life, as loss of WNT signaling activity results in the loss of the proliferative compartment and breakdown of the intestinal epithelium in neonatal and adult mice [2,84,217]. However, when WNT signaling is aberrantly activated, the intestinal crypts exhibit exacerbated proliferation [218,219,220]. A similar, yet less drastic, effect occurs when EGF signaling is inhibited in the intestines, which induces stem cell quiescence, while EGF activation results in aberrant proliferation [221,222,223]. The role of TGF-β is to negatively regulate proliferation in the intestine, striking a balance with the proliferative actions of WNT and EGF. Loss of TGF-β regulation also results in aberrant proliferation [5,11]. Taken together, disruption of the WNT–EGF–TGFβ interaction, such as by an APC, K-RAS, or SMAD4 mutation, results in an imbalance of the signaling pathways and confers a proliferative advantage upon the mutation-harboring cell.
The aberrant activation of WNT signaling leads to the constitutive formation of β-catenin/TCF4 transcriptional complexes, which enforce a crypt progenitor phenotype [103,104,224]. The downstream target genes of TCF4 include MYC and CCND1, which both facilitate progression into S-phase [89,90]. However, MYC appears to be the major target of WNT signaling, as MYC deletion rescues APC loss, despite high levels of β-catenin [111]. MYC lies at the core of cell cycle progression and facilitates the expression of genes associated with nucleotide biosynthesis, ribosome biogenesis, RNA processing, cell cycle progression, and DNA replication [225,226]. Additionally, MYC also represses the expression of key cell cycle inhibitors such as CDKN1A (p21) and CDKN2B (p15) [227,228]. The ability of TGF-β to counteract the WNT signal is centered on MYC, as MYC expression is repressed by SMAD3, which is necessary for TGF-β-induced G1 arrest [229,230]. Furthermore, TGF-β expresses CDKN1A and CDKN2B, the genes which are inhibited by MYC [197,198]. WNT and TGF-β, therefore, compete to regulate cell cycle progression at the MYC level, with TGF-β dominance resulting in p21 and p15 production and cell cycle arrest, while WNT dominance results in CDKN1A and CDKN2B repression. The APC mutation appears to give WNT a slight advantage. The increased levels of MYC, induced by activating WNT mutations, are complemented by activated RAS signaling via K-RAS [231]. Phosphorylated ELK1 leads to expression of FOS, a member of the AP-1 transcription factor [155,156]. The remaining component of AP-1, JUN, is phosphorylated by the JNK cascade of RAS signaling, which is also active in colon cancer [231]. Once formed, AP-1 leads to proliferation and survival by expressing genes such as CCND1 and repressing TP53 [232,233,234,235]. Therefore, WNT and RAS cooperate to express CCND1, by the concerted efforts of β-catenin/TCF4 and AP-1. It has been noted that WNT alone does not appear sufficient to fully express CCND1 [236]. Additionally, RAS signaling, acting via ERK, leads to the phosphorylation of MYC and FOS, enhancing their transcriptional output [150,154]. Furthermore, ERK phosphorylates the SMAD proteins, which inhibits their nuclear accumulation and, therefore, their signaling capabilities [237,238]. Oncogenic RAS, therefore, aids WNT signaling in CCND1 expression via AP-1 and MYC phosphorylation, helps to weaken TGF-β signaling by SMAD phosphorylation via ERK, and represses TP53 via AP-1. The high levels of cyclin D, from WNT and RAS, bind to CDK4, a MYC target gene, allowing for the formation of cyclin D and CDK4 complexes [239]. CyclinD/CDK4 phosphorylates pRb, releasing the E2F factors [240,241]. MYC then functions alongside the E2F factors to facilitate binding at the E2F gene promoters, facilitating the activation of the E2F transcriptional program [242]. E2F target genes are necessary for DNA replication and the transition from G1 to S, such as CCNE1 [243]. Cyclin E binds to CDK2, which further phosphorylates pRb, leading to the complete release of the E2F factors, and transition to the growth factor-independent stages of the cell cycle [214,244]. The concerted actions of WNT and RAS, therefore, generate a potent proliferative signal through the combined function of βcat/TCF4, MYC, and RAS/AP-1. Despite the combination of WNT and RAS, TGF-β appears to retain some proliferation inhibitory functions, as demonstrated by selection of TGF-β mutations. Its ability to repress MYC expression, as well as express CDKN1A and CDKN2B, may be the last checks on proliferation, which upon mutation of SMAD4 or one of the TGF-β receptors, results in the complete release of MYC and unregulated proliferation of cancerous cells (Figure 4).

7. The Intestinal Microbiota

The microorganisms which reside in the colon are often overlooked in regard to their influence on colorectal cancer tumorigenesis. These organisms can generate a profound impact on the gut by the alteration of chemical species, interaction with intestinal cells, or by the release of toxins [245,246,247]. Given that intestinal polyps extrude into the lumen of the colon, an interaction between cells within the polyps and gut-resident microbes is expected. The pathogen, C. difficile secretes toxins which bind the FZD receptors, inhibiting their ability to bind a WNT ligand [248,249]. This inhibition of WNT signaling may result in extended wound-healing and inflammation. However, the actions of bacteria are not solely deleterious. Probiotic strains can compete against virulent bacterial species, protecting the intestines from inflammation and damage [250]. Evidently, much work is to be done to understand the capability of bacteria to influence intestinal health.

8. Conclusions

In colon cancer, the mutations of the WNT, RAS, and TGF-β signaling pathways recapitulate the signaling arrangement present in the intestinal stem cells. This stochastic recapitulation of a similar functional program indicates that the coordinated activities of these pathways results in a potent growth-inducing stimulus. These efforts are, at times, overlapping, such as the expression of CCND1 through βcat/TCF4 and AP-1, whereas other efforts are complementary, such as MYC expression by WNT and MYC phosphorylation by RAS. TGF-β antagonizes the influences of WNT and RAS on the cell cycle by down-regulating MYC and up-regulating the cell cycle regulators CDKN1A and CDKN2B. The ability of TGF-β to regulate the cell cycle relies on the SMAD transcription factors, which are phosphorylated by the activated RAS pathway to inhibit their nuclear translocation. As colon cancer develops, mutations in the WNT pathway lead to an increase of MYC, which results in small adenoma formation. The coupling of oncogenic RAS to deregulated WNT leads to the formation of a large adenoma, as βcat/TCF4 and MYC are complemented by AP-1 of RAS. Loss of TGF-β, the developmentally designated gatekeeper, allows for the unrestrictive action of MYC and RAS. Taken together, the WNT, RAS, and TGF-β mutations function cooperatively to form a malignant cell.

Author Contributions

M.A.B. and T.R. contributed to the conception/design, writing. All authors have read and agreed to the published version of the manuscript.

Funding

M.A.B. and T.R. are supported by the Intramural Research Program of the National Cancer Institute of the National Institutes of Health.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gehart, H.; Clevers, H. Tales from the crypt: New insights into intestinal stem cells. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 19–34. [Google Scholar] [CrossRef] [PubMed]
  2. Korinek, V.; Barker, N.; Moerer, P.; Van Donselaar, E.; Huls, G.; Peters, P.J.; Clevers, H. Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nat. Genet. 1998, 19, 379–383. [Google Scholar] [CrossRef] [PubMed]
  3. Sato, T.; Vries, R.G.; Snippert, H.J.; Van De Wetering, M.; Barker, N.; Stange, D.E.; Van Es, J.H.; Abo, A.; Kujala, P.; Peters, P.J.; et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 2009, 459, 262–265. [Google Scholar] [CrossRef] [PubMed]
  4. van Es, J.H.; van Gijn, M.E.; Riccio, O. Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature 2005, 435, 959–963. [Google Scholar] [CrossRef]
  5. Haramis, A.-P.G.; Begthel, H.; Born, M.; van Es, J.; Jonkheer, S.; Offerhaus, G.J.A.; Clevers, H. De Novo Crypt Formation and Juvenile Polyposis on BMP Inhibition in Mouse Intestine. Science 2004, 303, 1684–1686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Sato, T.; Van Es, J.H.; Snippert, H.J.; Stange, D.E.; Vries, R.G.; van den Born, M.; Barker, N.; Shroyer, N.F.; Van De Wetering, M.; Clevers, H. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature 2011, 469, 415–418. [Google Scholar] [CrossRef] [Green Version]
  7. Rothenberg, M.E.; Nusse, Y.; Kalisky, T.; Lee, J.J.; Dalerba, P.; Scheeren, F.; Lobo, N.; Kulkarni, S.; Sim, S.; Qian, D.; et al. Identification of a cKit+ Colonic Crypt Base Secretory Cell That Supports Lgr5+ Stem Cells in Mice. Gastroenterology 2012, 142, 1195–1205.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Sasaki, N.; Sachs, N.; Wiebrands, K.; Ellenbroek, S.; Fumagalli, A.; Lyubimova, A.; Begthel, H.; Born, M.V.D.; van Es, J.H.; Karthaus, W.R.; et al. Reg4+ deep crypt secretory cells function as epithelial niche for Lgr5+ stem cells in colon. Proc. Natl. Acad. Sci. USA 2016, 113, E5399–E5407. [Google Scholar] [CrossRef] [Green Version]
  9. Farin, H.F.; Jordens, I.; Mosa, H.F.F.M.H.; Basak, O.; Korving, J.; Tauriello, D.V.F.; De Punder, K.; Angers, S.; Peters, K.D.P.P.J.; Maurice, M.; et al. Visualization of a short-range Wnt gradient in the intestinal stem-cell niche. Nature 2016, 530, 340–343. [Google Scholar] [CrossRef]
  10. Tian, H.; Biehs, B.; Chiu, C.; Siebel, C.W.; Wu, Y.; Costa, M.; de Sauvage, F.J.; Klein, O.D. Opposing Activities of Notch and Wnt Signaling Regulate Intestinal Stem Cells and Gut Homeostasis. Cell Rep. 2015, 11, 33–42. [Google Scholar] [CrossRef] [Green Version]
  11. He, X.C.; Zhang, J.; Tong, W.G. BMP signaling inhibits intestinal stem cell self-renewal through suppression of Wnt-beta-catenin signaling. Nat. Genet. 2004, 36, 1117–1121. [Google Scholar] [CrossRef] [Green Version]
  12. Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature 2012, 487, 330–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Vogelstein, B.; Fearon, E.R.; Hamilton, S.R.; Kern, S.E.; Preisinger, A.C.; Leppert, M.; Smits, A.M.; Bos, J.L. Genetic Alterations during Colorectal-Tumor Development. N. Engl. J. Med. 1988, 319, 525–532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Powell, S.M.; Zilz, N.; Beazer-Barclay, Y.; Bryan, T.M.; Hamilton, S.R.; Thibodeau, S.N.; Vogelstein, B.; Kinzler, K.W. APC mutations occur early during colorectal tumorigenesis. Nature 1992, 359, 235–237. [Google Scholar] [CrossRef] [PubMed]
  15. Smith, A.J.; Stern, H.S.; Penner, M.; Hay, K.; Mitri, A.; Bapat, B.V.; Gallinger, S. Somatic APC and K-ras codon 12 mutations in aberrant crypt foci from human colons. Cancer Res. 1994, 54, 5527–5530. [Google Scholar]
  16. Matano, M.; Date, S.; Shimokawa, M.; Takano, A.; Fujii, M.; Ohta, Y.; Watanabe, T.; Kanai, T.; Sato, T. Modeling colorectal cancer using CRISPR-Cas9–mediated engineering of human intestinal organoids. Nat. Med. 2015, 21, 256–262. [Google Scholar] [CrossRef]
  17. Drost, J.; Van Jaarsveld, R.H.; Ponsioen, B.; Zimberlin, C.; Van Boxtel, R.; Buijs, A.; Sachs, N.; Overmeer, R.M.; Offerhaus, G.J.; Begthel, H.; et al. Sequential cancer mutations in cultured human intestinal stem cells. Nature 2015, 521, 43–47. [Google Scholar] [CrossRef]
  18. Kusserow, A.; Pang, K.; Sturm, C.; Hrouda, M.; Lentfer, J.; Schmidt, H.; Technau, U.; Von Haeseler, A.; Hobmayer, B.; Martindale, M.Q.; et al. Unexpected complexity of the Wnt gene family in a sea anemone. Nature 2005, 433, 156–160. [Google Scholar] [CrossRef]
  19. Barker, N.; Van Es, J.H.; Kuipers, J.; Kujala, P.; Van Den Born, M.; Cozijnsen, M.; Haegebarth, A.; Korving, J.; Begthel, H.; Peters, P.J.; et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 2007, 449, 1003–1007. [Google Scholar] [CrossRef]
  20. Nusse, R.; Varmus, H.E. Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell 1982, 31, 99–109. [Google Scholar] [CrossRef]
  21. Nusse, R.; Brown, A.; Papkoff, J.; Scambler, P.; Shackleford, G.; McMahon, A.; Moon, R.; Varmus, H. A new nomenclature for int-1 and related genes: The Wnt gene family. Cell 1991, 64, 231. [Google Scholar] [CrossRef]
  22. Miller, J.R. The Wnts. Genome Biol. 2002, 3, 3001. [Google Scholar]
  23. Stark, K.; Vainio, S.; Vassileva, G.; McMahon, A.P. Epithelial transformation of metanephric mesenchyme in the developing kidney regulated by Wnt-4. Nature 1994, 372, 679–683. [Google Scholar] [CrossRef] [PubMed]
  24. Monkley, S.J.; Delaney, S.J.; Pennisi, D.J.; Christiansen, J.H.; Wainwright, B.J. Targeted disruption of the Wnt2 gene results in placentation defects. Development 1996, 122, 3343–3353. [Google Scholar] [CrossRef]
  25. Ikeya, M.; Lee, S.M.K.; Johnson, J.E.; McMahon, A.P.; Takada, S. Wnt signalling required for expansion of neural crest and CNS progenitors. Nature 1997, 389, 966–970. [Google Scholar] [CrossRef]
  26. Millar, S.E.; Willert, K.; Salinas, P.C.; Roelink, H.; Nusse, R.; Sussman, D.J.; Barsh, G.S. WNT Signaling in the Control of Hair Growth and Structure. Dev. Biol. 1999, 207, 133–149. [Google Scholar] [CrossRef] [Green Version]
  27. Yamaguchi, T.P.; Bradley, A.; McMahon, A.P.; Jones, S. A Wnt5a pathway underlies outgrowth of multiple structures in the vertebrate embryo. Development 1999, 126, 1211–1223. [Google Scholar] [CrossRef]
  28. van den Heuvel, M.; Harryman-Samos, C.; Klingensmith, J. Mutations in the segment polarity genes wingless and porcupine impair secretion of the wingless protein. EMBO J. 1993, 12, 5293–5302. [Google Scholar] [CrossRef] [PubMed]
  29. Kadowaki, T.; Wilder, E.; Klingensmith, J.; Zachary, K.; Perrimon, N. The segment polarity gene porcupine encodes a putative multitransmembrane protein involved in Wingless processing. Genes Dev. 1996, 10, 3116–3128. [Google Scholar] [CrossRef] [Green Version]
  30. Willert, K.; Brown, J.D.; Danenberg, E.; Duncan, A.W.; Weissman, I.L.; Reya, T.; Yates, J.R.; Nusse, R. Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature 2003, 423, 448–452. [Google Scholar] [CrossRef] [PubMed]
  31. Rios-Esteves, J.; Haugen, B.; Resh, M.D. Identification of Key Residues and Regions Important for Porcupine-mediated Wnt Acylation. J. Biol. Chem. 2014, 289, 17009–17019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Biechele, S.; Cox, B.J.; Rossant, J. Porcupine homolog is required for canonical Wnt signaling and gastrulation in mouse embryos. Dev. Biol. 2011, 355, 275–285. [Google Scholar] [CrossRef] [Green Version]
  33. Barrott, J.J.; Cash, G.M.; Smith, A.P.; Barrow, J.R.; Murtaugh, L.C. Deletion of mouse Porcn blocks Wnt ligand secretion and reveals an ectodermal etiology of human focal dermal hypoplasia/Goltz syndrome. Proc. Natl. Acad. Sci. USA 2011, 108, 12752–12757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Takada, R.; Satomi, Y.; Kurata, T.; Ueno, N.; Norioka, S.; Kondoh, H.; Takao, T.; Takada, S. Monounsaturated Fatty Acid Modification of Wnt Protein: Its Role in Wnt Secretion. Dev. Cell 2006, 11, 791–801. [Google Scholar] [CrossRef] [Green Version]
  35. Bänziger, C.; Soldini, D.; Schütt, C.; Zipperlen, P.; Hausmann, G.; Basler, K. Wntless, a Conserved Membrane Protein Dedicated to the Secretion of Wnt Proteins from Signaling Cells. Cell 2006, 125, 509–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Bartscherer, K.; Pelte, N.; Ingelfinger, D.; Boutros, M. Secretion of Wnt Ligands Requires Evi, a Conserved Transmembrane Protein. Cell 2006, 125, 523–533. [Google Scholar] [CrossRef] [Green Version]
  37. Goodman, R.M.; Thombre, S.; Firtina, Z.; Gray, D.; Betts, D.; Roebuck, J.; Spana, E.P.; Selva, E.M. Sprinter: A novel transmembrane protein required for Wg secretion and signaling. Development 2006, 133, 4901–4911. [Google Scholar] [CrossRef] [Green Version]
  38. Coombs, G.S.; Yu, J.; Canning, C.A.; Veltri, C.A.; Covey, T.M.; Cheong, J.K.; Utomo, V.; Banerjee, N.; Zhang, Z.H.; Jadulco, R.C.; et al. WLS-dependent secretion of WNT3A requires Ser209 acylation and vacuolar acidification. J. Cell Sci. 2010, 123, 3357–3367. [Google Scholar] [CrossRef] [Green Version]
  39. Herr, P.; Basler, K. Porcupine-mediated lipidation is required for Wnt recognition by Wls. Dev. Biol. 2012, 361, 392–402. [Google Scholar] [CrossRef] [Green Version]
  40. Nygaard, R.; Yu, J.; Kim, J.; Ross, D.R.; Parisi, G.; Clarke, O.B.; Virshup, D.M.; Mancia, F. Structural Basis of WLS/Evi-Mediated Wnt Transport and Secretion. Cell 2021, 184, 194–206.e14. [Google Scholar] [CrossRef]
  41. Baeg, G.; Lin, X.; Khare, N.; Baumgartner, S.; Perrimon, N. Heparan sulfate proteoglycans are critical for the organization of the extracellular distribution of Wingless. Development 2001, 128, 87–94. [Google Scholar] [CrossRef]
  42. Korkut, C.; Ataman, B.; Ramachandran, P.; Ashley, J.; Barria, R.; Gherbesi, N.; Budnik, V. Trans-Synaptic Transmission of Vesicular Wnt Signals through Evi/Wntless. Cell 2009, 139, 393–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Gross, J.C.; Chaudhary, V.; Bartscherer, K.; Boutros, M. Active Wnt proteins are secreted on exosomes. Nat. Cell Biol. 2012, 14, 1036–1045. [Google Scholar] [CrossRef] [PubMed]
  44. Alexandre, C.; Baena-Lopez, A.; Vincent, J.-P. Patterning and growth control by membrane-tethered Wingless. Nature 2014, 505, 180–185. [Google Scholar] [CrossRef]
  45. Stanganello, E.; Hagemann, A.; Mattes, B.; Sinner, C.; Meyen, D.; Weber, S.; Schug, A.; Raz, E.; Scholpp, S. Filopodia-based Wnt transport during vertebrate tissue patterning. Nat. Commun. 2015, 6, 5846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Bhanot, P.; Brink, M.; Samos, C.H.; Hsieh, J.-C.; Wang, Y.; Macke, J.P.; Andrew, D.; Nathans, J.; Nusse, R. A new member of the frizzled family from Drosophila functions as a Wingless receptor. Nature 1996, 382, 225–230. [Google Scholar] [CrossRef]
  47. Wehrli, M.; Dougan, S.T.; Caldwell, K.; Okeefe, L.V.; Schwartz, S.; Vaizel-Ohayon, D.; Schejter, E.D.; Tomlinson, A.; DiNardo, S. Arrow encodes an LDL-receptor-related protein essential for Wingless signalling. Nature 2000, 407, 527–530. [Google Scholar] [CrossRef] [PubMed]
  48. Tamai, K.; Semenov, M.; Kato, Y.; Spokony, R.; Liu, C.; Katsuyama, Y.; Hess, F.; Saint-Jeannet, J.-P.; He, X. LDL-receptor-related proteins in Wnt signal transduction. Nature 2000, 407, 530–535. [Google Scholar] [CrossRef] [PubMed]
  49. Tamai, K.; Zeng, X.; Liu, C.; Zhang, X.; Harada, Y.; Chang, Z.; He, X. A Mechanism for Wnt Coreceptor Activation. Mol. Cell 2004, 13, 149–156. [Google Scholar] [CrossRef]
  50. Janda, C.Y.; Waghray, D.; Levin, A.M.; Thomas, C.; Garcia, K.C. Structural Basis of Wnt Recognition by Frizzled. Science 2012, 337, 59–64. [Google Scholar] [CrossRef] [Green Version]
  51. Janda, C.Y.; Dang, L.T.; You, C.; Chang, J.; De Lau, W.; Zhong, Z.A.; Yan, K.S.; Marecic, O.; Siepe, D.; Li, X.; et al. Surrogate Wnt agonists that phenocopy canonical Wnt and β-catenin signalling. Nature 2017, 545, 234–237. [Google Scholar] [CrossRef] [PubMed]
  52. Bourhis, E.; Tam, C.; Franke, Y.; Bazan, J.F.; Ernst, J.; Hwang, J.; Costa, M.; Cochran, A.; Hannoush, R.N. Reconstitution of a Frizzled8·Wnt3a·LRP6 Signaling Complex Reveals Multiple Wnt and Dkk1 Binding Sites on LRP6. J. Biol. Chem. 2010, 285, 9172–9179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Yu, H.; Ye, X.; Guo, N.; Nathans, J. Frizzled 2 and frizzled 7 function redundantly in convergent extension and closure of the ventricular septum and palate: Evidence for a network of interacting genes. Development 2012, 139, 4383–4394. [Google Scholar] [CrossRef] [Green Version]
  54. Dijksterhuis, J.; Baljinnyam, B.; Stanger, K.; Sercan, H.O.; Ji, Y.; Andres, O.; Rubin, J.S.; Hannoush, R.N.; Schulte, G. Systematic Mapping of WNT-FZD Protein Interactions Reveals Functional Selectivity by Distinct WNT-FZD Pairs. J. Biol. Chem. 2015, 290, 6789–6798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Kakugawa, S.; Langton, P.F.; Zebisch, M.; Howell, S.A.; Chang, T.-H.; Liu, Y.; Feizi, T.; Bineva-Todd, G.; O’Reilly, N.; Snijders, B.; et al. Notum deacylates Wnt proteins to suppress signalling activity. Nature 2015, 519, 187–192. [Google Scholar] [CrossRef] [PubMed]
  56. Nusse, R.; Fuerer, C.; Ching, W.; Harnish, K.; Logan, C.; Zeng, A.; ten Berge, D.; Kalani, Y. Wnt Signaling and Stem Cell Control. Cold Spring Harb. Symp. Quant. Biol. 2008, 73, 59–66. [Google Scholar] [CrossRef] [Green Version]
  57. Glinka, A.; Wu, W.; Delius, H.; Monaghan, A.P.; Blumenstock, C.; Niehrs, C. Dickkopf-1 is a member of a new family of secreted proteins and functions in head induction. Nature 1998, 391, 357–362. [Google Scholar] [CrossRef] [PubMed]
  58. Leyns, L.; Bouwmeester, T.; Kim, S.-H.; Piccolo, S.; De Robertis, E. Frzb-1 Is a Secreted Antagonist of Wnt Signaling Expressed in the Spemann Organizer. Cell 1997, 88, 747–756. [Google Scholar] [CrossRef] [Green Version]
  59. Cruciat, C.-M.; Niehrs, C. Secreted and Transmembrane Wnt Inhibitors and Activators. Cold Spring Harb. Perspect. Biol. 2013, 5, a015081. [Google Scholar] [CrossRef] [Green Version]
  60. Hao, H.-X.; Xie, Y.; Zhang, Y.; Charlat, O.; Oster, E.; Avello, M.; Lei, H.; Mickanin, C.; Liu, D.; Ruffner, H.; et al. ZNRF3 promotes Wnt receptor turnover in an R-spondin-sensitive manner. Nature 2012, 485, 195–200. [Google Scholar] [CrossRef]
  61. Koo, B.-K.; Spit, M.; Jordens, I.; Low, T.Y.; Stange, D.; Van De Wetering, M.; Van Es, J.H.; Mohammed, S.; Heck, A.; Maurice, M.; et al. Tumour suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nature 2012, 488, 665–669. [Google Scholar] [CrossRef] [PubMed]
  62. Carmonm, K.S.; Gong, X.; Lin, Q. R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/beta-catenin signaling. Proc. Natl. Acad. Sci. USA 2011, 108, 11452–11457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Cliffe, A.; Hamada, F.; Bienz, M. A Role of Dishevelled in Relocating Axin to the Plasma Membrane during Wingless Signaling. Curr. Biol. 2003, 13, 960–966. [Google Scholar] [CrossRef]
  64. Bilić, J.; Huang, Y.-L.; Davidson, G.; Zimmermann, T.; Cruciat, C.-M.; Bienz, M.; Niehrs, C. Wnt Induces LRP6 Signalosomes and Promotes Dishevelled-Dependent LRP6 Phosphorylation. Science 2007, 316, 1619–1622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Zeng, X.; Huang, H.; Tamai, K.; Zhang, X.; Harada, Y.; Yokota, C.; Almeida, K.; Wang, J.; Doble, B.; Woodgett, J.; et al. Initiation of Wnt signaling: Control of Wnt coreceptor Lrp6 phosphorylation/activation via frizzled, dishevelled and axin functions. Development 2008, 135, 367–375. [Google Scholar] [CrossRef] [Green Version]
  66. Davidson, G.; Wu, W.; Shen, J. Casein kinase 1 gamma couples Wnt receptor activation to cytoplasmic signal transduction. Nature 2005, 438, 867–872. [Google Scholar] [CrossRef]
  67. Zeng, X.; Tamai, K.; Doble, B.; Li, S.; Huang, H.; Habas, R.; Okamura, H.; Woodgett, J.; He, X. A dual-kinase mechanism for Wnt co-receptor phosphorylation and activation. Nature 2005, 438, 873–877. [Google Scholar] [CrossRef] [Green Version]
  68. Li, V.; Ng, S.S.; Boersema, P.J.; Low, T.Y.; Karthaus, W.R.; Gerlach, J.; Mohammed, S.; Heck, A.; Maurice, M.; Mahmoudi, T.; et al. Wnt Signaling through Inhibition of β-Catenin Degradation in an Intact Axin1 Complex. Cell 2012, 149, 1245–1256. [Google Scholar] [CrossRef] [Green Version]
  69. Su, L.-K.; Vogelstein, B.; Kinzler, K.W. Association of the APC Tumor Suppressor Protein with Catenins. Science 1993, 262, 1734–1737. [Google Scholar] [CrossRef]
  70. Behrens, J.; Jerchow, B.-A.; Würtele, M.; Grimm, J.; Asbrand, C.; Wirtz, R.; Kühl, M.; Wedlich, D.; Birchmeier, W. Functional Interaction of an Axin Homolog, Conductin, with β-Catenin, APC, and GSK3β. Science 1998, 280, 596–599. [Google Scholar] [CrossRef]
  71. Liu, C.; Li, Y.; Semenov, M. Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell 2002, 108, 837–847. [Google Scholar] [CrossRef] [Green Version]
  72. Kitagawa, M.; Hatakeyama, S.; Shirane, M. An F-box protein, FWD1, mediates ubiquitin-dependent proteolysis of beta-catenin. EMBO J. 1999, 18, 2401–2410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Kishida, S.; Yamamoto, H.; Ikeda, S. Axin, a negative regulator of the wnt signaling pathway, directly interacts with adenomatous polyposis coli and regulates the stabilization of beta-catenin. J. Biol. Chem. 1998, 273, 10823–10826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Ikeda, S.; Kishida, S.; Yamamoto, H.; Murai, H.; Koyama, S.; Kikuchi, A. Axin, a negative regulator of the Wnt signaling pathway, forms a complex with GSK-3beta and beta -catenin and promotes GSK-3beta -dependent phosphorylation of beta -catenin. EMBO J. 1998, 17, 1371–1384. [Google Scholar] [CrossRef]
  75. Griffin, J.; del Viso, F.; Duncan, A.R.; Robson, A.; Hwang, W.; Kulkarni, S.; Liu, K.; Khokha, M.K. RAPGEF5 Regulates Nuclear Translocation of β-Catenin. Dev. Cell 2018, 44, 248–260.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Daniels, D.L.; Weis, W.I. Beta-catenin directly displaces Groucho/TLE repressors from Tcf/Lef in Wnt-mediated transcription activation. Nat. Struct. Mol. Biol. 2005, 12, 364–371. [Google Scholar] [CrossRef] [PubMed]
  77. Arce, L.; Pate, K.T.; Waterman, M.L. Groucho binds two conserved regions of LEF-1 for HDAC-dependent repression. BMC Cancer 2009, 9, 159. [Google Scholar] [CrossRef] [Green Version]
  78. Behrens, J.; von Kries, J.P.; Kühl, M. Functional interaction of beta-catenin with the transcription factor LEF-1. Nature 1996, 38, 638–642. [Google Scholar] [CrossRef]
  79. Molenaar, M.; van de Wetering, M.; Oosterwegel, M. XTcf-3 transcription factor mediates beta-catenin-induced axis formation in Xenopus embryos. Cell 1996, 86, 391–399. [Google Scholar] [CrossRef] [Green Version]
  80. van de Wetering, M.; Oosterwegel, M.; Dooijes, D. Identification and cloning of TCF-1, a T lymphocyte-specific transcription factor containing a sequence-specific HMG box. EMBO J. 1991, 10, 123–132. [Google Scholar] [CrossRef]
  81. Verbeek, S.; Izon, D.; Hofhuis, F.; Robanus-Maandag, E.; Riele, H.T.; Van De Watering, M.; Oosterwegel, M.; Wilson, A.; Macdonald, H.R.; Clevers, H. An HMG-box-containing T-cell factor required for thymocyte differentiation. Nature 1995, 374, 70–74. [Google Scholar] [CrossRef] [PubMed]
  82. Nguyen, H.; Rendl, M.; Fuchs, E. Tcf3 Governs Stem Cell Features and Represses Cell Fate Determination in Skin. Cell 2006, 127, 171–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Angus-Hill, M.L.; Elbert, K.M.; Hidalgo, J.; Capecchi, M.R. T-cell factor 4 functions as a tumor suppressor whose disruption modulates colon cell proliferation and tumorigenesis. Proc. Natl. Acad. Sci. USA 2011, 108, 4914–4919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. van Es, J.H.; Haegebarth, A.; Kujala, P. A critical role for the Wnt effector Tcf4 in adult intestinal homeostatic self-renewal. Mol. Cell Biol. 2014, 32, 1918–1927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. van Genderen, C.; Okamura, R.M.; Fariñas, I. Development of several organs that require inductive epithelial-mesenchymal interactions is impaired in LEF-1-deficient mice. Genes. Dev. 1994, 8, 2691–2703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Okamura, R.M.; Sigvardsson, M.; Galceran, J. Redundant regulation of T cell differentiation and TCRalpha gene expression by the transcription factors LEF-1 and TCF-1. Immunity 1998, 8, 11–20. [Google Scholar] [CrossRef] [Green Version]
  87. Oosterwegel, M.; Van De Wetering, M.; Timmerman, J.; Kruisbeek, A.; Destree, O.; Meijlink, F.; Clevers, H. Differential expression of the HMG box factors TCF-1 and LEF-1 during murine embryogenesis. Development 1993, 118, 439–448. [Google Scholar] [CrossRef]
  88. Van de Wetering, M.; Castrop, J.; Korinek, V. Extensive alternative splicing and dual promoter usage generate Tcf-1 protein isoforms with differential transcription control properties. Mol. Cell Biol. 1996, 16, 745–752. [Google Scholar] [CrossRef] [Green Version]
  89. He, T.-C.; Sparks, A.B.; Rago, C.; Hermeking, H.; Zawel, L.; da Costa, L.T.; Morin, P.J.; Vogelstein, B.; Kinzler, K.W. Identification of c- MYC as a Target of the APC Pathway. Science 1998, 281, 1509–1512. [Google Scholar] [CrossRef]
  90. Shtutman, M.; Zhurinsky, J.; Simcha, I. The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway. Proc. Natl. Acad. Sci. USA 1999, 96, 5522–5527. [Google Scholar] [CrossRef] [Green Version]
  91. Yan, D.; Wiesmann, M.; Rohan, M. Elevated expression of axin2 and hnkd mRNA provides evidence that Wnt/beta -catenin signaling is activated in human colon tumors. Proc. Natl. Acad. Sci. USA 2001, 98, 14973–14978. [Google Scholar] [CrossRef] [Green Version]
  92. Munemitsu, S.; Albert, I.; Souza, B.; Rubinfeld, B.; Polakis, P. Regulation of intracellular beta-catenin levels by the adenomatous polyposis coli (APC) tumor-suppressor protein. Proc. Natl. Acad. Sci. USA 1995, 92, 3046–3050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Rosin-Arbesfeld, R.; Townsley, F.; Bienz, M. The APC tumour suppressor has a nuclear export function. Nature 2000, 406, 1009–1012. [Google Scholar] [CrossRef]
  94. Rosin-Arbesfeld, R.; Cliffe, A.; Brabletz, T.; Bienz, M. Nuclear export of the APC tumour suppressor controls beta-catenin function in transcription. EMBO J. 2003, 22, 1101–1113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Groden, J.; Thliveris, A.; Samowitz, W.; Carlson, M.; Gelbert, L.; Albertsen, H.; Joslyn, G.; Stevens, J.; Spirio, L.; Robertson, M.; et al. Identification and characterization of the familial adenomatous polyposis coli gene. Cell 1991, 66, 589–600. [Google Scholar] [CrossRef]
  96. Morishita, E.C.; Murayama, K.; Kato-Murayama, M. Crystal structures of the armadillo repeat domain of adenomatous polyposis coli and its complex with the tyrosine-rich domain of Sam68. Structure 2011, 19, 1496–1508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Yost, C.; Torres, M.; Miller, J.R.; Huang, E.; Kimelman, D.; Moon, R.T. The axis-inducing activity, stability, and subcellular distribution of beta-catenin is regulated in Xenopus embryos by glycogen synthase kinase 3. Genes Dev. 1996, 10, 1443–1454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Aberle, H.; Bauer, A.; Stappert, J. β-catenin is a target for the ubiquitin-proteasome pathway. EMBO J. 1997, 16, 3797–3804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Cavallo, R.A.; Cox, R.T.; Moline, M.M.; Roose, J.; Polevoy, G.A.; Clevers, H.; Peifer, M.; Bejsovec, A. Drosophila Tcf and Groucho interact to repress Wingless signalling activity. Nature 1998, 395, 604–608. [Google Scholar] [CrossRef]
  100. Levanon, D.; Goldstein, R.E.; Bernstein, Y.; Tang, H.; Goldenberg, D.; Stifani, S.; Paroush, Z.; Groner, Y. Transcriptional repression by AML1 and LEF-1 is mediated by the TLE/Groucho corepressors. Proc. Natl. Acad. Sci. USA 1998, 95, 11590–11595. [Google Scholar] [CrossRef] [Green Version]
  101. Smith, K.J.; Johnson, K.A.; Bryan, T.M.; Hill, D.E.; Markowitz, S.; Willson, J.K.; Paraskeva, C.; Petersen, G.M.; Hamilton, S.R.; Vogelstein, B. The APC gene product in normal and tumor cells. Proc. Natl. Acad. Sci. USA 1993, 90, 2846–2850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Kinzler, K.W.; Vogelstein, B. Lessons from Hereditary Colorectal Cancer. Cell 1996, 87, 159–170. [Google Scholar] [CrossRef] [Green Version]
  103. Morin, P.J.; Sparks, A.B.; Korinek, V. Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science 1997, 275, 1787–1790. [Google Scholar] [CrossRef] [Green Version]
  104. Korinek, V.; Barker, N.; Morin, P.J. Constitutive transcriptional activation by a beta-catenin-Tcf complex in APC-/- colon carcinoma. Science 1997, 275, 1784–1787. [Google Scholar] [CrossRef] [Green Version]
  105. Levy, D.B.; Smith, K.J.; Beazer-Barclay, Y.; Hamilton, S.R.; Vogelstein, B.; Kinzler, K.W. Inactivation of both APC alleles in human and mouse tumors. Cancer Res. 1994, 54, 5953–5958. [Google Scholar]
  106. Luongo, C.; Moser, A.R.; Gledhill, S.; Dove, W.F. Loss of Apc+ in intestinal adenomas from Min mice. Cancer Res. 1994, 54, 5947–5952. [Google Scholar]
  107. Miyaki, M.; Seki, M.; Okamoto, M.; Yamanaka, A.; Maeda, Y.; Tanaka, K.; Kikuchi, R.; Iwama, T.; Ikeuchi, T.; Tonomura, A. Genetic changes and histopathological types in colorectal tumors from patients with familial adenomatous polyposis. Cancer Res. 1990, 50, 7166–7173. [Google Scholar] [PubMed]
  108. Miyaki, M.; Tanaka, K.; Kikuchi-Yanoshita, R.; Muraoka, M.; Konishi, M. Familial polyposis: Recent advances. Crit. Rev. Oncol. 1995, 19, 1–31. [Google Scholar] [CrossRef]
  109. Knudson, A.G. Antioncogenes and human cancer. Proc. Natl. Acad. Sci. USA 1993, 90, 10914–10921. [Google Scholar] [CrossRef] [Green Version]
  110. Sparks, A.B.; Morin, P.J.; Vogelstein, B.; Kinzler, K.W. Mutational analysis of the APC/beta-catenin/Tcf pathway in colorectal cancer. Cancer Res. 1998, 58, 1130–1134. [Google Scholar]
  111. Sansom, O.J.; Meniel, V.S.; Muncan, V.; Phesse, T.J.; Wilkins, J.A.; Reed, K.R.; Vass, J.K.; Athineos, D.; Clevers, H.; Clarke, A.R. Myc deletion rescues Apc deficiency in the small intestine. Nature 2007, 446, 676–679. [Google Scholar] [CrossRef] [PubMed]
  112. Downward, J. Targeting RAS signalling pathways in cancer therapy. Nat. Cancer 2003, 3, 11–22. [Google Scholar] [CrossRef] [PubMed]
  113. Bos, J.L. Ras oncogenes in human cancer: A review. Cancer Res. 1989, 49, 4682–4689. [Google Scholar]
  114. Cohen, S.; Carpenter, G.; King, L. Epidermal growth factor-receptor-protein kinase interactions. Co-purification of receptor and epidermal growth factor-enhanced phosphorylation activity. J. Biol. Chem. 1980, 255, 4834–4842. [Google Scholar] [CrossRef]
  115. Schechter, A.L.; Stern, D.F.; Vaidyanathan, L.; Decker, S.J.; Drebin, J.A.; Greene, M.I.; Weinberg, R.A. The neu oncogene: An erb-B-related gene encoding a 185,000-Mr tumour antigen. Nature 1984, 312, 513–516. [Google Scholar] [CrossRef]
  116. Kraus, M.H.; Issing, W.; Miki, T.; Popescu, N.C.; Aaronson, S.A. Isolation and characterization of ERBB3, a third member of the ERBB/epidermal growth factor receptor family: Evidence for overexpression in a subset of human mammary tumors. Proc. Natl. Acad. Sci. USA 1989, 86, 9193–9197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Plowman, G.D.; Culouscou, J.M.; Whitney, G.S.; Green, J.M.; Carlton, G.W.; Foy, L.; Neubauer, M.G.; Shoyab, M. Ligand-specific activation of HER4/p180erbB4, a fourth member of the epidermal growth factor receptor family. Proc. Natl. Acad. Sci. USA 1993, 90, 1746–1750. [Google Scholar] [CrossRef] [Green Version]
  118. Cohen, S.; Carpenter, G. Human epidermal growth factor: Isolation and chemical and biological properties. Proc. Natl. Acad. Sci. USA 1975, 72, 1317–1321. [Google Scholar] [CrossRef] [Green Version]
  119. Cohen, S.; Ushiro, H.; Stoscheck, C.; Chinkers, M. A native 170,000 epidermal growth factor receptor-kinase complex from shed plasma membrane vesicles. J. Biol. Chem. 1982, 257, 1523–1531. [Google Scholar] [CrossRef]
  120. Massagué, J. Transforming growth factor-alpha. A model for membrane-anchored growth factors. J. Biol. Chem. 1990, 15, 21393–21396. [Google Scholar] [CrossRef]
  121. Higashiyama, S.; Abraham, J.A.; Miller, J.; Fiddes, J.C.; Klagsbrun, M. A Heparin-Binding Growth Factor Secreted by Macrophage-Like Cells That Is Related to EGF. Science 1991, 251, 936–939. [Google Scholar] [CrossRef] [PubMed]
  122. Harris, R.C.; Chung, E.; Coffey, R.J. EGF receptor ligands. Exp. Cell Res. 2003, 284, 2–13. [Google Scholar] [CrossRef]
  123. Riethmacher, D.; Sonnenberg-Riethmacher, E.; Brinkmann, V.; Yamaai, T.; Lewin, G.R.; Birchmeier, C. Severe neuropathies in mice with targeted mutations in the ErbB3 receptor. Nature 1997, 389, 725–730. [Google Scholar] [CrossRef] [PubMed]
  124. Gassmann, M.; Casagranda, F.; Orioli, D.; Simon, H.; Lai, C.; Klein, R.; Lemke, G. Aberrant neural and cardiac development in mice lacking the ErbB4 neuregulin receptor. Nature 1995, 378, 390–394. [Google Scholar] [CrossRef] [PubMed]
  125. Stern, D.F.; Kamps, M.P. EGF-stimulated tyrosine phosphorylation of p185neu: A potential model for receptor interactions. EMBO J. 1988, 7, 995–1001. [Google Scholar] [CrossRef]
  126. King, C.R.; Borrello, I.; Bellot, F.; Comoglio, P.; Schlessinger, J. Egf binding to its receptor triggers a rapid tyrosine phosphorylation of the erbB-2 protein in the mammary tumor cell line SK-BR-3. EMBO J. 1988, 7, 1647–1651. [Google Scholar] [CrossRef]
  127. Yarden, Y.; Sliwkowski, M.X. Untangling the ErbB signalling network. Nat. Rev. Mol. Cell Biol. 2001, 2, 127–137. [Google Scholar] [CrossRef]
  128. Lemmon, M.; Schlessinger, J. Regulation of signal transduction and signal diversity by receptor oligomerization. Trends Biochem. Sci. 1994, 19, 459–463. [Google Scholar] [CrossRef]
  129. Yarden, Y.; Schlessinger, J. Epidermal growth factor induces rapid, reversible aggregation of the purified epidermal growth factor receptor. Biochemistry 1987, 26, 1443–1451. [Google Scholar] [CrossRef]
  130. Yarden, Y.; Schlessinger, J. Self-phosphorylation of epidermal growth factor receptor: Evidence for a model of intermolecular allosteric activation. Biochemistry 1987, 26, 1434–1442. [Google Scholar] [CrossRef]
  131. Ferguson, K.M.; Berger, M.; Mendrola, J.M.; Cho, H.-S.; Leahy, D.J.; A Lemmon, M. EGF Activates Its Receptor by Removing Interactions that Autoinhibit Ectodomain Dimerization. Mol. Cell 2003, 11, 507–517. [Google Scholar] [CrossRef]
  132. Burgess, A.W.; Cho, H.-S.; Eigenbrot, C.; Ferguson, K.M.; Garrett, T.P.J.; Leahy, D.J.; Lemmon, M.A.; Sliwkowski, M.X.; Ward, C.W.; Yokoyama, S. An Open-and-Shut Case? Recent Insights into the Activation of EGF/ErbB Receptors. Mol. Cell 2003, 12, 541–552. [Google Scholar] [CrossRef]
  133. Lowenstein, E.; Daly, R.; Batzer, A.; Li, W.; Margolis, B.; Lammers, R.; Ullrich, A.; Skolnik, E.; Bar-Sagi, D.; Schlessinger, J. The SH2 and SH3 domain-containing protein GRB2 links receptor tyrosine kinases to ras signaling. Cell 1992, 70, 431–442. [Google Scholar] [CrossRef]
  134. Buday, L.; Downward, J. Epidermal growth factor regulates p21ras through the formation of a complex of receptor, Grb2 adapter protein, and Sos nucleotide exchange factor. Cell 1993, 73, 611–620. [Google Scholar] [CrossRef]
  135. Gale, N.W.; Kaplan, S.; Lowenstein, E.J.; Schlessinger, J.; Bar-Sagi, D. Grb2 mediates the EGF-dependent activation of guanine nucleotide exchange on Ras. Nature 1993, 363, 88–92. [Google Scholar] [CrossRef] [PubMed]
  136. Chardin, P.; Camonis, J.H.; Gale, N.W.; Van Aelst, L.; Schlessinger, J.; Wigler, M.H.; Bar-Sagi, D. Human Sos1: A guanine nucleotide exchange factor for Ras that binds to GRB2. Science 1993, 260, 1338–1343. [Google Scholar] [CrossRef]
  137. Schlessinger, J. SH2/SH3 signaling proteins. Curr. Opin. Genet. Dev. 1994, 4, 25–30. [Google Scholar] [CrossRef]
  138. Boriack-Sjodin, P.A.; Margarit, S.M.; Bar-Sagi, D.; Kuriyan, J. The structural basis of the activation of Ras by Sos. Nature 1998, 394, 337–343. [Google Scholar] [CrossRef]
  139. Medema, R.H.; de Vries-Smits, A.M.; van der Zon, G.C.; A Maassen, J.; Bos, J.L. Ras activation by insulin and epidermal growth factor through enhanced exchange of guanine nucleotides on p21ras. Mol. Cell. Biol. 1993, 13, 155–162. [Google Scholar] [CrossRef]
  140. Bar-Sagi, D. The Sos (Son of sevenless) protein. Trends Endocrinol. Metab. 1994, 5, 165–169. [Google Scholar] [CrossRef]
  141. Moodie, S.A.; Wolfman, A. The 3Rs of life: Ras, Raf and growth regulation. Trends Genet. 1994, 10, 44–48. [Google Scholar] [CrossRef]
  142. Bruder, J.T.; Heidecker, G.; Rapp, U.R. Serum-, TPA-, and Ras-induced expression from Ap-1/Ets-driven promoters requires Raf-1 kinase. Genes Dev. 1992, 6, 545–556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Brtva, T.R.; Drugan, J.K.; Ghosh, S.; Terrell, R.S.; Campbell-Burk, S.; Bell, R.M.; Der, C.J. Two Distinct Raf Domains Mediate Interaction with Ras. J. Biol. Chem. 1995, 270, 9809–9812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Dent, P.; Haser, W.; Haystead, T.A.J.; Vincent, L.A.; Roberts, T.M.; Sturgill, T.W. Activation of Mitogen-Activated Protein Kinase Kinase by v-Raf in NIH 3T3 Cells and in Vitro. Science 1992, 257, 1404–1407. [Google Scholar] [CrossRef]
  145. Kyriakis, J.M.; App, H.; Zhang, X.-F.; Banerjee, P.; Brautigan, D.L.; Rapp, U.R.; Avruch, J. Raf-1 activates MAP kinase-kinase. Nature 1992, 358, 417–421. [Google Scholar] [CrossRef]
  146. Crews, C.M.; Erikson, R.L. Extracellular signals and reversible protein phosphorylation: What to Mek of it all. Cell 1993, 74, 215–217. [Google Scholar] [CrossRef]
  147. Howe, L.; Leevers, S.J.; Gómez, N.; Nakielny, S.; Cohen, P.; Marshall, C.J. Activation of the MAP kinase pathway by the protein kinase raf. Cell 1992, 71, 335–342. [Google Scholar] [CrossRef]
  148. Crews, C.M.; Alessandrini, A.; Erikson, R.L. Erks: Their fifteen minutes has arrived. Cell Growth Differ. Mol. Biol. J. Am. Assoc. Cancer Res. 1992, 3, 135–142. [Google Scholar]
  149. Payne, D.M.; Rossomando, A.J.; Martino, P.; Erickson, A.K.; Her, J.H.; Shabanowitz, J.; Hunt, D.F.; Weber, M.J.; Sturgill, T.W. Identification of the regulatory phosphorylation sites in pp42/mitogen-activated protein kinase (MAP kinase). EMBO J. 1991, 10, 885–892. [Google Scholar] [CrossRef]
  150. Chen, R.H.; Sarnecki, C.; Blenis, J. Nuclear localization and regulation of erk- and rsk-encoded protein kinases. Mol. Cell. Biol. 1992, 12, 915–927. [Google Scholar] [CrossRef]
  151. Gonzalez, A.F.; Seth, A.; Raden, D.L.; Bowman, D.S.; Fay, F.S.; Davis, R.J. Serum-induced translocation of mitogen-activated protein kinase to the cell surface ruffling membrane and the nucleus. J. Cell Biol. 1993, 122, 1089–1101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Karin, M.; Hunter, T. Transcriptional control by protein phosphorylation: Signal transmission from the cell surface to the nucleus. Curr. Biol. 1995, 5, 747–757. [Google Scholar] [CrossRef] [Green Version]
  153. Hunter, T. Signaling-2000 and beyond. Cell 2000, 100, 113–127. [Google Scholar] [CrossRef] [Green Version]
  154. Seth, A.; A Gonzalez, F.; Gupta, S.; Raden, D.L.; Davis, R.J. Signal transduction within the nucleus by mitogen-activated protein kinase. J. Biol. Chem. 1992, 267, 24796–24804. [Google Scholar] [CrossRef]
  155. Gille, H.; Kortenjann, M.; Thomae, O.; Moomaw, C.; Slaughter, C.; Cobb, M.H.; E Shaw, P. ERK phosphorylation potentiates Elk-1-mediated ternary complex formation and transactivation. EMBO J. 1995, 14, 951–962. [Google Scholar] [CrossRef]
  156. Kortenjann, M.; Thomae, O.; E Shaw, P. Inhibition of v-raf-dependent c-fos expression and transformation by a kinase-defective mutant of the mitogen-activated protein kinase Erk2. Mol. Cell. Biol. 1994, 14, 4815–4824. [Google Scholar] [CrossRef] [PubMed]
  157. Yordy, J.S.; Muise-Helmericks, R.C. Signal transduction and the Ets family of transcription factors. Oncogene 2000, 19, 6503–6513. [Google Scholar] [CrossRef]
  158. Schlessinger, J. Ligand-Induced, Receptor-Mediated Dimerization and Activation of EGF Receptor. Cell 2002, 110, 669–672. [Google Scholar] [CrossRef] [Green Version]
  159. Zhang, X.; Gureasko, J.; Shen, K.; Cole, P.A.; Kuriyan, J. An Allosteric Mechanism for Activation of the Kinase Domain of Epidermal Growth Factor Receptor. Cell 2006, 125, 1137–1149. [Google Scholar] [CrossRef] [Green Version]
  160. Jura, N.; Endres, N.F.; Engel, K.; Deindl, S.; Das, R.; Lamers, M.H.; Wemmer, D.E.; Zhang, X.; Kuriyan, J. Mechanism for Activation of the EGF Receptor Catalytic Domain by the Juxtamembrane Segment. Cell 2009, 137, 1293–1307. [Google Scholar] [CrossRef] [Green Version]
  161. Haigler, H.T.; A McKanna, J.; Cohen, S. Rapid stimulation of pinocytosis in human carcinoma cells A-431 by epidermal growth factor. J. Cell Biol. 1979, 83, 82–90. [Google Scholar] [CrossRef]
  162. Beguinot, L.; Lyall, R.M.; Willingham, M.C.; Pastan, I. Down-regulation of the epidermal growth factor receptor in KB cells is due to receptor internalization and subsequent degradation in lysosomes. Proc. Natl. Acad. Sci. USA 1984, 81, 2384–2388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Miller, K.; Beardmore, J.; Kanety, H.; Schlessinger, J.; Hopkins, C.R. Localization of the epidermal growth factor (EGF) receptor within the endosome of EGF-stimulated epidermoid carcinoma (A431) cells. J. Cell Biol. 1986, 102, 500–509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Boguski, M.S.; McCormick, F. Proteins regulating Ras and its relatives. Nature 1993, 366, 643–654. [Google Scholar] [CrossRef]
  165. Trahey, M.; McCormick, F. A cytoplasmic protein stimulates normal N-ras p21 GTPase, but does not affect oncogenic mutants. Science 1987, 238, 542–545. [Google Scholar] [CrossRef] [PubMed]
  166. Martin, G.A.; Yatani, A.; Clark, R.; Conroy, L.; Polakis, P.; Brown, A.M.; McCormick, F. Gap Domains Responsible for Ras P21-Dependent Inhibition of Muscarinic Atrial K + Channel Currents. Science 1992, 255, 192–194. [Google Scholar] [CrossRef] [PubMed]
  167. Hunter, J.C.; Manandhar, A.; Carrasco, M.A.; Gurbani, D.; Gondi, S.; Westover, K.D. Biochemical and Structural Analysis of Common Cancer-Associated KRAS Mutations. Mol. Cancer Res. 2015, 13, 1325–1335. [Google Scholar] [CrossRef] [Green Version]
  168. Smith, M.J.; Neel, B.G.; Ikura, M. NMR-based functional profiling of RASopathies and oncogenic RAS mutations. Proc. Natl. Acad. Sci. USA 2013, 110, 4574–4579. [Google Scholar] [CrossRef] [Green Version]
  169. Rajagopalan, H.; Bardelli, A.; Lengauer, C.; Kinzler, K.W.; Vogelstein, B.; Velculescu, V.E. Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature 2002, 418, 934. [Google Scholar] [CrossRef]
  170. Minden, A.; Lin, A.; Smeal, T.; Dérijard, B.; Cobb, M.; Davis, R.; Karin, M. c-Jun N-terminal phosphorylation correlates with activation of the JNK subgroup but not the ERK subgroup of mitogen-activated protein kinases. Mol. Cell. Biol. 1994, 14, 6683–6688. [Google Scholar] [CrossRef]
  171. Minden, A.; Lin, A.; McMahon, M.; Lange-Carter, C.; Dérijard, B.; Davis, R.J.; Johnson, G.L.; Karin, M. Differential Activation of ERK and JNK Mitogen-Activated Protein Kinases by Raf-1 and MEKK. Science 1994, 266, 1719–1723. [Google Scholar] [CrossRef]
  172. Davis, R.J. Signal transduction by the JNK group of MAP kinases. Cell 2000, 103, 239–252. [Google Scholar] [CrossRef] [Green Version]
  173. Massagué, J. TGFβ signalling in context. Nat. Rev. Mol. Cell Biol. 2012, 13, 616–630. [Google Scholar] [CrossRef]
  174. Vogelstein, B.; Papadopoulos, N.; Velculescu, V.E.; Zhou, S.; Diaz, L.A., Jr.; Kinzler, K.W. Cancer Genome Landscapes. Science 2013, 339, 1546–1558. [Google Scholar] [CrossRef] [PubMed]
  175. Moses, H.L.; Roberts, A.B.; Derynck, R. The Discovery and Early Days of TGF-beta: A Historical Perspective. Cold Spring Harb. Perspect. Biol. 2016, 8, a021865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Derynck, R.; Jarrett, J.A.; Chen, E.Y.; Eaton, D.H.; Bell, J.R.; Assoian, R.K.; Roberts, A.B.; Sporn, M.B.; Goeddel, D.V. Human transforming growth factor-beta complementary DNA sequence and expression in normal and transformed cells. Nature 1985, 316, 701–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Gentry, E.L.; Lioubin, M.N.; Purchio, A.F.; Marquardt, H. Molecular events in the processing of recombinant type 1 pre-pro-transforming growth factor beta to the mature polypeptide. Mol. Cell. Biol. 1988, 8, 4162–4168. [Google Scholar] [CrossRef]
  178. Kirsch, T.; Sebald, W.; Dreyer, M.K. Crystal structure of the BMP-2–BRIA ectodomain complex. Nat. Genet. 2000, 7, 492–496. [Google Scholar] [CrossRef]
  179. Massagué, J. Receptors for the TGF-beta family. Cell 1992, 69, 1067–7070. [Google Scholar] [CrossRef]
  180. Lin, H.Y.; Lodish, H.F. Receptors for the TGF-beta superfamily: Multiple polypeptides and serine/threonine kinases. Trends Cell Biol. 1993, 3, 14–19. [Google Scholar] [CrossRef]
  181. Kingsley, D.M. The TGF-beta superfamily: New members, new receptors, and new genetic tests of function in different organisms. Genes Dev. 1994, 8, 133–146. [Google Scholar] [CrossRef] [Green Version]
  182. Shi, Y.; Massagué, J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 2003, 113, 685–700. [Google Scholar] [CrossRef] [Green Version]
  183. Wrana, J.L. Signaling by the TGFβ superfamily. Cold Spring Harb Perspect Biol. 2013, 5, a011197. [Google Scholar] [CrossRef] [Green Version]
  184. Hardwick, J.C.; Van Den Brink, G.R.; Bleuming, S.A. Bone morphogenetic protein 2 is expressed by, and acts upon, mature epithelial cells in the colon. Gastroenterology 2004, 126, 111–121. [Google Scholar] [CrossRef] [PubMed]
  185. Liu, F.; Ventura, F.; Doody, J.; Massagué, J. Human type II receptor for bone morphogenic proteins (BMPs): Extension of the two-kinase receptor model to the BMPs. Mol. Cell. Biol. 1995, 15, 3479–3486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Massagué, J. TGF-beta signal transduction. Annu. Rev. Biochem. 1998, 67, 753–791. [Google Scholar] [CrossRef]
  187. Kretzschmar, M.; Liu, F.; Hata, A.; Doody, J.; Massagué, J. The TGF-beta family mediator Smad1 is phosphorylated directly and activated functionally by the BMP receptor kinase. Genes Dev. 1997, 11, 984–995. [Google Scholar] [CrossRef] [Green Version]
  188. Wrana, J.L.; Attisano, L.; Wieser, R. Mechanism of activation of the TGF-beta receptor. Nature 1994, 370, 341–377. [Google Scholar] [CrossRef]
  189. Wieser, R.; Wrana, J.L.; Massagué, J. GS domain mutations that constitutively activate T beta R-I, the downstream signaling component in the TGF-beta receptor complex. EMBO J. 1995, 14, 2199–2208. [Google Scholar] [CrossRef]
  190. Huse, M.; Muir, T.W.; Xu, L.; Chen, Y.-G.; Kuriyan, J.; Massagué, J. The TGF beta receptor activation process: An inhibitor- to substrate-binding switch. Mol. Cell 2001, 8, 671–682. [Google Scholar] [CrossRef]
  191. Attisano, L.; Wrana, J.L. Signal transduction by the TGF-beta superfamily. Science 2002, 296, 1646–1647. [Google Scholar] [CrossRef]
  192. Macías-Silva, M.; Abdollah, S.; Hoodless, P.A. MADR2 is a substrate of the TGFbeta receptor and its phosphorylation is required for nuclear accumulation and signaling. Cell 1996, 87, 1215–1224. [Google Scholar] [CrossRef] [Green Version]
  193. Liu, F.; Hata, A.; Baker, J.C.; Doody, J.; Cárcamo, J.; Harland, R.M.; Massague, J. A human Mad protein acting as a BMP-regulated transcriptional activator. Nature 1996, 381, 620–623. [Google Scholar] [CrossRef] [PubMed]
  194. Zhang, Y.; Feng, X.; We, R. Receptor-associated Mad homologues synergize as effectors of the TGF-beta response. Nature 1996, 383, 168–172. [Google Scholar] [CrossRef]
  195. Lagna, G.; Hata, A.; Hemmati-Brivanlou, A. Partnership between DPC4 and SMAD proteins in TGF-beta signalling pathways. Nature 1996, 383, 832–836. [Google Scholar] [CrossRef]
  196. Liu, F.; Pouponnot, C.; Massagué, J. Dual role of the Smad4/DPC4 tumor suppressor in TGFbeta-inducible transcriptional complexes. Genes. Dev. 1997, 11, 3157–3167. [Google Scholar] [CrossRef] [Green Version]
  197. Hannon, G.J.; Beach, D. p15INK4B is a potential effector of TGF-beta-induced cell cycle arrest. Nature 1994, 371, 257–361. [Google Scholar] [CrossRef]
  198. Datto, M.B.; Li, Y.; Panus, J.F.; Howe, D.J.; Xiong, Y.; Wang, X.F. Transforming growth factor beta induces the cyclin-dependent kinase inhibitor p21 through a p53-independent mechanism. Proc. Natl. Acad. Sci. USA 1995, 92, 5545–5549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Reynisdóttir, I.; Polyak, K.; Iavarone, A.; Massagué, J. Kip/Cip and Ink4 Cdk inhibitors cooperate to induce cell cycle arrest in response to TGF-beta. Genes Dev. 1995, 9, 1831–1845. [Google Scholar] [CrossRef] [Green Version]
  200. Zimmerman, L.B.; De Jesús-Escobar, J.M.; Harland, R.M. The Spemann Organizer Signal noggin Binds and Inactivates Bone Morphogenetic Protein 4. Cell 1996, 86, 599–606. [Google Scholar] [CrossRef] [Green Version]
  201. Groppe, J.; Greenwald, J.; Wiater, E.; Leon, J.M.R.; Economides, A.; Kwiatkowski, W.; Affolter, M.; Vale, W.W.; Belmonte, J.C.I.; Choe, S. Structural basis of BMP signalling inhibition by the cystine knot protein Noggin. Nature 2002, 420, 636–642. [Google Scholar] [CrossRef] [PubMed]
  202. Piccolo, S.; Sasai, Y.; Lu, B.; De Robertis, E. Dorsoventral Patterning in Xenopus: Inhibition of Ventral Signals by Direct Binding of Chordin to BMP-4. Cell 1996, 86, 589–598. [Google Scholar] [CrossRef] [Green Version]
  203. Chen, Y.G.; Liu, F.; Massague, J. Mechanism of TGFbeta receptor inhibition by FKBP12. EMBO J. 1997, 16, 3866–3876. [Google Scholar] [CrossRef]
  204. Imamura, T.; Takase, M.; Nishihara, A. Smad6 inhibits signalling by the TGF-beta superfamily. Nature 1997, 389, 622–626. [Google Scholar] [CrossRef]
  205. Nakao, A.; Afrakhte, M.; Morén, A. Identification of Smad7, a TGFbeta-inducible antagonist of TGF-beta signalling. Nature 1997, 389, 631–635. [Google Scholar] [CrossRef] [PubMed]
  206. MacGrogan, D.; Pegram, M.; Slamon, D.; Bookstein, R. Comparative mutational analysis of DPC4 (Smad4) in prostatic and colorectal carcinomas. Oncogene 1997, 15, 1111–1114. [Google Scholar] [CrossRef] [Green Version]
  207. Markowitz, S.; Wang, J.; Myeroff, L. Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science 1995, 268, 1336–1338. [Google Scholar] [CrossRef]
  208. Parsons, R.; Myeroff, L.L.; Liu, B.; Willson, J.K.; Markowitz, S.D.; Kinzler, K.W.; Vogelstein, B. Microsatellite instability and mutations of the transforming growth factor beta type II receptor gene in colorectal cancer. Cancer Res. 1995, 55, 5548–5550. [Google Scholar]
  209. Riggins, G.J.; Thiagalingam, S.; Rozenblum, E.; Weinstein, C.L.; Kern, S.E.; Hamilton, S.R.; Willson, J.K.; Markowitz, S.D.; Kinzler, K.W.; Vogelstein, B. Mad-related genes in the human. Nat. Genet. 1996, 13, 347–349. [Google Scholar] [CrossRef]
  210. Shi, Y.; Hata, A.; Lo, R.S.; Massague, J.; Pavletich, N.P. A structural basis for mutational inactivation of the tumour suppressor Smad4. Nature 1997, 388, 87–93. [Google Scholar] [CrossRef]
  211. Hata, A.; Lo, R.S.; Wotton, D.; Lagna, G.; Massague, J. Mutations increasing autoinhibition inactivate tumour suppressors Smad2 and Smad4. Nature 1997, 388, 82–87. [Google Scholar] [CrossRef]
  212. Batlle, E.; Massagué, J. Transforming Growth Factor-β Signaling in Immunity and Cancer. Immunity 2019, 50, 924–940. [Google Scholar] [CrossRef]
  213. Pardee, A.B. G 1 Events and Regulation of Cell Proliferation. Science 1989, 246, 603–608. [Google Scholar] [CrossRef] [PubMed]
  214. Blagosklonny, M.V.; Pardee, A.B. The Restriction Point of the Cell Cycle. Cell Cycle 2002, 1, 102–109. [Google Scholar] [CrossRef] [Green Version]
  215. Gong, J.; Traganos, F.; Darzynkiewicz, Z. Threshold expression of cyclin E but not D type cyclins characterizes normal and tumour cells entering S phase. Cell Prolif. 1995, 28, 337–346. [Google Scholar] [CrossRef]
  216. Ekholm, S.V.; Zickert, P.; Reed, S.I.; Zetterberg, A. Accumulation of Cyclin E Is Not a Prerequisite for Passage through the Restriction Point. Mol. Cell. Biol. 2001, 21, 3256–3265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Kuhnert, F.; Davis, C.R.; Wang, H.-T.; Chu, P.; Lee, M.; Yuan, J.; Nusse, R.; Kuo, C.J. Essential requirement for Wnt signaling in proliferation of adult small intestine and colon revealed by adenoviral expression of Dickkopf-1. Proc. Natl. Acad. Sci. USA 2004, 101, 266–271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Barker, N.; Ridgway, R.A.; Van Es, J.H.; Van De Wetering, M.; Begthel, H.; van den Born, M.; Danenberg, E.; Clarke, A.R.; Sansom, O.J.; Clevers, H. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature 2009, 457, 608–611. [Google Scholar] [CrossRef]
  219. Moser, A.; Luongo, C.; Gould, K.; McNeley, M.; Shoemaker, A.; Dove, W. ApcMin: A mouse model for intestinal and mammary tumorigenesis. Eur. J. Cancer 1995, 31, 1061–1064. [Google Scholar] [CrossRef]
  220. Dow, L.E.; O’Rourke, K.P.; Simon, J.; Tschaharganeh, D.F.; van Es, J.H.; Clevers, H.; Lowe, S.W. Apc Restoration Promotes Cellular Differentiation and Reestablishes Crypt Homeostasis in Colorectal Cancer. Cell 2015, 161, 1539–1552. [Google Scholar] [CrossRef] [Green Version]
  221. Basak, O.; Beumer, J.; Wiebrands, K.; Seno, H.; van Oudenaarden, A.; Clevers, H. Induced Quiescence of Lgr5+ Stem Cells in Intestinal Organoids Enables Differentiation of Hormone-Producing Enteroendocrine Cells. Cell Stem Cell 2017, 20, 177–190.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Wong, V.W.Y.; Stange, D.E.; Page, M.E.; Buczacki, S.; Wabik, A.; Itami, S.; van de Wetering, M.; Poulsom, R.; Wright, N.A.; Trotter, M.W.B.; et al. Lrig1 controls intestinal stem-cell homeostasis by negative regulation of ErbB signalling. Nature 2012, 14, 401–408. [Google Scholar] [CrossRef] [Green Version]
  223. Snippert, H.J.; Schepers, A.G.; Van Es, J.H.; Simons, B.; Clevers, H. Biased competition between Lgr5 intestinal stem cells driven by oncogenic mutation induces clonal expansion. EMBO Rep. 2014, 15, 62–69. [Google Scholar] [CrossRef]
  224. van de Wetering, M.; Sancho, E.; Verweij, C. The beta-catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells. Cell 2002, 111, 241–250. [Google Scholar] [CrossRef] [Green Version]
  225. Meyer, N.; Penn, L.Z. Reflecting on 25 years with MYC. Nat. Cancer 2008, 8, 976–990. [Google Scholar] [CrossRef] [PubMed]
  226. Dang, C.V. MYC on the Path to Cancer. Cell 2012, 149, 22–35. [Google Scholar] [CrossRef] [Green Version]
  227. Claassen, G.F.; Hann, S.R. A role for transcriptional repression of p21CIP1 by c-Myc in overcoming transforming growth factor beta -induced cell-cycle arrest. Proc. Natl. Acad. Sci. USA 2000, 97, 9498–9503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Staller, P.; Peukert, K.; Kiermaier, A.; Seoane, J.; Lukas, J.; Karsunky, H.; Möröy, T.; Bartek, J.; Massague, J.; Hänel, F.; et al. Repression of p15INK4b expression by Myc through association with Miz-1. Nat. Cell Biol. 2001, 3, 392–399. [Google Scholar] [CrossRef]
  229. Chen, C.-R.; Kang, Y.; Siegel, P.M.; Massagué, J. E2F4/5 and p107 as Smad cofactors linking the TGFbeta receptor to c-myc repression. Cell 2002, 110, 19–32. [Google Scholar] [CrossRef] [Green Version]
  230. Warner, B.J.; Blain, S.W.; Seoane, J. Myc downregulation by transforming growth factor beta required for activation of the p15(Ink4b) G(1) arrest pathway. Mol. Cell Biol. 1999, 19, 5913–5922. [Google Scholar] [CrossRef] [Green Version]
  231. Nateri, A.S.; Spencer-Dene, B.; Behrens, A. Interaction of phosphorylated c-Jun with TCF4 regulates intestinal cancer development. Nature 2005, 437, 281–285. [Google Scholar] [CrossRef]
  232. Albanese, C.; Johnson, J.; Watanabe, G. Transforming p21ras mutants and c-Ets-2 activate the cyclin D1 promoter through distinguishable regions. J. Biol. Chem. 1995, 270, 23589–23597. [Google Scholar] [CrossRef] [Green Version]
  233. Bakiri, L.; Lallemand, D.; Bossy-Wetzel, E.; Yaniv, M. Cell cycle-dependent variations in c-Jun and JunB phosphorylation: A role in the control of cyclin D1 expression. EMBO J. 2000, 19, 2056–2068. [Google Scholar] [CrossRef] [PubMed]
  234. Schreiber, M.; Kolbus, A.; Piu, F.; Szabowski, A.; Möhle-Steinlein, U.; Tian, J.; Karin, M.; Angel, P.; Wagner, E.F. Control of cell cycle progression by c-Jun is p53 dependent. Genes Dev. 1999, 13, 607–619. [Google Scholar] [CrossRef] [PubMed]
  235. Shaulian, E.; Karin, M. AP-1 in cell proliferation and survival. Oncogene 2001, 20, 2390–2400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Sansom, O.J.; Reed, K.R.; van de Wetering, M. Cyclin D1 is not an immediate target of beta-catenin following Apc loss in the intestine. J. Biol. Chem. 2005, 280, 28463–28467. [Google Scholar] [CrossRef] [Green Version]
  237. Kretzschmar, M.; Doody, J.; Massagué, J. Opposing BMP and EGF signalling pathways converge on the TGF-beta family mediator Smad1. Nature 1997, 389, 618–622. [Google Scholar] [CrossRef]
  238. Kretzschmar, M.; Doody, J.; Timokhina, I.; Massague, J. A mechanism of repression of TGFbeta / Smad signaling by oncogenic Ras. Genes Dev. 1999, 13, 804–816. [Google Scholar] [CrossRef]
  239. Hermeking, H.; Rago, C.; Schuhmacher, M.; Li, Q.; Barrett, J.F.; Obaya-Gonzalez, A.J.; O’Connell, B.C.; Mateyak, M.; Tam, W.; Kohlhuber, F.; et al. Identification of CDK4 as a target of c-MYC. Proc. Natl. Acad. Sci. USA 2000, 97, 2229–2234. [Google Scholar] [CrossRef] [Green Version]
  240. Hinds, P.W.; Mittnacht, S.; Dulic, V.; Arnold, A.; Reed, S.I.; Weinberg, R.A. Regulation of retinoblastoma protein functions by ectopic expression of human cyclins. Cell 1992, 70, 993–1006. [Google Scholar] [CrossRef]
  241. Harbour, J.W.; Dean, D.C. The Rb/E2F pathway: Expanding roles and emerging paradigms. Genes Dev. 2000, 14, 2393–2409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Leung, J.Y.; Ehmann, G.L.; Giangrande, P.H.; Nevins, J.R. A role for Myc in facilitating transcription activation by E2F1. Oncogene 2008, 27, 4172–4179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Ohtani, K.; DeGregori, J.; Nevins, J.R. Regulation of the cyclin E gene by transcription factor E2F1. Proc. Natl. Acad. Sci. USA 1995, 92, 12146–12150. [Google Scholar] [CrossRef] [Green Version]
  244. Bartek, J.; Bartkova, J.; Lukas, J. The retinoblastoma protein pathway and the restriction point. Curr. Opin. Cell Biol. 1996, 8, 805–814. [Google Scholar] [CrossRef]
  245. Reddy, B.S.; Narisawa, T.; Wright, P.; Vukusich, D.; Weisburger, J.H.; Wynder, E.L. Colon carcinogenesis with azoxymethane and dimethylhydrazine in germ-free rats. Cancer Res. 1975, 35, 287–290. [Google Scholar] [PubMed]
  246. Li, L.; Li, X.; Sun, Y.; Jiang, K.; Wang, B.; Cao, H. 626–Gut Microbiota from Colorectal Cancer Patients Enhances the Progression of Intestinal Adenoma in Apc Min/+Mice. Gastroenterology 2019, 156, 301–315. [Google Scholar] [CrossRef]
  247. Wu, S.; Rhee, K.-J.; Albesiano, E.; Rabizadeh, S.; Wu, X.; Yen, H.-R.; Huso, D.L.; Brancati, F.L.; Wick, E.; McAllister, F.; et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat. Med. 2009, 15, 1016–1022. [Google Scholar] [CrossRef]
  248. Bezerra, L.B.; Faria, F.B.; da Graça, A.N. Clostridium difficile toxin A attenuates Wnt/β-catenin signaling in intestinal epithelial cells. Infect Immun. 2014, 82, 2680–2687. [Google Scholar] [CrossRef] [Green Version]
  249. Tao, L.; Zhang, J.; Meraner, P.; Tovaglieri, A.; Wu, X.; Gerhard, R.; Zhang, X.; Stallcup, W.B.; Miao, J.; He, X.Z.X.; et al. Frizzled proteins are colonic epithelial receptors for C. difficile toxin B. Nature 2016, 538, 350–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  250. Derwa, Y.; Gracie, D.J.; Hamlin, P.J.; Ford, A.C. Systematic review with meta-analysis: The efficacy of probiotics in inflammatory bowel disease. Aliment. Pharmacol. Ther. 2017, 46, 389–400. [Google Scholar] [CrossRef]
Figure 1. The WNT Signaling Pathway. In the WNT ON state (left), an R-spondin ligand (RSPO) binds LGR5 and RNF43/ZNRF3, preventing the inhibition of FZD. FZD is, therefore, able to bind a WNT ligand and LRP5/6 to form a co-receptor complex. DVL and AXIN, as well as the rest of the DC, is recruited to WNT–FZD–LRP at the plasma membrane. β-catenin may be phosphorylated within the sequestered DC, however, it cannot be ubiquitinated by β-TrCP, and is not degraded by the proteasome. The DCs become saturated with β-catenin, and newly synthesized β-catenin accumulates in the cytoplasm. β-catenin then migrates to the nucleus where it binds the TCF/LEF family of transcription factors (TCF4 in the intestines). The potent transcriptional activation domain of β-catenin, complexed with TCF4, results in the expression of WNT signaling target genes such as MYC, CCND1, and CD44. In the WNT OFF state (center), no WNT or RSPO ligands are present, and FZD is inhibited by RNF43/ZNRF3. The DC remains in the cytoplasm where it actively phosphorylates β-catenin. Phosphorylated β-catenin is recognized by β-TrCP and ubiquitinated, resulting in degradation of β-catenin (not diagrammed). β-catenin is unable to accumulate in the cytoplasm. The TCF/LEF factors are bound by the inhibitory TLE factors and repress target gene expression. In Colon Cancer (right), APC is truncated due to mutation, which compromises the ability of the DC to target β-catenin for degradation. The upstream components of the pathway are in grey as their functional role, to regulate β-catenin, has been compromised by APC truncation. β-catenin accumulates in the cytoplasm, after which it migrates to the nucleus. β-catenin binds the TCF/LEF transcription factors and constitutively activates the expression of WNT signaling target genes which leads to proliferation and the maintenance of stemness.
Figure 1. The WNT Signaling Pathway. In the WNT ON state (left), an R-spondin ligand (RSPO) binds LGR5 and RNF43/ZNRF3, preventing the inhibition of FZD. FZD is, therefore, able to bind a WNT ligand and LRP5/6 to form a co-receptor complex. DVL and AXIN, as well as the rest of the DC, is recruited to WNT–FZD–LRP at the plasma membrane. β-catenin may be phosphorylated within the sequestered DC, however, it cannot be ubiquitinated by β-TrCP, and is not degraded by the proteasome. The DCs become saturated with β-catenin, and newly synthesized β-catenin accumulates in the cytoplasm. β-catenin then migrates to the nucleus where it binds the TCF/LEF family of transcription factors (TCF4 in the intestines). The potent transcriptional activation domain of β-catenin, complexed with TCF4, results in the expression of WNT signaling target genes such as MYC, CCND1, and CD44. In the WNT OFF state (center), no WNT or RSPO ligands are present, and FZD is inhibited by RNF43/ZNRF3. The DC remains in the cytoplasm where it actively phosphorylates β-catenin. Phosphorylated β-catenin is recognized by β-TrCP and ubiquitinated, resulting in degradation of β-catenin (not diagrammed). β-catenin is unable to accumulate in the cytoplasm. The TCF/LEF factors are bound by the inhibitory TLE factors and repress target gene expression. In Colon Cancer (right), APC is truncated due to mutation, which compromises the ability of the DC to target β-catenin for degradation. The upstream components of the pathway are in grey as their functional role, to regulate β-catenin, has been compromised by APC truncation. β-catenin accumulates in the cytoplasm, after which it migrates to the nucleus. β-catenin binds the TCF/LEF transcription factors and constitutively activates the expression of WNT signaling target genes which leads to proliferation and the maintenance of stemness.
Cancers 14 00784 g001
Figure 2. The RAS-MAPK Signaling Pathway. In the RAS ON state (left), EGF binds to EGFR with 2:2 stoichiometry at the plasma membrane. EGF binding results in dimerization of the receptors, and their autophosphorylation. GRB2 binds to the phosphorylated tyrosine on EGFR via its SH2 domain, and subsequently recruits the rasGEF, SOS. SOS catalyzes the exchange of GDP for GTP on RAS, activating it. Activated RAS then activates RAF, which phosphorylates MEK, which, in turn, phosphorylates ERK. Phosphorylated ERK travels to the nucleus, though some remains in the cytoplasm, resulting in the phosphorylation of cytoplasmic and nuclear targets. ERK phosphorylates the transcription factor ELK, which allows it to form a ternary complex with serum response factor (SRF) at serum response elements (SREs). The complex then drives the expression of growth factor response genes, such as FOS, a component of the AP-1 transcription factor. In the RAS OFF state (center), no EGF or EGF-like ligand is present and EGFR remains unbound, its dimerization inhibited by its conformation. GTP bound to RAS is hydrolyzed to GDP by rasGAP, inactivating it. RAS which is bound to GDP, remains so. GRB2 and SOS remain unbound to EGFR, and RAF is inactive. MEK and ERK remain unphosphorylated, with some ERK remaining in the nucleus. ELK is unphosphorylated and is unable to form a ternary complex with SRF at the SRE. In Colon Cancer (right), K-RAS is mutated, at codon 12, which renders it refractory to GTP–GDP exchange by rasGAP, and rasGAP is, therefore, in grey. The actions of the upstream components EGFR, GRB2, and SOS are also in grey as their purpose, promoting the GTP bound state of RAS, has been achieved. RAF–MEK–ERK is active, inducing the activity of ELK and others.
Figure 2. The RAS-MAPK Signaling Pathway. In the RAS ON state (left), EGF binds to EGFR with 2:2 stoichiometry at the plasma membrane. EGF binding results in dimerization of the receptors, and their autophosphorylation. GRB2 binds to the phosphorylated tyrosine on EGFR via its SH2 domain, and subsequently recruits the rasGEF, SOS. SOS catalyzes the exchange of GDP for GTP on RAS, activating it. Activated RAS then activates RAF, which phosphorylates MEK, which, in turn, phosphorylates ERK. Phosphorylated ERK travels to the nucleus, though some remains in the cytoplasm, resulting in the phosphorylation of cytoplasmic and nuclear targets. ERK phosphorylates the transcription factor ELK, which allows it to form a ternary complex with serum response factor (SRF) at serum response elements (SREs). The complex then drives the expression of growth factor response genes, such as FOS, a component of the AP-1 transcription factor. In the RAS OFF state (center), no EGF or EGF-like ligand is present and EGFR remains unbound, its dimerization inhibited by its conformation. GTP bound to RAS is hydrolyzed to GDP by rasGAP, inactivating it. RAS which is bound to GDP, remains so. GRB2 and SOS remain unbound to EGFR, and RAF is inactive. MEK and ERK remain unphosphorylated, with some ERK remaining in the nucleus. ELK is unphosphorylated and is unable to form a ternary complex with SRF at the SRE. In Colon Cancer (right), K-RAS is mutated, at codon 12, which renders it refractory to GTP–GDP exchange by rasGAP, and rasGAP is, therefore, in grey. The actions of the upstream components EGFR, GRB2, and SOS are also in grey as their purpose, promoting the GTP bound state of RAS, has been achieved. RAF–MEK–ERK is active, inducing the activity of ELK and others.
Cancers 14 00784 g002
Figure 3. The TGF-β Signaling Pathway. In the TGF-β ON state (left), a BMP dimer binds to two BMPRI and two BMPRII to from a hetero-tetrameric complex. The Type II receptor phosphorylates the Type I receptor (not shown), which removes the inhibitory FKBP12 protein. An R-SMAD, such as SMAD1, binds the Type I receptor and is phosphorylated. The phosphorylated R-SMAD then forms a hetero-trimeric complex with SMAD4, the Co-SMAD and another SMAD1. The complex migrates to the nucleus where it binds the DNA and influences target gene expression. In the TGF-β OFF state (center), the BMP dimer is bound by Noggin which blocks binding to the Type II or Type I BMP receptors. The receptors remain unassociated, which prevents the phosphorylation of BMPRI. Therefore, BMPRI remains bound by FKBP12, which inhibits the phosphorylation of the R-SMAD. SMAD4 does not form a complex with the R-SMAD. In Colon Cancer (right), with a mutated SMAD4, the BMP ligand remains capable of inducing hetero-tetramer formation and phosphorylation of the R-SMAD. However, the mutation inhibits formation of the Co-SMAD::R-SMAD complex and the R-SMAD and Co-SMAD is therefore in grey. SMAD1 may migrate to the nucleus alone, however the intended function of the Co-SMAD::R-SMAD complex is not performed.
Figure 3. The TGF-β Signaling Pathway. In the TGF-β ON state (left), a BMP dimer binds to two BMPRI and two BMPRII to from a hetero-tetrameric complex. The Type II receptor phosphorylates the Type I receptor (not shown), which removes the inhibitory FKBP12 protein. An R-SMAD, such as SMAD1, binds the Type I receptor and is phosphorylated. The phosphorylated R-SMAD then forms a hetero-trimeric complex with SMAD4, the Co-SMAD and another SMAD1. The complex migrates to the nucleus where it binds the DNA and influences target gene expression. In the TGF-β OFF state (center), the BMP dimer is bound by Noggin which blocks binding to the Type II or Type I BMP receptors. The receptors remain unassociated, which prevents the phosphorylation of BMPRI. Therefore, BMPRI remains bound by FKBP12, which inhibits the phosphorylation of the R-SMAD. SMAD4 does not form a complex with the R-SMAD. In Colon Cancer (right), with a mutated SMAD4, the BMP ligand remains capable of inducing hetero-tetramer formation and phosphorylation of the R-SMAD. However, the mutation inhibits formation of the Co-SMAD::R-SMAD complex and the R-SMAD and Co-SMAD is therefore in grey. SMAD1 may migrate to the nucleus alone, however the intended function of the Co-SMAD::R-SMAD complex is not performed.
Cancers 14 00784 g003
Figure 4. Signaling Overview in Sporadic Colon Cancer. Activated WNT signaling, via mutations in APC or CTNNB1, results in constitutive formation of βcat/TCF4 complexes, which drive WNT target gene expression, such as MYC and CCND1. Activated RAS signaling, via mutations in K-RAS, result in activation of ERK, which phosphorylates MYC, as well as the R-SMADs, inhibiting SMAD migration to the nucleus (not shown). ERK also phosphorylates ELK, which results in expression of FOS. JUN is regulated by the JNK cascade (not shown), and binds to FOS, forming the AP-1 transcription factor. AP-1 and βcat/TCF4 drive expression of CCND1. Cyclin D1 binds to CDK4 or CDK6 and phosphorylates pRb, causing the release of E2F. E2F, with MYC, drives the expression of other E2F genes and CCNE1. Cyclin E binds to CDK2, which fully phosphorylates pRb, leading to progression into S-phase. The TGF-β pathway is inactivated due to mutation, though low levels of phosphorylated R-SMAD may be found in the nucleus. The cell cycle inhibitors, p15 and p21, which would be expressed given active TGF-β, cannot inhibit CDK4/6 or CDK2, and are in grey.
Figure 4. Signaling Overview in Sporadic Colon Cancer. Activated WNT signaling, via mutations in APC or CTNNB1, results in constitutive formation of βcat/TCF4 complexes, which drive WNT target gene expression, such as MYC and CCND1. Activated RAS signaling, via mutations in K-RAS, result in activation of ERK, which phosphorylates MYC, as well as the R-SMADs, inhibiting SMAD migration to the nucleus (not shown). ERK also phosphorylates ELK, which results in expression of FOS. JUN is regulated by the JNK cascade (not shown), and binds to FOS, forming the AP-1 transcription factor. AP-1 and βcat/TCF4 drive expression of CCND1. Cyclin D1 binds to CDK4 or CDK6 and phosphorylates pRb, causing the release of E2F. E2F, with MYC, drives the expression of other E2F genes and CCNE1. Cyclin E binds to CDK2, which fully phosphorylates pRb, leading to progression into S-phase. The TGF-β pathway is inactivated due to mutation, though low levels of phosphorylated R-SMAD may be found in the nucleus. The cell cycle inhibitors, p15 and p21, which would be expressed given active TGF-β, cannot inhibit CDK4/6 or CDK2, and are in grey.
Cancers 14 00784 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Brown, M.A.; Ried, T. Shifting the Focus of Signaling Abnormalities in Colon Cancer. Cancers 2022, 14, 784. https://doi.org/10.3390/cancers14030784

AMA Style

Brown MA, Ried T. Shifting the Focus of Signaling Abnormalities in Colon Cancer. Cancers. 2022; 14(3):784. https://doi.org/10.3390/cancers14030784

Chicago/Turabian Style

Brown, Markus A., and Thomas Ried. 2022. "Shifting the Focus of Signaling Abnormalities in Colon Cancer" Cancers 14, no. 3: 784. https://doi.org/10.3390/cancers14030784

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop