Next Article in Journal
Cortical Bone Loss and Fragility in a 2-Month Triple Transgenic Mouse Model of Alzheimer’s Disease
Previous Article in Journal
Viewing Inflammation and Immunoregulation Under the Calpain System Lens
Previous Article in Special Issue
Claudin18.2 as a Promising Therapeutic Target in Gastric Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Two-Way Role of Jagged1 in Cancer: A Focus on CRC

by
Sabrina Zema
1,*,
Francesca Di Fazio
1,
Rocco Palermo
1,
Claudio Talora
1 and
Diana Bellavia
1,2,*
1
Department of Molecular Medicine, La Sapienza University of Rome, 00161 Rome, Italy
2
Istituto Pasteur Italia, Fondazione Cenci-Bolognetti, La Sapienza University of Rome, 00161 Rome, Italy
*
Authors to whom correspondence should be addressed.
Cells 2025, 14(22), 1815; https://doi.org/10.3390/cells14221815
Submission received: 20 October 2025 / Revised: 14 November 2025 / Accepted: 17 November 2025 / Published: 19 November 2025

Highlights

What are the main findings?
  • Jagged1 is cleaved, releasing an intracellular domain (Jag1-ICD).
  • Jag1-ICD acquires oncogenic properties in CRC.
What are the implications of the main findings?
  • Jagged1 is a novel oncogenic driver that contributes to the multistep genetic model underlying the adenoma-to-carcinoma sequence in CRC.
  • Understanding the two-way role of Jagged1 could lead to new strategies for addressing drug resistance in CRC.

Abstract

Colorectal cancer (CRC) remains one of the most prevalent and lethal malignancies. Accumulating genetic evidence supports a multistep model of tumor progression, in which early APC loss leads to chromosomal instability and adenoma formation, followed by activating mutations in KRAS that synergize with β-catenin signaling to promote tumor growth and invasion. Among the downstream effectors of these pathways, the Notch ligand Jagged1 has emerged as a critical mediator of CRC progression and chemoresistance. Jagged1 is not only a transcriptional target of the Wnt/β-catenin axis but also undergoes proteolytic cleavage via the KRAS/ERK/ADAM17 signaling cascade, generating a nuclear Jagged1 intracellular domain (Jag1-ICD) that drives reverse signaling. This dual functionality, activating canonical Notch signaling and initiating reverse nuclear signaling, positions Jagged1 as a key oncogenic driver in CRC. In this review, we first summarize the role of Jagged1 as an integral part of canonical Notch signaling. We then focus on the non-canonical Jagged1 reverse signaling function in cancer, with a particular emphasis on CRC. We underscore the dual role of Jagged1 in tumor biology and propose that it functions as a novel oncogene within the adenoma-to-carcinoma sequence, supporting CRC development and drug resistance via non-canonical mechanisms.

1. Introduction

Colorectal cancer (CRC) is the second leading cause of cancer-related mortality worldwide, constituting a significant global public health concern [1]. CRC is a heterogeneous disease resulting from a complex interplay between genetic predisposition and environmental influences. It arises through the cumulative accumulation of genetic mutations and progressive epigenetic alterations in pathways controlling cell proliferation, differentiation, and apoptosis. Ultimately, these alterations allow malignant cells to bypass normal mechanisms of growth control [2]. The Notch pathway is one of the various signaling pathways associated with the onset of CRC [3,4,5,6]. The aberrant activation of Notch has been associated with poorer prognosis and metastasis of CRC [7,8]. It is known that the Notch pathway is involved in epithelial–mesenchymal transition (EMT) events [7,9], in modulating the tumor microenvironment (TME) [10], and in regulating cancer stem cells (CSCs) self-renewal and survival [11,12] through preferential interactions between the ligand–receptor pair Jagged1 and Notch1. Elevated Jagged1 expression has been detected in CRC tissues and is significantly associated with poor differentiation, advanced TNM (Tumor, Node, Metastasis) stage, and lymph node metastasis [5,13,14]. The role of Jagged1 in sustaining the proliferation and invasion of CRC cells has been the object of extensive research [15,16]. In addition to the canonical role of Jagged1 as a ligand of the Notch receptor, a plethora of evidence has documented that Jagged1 undergoes sequential proteolytic cleavage, ultimately leading to the release of its intracellular domain (Jag1-ICD) capable of activating its own signaling, named as “reverse signaling”. This intracellular domain has been shown to act as a nuclear oncogene, empowering transcriptional complexes in a Notch-dependent or -independent manner [17,18,19,20]. In the context of CRC, Jag1-ICD-induced reverse signaling is positively regulated by the KRAS/ERK/ADAM17 axis. This regulatory mechanism is crucial for sustaining various hallmarks of cancer cell behavior, including proliferation, EMT, invasion/migration, and drug resistance [21,22]. A comprehensive understanding of the role of Jagged1, which functions both as a ligand in canonical Notch signaling and as a nuclear oncogene through the Jag1-ICD-induced non-canonical pathway, may drive a paradigm shift in addressing drug resistance in this challenging malignancy.

2. Colorectal Cancer: An Overview

Colorectal cancer represents a significant global public health challenge, ranking as the second leading cause of cancer-related mortality worldwide among both men and women, and the third in terms of incidence [1]. CRC is characterized by a median age at diagnosis of 68 years in males and 72 years in females, with a 60% survival within 5 years upon their diagnosis [23]. Over the past decades, the primary objective has been the early-stage diagnosis of CRC, and the implementation of comprehensive screening programs has significantly contributed to reducing CRC-related mortality, primarily by enabling the early detection and treatment of malignant and premalignant lesions [23,24].
CRC arises through the cumulative accumulation of genetic mutations and progressive epigenetic alterations in pathways controlling cell proliferation, differentiation, and apoptosis, ultimately allowing malignant cells to bypass normal mechanisms of growth control. The stepwise progression from normal mucosa to adenoma and, eventually, to carcinoma is primarily driven by the dysregulated proliferation of colonocytes—the epithelial cells lining the colon and rectum [2,25]. Age, family history, hereditary syndromes, such as Familial Adenomatous Polyposis (FAP) or hereditary non-polyposis colorectal cancer also known as Lynch syndrome, and chronic inflammatory bowel disease (i.e., ulcerative colitis and Crohn’s disease) are considered the main risk factors for CRC [26,27]. Genetic mutations that are inherited through the germline are a contributing factor to hereditary forms of CRC. However, the majority of CRC cases are sporadic and result from the accumulation of somatic mutations influenced by environmental and lifestyle factors, including diet, physical inactivity, and exposure to carcinogens. It is estimated that 70% of cases of CRC are sporadic, with only 5% being associated with hereditary conditions, such as Lynch syndrome or FAP. The remaining 25% exhibit a familial disposition with no associated or known germline mutation [28,29]. The inter-tumoral heterogeneity, which is characteristic of CRC, is supported by the existence of different sequences of genomic and epigenomic alterations in different patients. The alterations manifest as a wide spectrum of neoplastic conditions, ranging from benign lesions to invasive carcinomas. These alterations are reflected at the macroscopic level by the emergence of different precursor lesions. There is a broad consensus in the scientific community that the majority of cases of colorectal cancer have their origins in aberrant crypt foci, the earliest microscopic precursors to colorectal cancer that can be classified as either serrated or non-serrated (adenomas), based on their appearance and molecular markers [25,30]. Adenomas have been identified in up to one-third of all surgical specimens that have been resected for CRC. Sporadic adenomas exhibit a histological similarity to adenomas arising from germline mutations in FAP, a condition widely acknowledged as premalignant [28,31]. The majority of both sporadic and hereditary CRCs arise through a series of sequential genetic alterations, with mutations in APC, KRAS, and TP53 representing key early events in CRC tumorigenesis. Loss-of-function mutations in the tumor suppressor gene APC initiate neoplastic transformation, while subsequent activating mutations in the oncogene KRAS are frequently associated with the progression from benign adenoma to dysplastic adenocarcinoma [32]. APC is the key tumor suppressor protein and is mutated in ∼80% of sporadic cancers, and germline heterozygosity of this gene leads to FAP [33,34,35]. The loss of APC function, a key negative regulator of Wnt signaling, results in constitutive activation of the β-catenin/TCF transcriptional complex, leading to sustained expression of Wnt/TCF target genes such as MYC and CCND1 [36,37,38]. Mutations in CTNNB1, the gene which encodes β-catenin, result in resistance to degradation via phosphorylation sites [39].
Constitutive KRAS mutations, found in about 50% of all CRCs and in advanced adenomas [40,41,42,43,44,45,46], drive persistent activation of the RAF/MEK/ERK mitogen-activated protein kinase (MAPK) cascade and the PI3K/AKT pathway independently of upstream signals, including the epidermal growth factor receptor (EGFR) [47,48]. This process has been demonstrated to promote uncontrolled cell growth. The RAF-MEK-ERK pathway has been shown to play a pivotal role in cell cycle regulation and intestinal tumorigenesis [49]. Interestingly, KRAS driver mutations confer a continuous “on” signal to downstream pathways, promoting cell proliferation, inhibiting apoptosis, and contributing to drug resistance, making it a critical factor in the pathogenesis of colorectal cancer [41,47,50]. Notably, 30–60% of CRC samples harbor concurrent mutations in the WNT and KRAS [25,48,51,52,53,54]. Specifically, CRC is synergistically induced by both APC loss of function and activated KRAS mutations, inducing cell proliferation and transformation [49]. APC mutation can stabilize both β-catenin and RAS (especially mutant KRAS) proteins, leading to tumor initiation and progression. This effect is mediated by direct interaction between β-catenin and KRAS proteins, an interaction that plays a regulatory function for the crosstalk between APC/β-catenin and KRAS/ERK signaling pathways [49]. Of note, RAS proteins are subject to polyubiquitination-mediated proteasomal degradation, a process orchestrated by glycogen synthase kinase 3 beta (GSK3β), promoting the recruitment of the β-TrCP E3 ligase adaptor [55,56]. In the resting state, APC protein forms a degradative complex with various components, including glycogen synthase kinase 3 beta (GSK3β), which binds to and phosphorylates β-catenin, a prerequisite for its ubiquitination and subsequent proteasomal degradation [36,39,57,58]. Defective APC alleles or β-catenin somatic mutations favor a strong β-catenin stabilization that can directly interact with RAS at the region containing the GSK3β phosphorylation sites, blocking GSK3β-mediated RAS degradation [59]. The co-stabilization of β-catenin and RAS, particularly the mutant form of KRAS, through APC mutations synergistically promotes the growth of CRC [36]. Elevated levels of both β-catenin and RAS are observed in CRC patient tissues, suggesting their pathological significance in tumor progression [51,56].
The development of additional mutations is a required step towards colon carcinogenesis. Allelic deletions of chromosome 17p and 18q usually occur at a later stage of tumorigenesis, causing the inactivation of the TP53 tumor suppressor, driving the transition from late-stage adenoma to invasive carcinoma [60]. Once carcinomas have formed, tumors invariably continue to progress. Furthermore, the loss of suppressor genes that accumulate on additional chromosomes is directly correlated with the ability of the carcinomas to metastasize and cause death [25].
Preneoplastic lesions from the serrated carcinogenesis pathway represent a heterogeneous group of colorectal lesions that include hyperplastic polyps (HPs), sessile serrated adenoma (SSA), traditional serrated adenoma (TSA) and mixed polyps [30]. Conversely to CRC that arises via the conventional pathway, serrated CRCs are rarely characterized by mutations in the APC and KRAS genes. Generally, serrated CRCs are enriched in the BRAFV600E activating mutation and are strongly associated with different genomic/epigenomic features: microsatellite instability (MSI) and the CpG island methylator phenotype (CIMP) [61]. It has been hypothesized that the BRAFV600E mutation plays a pivotal initiating role in early serrated lesions. This assertion is supported by the observation that the mutation has been detected in over 60% of precursor hyperplastic (HPs) or serrated crypt foci, while only 6% of non-serrated lesions have shown similar mutations [62]. In general, sessile serrated adenoma (SSA) emerges from mutations in DNA mismatch repair (MMR) genes, MSI-High, which is correlated with a high CIMP, resulting in high DNA methylation. Traditional serrated adenomas (TSA) characteristically exhibit low methylation and a stable microsatellite profile (MSS), with mutations that occur in KRAS or BRAF, and are predominantly characterized by an elevated level of activation of the TGFβ pathway [63].
In 2015, Guinney and colleagues proposed a consensus molecular classification system that allows the categorization of most tumors into one of four consensus molecular subtypes (CMS 1–4), based on the integration of genomic and transcriptomic features of colorectal tumors [64]. These subtypes reflect significant biological differences in gene expression-based molecular subtypes. The presence of marked differences in the intrinsic biological underpinnings of each subtype provides substantial support for the new taxonomy of CRC [64] (Table 1). Several studies have been conducted with the objective of enhancing the CMS in order to facilitate more precise prognostication, with a view to ensuring the more precise treatment of CRC. This necessitates a high degree of confidence in the CMS classification method [65,66].
In the current era of personalized medicine, accurate determination of a patient’s mutational profile is essential to guide the selection of the most effective and targeted therapies, while minimizing the risk of chemoresistance development. CRC harboring KRAS mutations is generally associated with poor prognosis, and in recent years, significant research efforts have focused on identifying effective therapeutic strategies targeting KRAS and its downstream pathways. Despite its clinical relevance, the biochemical complexity and structural characteristics of KRAS proteins have long impeded the development of direct inhibitors, delaying major therapeutic breakthroughs [67]. Historically, research has concentrated on targeting molecules within the RAS signaling cascade, particularly the MAPK pathway [68]. Numerous MEK inhibitors have been developed and evaluated both as monotherapy and in combination with other agents. However, MEK inhibitors such as trametinib [69] and cobimetinib [70], whether used alone or in combination with chemotherapy, PI3K/mTOR inhibitors [71], EGFR inhibitors [72] and AKT inhibitors [73], have demonstrated limited clinical efficacy in patients with advanced CRC [67]. Similarly, inhibitors targeting downstream effectors such as ERK and cyclin-dependent kinases (CDKs) have shown only modest results when used as monotherapy [74]. This has prompted the need for alternative approaches to KRAS inhibition. A major turning point arrived when a novel druggable pocket was identified below the switch II region of the KRASG12C mutant [75,76]. This discovery led to the development of allosteric inhibitors, small molecules designed to covalently bind to the G12C mutant, locking KRAS in its inactive GDP-bound (OFF) state without directly targeting GTP binding [77,78,79,80,81]. Several allosteric inhibitors targeting KRASG12C have been developed, including sotorasib [82] and adagrasib [83]. However, preclinical models have shown that selective inhibition of KRASG12C in CRC is hindered by the emergence of treatment resistance, primarily driven by upstream reactivation of the EGFR pathway [84]. Notably, approximately 40% of metastatic CRC (mCRC) cases harbor activating KRAS mutations, with G12D (30–36%), G12V (20–22%), and G13D (15–18%) being the most prevalent, while the KRASG12C variant accounts for only ~3% of cases [85].
Traditional chemotherapy remains a key component of CRC treatment. Combination regimens involving KRAS inhibitors and agents such as 5-FU, oxaliplatin (OXA), or irinotecan are being studied. Additionally, anti-angiogenic agents like bevacizumab may enhance the delivery and efficacy of targeted therapies, although clinical data in this setting are currently limited [86]. The identification of novel resistance mechanisms has prompted the development of innovative therapeutic strategies to overcome acquired resistance. Combination therapies represent a promising strategy to improve outcomes in KRASmut CRC. Dual inhibition of parallel signaling pathways, targeting upstream activators, and integrating immunotherapy or chemotherapy may help overcome intrinsic resistance and extend clinical benefits. Further research and biomarker-driven trials are needed to optimize these approaches.

3. Jagged1 as a Component of the Canonical Notch Pathway

The human JAG1 gene is located on the short arm of chromosome 20 at 20p12.2 and consists of 26 exons, encoding a protein comprising 1218 amino acids [87]. Jagged1 is a single-pass transmembrane ligand belonging to the Delta/Serrate/Lag-2 (DSL) family, characterized by Delta-like proteins (DLL1, 3 and 4) and Jagged proteins (Jagged1 and 2). These ligands primarily mediate the transactivation of the highly conserved Notch receptors (Notch1–4), via direct cell-to-cell contact.
All Notch proteins share a conserved basic structure. The extracellular domain is composed of 29–36 epidermal growth factor EGF-like repeats, specifically 36 in Notch1 and Notch2, 34 in Notch3, and 29 in Notch4, followed by three Lin12-Notch repeats (LNRs) and a membrane-proximal negative regulatory region (NRR). This is succeeded by a single transmembrane domain and an intracellular domain (Notch-ICD). The Notch-ICD comprises three conserved motifs present in all Notch orthologs: the RAM (RBP-Jκ-associated module) domain, a central ankyrin repeat domain (ANK) composed of seven repeats, and a C-terminal PEST domain, which is rich in proline (P), glutamic acid (E), serine (S), and threonine (T) residues. Notably, a complete transactivation domain (TAD) is present only in Notch1 and 2, distinguishing them functionally from Notch3 and Notch4 [88].
Similarly, Jagged1 is a type 1 cell surface protein composed of three major domains with modular architecture: a large extracellular domain (Jag1-ECD), a transmembrane domain (Jag1-TM), and a short intracellular domain (Jag1-ICD) located at the C-terminal region. Within the extracellular domain, a C2 phospholipid-binding domain is required to anchor phospholipid bilayers and provides Notch receptor interaction sites. This domain contributes to Notch activation through a process of N-glycosylation, a post-translational modification that ensures the proper spatial orientation of Jagged1 for effective Notch signaling. The functional significance of this modification is underscored by studies showing that mutations disrupting the glycosylation site markedly impair Jagged1-mediated activation of Notch signaling [89,90]. In addition, the extracellular region of Jagged1 contains a disulfide-rich DSL domain and 16 EGF-like repeats, which are necessary for Jagged1 binding to the EGF repeats of the Notch receptors. As well, a cysteine-rich domain (CRD) on the membrane-proximal side contributes to protein stability and protein–protein interactions [91]. The interaction between Jagged1 and Notch1 primarily involves the C2–EGF3 region of Jagged1 and the EGF11–EGF12 plus NRRs of Notch1, which are necessary for ligand-dependent Notch activation [92,93]. Beyond these well-established domains, other sites may play a direct role in mediating Notch/Jagged1 inter- and intramolecular interaction regions. Notably, the Notch1 NRR is sufficient for binding to Jagged1 C2–EGF3, supporting the idea of complex multi-site interactions [94]. Furthermore, Jagged1 undergoes conformational changes upon binding to Notch, forming a catch bond that prolongs the interaction for Notch activation under mechanical force [92]. The prevailing model for the canonical Notch activation mechanism is believed to rely on mechanical pulling forces generated by the signal-sending cell via ligand endocytosis, which exposes the Notch receptor to proteolytic cleavage [95,96]. Before membrane localization, Notch receptors undergo S1 cleavage by furin-like convertases and are glycosylated in the Golgi apparatus, resulting in the formation of a heterodimeric receptor [97,98]. Upon ligand-receptor interaction, the ligand can trigger Notch conformational change that favors proteolytic cleavage mediated by A-Disintegrin And Metalloprotease 10 (ADAM-10) in the juxtamembrane region (S2 cleavage), resulting in internalization of the ligand together with the Notch extracellular domain (Notch-ECD) [95,96,99,100,101,102,103]. This process involves sequential proteolysis, so the S2 cleavage is followed by S3 cleavage within the transmembrane domain by the PS/γ-secretase complex, which ends in the release of its intracellular domain (Notch-ICD), leading to the activation of Notch canonical signalling pathways. Once released, the Notch-ICD moves directly into the nucleus where it binds the transcriptional effector CSL (CBF1/RBP-Jκ/Su(H)/Lag-1), inducing a conformational change that displaces co-repressors and recruits co-activators, including Mastermind-like proteins (MAML1, 2, and 3 in mammals). MAML1 co-activator recognizes the Notch-ICD/CSL interface, forming a minimal ternary complex necessary to activate several downstream effectors, including genes in the Hairy/Enhancer of Split and bearded complexes (Hes and Hey families) [104], pre-T cell antigen receptor alpha (PTCRA) [105,106], MYC [107], and the JAG1, inside the Notch pathway, thus establishing a positive feedback loop [20,108] (Figure 1, canonical Notch signaling). The Notch pathway plays a central role in developmental processes, including neurogenesis [109,110], angiogenesis [111], T cell lineage commitment and maturation [112,113,114] and in determining cell fate of numerous tissues and organs, such as heart, lung, and colon [115]. Through regulation of cell proliferation, apoptosis, and differentiation, Notch signaling maintains tissue homeostasis and contributes to pathophysiological processes when dysregulated [97,116]. Given its highly pleiotropic functions, it is not surprising that dysregulation of the Notch signaling pathway is implicated in a broad range of pathological conditions, including developmental disorders [117,118], various progressive neurodegenerative diseases, including Alzheimer’s, multiple sclerosis, and amyotrophic lateral sclerosis [119], as well as numerous cancers. It is known that Notch signaling plays either an oncogenic or a tumor suppressor role depending on tissue context [120]. Notably, aberrant Notch1 signaling was originally associated with rare cases of T-cell acute lymphoblastic leukemia (T-ALL) in humans. The oncogenic role of Notch was first recognized following the discovery of a t(7;9)(q34;q34.3) chromosomal translocation, which affects the NOTCH1 gene in T-ALL [121]. Afterward, somatic activating mutations of Notch1 [122,123] or Notch3 [124] were identified in several cases of human T-ALL. The non-redundant role of Notch3 receptor in T-ALL pathogenesis was strongly demonstrated by in vivo mouse models [106,125,126,127,128]. Following these discoveries, activating deregulation of Notch receptors was found in other hematological malignancies, including B-cell chronic lymphocytic leukemia (B-CLL) [129] and in a variety of solid cancers, such as colorectal cancer, breast cancer, and ovarian cancer [98,115,130]. In these contexts, Notch frequently cross-talks with other oncogenic pathways, further enhancing its pathological impact [131,132]. Dysregulation of Notch signaling may arise from gain-of-function mutations in Notch receptors, commonly observed in hematologic cancers [133,134] or from loss-of-function mutations, as reported in squamous cell carcinomas [135,136,137,138]. Similarly, in small-cell lung cancer (SCLC) and in Glioma, Notch signaling acts as a tumor suppressor pathway, with loss-of-function mutations in Notch family genes [139,140,141], as well as in bladder cancer [142,143]. In Neuroendocrine Tumors (NETs), Notch expression is reduced, with concomitant mutations in Notch pathway components [144,145]; scientific evidence shows that Notch1 activation inhibits cell proliferation, indicating a tumor suppressive role [146]. Due to the complex nature of the Notch pathway, it is not always so easy to discern its role in a pathological context. Indeed, in both Pancreatic Ductal Carcinoma (PDAC) and acute myeloid leukemia (AML), different studies reported a role for both [147], suggesting a controversial role of the pathway during carcinogenesis.
Additionally, overexpression of the Jagged1 ligand can lead to ligand-dependent hyperactivation of the Notch pathway, representing another key mechanism of aberrant Notch signaling in cancer. Jagged1 is frequently overexpressed across multiple tumor types, and its transcription is regulated by several oncogenic signaling pathways, including Wnt/β-catenin [148], IL-6/STAT3 [149], TGF-β [150], NF-KB [151], SOX12 promoting stem cell-like phenotypes [152], and even Notch signalling itself [20,108]. This underscores a regulatory loop wherein Jagged1 is not only a downstream effector of these pathways, but may also amplify oncogenic signaling, independently of Notch. Clinically, Jagged1 overexpression is strongly associated with poor prognosis, high tumor grade, and increased metastatic potential in a variety of human cancers, including prostate cancer [153], tongue squamous cell carcinoma [154], renal cell carcinoma [155], breast cancer [156], pancreatic cancer [157], multiple myeloma [158,159] and ovarian cancer [160].
Interestingly, Jagged1 has the capacity to modulate tumor biology via mechanisms that are either dependent on, or independent of, canonical Notch signaling.

4. The Relevance of Jagged1 Intracellular Domain: From Development to Cancer

The commonly accepted scenario is based on the idea that the Jagged1 dysregulated expression contributes to tumorigenesis primarily through canonical trans-activation of Notch signaling in neighboring cells [161]. However, Jagged1 can also initiate a non-canonical reverse signaling through the release of the intracellular fragment within ligand-expressing cells. These dual roles underscore the multifaceted nature of Jagged1 activity, highlighting its ability to modulate both Notch-dependent and Notch-independent pathways, thereby contributing to a more intricate and context-dependent regulatory.
Similarly to the Notch receptor, Jagged1 undergoes sequential proteolytic processing. The first cleavage occurs in the juxtamembrane region and is mediated by ADAM-17/TACE, resulting in the shedding of the soluble Jagged1 extracellular domain (sJag1-ECD) and the generation of a membrane-tethered C-terminal fragment (Jag1-TMICD) [162,163]. This is followed by intramembrane proteolysis catalyzed by the presenilin/γ-secretase complex, which releases the intracellular domain of Jagged1 (Jag1-ICD). Once liberated, Jag1-ICD translocates into the nucleus, where it functions as a signaling molecule, potentially regulating gene expression independently of canonical Notch activation [164] (Figure 1, non-canonical Jagged1 reverse signaling).
The role of the soluble Jagged1 extracellular domain (sJag1-ECD) in Notch signaling remains controversial, as it has been shown to function as either an agonist or an antagonist, depending on the cellular context. Several studies suggest that sJag1-ECD can inhibit Notch signaling by competitively binding to Notch receptors, thereby blocking the interaction with membrane-bound ligands on signal-sending cells [165,166,167,168,169], while it can also exhibit Notch-activating functions in a paracrine manner [167,170,171,172].
Growing evidence indicates that Jag1-ICD–mediated reverse signaling plays critical roles in diverse biological contexts. In cardiac tissue, Jag1-ICD contributes to neonatal cardiomyocyte differentiation by inhibiting Notch1 processing and downstream target gene expression (HES1 and HEY1/2) [173]. In the developing mammalian lens, nuclear Jag1-ICD has been shown to promote its own gene transcription, suggesting a positive feedback mechanism [174], as well as Jagged1 also plays a regulatory role in steroidogenesis within testicular Leydig cells, where it fine-tunes hormone production during testis development [175]. Interestingly, pioneering work from Capobianco’s lab demonstrates that the intracellular domain of Jagged1 can induce cellular transformation in a dose-dependent manner [164]. This effect requires a highly conserved PDZ-ligand motif (RMEYIV), located in the C terminus of Jagged1. Interestingly, this motif interacts with the PDZ domain of afadin (AF-6/MLLT4), an actin filament-binding protein localized at adherens junctions [176,177]. Notably, afadin plays a central role in crosstalk between multiple pathways, including Wnt/Wingless, Ras/MAPK, and Notch, by physically interacting with Dishevelled, Ras, and Notch proteins [178]. The PDZ-dependent interaction between Jagged1 and AF6 suggests that Jagged1 can recruit intracellular signaling complexes independent of Notch receptor engagement. While AF6 may serve as a scaffold linking Jagged1 to Ras pathway components, its exact role in Jagged1-mediated transformation remains to be fully defined. Importantly, mutations in the PDZ-ligand motif do not impair Jagged1’s ability to activate canonical Notch signaling in neighboring cells but abolish its capacity to mediate reverse signaling, highlighting the functional specificity of this motif [164]. These findings strongly support the existence of a PDZ-dependent, reverse signaling pathway initiated by Jagged1, which contributes to a bidirectional signaling model. Significantly, elevated Jagged1 expression correlates with enhanced cellular transformation, providing mechanistic evidence for its oncogenic potential via reverse signaling. The PDZ-ligand motif is essential for downstream activation of the AP-1 transcription factor, leading to upregulation of Jagged1 and Notch3 mRNA levels [163,164]. These events link aberrant Jagged1 expression with tumorigenesis, in a Notch-dependent or Notch-independent manner. Likewise, a positive feedback loop between Notch3 and Jagged1 has been described in ovarian cancer, where their co-expression forms a functional signaling network [160]. Jagged1 is a transcriptional target of both Notch3-ICD and β-catenin, and both pathways can sustain Notch3 signaling and promote ovarian carcinoma progression by supporting tumor cell adhesion and growth [20,108,179]. Accordingly, aberrant Notch3/Jagged1 cis-expression, inside the same cell, has been shown in T-ALL. In this context, Jagged1 undergoes constitutive, lipid raft-associated processing mediated by ADAM17, leading to the release of sJag1-ECD into the conditioned medium (CM) and bloodstream of Notch3-ICD transgenic mice, strongly activating Notch signaling in adjacent cells [20]. The sequential Jagged1 cleavage mediated by γ-secretase leads to the release of its intracellular domain that translocates into the nucleus, where it integrates into the Notch transcriptional activation complex by interacting directly with Notch-ICD and the RBP-Jκ transcription factor. This interaction enhances Notch signaling activity and promotes the transcriptional activation of Jagged1 itself. Notch3-ICD-transcriptional complex shows the ability to specifically activate the Jagged1 promoter-1351/-237, which contains a canonical RBP-Jk binding site. Jag1-ICD protein cooperates with Notch3-ICD/RBP-Jk/MAML1 transcriptional complex to drive the activation of its own promoter, strengthening the activity of Notch3-driven transcriptional complex in triggering its own transcription [20]. This complex activates genes such as PTCRA and JAG1, further reinforcing the loop in an autocrine fashion in immature T cell lines and sustains survival, proliferation, and invasion, contributing to the development and progression of Notch-dependent T-ALL [20], suggesting an oncogenic role for Jag1-ICD in a Notch-dependent manner (Figure 2, left panel). Strong evidence suggests that inside the nucleus Jag1-ICD plays a role as a co-activator, reinforcing several transcriptional complexes and addressing specific Jagged1 target genes. In chronic lymphocytic leukemia (CLL), Jagged1 undergoes proteolytic activation in signaling-sending cells, triggering Notch activation through autocrine/paracrine loops, associated with biological effects and sJag1-ECD is detected in CM from CLL cultures and in patient plasma. Notably, interleukin-4 (IL-4) upregulates Jagged1 expression and promotes Jagged1 processing via activation of the phosphatidylinositol 3-kinase δ (PI3Kδ)/AKT signaling pathway [17] (Figure 2, central panel). Interestingly, Jag1-ICD can also regulate tumorigenesis through a Notch-independent mechanism. Specifically, it interacts with a transcriptional complex composed of DDX17, SMAD3, and TGIF2, which drives the expression of SOX2, a key regulator of cancer stem cell properties. This upregulation of SOX2 contributes to the acquisition of stem-like features in astrocytes, including enhanced tumorigenic potential, invasiveness, and resistance to anticancer therapies, thereby promoting oncogenic transformation [18]. Mechanistically, Jag1-ICD/DDX17 complex binds to DNA via the transcription factor SMAD3. Therefore, cells overexpressing Jagged1 generate an accumulation of Jag1-ICD, which activates a transcriptional complex using SMAD3 as a molecular hub, independent of Notch receptor signaling. Interestingly, deletion of the PDZL domain of Jag1-ICD does not affect Jag1-ICD/DDX17 binding, suggesting that PDZL may be essential to participate in the Notch-dependent transcriptional complex. Of note, a molecular crosstalk between Notch and TGF-β pathways is dependent on Jag1-ICD transcriptional complex, which can sustain oncogenic transformation [18]. Moreover, Jag1-ICD enhances invasive phenotypes of glioblastoma cells by transcriptionally activating EMT-related genes, especially TWIST1. The Jag1-ICD/SMAD3–TWIST1 axis represents a novel regulatory pathway that promotes invasive phenotypes in cancer cells, driving brain tumor invasion through a mechanism distinct from canonical TGF-β signaling [19]. In prostate cancer, Jag1-ICD has been shown to upregulate the expression of androgen receptor variants (AR-Vs) and to enhance AR transactivation under both androgen-dependent and -independent conditions [180]. Moreover, Jag1-ICD promotes the expression of CSC markers, such as CD133, and pluripotency-associated factors, including NANOG and OCT3/4. Functionally, Jag1-ICD increases the migratory capacity of prostate cancer cells and enhances tumorigenic potential in vivo, indicating that Jag1-ICD contributes to the acquisition of an aggressive prostate cancer phenotype characterized by AR positivity, elevated CD133 expression, and enhanced self-renewal and survival properties [180] (Figure 2, right panel).
These findings provide strong evidence that Jag1-ICD functions as an oncogenic co-activator within diverse transcriptional complexes, thereby empowering distinct signaling pathways that drive oncogenic transformation. Unlike the canonical Notch pathway that operates through unidirectional signaling from ligand-expressing to receptor-expressing cells, Jagged1 is also processed into a nuclear Jag1-ICD fragment that initiates a reverse signaling cascade within the signal-sending cell. This bidirectional signaling model amplifies the functional role of several oncogenic pathways and positions Jagged1 as a key regulator in both physiological and pathological contexts.

5. The Canonical and Non-Canonical Role of Jagged1 Ligand in CRC

5.1. The Canonical Notch Signaling in CRC

In the pathological context of CRC, several studies have reported the upregulation of components of the Notch signaling pathways, including its ligands [13,181] and receptors [3,4,5,6]. Physiologically, Notch signaling is required for the development and homeostasis of normal intestinal epithelia, in the differentiation of colonic goblet cells and stem cells [115]. The aberrant activation of Notch is associated, in patients, with poorer prognosis and metastasis of CRC [7,8]. Abnormal Notch signaling promotes the invasion and metastasis of CRC cells. In particular, Notch1 promotes the recruitment of neutrophils and induces the transcriptional expression of TGF-β2, thereby leading to the activation of its own signaling [7]. Elevated Notch1 expression has been closely associated with lymph node metastasis, tumor stage, depth of infiltration, and histological differentiation in CRC patients [182]. In CRC, Notch1 and Notch2 have opposite roles in determination of the tumor biological behavior; Notch1 and Notch2 are independent adverse prognostic predictors, with a synergistic effect of positive Notch1 and negative Notch2 co-expression on predicting poor overall survival [4,183,184]. Notch3 is also found to be upregulated in CRC, compared to healthy tissue, and is associated with tumor recurrence [185] and higher expression of Notch3 is associated with increased tumor growth rate [186]. The overexpression rate of nuclear Notch3 in CRC was 38%, and nuclear Notch3 expression was correlated to distant relapse-free survival in patients affected with stage II and III CRC [185]. Furthermore, the co-expression of nuclear Notch3 and Notch1 predicted a worse prognosis than negative subtypes. Notch3 expression is positively correlated with the expression of macrophage recruitment-related cytokines in colon tumor tissues. Specifically, Notch3 enhances the progression of CRC by increasing the infiltration of macrophages and myeloid-derived suppressor cells (MDSCs) to promote the immunosuppressive TME [187]. Moreover, Notch3 regulates DNA repair within CRC cells to sustain chemoresistance events [188]. Interestingly, Sharma and colleagues found in CRC patients Hypomethylation of Notch 2 and 3 receptors in a small cohort of CRC patients [189]. The upregulation of Notch 2 and Notch 3 was associated with high-grade tumors, advanced stage and presence of lymph node metastasis [189]. Accordingly, the Notch pathway is actively involved in EMT events; indeed, the interaction between Notch, the transcription factors Slug and Snail, and TGF-β is critical for EMT [7,9]. Furthermore, epithelial Notch1 activation is enriched in aggressive colorectal cancer subtypes, where it potentiates TGF-β signaling. This interaction promotes neutrophil recruitment and contributes to immunosuppression within the metastatic TME, particularly in KRASG12D-driven serrated CRC [7]. Aberrant Notch signaling plays a pivotal role in modulating the TME in CRC, thereby impacting both tumor progression and therapeutic outcomes. Dysregulation of the Notch pathway influences the differentiation and functional polarization of MDSCs and tumor-associated macrophages (TAMs), two key immunosuppressive cell populations within the TME that facilitate immune evasion and support tumor growth [10]. Jagged1–Notch1 signaling has been identified as a key pathway within the TME, driving the generation of CD8+ CXCL13+ T cells mediated by melanoma cell adhesion molecule (MCAM)-expressing fibroblasts [190]. In inflammatory bowel disease-associated CRC, elevated levels of Claudin-1 (CLDN1) activate Notch signaling, which subsequently triggers the PI3K/Akt pathway. This cascade leads to β-catenin phosphorylation and promotes hyperproliferation of CRC cells [191]. Additionally, Notch1 can induce chemoresistance events in response to 5-FU, OXA, or irinotecan treatment [192], in particular, through the upregulation of MRP1 and BCL2 antiapoptotic proteins [193]. In addition, the Notch signaling pathway plays a critical role in the self-renewal and differentiation of intestinal epithelial stem and progenitor cells [26]. The aberrant activation of Notch signaling is involved in the modulation of stemness in CRC cells [11]. In colorectal tumors, CSCs exhibit a 10- to 30-fold increase in Notch signaling activity compared to non-stem cancer cells. Notch has been identified as a fundamental regulator of CSC self-renewal and survival, in part through the inhibition of apoptosis. Mechanistically, Notch suppresses the expression of the cell cycle inhibitor p27 and the pro-differentiation transcription factor ATOH1 [12]. The deletion of Notch1 and Notch2 or the pharmacological inhibition of Notch signaling with a γ-secretase inhibitor triggers colon columnar stem cells to differentiate into goblet secretory cells in the murine model [194,195]. Accordingly, genes of canonical Notch signaling components (i.e., JAG1, JAG2 and NOTCH1) and Notch target genes (i.e., HES1, HES4 and HES6) are all significantly higher in CSCs [12]. In particular, the Jagged1/Notch1/Hes1 axis plays a crucial role in the maintenance and viability of CSCs through the inhibition of apoptosis and cell cycle arrest. The pharmacological inhibition of Notch by γ-secretase inhibitor induces the activation of the intrinsic apoptotic pathway, causing cleavage of caspase-3 and increasing levels of proteins responsible for cell cycle arrest, like ATOH1, p27, and p57 [12]. Of note, the same results are obtained through the inhibition of ADAM17 by MEDI3622 or TAPI-2, which have shown similar negative effects on self-renewal of colorectal CSCs [196,197,198]. In particular, the ADAM17 inhibitor ZLDI-8 inhibits the proliferation of CRC and improves the anti-tumor and anti-metastasis activity of 5-fluorouracil (5-FU) or irinotecan by reversing Notch and EMT pathways, both in vitro and in vivo [197]. These findings highlight the pivotal role of Notch signaling in the formation and maintenance of CSCs, which contribute to tumorigenesis and metastasis.

5.2. The Canonical Role of Jagged1 Ligand in CRC

Several reports have highlighted that Jagged1 expression is higher in CRC tissues than in adjacent nontumor colon tissues and that its expression correlates with low differentiation degree, advanced TNM stage, and lymph node metastasis [5,13]. The role of Jagged11 in sustaining the proliferation and invasion of CRC cells and tumor growth has been extensively studied [15,16]. In the context of CRC, deletion of a single JAG1 allele in an APC mutant background significantly reduces tumor burden, which is associated with decreased levels of active Notch1. Moreover, Jagged1 is overexpressed in the majority of CRC cases and is considered a key contributor to the constitutive activation of the Notch signaling pathway [15]. Analysis of circulating mRNA derived from blood cells and serum of patients with mCRC showed that JAG1 upregulation in both serum and blood of mCRC patients correlated with high discrimination ability, suggesting that Jagged1 could be a potential non-invasive biomarker for the diagnosis and/or prognosis of patients with mCRC [199].
The pivotal role of the Notch pathway in CRC is due to the combined expression of the ligand/receptor couple, Jagged1/Notch1. The constitutive activation of Notch in colon tumor cell lines resulted in increased expression of EMT and stemness-associated proteins, such as CD44, Slug, Smad-3, and induction of Jagged1 expression [9]. In the regulation of CRC stemness, Jagged1 acts as the primary Notch ligand. In APC-deficient adenomas, the deletion of JAG1 disrupts stem cell niche formation [200]. In addition, cytoplasmic Jagged1 expression correlates with Notch3 expression in tumor cells [26,186].
In addition, it has been demonstrated that Jagged1/Notch signaling activated by Wnt/β-catenin signaling promotes the colon sphere formation (3D culture assay to measure the stem-like, self-renewal ability of colon cancer cells) by CRC cells and tumor vasculogenesis [148,161,201]. Specifically, β-catenin directly controls the transcriptional activation of Jagged1. In a mouse model APCMin/+ crossed with Jag1+/δ mice, the growth and size of polyps are significantly reduced, suggesting a pivotal role of Notch during tumorigenesis induced by nuclear β-catenin. The activation of Notch signaling occurs by β-catenin-mediated up-regulation of Jagged1 and is required for tumorigenesis in the intestine [148]. Moreover, in CRC cells, progastrin sequentially activated the transcription of Wnt and Notch target genes, suggesting a feedback regulation from Notch toward Wnt signaling [202]. The Jagged1 expression progastrin-induced activates Tcf-4 activity, maintaining the concomitant activation of Wnt and Notch pathways in CRC cells [202]. The transcriptional activity of β-catenin to the JAG1 promoter depends on the histone demethylases KDM4C [201]. β-Catenin bound to the KDM4C promoter, and the binding of β-catenin and KDM4C onto the JAG1 promoter is essential during colon sphere formation, suggesting that KDM4C maintains the sphere-forming capacity in CRC by mediating the β-catenin-dependent transcription of JAG1 in a feed-forward manner [201]. The synergy between Notch and Wnt signaling can provide a developmental context that is favorable for the accumulation of oncogenic mutations, in which aberrant Notch activation results in hyperplastic conditions, suggesting a preneoplastic state, in which the occurrence of secondary mutations increases the possibility of developing a malignancy [3].
The deletion of Jagged1 intestine-specific, in the mouse model APCMin/+, has been shown to prevent tumor formation, reduce the expression of intestinal stem cell markers, and inhibit tumor spheroid growth, without affecting normal intestinal homeostasis [203]. Notably, spheroids derived from model APCMin/+ are characterized by lower levels of the N-acetylglucosaminyltransferases Manic Fringe (MFNG), compared to the non-tumoral-derived organoids, while Jagged1 levels were comparable. MFNG fine-tune Jagged–Notch binding specificity and strength [204], which enhance Delta ligand signaling while attenuating cellular responsiveness to Jagged ligands [205,206,207]. These observations suggest that the downmodulation of MFNG in tumor tissues could switch Notch activation from DLL ligands to Jagged1. Moreover, in CRC patients with high Jagged1 expression, the lower expression of MFNG is significantly associated with poor CRC prognosis, suggesting that the Jagged1-high/MFNG-low pattern highlights a CRC subset that could benefit from Jagged1 inhibition. In fact, the inhibition of Jagged1 by blocking antibody prevents tumor initiation in mice and reduces patient-derived tumor orthoxenograft growth without affecting normal intestinal mucosa, targeting exclusively the tumor cells and avoiding the side effects in the normal gut [203].
Finally, the Jagged1/Notch signaling pathway in CRC is positively regulated by the activity of APEX1 as an upstream activator [208]. APEX1 overexpression in human colon cancer cell lines induces cell proliferation, anchorage-independent growth, migration, invasion, and angiogenesis both in vitro and in vivo. APEX1 exerts its oncogenic effect by upregulating Jagged1 transcription, activating Notch signaling. Furthermore, APEX1 expression was associated with Jagged1 in tissues from colon cancer patients [208]. Moreover, the concomitant expression of APEX1 and Jagged1 is associated with chemoresistance toward 5-FU, OXA, and irinotecan [209]. The analysis of tissue from CRC patients highlighted that high expression of Jagged1 is associated with a significantly low response to chemotherapy. The authors suggest that the overexpression of Jagged1 by APEX1 could represent a predictor of response to chemotherapy and of poor prognosis, and the combined expression of both proteins could be a therapeutic target for chemotherapy of advanced CRC [209] (Figure 3).

5.3. The Non-Canonical Role of Jagged1 in CRC

As described above, the majority of both sporadic and hereditary CRCs arise through a series of sequential genetic alterations, with mutations in APC, KRAS, and TP53 representing key early events in CRC tumorigenesis. The uncontrolled activation of KRAS is a hallmark event in CRC development, progression and metastasis, able to trigger multiple downstream pathways, including the RAF/MEK/ERK MAPK cascades, involved in intestinal tumorigenesis [52]. Increasing evidence further suggests that oncogenic KRAS regulates ADAM17 activity and the shedding of growth factor ligands in a MEK/ERK-dependent manner, through direct ERK/ADAM17 interaction [47,210,211]. Specifically, the pro-tumorigenic function of ADAM17 relies on its threonine phosphorylation mediated by p38, which promotes the release of its substrate, soluble IL-6R, thereby activating IL-6 trans-signaling via the ERK1/2 MAPK pathway [211]. Moreover, MAPKs play a pivotal role in controlling the shedding of membrane-bound proteins. In particular, the cytosolic tail of the TACE/ADAM17 enzyme is phosphorylated by ERK at threonine 735, a post-translational modification essential for its catalytic activity [212].
Jagged1 is a direct transcriptional target of the β-catenin/TCF complex, leading to its robust upregulation in CRC, which can contribute to tumor development and progression, activating the canonical Notch signaling pathway [148]. Accordingly, the Notch ligand Jagged1 is aberrantly expressed in about 50% of human CRC [14] and its expression levels correlate with poor prognosis, chemoresistance, and recurrence [13]. In addition, combined mutations in the Wnt/β-catenin and KRAS pathways synergistically amplify downstream signaling events, ultimately converging in the activation of the Jagged1 protein, with the release of functional fragments, the soluble Jag1-ECD, and the Jagged1 intracellular domain, which sustain malignant traits and tumor progression in CRC [21]. Strong evidence demonstrates that Kras can regulate ADAM17 activity in a MEK/ERK-dependent manner, inducing a KRAS/ERK/ADAM17 signalling axis constitutively activated in CRC [210]. In this regard, the soluble Jag1-ECD, derived from endothelial cells (ECs), has been shown to promote colorectal cancer progression in a paracrine manner. EC-secreted Jag1-ECD activates Notch signaling in CRC cells via ADAM17-dependent mechanisms, thereby enhancing metastatic potential in an in vivo mouse model. Furthermore, Jag1-ECD contributes to the acquisition of CSCs characteristics and resistance to chemotherapy [213]. The soluble form of Jagged1, originated by the cleavage of ADAM17 in endothelial cells, increases the tumorigenic potential of neighboring CRC cells, which in turn express stemness markers (i.e., CD133, EPCAM and ALDH activity). Both Jagged1 and Jagged2 soluble forms are released from the extracellular membrane to promote CSC phenotype through Notch1 activation [196].
Moreover, Kras-induced ADAM17 sheddase activity induces extensive Jagged1 processing, supporting the existence of a direct link between the aberrant activation of the KRAS/ERK pathway and the Jagged1 processing in CRC [21]. Therefore, Jagged1 undergoes sequential proteolytic cleavages, ultimately resulting in the release of its intracellular domain, Jag1-ICD. Once released, Jag1-ICD translocates to the nucleus, where it initiates a distinct signaling cascade through interaction with the CSL/RBP-Jκ transcription factor, directly regulating SNAI1 and SNAI2 promoter activity [21]. The constitutive processing of full-length Jagged1 into Jag1-ICD thus represents a critical oncogenic event, effectively converting a proto-oncogene into a functional nuclear oncogene. This event endows cancer cells with the ability to sustain key malignant processes, including proliferation, invasion, migration, and chemoresistance [21,22]. Accordingly, the oncogenic role of Jag1-ICD can be inhibited in vivo by using the TAPI-2 compound, an inhibitor of ADAM17 activity [20,21]. Collectively, these processes are orchestrated by the constitutively active KRAS/ERK/ADAM17 signaling axis in CRC harboring KRAS mutations, which identifies Jagged1 as both a proteolytic substrate and a terminal effector of KRAS signaling. Notably, the most widely used anticancer agents in CRC therapy, OXA and 5-FU, often give rise to chemoresistant cancer cell subpopulations through intrinsic or acquired mechanisms [214,215]. These treatments also induce robust Jag1-ICD activation via ERK1/2 signaling, leading to the selection of drug-resistant cells protected from apoptosis through the upregulation of Jag1-ICD–dependent pro-survival targets, including IAP1, IAP2, XIAP, BCL-XL, and MCL1. This mechanism establishes an intrinsic form of chemoresistance in which Jag1-ICD functions as a nuclear effector downstream of β-catenin and KRAS [22].
Consistently, silencing of Jagged1 in OXA- or 5-FU–resistant colorectal cancer subpopulations restores their sensitivity to chemotherapy, confirming that drug response is Jag1-ICD–dependent [22]. These findings suggest that Jagged1 may serve as a predictive molecular marker for chemotherapy response. Collectively, these observations highlight the central role of Jagged1 in colorectal cancer biology and therapy resistance.
Interestingly, γ-secretase inhibitors (GSIs) were widely used to inhibit Notch activation, and they have been progressively recognized as potential anticancer drugs in patients with solid tumors, including sarcoma [216], breast cancer [217], desmoid tumors [218], or T-cell acute lymphoblastic leukemia [130,219,220]. In CRC, the therapeutic effects of GSIs remain controversial. However, the clinical application of GSIs is limited by associated toxicities, primarily due to goblet cell metaplasia and the depletion of intestinal stem cells, which remain significant concerns requiring further investigation. On one hand, GSIs have been explored as potential chemotherapeutic agents, with several studies demonstrating their ability to enhance OXA sensitivity in CRC cells [192,221,222]. On the other hand, conflicting evidence suggests that GSIs may attenuate OXA-induced apoptosis [223]. Moreover, the oral GSI RO4929097 was evaluated in a phase II clinical trial in patients with refractory metastatic CRC but showed no significant clinical efficacy [224]. Nonetheless, γ-secretase activity is not specific to Notch receptors alone and can also process other substrates, potentially leading to additional off-target effects and undesirable side effects. The effects of GSIs, OXA, and 5-FU, administered individually or in combination, have been evaluated in colorectal CRC cell lines harboring KRAS mutations. These studies demonstrate that GSIs not only suppress canonical Notch signaling but also trigger robust activation of Jagged1 reverse signaling through the MAPK/ERK1/2 pathway. This activation enhances the release of the Jag1-ICD, which exerts oncogenic effects independently of Notch receptor signaling. GSIs enhance cellular proliferation, acting as tumor-promoting agents through the processing of Jagged1. In addition, treatment with OXA and 5-FU promotes robust Jag1-ICD processing through ERK1/2 activation, resulting in the upregulation of Jag1-ICD-dependent pro-survival targets and conferring resistance to apoptosis in KRASmut CRC cells [22]. Evidence also supports a synergistic effect induced by GSIs and chemotherapeutic agents (OXA or 5-FU) in sustaining Jag1-ICD–mediated multidrug resistance. These findings reveal a novel mechanism of acquired drug resistance in KRAS-mutant CRC, wherein Jag1-ICD functions as a novel nuclear effector downstream of the KRAS signaling pathway [21,22] (Figure 3).

6. Conclusions

Colorectal cancer is one of the most commonly diagnosed malignancies and remains a major cause of cancer-related mortality worldwide. CRC is a genetically and molecularly heterogeneous disease, driven by a series of sequential alterations in key signaling pathways that govern tumor initiation, progression, and resistance to therapy.
Recent studies have identified the Notch ligand Jagged1 as a critical contributor to CRC progression and chemoresistance. Overexpression of Jagged1 has been consistently associated with poor prognosis. Remarkably, Jagged1 may influence tumor biology through both canonical and non-canonical mechanisms. While classically defined as a Notch ligand mediating canonical cell-to-cell signaling, emerging evidence indicates that Jagged1 can also initiate autonomous signaling within signal-sending cells, thereby contributing to non-canonical, Notch-independent pathways. This non-canonical, Notch-independent function adds a further layer of complexity to the role of Jagged1 in cancer biology. Altogether, these observations underscore the multifaced role of Jagged1, including its additional ability to initiate a robust reverse signaling pathway driven by the KRAS/ERK/ADAM17 signaling axis. In this context, Jagged1 acts as a downstream effector of oncogenic KRAS signaling, ultimately leading to the release of the nuclear Jagged1 intracellular domain, which has been directly implicated in progression and drug resistance phenomena. We propose that Jagged1 functions as a novel oncogenic driver that contributes to the multistep genetic model underlying the adenoma-to-carcinoma sequence in CRC. This leaves open the possibility that targeting Jagged1 may represent a promising therapeutic strategy to overcome chemoresistance and improve clinical outcomes in CRC patients.

Author Contributions

Conceptualization, D.B.; literature search & data curation, D.B. and S.Z.; writing—original draft preparation, D.B. and S.Z.; writing—review and editing, D.B., S.Z. and F.D.F.; critical revision of intellectual content and supervision, R.P. and C.T.; visualization, F.D.F.; funding acquisition, D.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Italian University and Research Ministry (PRIN-2022 20223NY37M) to D.B. We also acknowledge Sapienza University Research funding (RG123188B397E163) to D.B.; Istituto Pasteur Italia—Fondazione Cenci Bolognetti—(Call 2020_Anna Tramontano_under 60) to D.B.; and “European Union—NextGenerationEU through the Italian Ministry of University and Research under PNRR—M4C2-I1.3 Project PE_00000019 “HEAL ITALIA” to D.B. CUP B53C22004000006.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We thank the “Fondazione Umberto Veronesi” for supporting the post-doctoral fellowship of Sabrina Zema. We also acknowledge the Italian Ministry of University and Research (MUR)—Grant Dipartimenti di Eccellenza 2023–2027.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the writing of the manuscript; or in the decision to publish.

Abbreviations

The following abbreviations are used in this manuscript:
5-FU5-fluorouracil
ANKAnkyrin repeat domain
APCAdenomatous polyposis coli
CIMPCpG island methylator phenotype
CINChromosomal instability
CLLChronic lymphocytic leukemia
CMSConsensus molecular subtypes
CRCColorectal cancer
CRDCysteine-rich domain
CSCCancer stem cell
CSLCBF1/RBP-Jκ/Su(H)/Lag-1
DLLDelta-like proteins
DSLDelta/Serrate/Lag-2
ECDExtracellular domain
EGFEpidermal growth factor
EGFREpidermal growth factor receptor
EMTEpithelial–mesenchymal transition
FAPFamilial Adenomatous Polyposis
GSIγ-secretase inhibitor
ICDIntracellular domain
LNRLin12-Notch repeats
MAPKMitogen-activated protein kinase
mCRCMetastatic CRC
MDSCMyeloid-derived suppressor cells
MFNGManic Fringe
MMRMismatch repair
MSIMicrosatellite instability
NRRNegative regulatory region
OXAOxaliplatin
PI3KPhosphatidylinositol 3-kinase
RAMRBP-Jκ-associated module
T-ALLT-cell acute lymphoblastic leukemia
TAMTumor-associated macrophages
TMETumor microenvironment
TMICDMembrane-tethered intracellular fragment
TNMTumor, Node, Metastasis

References

  1. Morgan, E.; Arnold, M.; Gini, A.; Lorenzoni, V.; Cabasag, C.J.; Laversanne, M.; Vignat, J.; Ferlay, J.; Murphy, N.; Bray, F. Global Burden of Colorectal Cancer in 2020 and 2040: Incidence and Mortality Estimates from GLOBOCAN. Gut 2023, 72, 338–344. [Google Scholar] [CrossRef]
  2. Li, J.; Pan, J.; Wang, L.; Ji, G.; Dang, Y. Colorectal Cancer: Pathogenesis and Targeted Therapy. MedComm 2025, 6, e70127. [Google Scholar] [CrossRef]
  3. Fre, S.; Pallavi, S.K.; Huyghe, M.; Laé, M.; Janssen, K.-P.; Robine, S.; Artavanis-Tsakonas, S.; Louvard, D. Notch and Wnt Signals Cooperatively Control Cell Proliferation and Tumorigenesis in the Intestine. Proc. Natl. Acad. Sci. USA 2009, 106, 6309–6314. [Google Scholar] [CrossRef]
  4. Chu, D.; Wang, W.; Xie, H.; Li, Y.; Dong, G.; Xu, C.; Chen, D.; Zheng, J.; Li, M.; Lu, Z.; et al. Notch1 Expression in Colorectal Carcinoma Determines Tumor Differentiation Status. J. Gastrointest. Surg. 2009, 13, 253–260. [Google Scholar] [CrossRef]
  5. Zheng, C.-G.; Chen, R.; Xie, J.-B.; Liu, C.-B.; Jin, Z.; Jin, C. Immunohistochemical Expression of Notch1, Jagged1, NF-ΚB and MMP-9 in Colorectal Cancer Patients and the Relationship to Clinicopathological Parameters. Cancer Biomark. 2015, 15, 889–897. [Google Scholar] [CrossRef] [PubMed]
  6. Reedijk, M.; Odorcic, S.; Zhang, H.; Chetty, R.; Tennert, C.; Dickson, B.C.; Lockwood, G.; Gallinger, S.; Egan, S.E. Activation of Notch Signaling in Human Colon Adenocarcinoma. Int. J. Oncol. 2008, 33, 1223–1229. [Google Scholar] [CrossRef] [PubMed]
  7. Jackstadt, R.; van Hooff, S.R.; Leach, J.D.; Cortes-Lavaud, X.; Lohuis, J.O.; Ridgway, R.A.; Wouters, V.M.; Roper, J.; Kendall, T.J.; Roxburgh, C.S.; et al. Epithelial NOTCH Signaling Rewires the Tumor Microenvironment of Colorectal Cancer to Drive Poor-Prognosis Subtypes and Metastasis. Cancer Cell 2019, 36, 319–336.e7. [Google Scholar] [CrossRef] [PubMed]
  8. Gonulcu, S.C.; Unal, B.; Bassorgun, I.C.; Ozcan, M.; Coskun, H.S.; Elpek, G.O. Expression of Notch Pathway Components (Numb, Itch, and Siah-1) in Colorectal Tumors: A Clinicopathological Study. World J. Gastroenterol. 2020, 26, 3814–3833. [Google Scholar] [CrossRef]
  9. Fender, A.W.; Nutter, J.M.; Fitzgerald, T.L.; Bertrand, F.E.; Sigounas, G. Notch-1 Promotes Stemness and Epithelial to Mesenchymal Transition in Colorectal Cancer. J. Cell. Biochem. 2015, 116, 2517–2527. [Google Scholar] [CrossRef]
  10. Zhao, J.L.; Ye, Y.C.; Gao, C.C.; Wang, L.; Ren, K.X.; Jiang, R.; Hu, S.J.; Liang, S.Q.; Bai, J.; Liang, J.L.; et al. Notch-Mediated Lactate Metabolism Regulates MDSC Development through the Hes1/MCT2/c-Jun Axis. Cell Rep. 2022, 38, 110451. [Google Scholar] [CrossRef]
  11. Fre, S.; Huyghe, M.; Mourikis, P.; Robine, S.; Louvard, D.; Artavanis-Tsakonas, S. Notch Signals Control the Fate of Immature Progenitor Cells in the Intestine. Nature 2005, 435, 964–968. [Google Scholar] [CrossRef]
  12. Sikandar, S.S.; Pate, K.T.; Anderson, S.; Dizon, D.; Edwards, R.A.; Waterman, M.L.; Lipkin, S.M. NOTCH Signaling Is Required for Formation and Self-Renewal of Tumor-Initiating Cells and for Repression of Secretory Cell Differentiation in Colon Cancer. Cancer Res. 2010, 70, 1469–1478. [Google Scholar] [CrossRef] [PubMed]
  13. Sugiyama, M.; Oki, E.; Nakaji, Y.; Tsutsumi, S.; Ono, N.; Nakanishi, R.; Sugiyama, M.; Nakashima, Y.; Sonoda, H.; Ohgaki, K.; et al. High Expression of the Notch Ligand Jagged-1 Is Associated with Poor Prognosis after Surgery for Colorectal Cancer. Cancer Sci. 2016, 107, 1705–1716. [Google Scholar] [CrossRef]
  14. Guilmeau, S.; Flandez, M.; Mariadason, J.M.; Augenlicht, L.H. Heterogeneity of Jagged1 Expression in Human and Mouse Intestinal Tumors: Implications for Targeting Notch Signaling. Oncogene 2010, 29, 992–1002. [Google Scholar] [CrossRef]
  15. Dai, Y.; Wilson, G.; Huang, B.; Peng, M.; Teng, G.; Zhang, D.; Zhang, R.; Ebert, M.P.A.; Chen, J.; Wong, B.C.Y.; et al. Silencing of Jagged1 Inhibits Cell Growth and Invasion in Colorectal Cancer. Cell Death Dis. 2014, 5, e1170. [Google Scholar] [CrossRef] [PubMed]
  16. Tan, Y.; Peng, J.; Wei, D.; Chen, P.; Zhao, Y. Effect of Jagged1 on the Proliferation and Migration of Colon Cancer Cells. Exp. Ther. Med. 2012, 4, 89–92. [Google Scholar] [CrossRef] [PubMed]
  17. De Falco, F.; Del Papa, B.; Baldoni, S.; Sabatini, R.; Falzetti, F.; Di Ianni, M.; Martelli, M.P.; Mezzasoma, F.; Pelullo, M.; Marconi, P.; et al. IL-4-Dependent Jagged1 Expression/Processing Is Associated with Survival of Chronic Lymphocytic Leukemia Cells but Not with Notch Activation. Cell Death Dis. 2018, 9, 1160. [Google Scholar] [CrossRef]
  18. Kim, E.J.; Kim, J.Y.; Kim, S.O.; Hong, N.; Choi, S.H.; Park, M.G.; Jang, J.; Ham, S.W.; Seo, S.; Lee, S.Y.; et al. The Oncogenic JAG1 Intracellular Domain Is a Transcriptional Cofactor That Acts in Concert with DDX17/SMAD3/TGIF2. Cell Rep. 2022, 41, 111626. [Google Scholar] [CrossRef]
  19. Kim, J.Y.; Hong, N.; Park, S.; Ham, S.W.; Kim, E.J.; Kim, S.O.; Jang, J.; Kim, Y.; Kim, J.K.; Kim, S.C.; et al. Jagged1 Intracellular Domain/SMAD3 Complex Transcriptionally Regulates TWIST1 to Drive Glioma Invasion. Cell Death Dis. 2023, 14, 822. [Google Scholar] [CrossRef]
  20. Pelullo, M.; Quaranta, R.; Talora, C.; Checquolo, S.; Cialfi, S.; Felli, M.P.; te Kronnie, G.; Borga, C.; Besharat, Z.M.; Palermo, R.; et al. Notch3/Jagged1 Circuitry Reinforces Notch Signaling and Sustains T-ALL. Neoplasia 2014, 16, 1007–1017. [Google Scholar] [CrossRef]
  21. Pelullo, M.; Nardozza, F.; Zema, S.; Quaranta, R.; Nicoletti, C.; Besharat, Z.M.Z.M.; Felli, M.P.M.P.; Cerbelli, B.; D’Amati, G.; Palermo, R.; et al. Kras/ADAM17-Dependent Jag1-ICD Reverse Signalling Sustains Colorectal Cancer Progression and Chemoresistance. Cancer Res. 2019, 79, 5575–5586. [Google Scholar] [CrossRef]
  22. Pelullo, M.; Zema, S.; De Carolis, M.; Cialfi, S.; Giuli, M.V.; Palermo, R.; Capalbo, C.; Giannini, G.; Screpanti, I.; Checquolo, S.; et al. 5FU/Oxaliplatin-Induced Jagged1 Cleavage Counteracts Apoptosis Induction in Colorectal Cancer: A Novel Mechanism of Intrinsic Drug Resistance. Front. Oncol. 2022, 12, 918763. [Google Scholar] [CrossRef] [PubMed]
  23. Patel, S.G.; Karlitz, J.J.; Yen, T.; Lieu, C.H.; Boland, C.R. The Rising Tide of Early-Onset Colorectal Cancer: A Comprehensive Review of Epidemiology, Clinical Features, Biology, Risk Factors, Prevention, and Early Detection. Lancet Gastroenterol. Hepatol. 2022, 7, 262–274. [Google Scholar] [CrossRef] [PubMed]
  24. Siegel, R.L.; Wagle, N.S.; Cercek, A.; Smith, R.A.; Jemal, A. Colorectal Cancer Statistics, 2023. CA Cancer J. Clin. 2023, 73, 233–254. [Google Scholar] [CrossRef]
  25. Fearon, E.R.; Vogelstein, B. A Genetic Model for Colorectal Tumorigenesis. Cell 1990, 61, 759–767. [Google Scholar] [CrossRef] [PubMed]
  26. Brisset, M.; Mehlen, P.; Meurette, O.; Hollande, F. Notch Receptor/Ligand Diversity: Contribution to Colorectal Cancer Stem Cell Heterogeneity. Front. Cell Dev. Biol. 2023, 11, 1231416. [Google Scholar] [CrossRef]
  27. Mundade, R.; Imperiale, T.F.; Prabhu, L.; Loehrer, P.J.; Lu, T. Genetic Pathways, Prevention, and Treatment of Sporadic Colorectal Cancer. Oncoscience 2014, 1, 400–406. [Google Scholar] [CrossRef]
  28. Weledji, E.P. The Etiology and Pathogenesis of Colorectal Cancer. Clin. Oncol. 2024, 9, 2046. [Google Scholar]
  29. Kastrinos, F.; Syngal, S. Inherited Colorectal Cancer Syndromes. Cancer J. 2011, 17, 405–415. [Google Scholar] [CrossRef]
  30. Yamane, L.; Scapulatempo-Neto, C.; Reis, R.M.; Guimarães, D.P. Serrated Pathway in Colorectal Carcinogenesis. World J. Gastroenterol. 2014, 20, 2634–2640. [Google Scholar] [CrossRef]
  31. Leslie, A.; Carey, F.A.; Pratt, N.R.; Steele, R.J.C. The Colorectal Adenoma-Carcinoma Sequence. Br. J. Surg. 2002, 89, 845–860. [Google Scholar] [CrossRef]
  32. Vogelstein, B.; Fearon, E.R.; Hamilton, S.R.; Kern, S.E.; Preisinger, A.C.; Leppert, M.; Smits, A.M.M.; Bos, J.L. Genetic Alterations during Colorectal-Tumor Development. N. Engl. J. Med. 1988, 319, 525–532. [Google Scholar] [CrossRef]
  33. Miyoshi, Y.; Nagase, H.; Horii, H.A.A.; Shigetoshi Ichii, S.N.; Aoki, T.; Miki, Y.; Mori, T.; Nakamura, Y. Somatic Mutations of the APC Gene in Colorectal Tumors: Mutation Cluster Region in the APC Gene. Hum. Mol. Genet. 1992, 1, 229–233. [Google Scholar] [CrossRef]
  34. Kinzler, K.W.; Nilbert, M.C.; Su, L.K.; Vogelstein, B.; Bryan, T.M.; Levy, D.B.; Smith, K.J.; Preisinger, A.C.; Hedge, P.; McKechnie, D.; et al. Identification of FAP Locus Genes from Chromosome 5q21. Science 1991, 253, 661–665. [Google Scholar] [CrossRef] [PubMed]
  35. Nagase, H.; Miyoshi, Y.; Horii, A.; Aoki, T.; Petersen, G.M.; Vogelstein, B.; Maher, E.; Ogawa, M.; Maruyama, M.; Utsunomiya, J.; et al. Screening for Germ-line Mutations in Familial Adenomatous Polyposis Patients: 61 New Patients and a Summary of 150 Unrelated Patients. Hum. Mutat. 1992, 1, 467–473. [Google Scholar] [CrossRef]
  36. Korinek, V.; Barker, N.; Morin, P.J.; Van Wichen, D.; De Weger, R.; Kinzler, K.W.; Vogelstein, B.; Clevers, H. Constitutive Transcriptional Activation by a β-Catenin-Tcf Complex in APC(−/−) Colon Carcinoma. Science 1997, 275, 1784–1787. [Google Scholar] [CrossRef]
  37. Van de Wetering, M.; Sancho, E.; Verweij, C.; De Lau, W.; Oving, I.; Hurlstone, A.; Van der Horn, K.; Batlle, E.; Coudreuse, D.; Haramis, A.P.; et al. The β-Catenin/TCF-4 Complex Imposes a Crypt Progenitor Phenotype on Colorectal Cancer Cells. Cell 2002, 111, 241–250. [Google Scholar] [CrossRef]
  38. Li, Q.; Geng, S.; Luo, H.; Wang, W.; Mo, Y.-Q.; Luo, Q.; Wang, L.; Song, G.-B.; Sheng, J.-P.; Xu, B. Signaling Pathways Involved in Colorectal Cancer: Pathogenesis and Targeted Therapy. Signal Transduct. Target. Ther. 2024, 9, 266. [Google Scholar] [CrossRef] [PubMed]
  39. Morin, P.J.; Sparks, A.B.; Korinek, V.; Barker, N.; Clevers, H.; Vogelstein, B.; Kinzler, K.W. Activation of β-Catenin-Tcf Signaling in Colon Cancer by Mutations in β-Catenin or APC. Science 1997, 275, 1787–1790. [Google Scholar] [CrossRef]
  40. Worthley, D.L.; Leggett, B.A. Colorectal Cancer: Molecular Features and Clinical Opportunities. Clin. Biochem. Rev. 2010, 31, 31–38. [Google Scholar] [PubMed]
  41. Pretlow, T.P.; Pretlow, T.G. Mutant KRAS in Aberrant Crypt Foci (ACF): Initiation of Colorectal Cancer? Biochim. Et Biophys. Acta (BBA)—Rev. Cancer 2005, 1756, 83–96. [Google Scholar] [CrossRef] [PubMed]
  42. Wood, L.D.; Parsons, D.W.; Jones, S.; Lin, J.; Sjöblom, T.; Leary, R.J.; Shen, D.; Boca, S.M.; Barber, T.; Ptak, J.; et al. The Genomic Landscapes of Human Breast and Colorectal Cancers. Science 2007, 318, 1108–1113. [Google Scholar] [CrossRef] [PubMed]
  43. Frattini, M.; Balestra, D.; Suardi, S.; Oggionni, M.; Alberici, P.; Radice, P.; Costa, A.; Daidone, M.G.; Leo, E.; Pilotti, S.; et al. Different Genetic Features Associated with Colon and Rectal Carcinogenesis. Clin. Cancer Res. 2004, 10, 4015–4021. [Google Scholar] [CrossRef]
  44. Vasovcak, P.; Pavlikova, K.; Sedlacek, Z.; Skapa, P.; Kouda, M.; Hoch, J.; Krepelova, A. Molecular Genetic Analysis of 103 Sporadic Colorectal Tumours in Czech Patients. PLoS ONE 2011, 6, e24114. [Google Scholar] [CrossRef]
  45. Sjöblom, T.; Jones, S.; Wood, L.D.; Parsons, D.W.; Lin, J.; Barber, T.D.; Mandelker, D.; Leary, R.J.; Ptak, J.; Silliman, N.; et al. The Consensus Coding Sequences of Human Breast and Colorectal Cancers. Science 2006, 314, 268–274. [Google Scholar] [CrossRef]
  46. Fabien, N.; Paulin, C.; Dubois, P.M.; Santoro, M.; Grieco, M.; Berger, N.; Fusco, A.; Galvain, D.; Barbier, Y. Detection of RET Oncogene Activation in Human Papillary Thyroid Carcinomas by in Situ Hybridisation. Br. J. Cancer 1992, 66, 1094–1098. [Google Scholar] [CrossRef]
  47. Van Schaeybroeck, S.; Kyula, J.N.; Fenton, A.; Fenning, C.S.; Sasazuki, T.; Shirasawa, S.; Longley, D.B.; Johnston, P.G. Oncogenic Kras Promotes Chemotherapy-Induced Growth Factor Shedding via ADAM17. Cancer Res. 2011, 71, 1071–1080. [Google Scholar] [CrossRef]
  48. Mologni, L.; Brussolo, S.; Ceccon, M.; Gambacorti-Passerini, C. Synergistic Effects of Combined Wnt/KRAS Inhibition in Colorectal Cancer Cells. PLoS ONE 2012, 7, e51449. [Google Scholar] [CrossRef]
  49. Lee, S.K.; Hwang, J.H.; Choi, K.Y. Interaction of the Wnt/β-Catenin and RAS-ERK Pathways Involving Co-Stabilization of Both β-Catenin and RAS Plays Important Roles in the Colorectal Tumorigenesis. Adv. Biol. Regul. 2018, 68, 46–54. [Google Scholar] [CrossRef]
  50. Saucier, C.; Rivard, N. Epithelial Cell Signalling in Colorectal Cancer Metastasis. In Metastasis of Colorectal Cancer; Springer: Dordrecht, The Netherlands, 2010. [Google Scholar]
  51. Moon, B.-S.; Jeong, W.-J.; Park, J.; Kim, T.I.; Min, D.S.; Choi, K.-Y. Role of Oncogenic K-Ras in Cancer Stem Cell Activation by Aberrant Wnt/β-Catenin Signaling. J. Natl. Cancer Inst. 2014, 106, djt373. [Google Scholar] [CrossRef] [PubMed]
  52. Lemieux, E.; Cagnol, S.; Beaudry, K.; Carrier, J.; Rivard, N. Oncogenic KRAS Signalling Promotes the Wnt/β-Catenin Pathway through LRP6 in Colorectal Cancer. Oncogene 2015, 34, 4914–4927. [Google Scholar] [CrossRef]
  53. D’Abaco, G.M.; Whitehead, R.H.; Burgess, A.W. Synergy between Apc Min and an Activated Ras Mutation Is Sufficient To Induce Colon Carcinomas. Mol. Cell. Biol. 1996, 16, 884–891. [Google Scholar] [CrossRef]
  54. Zeller, E.; Hammer, K.; Kirschnick, M.; Braeuning, A. Mechanisms of RAS/β-Catenin Interactions. Arch. Toxicol. 2013, 87, 611–632. [Google Scholar] [CrossRef]
  55. Kim, S.E.; Yoon, J.Y.; Jeong, W.J.; Jeon, S.H.; Park, Y.; Yoon, J.B.; Park, Y.N.; Kim, H.; Choi, K.Y. H-Ras Is Degraded by Wnt/β-Catenin Signaling via β-TrCP-Mediated Polyubiquitylation. J. Cell Sci. 2009, 122, 842–848. [Google Scholar] [CrossRef]
  56. Jeong, W.J.; Yoon, J.; Park, J.C.; Lee, S.H.; Lee, S.H.; Kaduwal, S.; Kim, H.; Yoon, J.B.; Choi, K.Y. Ras Stabilization through Aberrant Activation of Wnt/β-Catenin Signaling Promotes Intestinal Tumorigenesis. Sci. Signal 2012, 5, ra30. [Google Scholar] [CrossRef]
  57. Rubinfeld, B.; Albert, I.; Porfiri, E.; Fiol, C.; Munemitsu, S.; Polakis, P. Binding of GSK3β to the APC-β-Catenin Complex and Regulation of Complex Assembly. Science 1996, 272, 1023–1026. [Google Scholar] [CrossRef]
  58. 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] [PubMed]
  59. Lee, S.; Jeong, W.; Cho, Y.; Cha, P.; Yoon, J.; Ro, E.J.; Choi, S.; Oh, J.; Heo, Y.; Kim, H.; et al. Β-Catenin- RAS Interaction Serves as a Molecular Switch for RAS Degradation via GSK 3β. EMBO Rep. 2018, 19, e46060. [Google Scholar] [CrossRef] [PubMed]
  60. Baker, S.J.; Preisinger, A.C.; Jessup, J.M.; Paraskeva, C.; Markowitz, S.; Willson, J.K.V.; Hamilton, S.; Vogelstein, B. P53 Gene Mutations Occur in Combination with 17p Allelic Deletions as Late Events in Colorectal Tumorigenesis. Cancer Res. 1990, 50, 7717–7722. [Google Scholar] [PubMed]
  61. Aiderus, A.; Barker, N.; Tergaonkar, V. Serrated Colorectal Cancer: Preclinical Models and Molecular Pathways. Trends Cancer 2024, 10, 76–91. [Google Scholar] [CrossRef]
  62. Rosenberg, D.W.; Yang, S.; Pleau, D.C.; Greenspan, E.J.; Stevens, R.G.; Rajan, T.V.; Heinen, C.D.; Levine, J.; Zhou, Y.; O’Brien, M.J. Mutations in BRAF and KRAS Differentially Distinguish Serrated versus Non-Serrated Hyperplastic Aberrant Crypt Foci in Humans. Cancer Res. 2007, 67, 3551–3554. [Google Scholar] [CrossRef]
  63. Nguyen, L.H.; Goel, A.; Chung, D.C. Pathways of Colorectal Carcinogenesis. Gastroenterology 2020, 158, 291–302. [Google Scholar] [CrossRef]
  64. Guinney, J.; Dienstmann, R.; Wang, X.; De Reyniès, A.; Schlicker, A.; Soneson, C.; Marisa, L.; Roepman, P.; Nyamundanda, G.; Angelino, P.; et al. The Consensus Molecular Subtypes of Colorectal Cancer. Nat. Med. 2015, 21, 1350–1356. [Google Scholar] [CrossRef] [PubMed]
  65. Dienstmann, R.; Vermeulen, L.; Guinney, J.; Kopetz, S.; Tejpar, S.; Tabernero, J. Consensus Molecular Subtypes and the Evolution of Precision Medicine in Colorectal Cancer. Nat. Rev. Cancer 2017, 17, 79–92. [Google Scholar] [CrossRef]
  66. Menter, D.G.; Davis, J.S.; Broom, B.M.; Overman, M.J.; Morris, J.; Kopetz, S. Back to the Colorectal Cancer Consensus Molecular Subtype Future. Curr. Gastroenterol. Rep. 2019, 21, 5. [Google Scholar] [CrossRef]
  67. Zhang, M.; Wu, D.; Tang, Y.; Zhang, L.; Zhang, S.; Li, W.; Li, N.; Yan, X. Targeting KRAS in Colorectal Cancer (Review). Mol. Clin. Oncol. 2025, 23, 28. [Google Scholar] [CrossRef]
  68. Bteich, F.; Mohammadi, M.; Li, T.; Bhat, M.A.; Sofianidi, A.; Wei, N.; Kuang, C. Targeting KRAS in Colorectal Cancer: A Bench to Bedside Review. Int. J. Mol. Sci. 2023, 24, 12030. [Google Scholar] [CrossRef] [PubMed]
  69. Infante, J.R.; Fecher, L.A.; Falchook, G.S.; Nallapareddy, S.; Gordon, M.S.; Becerra, C.; DeMarini, D.J.; Cox, D.S.; Xu, Y.; Morris, S.R.; et al. Safety, Pharmacokinetic, Pharmacodynamic, and Efficacy Data for the Oral MEK Inhibitor Trametinib: A Phase 1 Dose-Escalation Trial. Lancet Oncol. 2012, 13, 773–781. [Google Scholar] [CrossRef] [PubMed]
  70. Rosen, L.S.; LoRusso, P.; Ma, W.W.; Goldman, J.W.; Weise, A.; Colevas, A.D.; Adjei, A.; Yazji, S.; Shen, A.; Johnston, S.; et al. A First-in-Human Phase I Study to Evaluate the MEK1/2 Inhibitor, Cobimetinib, Administered Daily in Patients with Advanced Solid Tumors. Investig. New Drugs 2016, 34, 740–749. [Google Scholar] [CrossRef]
  71. Grilley-Olson, J.E.; Bedard, P.L.; Fasolo, A.; Cornfeld, M.; Cartee, L.; Razak, A.R.A.; Stayner, L.A.; Wu, Y.; Greenwood, R.; Singh, R.; et al. A Phase Ib Dose-Escalation Study of the MEK Inhibitor Trametinib in Combination with the PI3K/MTOR Inhibitor GSK2126458 in Patients with Advanced Solid Tumors. Investig. New Drugs 2016, 34, 740–749. [Google Scholar] [CrossRef]
  72. Deming, D.A.; Cavalcante, L.L.; Lubner, S.J.; Mulkerin, D.L.; Loconte, N.K.; Eickhoff, J.C.; Kolesar, J.M.; Fioravanti, S.; Greten, T.F.; Compton, K.; et al. A Phase i Study of Selumetinib (AZD6244/ARRY-142866), a MEK1/2 Inhibitor, in Combination with Cetuximab in Refractory Solid Tumors and KRAS Mutant Colorectal Cancer. Investig. New Drugs 2016, 34, 168–175. [Google Scholar] [CrossRef]
  73. Weisner, J.; Landel, I.; Reintjes, C.; Uhlenbrock, N.; Trajkovic-Arsic, M.; Dienstbier, N.; Hardick, J.; Ladigan, S.; Lindemann, M.; Smith, S.; et al. Preclinical Efficacy of Covalent-Allosteric AKT Inhibitor Borussertib in Combination with Trametinib in KRAS-Mutant Pancreatic and Colorectal Cancer. Cancer Res. 2019, 79, 2367–2378. [Google Scholar] [CrossRef] [PubMed]
  74. Sorokin, A.V.; Marie, P.K.; Bitner, L.; Syed, M.; Woods, M.; Manyam, G.; Kwong, L.N.; Johnson, B.; Morris, V.K.; Jones, P.; et al. Targeting RAS Mutant Colorectal Cancer with Dual Inhibition of MEK and CDK4/6. Cancer Res. 2022, 82, 3335–3344. [Google Scholar] [CrossRef]
  75. Sun, Q.; Burke, J.P.; Phan, J.; Burns, M.C.; Olejniczak, E.T.; Waterson, A.G.; Lee, T.; Rossanese, O.W.; Fesik, S.W. Discovery of Small Molecules That Bind to K-Ras and Inhibit Sos-Mediated Activation. Angew. Chem. Int. Ed. 2012, 51, 6140–6143. [Google Scholar] [CrossRef]
  76. Maurer, T.; Garrenton, L.S.; Oh, A.; Pitts, K.; Anderson, D.J.; Skelton, N.J.; Fauber, B.P.; Pan, B.; Malek, S.; Stokoe, D.; et al. Small-Molecule Ligands Bind to a Distinct Pocket in Ras and Inhibit SOS-Mediated Nucleotide Exchange Activity. Proc. Natl. Acad. Sci. USA 2012, 109, 5299–5304. [Google Scholar] [CrossRef]
  77. Cruz-Migoni, A.; Canning, P.; Quevedo, C.E.; Bataille, C.J.R.; Bery, N.; Miller, A.; Russell, A.J.; Phillips, S.E.V.; Carr, S.B.; Rabbitts, T.H. Structure-Based Development of New RAS-Effector Inhibitors from a Combination of Active and Inactive RAS-Binding Compounds. Proc. Natl. Acad. Sci. USA 2019, 116, 2545–2550. [Google Scholar] [CrossRef]
  78. Quevedo, C.E.; Cruz-Migoni, A.; Bery, N.; Miller, A.; Tanaka, T.; Petch, D.; Bataille, C.J.R.; Lee, L.Y.W.; Fallon, P.S.; Tulmin, H.; et al. Small Molecule Inhibitors of RAS-Effector Protein Interactions Derived Using an Intracellular Antibody Fragment. Nat. Commun. 2018, 9, 3169. [Google Scholar] [CrossRef]
  79. Ostrem, J.M.; Peters, U.; Sos, M.L.; Wells, J.A.; Shokat, K.M. K-Ras(G12C) Inhibitors Allosterically Control GTP Affinity and Effector Interactions. Nature 2013, 503, 548–551. [Google Scholar] [CrossRef]
  80. Janes, M.R.; Zhang, J.; Li, L.S.; Hansen, R.; Peters, U.; Guo, X.; Chen, Y.; Babbar, A.; Firdaus, S.J.; Darjania, L.; et al. Targeting KRAS Mutant Cancers with a Covalent G12C-Specific Inhibitor. Cell 2018, 172, 578–589.e17. [Google Scholar] [CrossRef] [PubMed]
  81. Kessler, D.; Gmachl, M.; Mantoulidis, A.; Martin, L.J.; Zoephel, A.; Mayer, M.; Gollner, A.; Covini, D.; Fischer, S.; Gerstberger, T.; et al. Drugging an Undruggable Pocket on KRAS. Proc. Natl. Acad. Sci. USA 2019, 116, 15823–15829. [Google Scholar] [CrossRef] [PubMed]
  82. Hong, D.S.; Fakih, M.G.; Strickler, J.H.; Desai, J.; Durm, G.A.; Shapiro, G.I.; Falchook, G.S.; Price, T.J.; Sacher, A.; Denlinger, C.S.; et al. KRAS G12C Inhibition with Sotorasib in Advanced Solid Tumors. N. Engl. J. Med. 2020, 383, 1207–1217. [Google Scholar] [CrossRef] [PubMed]
  83. Ou, S.H.I.; Jänne, P.A.; Leal, T.A.; Rybkin, I.I.; Sabari, J.K.; Barve, M.A.; Bazhenova, L.; Johnson, M.L.; Velastegui, K.L.; Cilliers, C.; et al. First-in-Human Phase I/IB Dose-Finding Study of Adagrasib (MRTX849) in Patients with Advanced KRAS G12CSolid Tumors (KRYSTAL-1). J. Clin. Oncol. 2022, 40, 2530–2538. [Google Scholar] [CrossRef]
  84. Amodio, V.; Yaeger, R.; Arcella, P.; Cancelliere, C.; Lamba, S.; Lorenzato, A.; Arena, S.; Montone, M.; Mussolin, B.; Bian, Y.; et al. Egfr Blockade Reverts Resistance to Krasg12c Inhibition in Colorectal Cancer. Cancer Discov. 2020, 10, 1129–1139. [Google Scholar] [CrossRef]
  85. Pellatt, A.J.; Bhamidipati, D.; Subbiah, V. Ready, Set, Go: Setting Off on the Mission to Target KRAS in Colorectal Cancer. JCO Oncol. Pr. 2024, 20, 1289–1292. [Google Scholar] [CrossRef]
  86. Kumar, A.; Gautam, V.; Sandhu, A.; Rawat, K.; Sharma, A.; Saha, L. Current and Emerging Therapeutic Approaches for Colorectal Cancer: A Comprehensive Review. World J. Gastrointest. Surg. 2023, 15, 495–519. [Google Scholar] [CrossRef]
  87. Oda, T.; Elkahloun, A.G.; Pike, B.L.; Okajima, K.; Krantz, I.D.; Genin, A.; Piccoli, D.A.; Meltzer, P.S.; Spinner, N.B.; Collins, F.S.; et al. Mutations in the Human Jagged1 Gene Are Responsible for Alagille Syndrome. Nat. Genet. 1997, 16, 235–242. [Google Scholar] [CrossRef] [PubMed]
  88. Bellavia, D.; Checquolo, S.; Campese, A.F.; Felli, M.P.; Gulino, A.; Screpanti, I. Notch3: From Subtle Structural Differences to Functional Diversity. Oncogene 2008, 27, 5092–5098. [Google Scholar] [CrossRef]
  89. Taylor, P.; Takeuchi, H.; Sheppard, D.; Chillakuri, C.; Lea, S.M.; Haltiwanger, R.S.; Handford, P.A. Fringe-Mediated Extension of O-Linked Fucose in the Ligand-Binding Region of Notch1 Increases Binding to Mammalian Notch Ligands. Proc. Natl. Acad. Sci. USA 2014, 111, 7290–7295. [Google Scholar] [CrossRef] [PubMed]
  90. Meng, Y.; Sanlidag, S.; Jensen, S.A.; Burnap, S.A.; Struwe, W.B.; Larsen, A.H.; Feng, X.; Mittal, S.; Sansom, M.S.P.; Sahlgren, C.; et al. An N-Glycan on the C2 Domain of JAGGED1 Is Important for Notch Activation. Sci. Signal 2022, 15, eabo3507. [Google Scholar] [CrossRef]
  91. Grochowski, C.M.; Loomes, K.M.; Spinner, N.B. Jagged1 (JAG1): Structure, Expression, and Disease Associations. Gene 2016, 576, 381–384. [Google Scholar] [CrossRef]
  92. Luca, V.C.; Kim, B.C.; Ge, C.; Kakuda, S.; Wu, D.; Roein-Peikar, M.; Haltiwanger, R.S.; Zhu, C.; Ha, T.; Garcia, K.C. Notch-Jagged Complex Structure Implicates a Catch Bond in Tuning Ligand Sensitivity. Science 2017, 355, 1320–1324. [Google Scholar] [CrossRef]
  93. Gordon, W.R.; Vardar-Ulu, D.; Histen, G.; Sanchez-Irizarry, C.; Aster, J.C.; Blacklow, S.C. Structural Basis for Autoinhibition of Notch. Nat. Struct. Mol. Biol. 2007, 14, 295–300. [Google Scholar] [CrossRef]
  94. Zeronian, M.R.; Klykov, O.; Portell i de Montserrat, J.; Konijnenberg, M.J.; Gaur, A.; Scheltema, R.A.; Janssen, B.J.C. Notch–Jagged Signaling Complex Defined by an Interaction Mosaic. Proc. Natl. Acad. Sci. USA 2021, 118, e2102502118. [Google Scholar] [CrossRef] [PubMed]
  95. Lovendahl, K.N.; Blacklow, S.C.; Gordon, W.R. The Molecular Mechanism of Notch Activation. In Advances in Experimental Medicine and Biology; Springer: Cham, Switzerland, 2018; Volume 1066. [Google Scholar]
  96. Langridge, P.D.; Struhl, G. Epsin-Dependent Ligand Endocytosis Activates Notch by Force. Cell 2017, 171, 1383–1396. [Google Scholar] [CrossRef]
  97. Artavanis-Tsakonas, S.; Rand, M.D.; Lake, R.J. Notch Signaling: Cell Fate Control and Signal Integration in Development. Science 1999, 284, 770–776. [Google Scholar] [CrossRef] [PubMed]
  98. Palermo, R.; Checquolo, S.; Bellavia, D.; Screpanti, C.T. and I. The Molecular Basis of Notch Signaling Regulation: A Complex Simplicity. Curr. Mol. Med. 2014, 14, 34–44. [Google Scholar] [CrossRef]
  99. Shimizu, K.; Chiba, S.; Kumano, K.; Hosoya, N.; Takahashi, T.; Kanda, Y.; Hamada, Y.; Yazaki, Y.; Hirai, H. Mouse Jagged1 Physically Interacts with Notch2 and Other Notch Receptors. Assessment by Quantitative Methods. J. Biol. Chem. 1999, 274, 32961–32969. [Google Scholar] [CrossRef] [PubMed]
  100. Shimizu, K.; Chiba, S.; Hosoya, N.; Kumano, K.; Saito, T.; Kurokawa, M.; Kanda, Y.; Hamada, Y.; Hirai, H. Binding of Delta1, Jagged1, and Jagged2 to Notch2 Rapidly Induces Cleavage, Nuclear Translocation, and Hyperphosphorylation of Notch2. Mol. Cell Biol. 2000, 20, 6913–6922. [Google Scholar] [CrossRef]
  101. Chapman, G.; Major, J.A.; Iyer, K.; James, A.C.; Pursglove, S.E.; Moreau, J.L.M.; Dunwoodie, S.L. Notch1 Endocytosis Is Induced by Ligand and Is Required for Signal Transduction. Biochim. Biophys. Acta Mol. Cell Res. 2016, 1863, 166–177. [Google Scholar] [CrossRef]
  102. Parks, A.L.; Klueg, K.M.; Stout, J.R.; Muskavitch, M.A.T. Ligand Endocytosis Drives Receptor Dissociation and Activation in the Notch Pathway. Development 2000, 127, 1373–1385. [Google Scholar] [CrossRef]
  103. Weinmaster, G. The Ins and Outs of Notch Signaling. Mol. Cell. Neurosci. 1997, 9, 91–102. [Google Scholar] [CrossRef] [PubMed]
  104. Kopan, R.; Ilagan, M.X.G. The Canonical Notch Signaling Pathway: Unfolding the Activation Mechanism. Cell 2009, 137, 216–233. [Google Scholar] [CrossRef]
  105. Talora, C.; Campese, A.F.; Bellavia, D.; Pascucci, M.; Checquolo, S.; Groppioni, M.; Frati, L.; von Boehmer, H.; Gulino, A.; Screpanti, I. Pre-TCR-Triggered ERK Signalling-Dependent Downregulation of E2A Activity in Notch3-Induced T-Cell Lymphoma. EMBO Rep. 2003, 4, 1067–1071. [Google Scholar] [CrossRef]
  106. Bellavia, D.; Campese, A.F.; Checquolo, S.; Balestri, A.; Biondi, A.; Cazzaniga, G.; Lendahl, U.; Fehling, H.J.; Hayday, A.C.; Frati, L.; et al. Combined Expression of PTα and Notch3 in T Cell Leukemia Identifies the Requirement of PreTCR for Leukemogenesis. Proc. Natl. Acad. Sci. USA 2002, 99, 3788–3793. [Google Scholar] [CrossRef]
  107. Petrovic, J.; Zhou, Y.; Fasolino, M.; Goldman, N.; Schwartz, G.W.; Mumbach, M.R.; Nguyen, S.C.; Rome, K.S.; Sela, Y.; Zapataro, Z.; et al. Oncogenic Notch Promotes Long-Range Regulatory Interactions within Hyperconnected 3D Cliques. Mol. Cell 2019, 73, 1174–1190.e12. [Google Scholar] [CrossRef]
  108. Chen, X.; Stoeck, A.; Lee, S.J.; Shih, I.-M.; Wang, M.M.; Wang, T.-L. Jagged1 Expression Regulated by Notch3 and Wnt/β-Catenin Signaling Pathways in Ovarian Cancer. Oncotarget 2010, 1, 210–218. [Google Scholar] [CrossRef]
  109. Zhang, R.; Engler, A.; Taylor, V. Notch: An Interactive Player in Neurogenesis and Disease. Cell Tissue Res. 2018, 371, 73–89. [Google Scholar] [CrossRef] [PubMed]
  110. Louvi, A.; Artavanis-Tsakonas, S. Notch Signalling in Vertebrate Neural Development. Nat. Rev. Neurosci. 2006, 7, 93–102. [Google Scholar] [CrossRef] [PubMed]
  111. Akil, A.; Gutiérrez-García, A.K.; Guenter, R.; Rose, J.B.; Beck, A.W.; Chen, H.; Ren, B. Notch Signaling in Vascular Endothelial Cells, Angiogenesis, and Tumor Progression: An Update and Prospective. Front. Cell Dev. Biol. 2021, 9, 642352. [Google Scholar] [CrossRef]
  112. Pui, J.C.; Allman, D.; Xu, L.; DeRocco, S.; Karnell, F.G.; Bakkour, S.; Lee, J.Y.; Kadesch, T.; Hardy, R.R.; Aster, J.C.; et al. Notch1 Expression in Early Lymphopoiesis Influences B versus T Lineage Determination. Immunity 1999, 11, 299–308. [Google Scholar] [CrossRef]
  113. Radtke, F.; Wilson, A.; Stark, G.; Bauer, M.; Van Meerwijk, J.; MacDonald, H.R.; Aguet, M. Deficient T Cell Fate Specification in Mice with an Induced Inactivation of Notch1. Immunity 1999, 10, 547–558. [Google Scholar] [CrossRef] [PubMed]
  114. Felli, M.P.; Maroder, M.; Mitsiadis, T.A.; Campese, A.F.; Bellavia, D.; Vacca, A.; Mann, R.S.; Frati, L.; Lendahl, U.; Gulino, A.; et al. Expression Pattern of Notch1, 2 and 3 and Jagged1 and 2 in Lymphoid and Stromal Thymus Components: Distinct Ligand-Receptor Interactions in Intrathymic T Cell Development. Int. Immunol. 1999, 11, 1017–1025. [Google Scholar] [CrossRef]
  115. Zhou, B.; Lin, W.; Long, Y.; Yang, Y.; Zhang, H.; Wu, K.; Chu, Q. Notch Signaling Pathway: Architecture, Disease, and Therapeutics. Signal Transduct. Target. Ther. 2022, 7, 95. [Google Scholar] [CrossRef]
  116. Brai, E.; Alina Raio, N.; Alberi, L. Notch1 Hallmarks Fibrillary Depositions in Sporadic Alzheimer’s Disease. Acta Neuropathol. Commun. 2016, 4, 64. [Google Scholar] [CrossRef]
  117. Mašek, J.; Andersson, E.R. The Developmental Biology of Genetic Notch Disorders. Development 2017, 144, 1743–1763. [Google Scholar] [CrossRef]
  118. Siebel, C.; Lendahl, U. Notch Signaling in Development, Tissue Homeostasis, and Disease. Physiol. Rev. 2017, 97, 1235–1294. [Google Scholar] [CrossRef] [PubMed]
  119. Ho, D.M.; Artavanis-Tsakonas, S.; Louvi, A. The Notch Pathway in CNS Homeostasis and Neurodegeneration. Wiley Interdiscip. Rev. Dev. Biol. 2020, 9, e358. [Google Scholar] [CrossRef] [PubMed]
  120. Shi, Q.; Xue, C.; Zeng, Y.; Yuan, X.; Chu, Q.; Jiang, S.; Wang, J.; Zhang, Y.; Zhu, D.; Li, L. Notch Signaling Pathway in Cancer: From Mechanistic Insights to Targeted Therapies. Signal Transduct. Target. Ther. 2024, 9, 128. [Google Scholar] [CrossRef]
  121. Ellisen, L.W.; Bird, J.; West, D.C.; Soreng, A.L.; Reynolds, T.C.; Smith, S.D.; Sklar, J. TAN-1, the Human Homolog of the Drosophila Notch Gene, Is Broken by Chromosomal Translocations in T Lymphoblastic Neoplasms. Cell 1991, 66, 649–661. [Google Scholar] [CrossRef]
  122. Weng, A.P.; Ferrando, A.A.; Lee, W.; Morris, J.P.; Silverman, L.B.; Sanchez-Irizarry, C.; Blacklow, S.C.; Look, A.T.; Aster, J.C. Activating Mutations of NOTCH1 in Human T Cell Acute Lymphoblastic Leukemia. Science 2004, 306, 269–271. [Google Scholar] [CrossRef]
  123. Pear, W.S.; Aster, J.C.; Scott, M.L.; Hasserjian, R.P.; Soffer, B.; Sklar, J.; Baltimore, D. Exclusive Development of T Cell Neoplasms in Mice Transplanted with Bone Marrow Expressing Activated Notch Alleles. J. Exp. Med. 1996, 183, 2283–2291. [Google Scholar] [CrossRef] [PubMed]
  124. Bernasconi-Elias, P.; Hu, T.; Jenkins, D.; Firestone, B.; Gans, S.; Kurth, E.; Capodieci, P.; Deplazes-Lauber, J.; Petropoulos, K.; Thiel, P.; et al. Characterization of Activating Mutations of NOTCH3 in T-Cell Acute Lymphoblastic Leukemia and Anti-Leukemic Activity of NOTCH3 Inhibitory Antibodies. Oncogene 2016, 35, 6077–6086. [Google Scholar] [CrossRef]
  125. Bellavia, D.; Campese, A.F.; Alesse, E.; Vacca, A.; Felli, M.P.; Balestri, A.; Stoppacciaro, A.; Tiveron, C.; Tatangelo, L.; Giovarelli, M.; et al. Constitutive Activation of NF-KappaB and T-Cell Leukemia/Lymphoma in Notch3 Transgenic Mice. EMBO J. 2000, 19, 3337–3348. [Google Scholar] [CrossRef]
  126. Giuli, M.V.; Diluvio, G.; Giuliani, E.; Franciosa, G.; Di Magno, L.; Pignataro, M.G.; Tottone, L.; Nicoletti, C.; Besharat, Z.M.; Peruzzi, G.; et al. Notch3 Contributes to T-Cell Leukemia Growth via Regulation of the Unfolded Protein Response. Oncogenesis 2020, 9, 93. [Google Scholar] [CrossRef]
  127. Bellavia, D.; Campese, A.F.; Vacca, A.; Gulino, A.; Screpanti, I. Notch3, Another Notch in T Cell Development. Semin. Immunol. 2003, 15, 107–112. [Google Scholar] [CrossRef]
  128. Campese, A.F.; Grazioli, P.; Colantoni, S.; Anastasi, E.; Mecarozzi, M.; Checquolo, S.; De Luca, G.; Bellavia, D.; Frati, L.; Gulino, A.; et al. Notch3 and PTα/Pre-TCR Sustain the in Vivo Function of Naturally Occurring Regulatory T Cells. Int. Immunol. 2009, 21, 727–743. [Google Scholar] [CrossRef]
  129. Tardivon, D.; Antoszewski, M.; Zangger, N.; Nkosi, M.; Sordet-Dessimoz, J.; Hendriks, R.; Koch, U.; Radtke, F. Notch Signaling Promotes Disease Initiation and Progression in Murine Chronic Lymphocytic Leukemia. Blood 2021, 137, 3079–3092. [Google Scholar] [CrossRef] [PubMed]
  130. Bellavia, D.; Palermo, R.; Felli, M.P.; Screpanti, I.; Checquolo, S. Notch Signaling as a Therapeutic Target for Acute Lymphoblastic Leukemia. Expert Opin. Ther. Targets 2018, 22, 331–342. [Google Scholar] [CrossRef] [PubMed]
  131. Pelullo, M.; Zema, S.; Nardozza, F.; Checquolo, S.; Screpanti, I.; Bellavia, D. Wnt, Notch, and TGF-β Pathways Impinge on Hedgehog Signaling Complexity: An Open Window on Cancer. Front. Genet. 2019, 10, 711. [Google Scholar] [CrossRef]
  132. Zema, S.; Pelullo, M.; Nardozza, F.; Felli, M.P.; Screpanti, I.; Bellavia, D. A Dynamic Role of Mastermind-Like 1: A Journey Through the Main (Path)Ways Between Development and Cancer. Front. Cell Dev. Biol. 2020, 8, 1615. [Google Scholar] [CrossRef]
  133. Gragnani, L.; Lorini, S.; Marri, S.; Zignego, A.L. Role of Notch Receptors in Hematologic Malignancies. Cells 2021, 10, 16. [Google Scholar] [CrossRef]
  134. Sorrentino, C.; Cuneo, A.; Roti, G. Therapeutic Targeting of Notch Signaling Pathway in Hematological Malignancies. Mediterr. J. Hematol. Infect. Dis. 2019, 11, e2019037. [Google Scholar] [CrossRef]
  135. Nicolas, M.; Wolfer, A.; Raj, K.; Kummer, J.A.; Mill, P.; van Noort, M.; Hui, C.-C.; Clevers, H.; Dotto, G.P.; Radtke, F. Notch1 Functions as a Tumor Suppressor in Mouse Skin. Nat. Genet. 2003, 33, 416–421. [Google Scholar] [CrossRef]
  136. Wang, N.J.; Sanborn, Z.; Arnett, K.L.; Bayston, L.J.; Liao, W.; Proby, C.M.; Leigh, I.M.; Collisson, E.A.; Gordon, P.B.; Jakkula, L.; et al. Loss-of-Function Mutations in Notch Receptors in Cutaneous and Lung Squamous Cell Carcinoma. Proc. Natl. Acad. Sci. USA 2011, 108, 17761–17766. [Google Scholar] [CrossRef] [PubMed]
  137. Lefort, K.; Mandinova, A.; Ostano, P.; Kolev, V.; Calpini, V.; Kolfschoten, I.; Devgan, V.; Lieb, J.; Raffoul, W.; Hohl, D.; et al. Notch1 Is a P53 Target Gene Involved in Human Keratinocyte Tumor Suppression through Negative Regulation of ROCK1/2 and MRCKα Kinases. Genes. Dev. 2007, 21, 562–577. [Google Scholar] [CrossRef]
  138. Agrawal, N.; Frederick, M.J.; Pickering, C.R.; Bettegowda, C.; Chang, K.; Li, R.J.; Fakhry, C.; Xie, T.-X.; Zhang, J.; Wang, J.; et al. Exome Sequencing of Head and Neck Squamous Cell Carcinoma Reveals Inactivating Mutations in NOTCH1. Science 2011, 333, 1154–1157. [Google Scholar] [CrossRef]
  139. The Cancer Genome Atlas Research Network. Comprehensive, Integrative Genomic Analysis of Diffuse Lower-Grade Gliomas. N. Engl. J. Med. 2015, 372, 2481–2498. [Google Scholar] [CrossRef] [PubMed]
  140. Suzuki, H.; Aoki, K.; Chiba, K.; Sato, Y.; Shiozawa, Y.; Shiraishi, Y.; Shimamura, T.; Niida, A.; Motomura, K.; Ohka, F.; et al. Mutational Landscape and Clonal Architecture in Grade II and III Gliomas. Nat. Genet. 2015, 47, 458–468. [Google Scholar] [CrossRef] [PubMed]
  141. George, J.; Lim, J.S.; Jang, S.J.; Cun, Y.; Ozretia, L.; Kong, G.; Leenders, F.; Lu, X.; Fernández-Cuesta, L.; Bosco, G.; et al. Comprehensive Genomic Profiles of Small Cell Lung Cancer. Nature 2015, 524, 47–53. [Google Scholar] [CrossRef]
  142. Maraver, A.; Fernandez-Marcos, P.J.; Cash, T.P.; Mendez-Pertuz, M.; Dueñas, M.; Maietta, P.; Martinelli, P.; Muñoz-Martin, M.; Martínez-Fernández, M.; Cañamero, M.; et al. NOTCH Pathway Inactivation Promotes Bladder Cancer Progression. J. Clin. Investig. 2015, 125, 824–830. [Google Scholar] [CrossRef]
  143. Rampias, T.; Vgenopoulou, P.; Avgeris, M.; Polyzos, A.; Stravodimos, K.; Valavanis, C.; Scorilas, A.; Klinakis, A. A New Tumor Suppressor Role for the Notch Pathway in Bladder Cancer. Nat. Med. 2014, 20, 1199–1205. [Google Scholar] [CrossRef]
  144. Rekhtman, N.; Pietanza, M.C.; Hellmann, M.D.; Naidoo, J.; Arora, A.; Won, H.; Halpenny, D.F.; Wang, H.; Tian, S.K.; Litvak, A.M.; et al. Next-Generation Sequencing of Pulmonary Large Cell Neuroendocrine Carcinoma Reveals Small Cell Carcinoma-like and Non-Small Cell Carcinoma-like Subsets. Clin. Cancer Res. 2016, 22, 3618–3629. [Google Scholar] [CrossRef]
  145. Pecora, G.; Mancini, C.; Mazzilli, R.; Zamponi, V.; Telese, S.; Scalera, S.; Maugeri-Saccà, M.; Ciuffreda, L.; De Nicola, F.; Fanciulli, M.; et al. Genetic Insight into Lung Neuroendocrine Tumors: Notch and Wnt Signaling Pathways as Potential Targets. J. Transl. Med. 2025, 23, 538. [Google Scholar] [CrossRef]
  146. Kunnimalaiyaan, M.; Vaccaro, A.M.; Ndiaye, M.A.; Chen, H. Overexpression of the NOTCH1 Intracellular Domain Inhibits Cell Proliferation and Alters the Neuroendocrine Phenotype of Medullary Thyroid Cancer Cells. J. Biol. Chem. 2006, 281, 39819–39830. [Google Scholar] [CrossRef]
  147. Nowell, C.S.; Radtke, F. Notch as a Tumour Suppressor. Nat. Rev. Cancer 2017, 17, 145–159. [Google Scholar] [CrossRef] [PubMed]
  148. Rodilla, V.; Villanueva, A.; Obrador-Hevia, A.; Robert-Moreno, A.; Fernández-Majada, V.; Grilli, A.; López-Bigas, N.; Bellora, N.; Albà, M.M.; Torres, F.; et al. Jagged1 Is the Pathological Link between Wnt and Notch Pathways in Colorectal Cancer. Proc. Natl. Acad. Sci. USA 2009, 106, 6315–6320. [Google Scholar] [CrossRef] [PubMed]
  149. Sansone, P.; Storci, G.; Tavolari, S.; Guarnieri, T.; Giovannini, C.; Taffurelli, M.; Ceccarelli, C.; Santini, D.; Paterini, P.; Marcu, K.B.; et al. IL-6 Triggers Malignant Features in Mammospheres from Human Ductal Breast Carcinoma and Normal Mammary Gland. J. Clin. Investig. 2007, 17, 3988–4002. [Google Scholar] [CrossRef]
  150. Zavadil, J.; Cermak, L.; Soto-Nieves, N.; Böttinger, E.P. Integration of TGF-Beta/Smad and Jagged1/Notch Signalling in Epithelial-to-Mesenchymal Transition. EMBO J. 2004, 23, 1155–1165. [Google Scholar] [CrossRef] [PubMed]
  151. Yamamoto, M.; Taguchi, Y.; Ito-Kureha, T.; Semba, K.; Yamaguchi, N.; Inoue, J. NF-ΚB Non-Cell-Autonomously Regulates Cancer Stem Cell Populations in the Basal-like Breast Cancer Subtype. Nat. Commun. 2013, 4, 2299. [Google Scholar] [CrossRef]
  152. Zhang, W.; Yu, F.; Weng, J.; Zheng, Y.; Lin, J.; Qi, T.; Wei, Y.; Wang, D.; Zeng, H. SOX12 Promotes Stem Cell-Like Phenotypes and Osteosarcoma Tumor Growth by Upregulating JAGGED1. Stem Cells Int. 2021, 2021, 9941733. [Google Scholar] [CrossRef]
  153. Santagata, S.; Demichelis, F.; Riva, A.; Varambally, S.; Hofer, M.D.; Kutok, J.L.; Kim, R.; Tang, J.; Montie, J.E.; Chinnaiyan, A.M.; et al. JAGGED1 Expression Is Associated with Prostate Cancer Metastasis and Recurrence. Cancer Res. 2004, 64, 6854–6857. [Google Scholar] [CrossRef]
  154. Zhang, T.; Liu, H.; Liang, Y.; Liang, L.; Zheng, G.; Huang, H.; Wu, J.; Liao, G. Suppression of Tongue Squamous Cell Carcinoma Growth by Inhibition of Jagged1 in Vitro and in Vivo. J. Oral. Pathol. Med. 2013, 42, 322–331. [Google Scholar] [CrossRef]
  155. Wu, K.; Xu, L.; Zhang, L.; Lin, Z.; Hou, J. High Jagged1 Expression Predicts Poor Outcome in Clear Cell Renal Cell Carcinoma. Jpn. J. Clin. Oncol. 2010, 41, 411–416. [Google Scholar] [CrossRef]
  156. Reedijk, M.; Pinnaduwage, D.; Dickson, B.C.; Mulligan, A.M.; Zhang, H.; Bull, S.B.; O’Malley, F.P.; Egan, S.E.; Andrulis, I.L. JAG1 Expression Is Associated with a Basal Phenotype and Recurrence in Lymph Node-Negative Breast Cancer. Breast Cancer Res. Treat. 2008, 111, 439–448. [Google Scholar] [CrossRef]
  157. Lee, J.; Lee, J.; Kim, J.H. Association of Jagged1 Expression with Malignancy and Prognosis in Human Pancreatic Cancer. Cell. Oncol. 2020, 43, 821–834. [Google Scholar] [CrossRef]
  158. Platonova, N.; Lazzari, E.; Colombo, M.; Falleni, M.; Tosi, D.; Giannandrea, D.; Citro, V.; Casati, L.; Ronchetti, D.; Bolli, N.; et al. The Potential of JAG Ligands as Therapeutic Targets and Predictive Biomarkers in Multiple Myeloma. Int. J. Mol. Sci. 2023, 24, 14558. [Google Scholar] [CrossRef] [PubMed]
  159. Colombo, M.; Garavelli, S.; Mazzola, M.; Platonova, N.; Giannandrea, D.; Colella, R.; Apicella, L.; Lancellotti, M.; Lesma, E.; Ancona, S.; et al. Multiple Myeloma Exploits Jagged1 and Jagged2 to Promote Intrinsic and Bone Marrow-Dependent Drug Resistance. Haematologica 2020, 105, 1925–1936. [Google Scholar] [CrossRef] [PubMed]
  160. Choi, J.-H.; Park, J.T.; Davidson, B.; Morin, P.J.; Shih, I.-M.; Wang, T.-L. Jagged-1 and Notch3 Juxtacrine Loop Regulates Ovarian Tumor Growth and Adhesion. Cancer Res. 2008, 68, 5716–5723. [Google Scholar] [CrossRef] [PubMed]
  161. Xiu, M.-X.; Liu, Y.-M.; Kuang, B.-H. The Oncogenic Role of Jagged1/Notch Signaling in Cancer. Biomed. Pharmacother. 2020, 129, 110416. [Google Scholar] [CrossRef]
  162. Qi, H.; Rand, M.D.; Wu, X.; Sestan, N.; Wang, W.; Rakic, P.; Xu, T.; Artavanis-Tsakonas, S. Processing of the Notch Ligand Delta by the Metalloprotease Kuzbanian. Science 1999, 283, 91–94. [Google Scholar] [CrossRef]
  163. LaVoie, M.J.; Selkoe, D.J. The Notch Ligands, Jagged and Delta, Are Sequentially Processed by α-Secretase and Presenilin/γ-Secretase and Release Signaling Fragments. J. Biol. Chem. 2003, 278, 34427–34437. [Google Scholar] [CrossRef]
  164. Ascano, J.M.; Beverly, L.J.; Capobianco, A.J. The C-Terminal PDZ-Ligand of JAGGED1 Is Essential for Cellular Transformation. J. Biol. Chem. 2003, 278, 8771–8779. [Google Scholar] [CrossRef]
  165. Caolo, V.; Schulten, H.M.; Zhuang, Z.W.; Murakami, M.; Wagenaar, A.; Verbruggen, S.; Molin, D.G.M.; Post, M.J. Soluble Jagged-1 Inhibits Neointima Formation by Attenuating Notch-Herp2 Signaling. Arter. Thromb. Vasc. Biol. 2011, 31, 1059–1065. [Google Scholar] [CrossRef]
  166. Xiao, Y.; Gong, D.; Wang, W. Soluble Jagged1 Inhibits Pulmonary Hypertension by Attenuating Notch Signaling. Arter. Thromb. Vasc. Biol. 2013, 33, 2733–2739. [Google Scholar] [CrossRef] [PubMed]
  167. Urs, S.; Roudabush, A.; O’Neill, C.F.; Pinz, I.; Prudovsky, I.; Kacer, D.; Tang, Y.; Liaw, L.; Small, D.J. Soluble Forms of the Notch Ligands Delta1 and Jagged1 Promote in Vivo Tumorigenicity in NIH3T3 Fibroblasts with Distinct Phenotypes. Am. J. Pathol. 2008, 173, 865–878. [Google Scholar] [CrossRef]
  168. Sun, J.; Luo, Z.; Wang, G.; Wang, Y.; Wang, Y.; Olmedo, M.; Morandi, M.M.; Barton, S.; Kevil, C.G.; Shu, B.; et al. Notch Ligand Jagged1 Promotes Mesenchymal Stromal Cell-Based Cartilage Repair. Exp. Mol. Med. 2018, 50, 1–10. [Google Scholar] [CrossRef] [PubMed]
  169. Aho, S. Soluble Form of Jagged1: Unique Product of Epithelial Keratinocytes and a Regulator of Keratinocyte Differentiation. J. Cell Biochem. 2004, 92, 1271–1281. [Google Scholar] [CrossRef] [PubMed]
  170. Karanu, F.N.; Murdoch, B.; Gallacher, L.; Wu, D.M.; Koremoto, M.; Sakano, S.; Bhatia, M. The Notch Ligand Jagged-1 Represents a Novel Growth Factor of Human Hematopoietic Stem Cells. J. Exp. Med. 2000, 192, 1365–1372. [Google Scholar] [CrossRef]
  171. Masuya, M.; Katayama, N.; Hoshino, N.; Nishikawa, H.; Sakano, S.; Araki, H.; Mitani, H.; Suzuki, H.; Miyashita, H.; Kobayashi, K.; et al. The Soluble Notch Ligand, Jagged-1, Inhibits Proliferation of CD34+ Macrophage Progenitors. Int. J. Hematol. 2002, 75, 269–276. [Google Scholar] [CrossRef]
  172. Campese, A.F.; Grazioli, P.; de Cesaris, P.; Riccioli, A.; Bellavia, D.; Pelullo, M.; Padula, F.; Noce, C.; Verkhovskaia, S.; Filippini, A.; et al. Mouse Sertoli Cells Sustain De Novo Generation of Regulatory T Cells by Triggering the Notch Pathway Through Soluble JAGGED1. Biol. Reprod. 2014, 90, 53. [Google Scholar] [CrossRef]
  173. Metrich, M.; Bezdek Pomey, A.; Berthonneche, C.; Sarre, A.; Nemir, M.; Pedrazzini, T. Jagged1 Intracellular Domain-Mediated Inhibition of Notch1 Signalling Regulates Cardiac Homeostasis in the Postnatal Heart. Cardiovasc. Res. 2015, 108, 74–86. [Google Scholar] [CrossRef]
  174. Azimi, M.; Brown, N.L. Jagged1 Protein Processing in the Developing Mammalian Lens. Biol. Open 2019, 8, bio041095. [Google Scholar] [CrossRef]
  175. Kumar, S.; Park, H.S.; Lee, K. Jagged1 Intracellular Domain Modulates Steroidogenesis in Testicular Leydig Cells. PLoS ONE 2020, 15, e0244553. [Google Scholar] [CrossRef]
  176. Hock, B.; Böhme, B.; Karn, T.; Yamamoto, T.; Kaibuchi, K.; Holtrich, U.; Holland, S.; Pawson, T.; Rübsamen-Waigmann, H.; Strebhardt, K. PDZ-Domain-Mediated Interaction of the Eph-Related Receptor Tyrosine Kinase EphB3 and the Ras-Binding Protein AF6 Depends on the Kinase Activity of the Receptor. Proc. Natl. Acad. Sci. USA 1998, 95, 9779–9784. [Google Scholar] [CrossRef]
  177. Popovic, M.; Bella, J.; Zlatev, V.; Hodnik, V.; Anderluh, G.; Barlow, P.N.; Pintar, A.; Pongor, S. The Interaction of Jagged-1 Cytoplasmic Tail with Afadin PDZ Domain Is Local, Folding-Independent, and Tuned by Phosphorylation. J. Mol. Recognit. 2011, 24, 245–253. [Google Scholar] [CrossRef]
  178. Carmena, A.; Speicher, S.; Baylies, M. The PDZ Protein Canoe/AF-6 Links Ras-MAPK, Notch and Wingless/Wnt Signaling Pathways by Directly Interacting with Ras, Notch and Dishevelled. PLoS ONE 2006, 1, e66. [Google Scholar] [CrossRef]
  179. Liu, H.; Kennard, S.; Lilly, B. NOTCH3 Expression Is Induced in Mural Cells through an Autoregulatory Loop That Requires Endothelial-Expressed JAGGED1. Circ. Res. 2009, 104, 466–475. [Google Scholar] [CrossRef]
  180. Tran, T.T.; Lee, K. JAG1 Intracellular Domain Enhances AR Expression and Signaling and Promotes Stem-like Properties in Prostate Cancer Cells. Cancers 2022, 14, 5714. [Google Scholar] [CrossRef] [PubMed]
  181. He, W.; Tang, J.; Li, W.; Li, Y.; Mei, Y.; He, L.; Zhong, K.; Xu, R. Mutual Regulation of JAG2 and PRAF2 Promotes Migration and Invasion of Colorectal Cancer Cells Uncoupled from Epithelial-Mesenchymal Transition. Cancer Cell Int. 2019, 19, 160. [Google Scholar] [CrossRef] [PubMed]
  182. Li, G.; Zhou, Z.; Zhou, H.; Zhao, L.; Chen, D.; Chen, H.; Zou, H.; Qi, Y.; Jia, W.; Pang, L. The Expression Profile and Clinicopathological Significance of Notch1 in Patients with Colorectal Cancer: A Meta-Analysis. Future Oncol. 2017, 13, 2103–2118. [Google Scholar] [CrossRef] [PubMed]
  183. Chu, D.; Zheng, J.; Wang, W.; Zhao, Q.; Li, Y.; Li, J.; Xie, H.; Zhang, H.; Dong, G.; Xu, C.; et al. Notch2 Expression Is Decreased in Colorectal Cancer and Related to Tumor Differentiation Status. Ann. Surg. Oncol. 2009, 16, 3259–3266. [Google Scholar] [CrossRef]
  184. Chu, D.; Zhang, Z.; Zhou, Y.; Wang, W.; Li, Y.; Zhang, H.; Dong, G.; Zhao, Q.; Ji, G. Notch1 and Notch2 Have Opposite Prognostic Effects on Patients with Colorectal Cancer. Ann. Oncol. 2011, 22, 2440–2447. [Google Scholar] [CrossRef] [PubMed]
  185. Ozawa, T.; Kazama, S.; Akiyoshi, T.; Murono, K.; Yoneyama, S.; Tanaka, T.; Tanaka, J.; Kiyomatsu, T.; Kawai, K.; Nozawa, H.; et al. Nuclear Notch3 Expression Is Associated with Tumor Recurrence in Patients with Stage II and III Colorectal Cancer. Ann. Surg. Oncol. 2014, 21, 2650–2658. [Google Scholar] [CrossRef] [PubMed]
  186. Serafin, V.; Persano, L.; Moserle, L.; Esposito, G.; Ghisi, M.; Curtarello, M.; Bonanno, L.; Masiero, M.; Ribatti, D.; Stürzl, M.; et al. Notch3 Signalling Promotes Tumour Growth in Colorectal Cancer. J. Pathol. 2011, 224, 448–460. [Google Scholar] [CrossRef] [PubMed]
  187. Huang, K.; Luo, W.; Fang, J.; Yu, C.; Liu, G.; Yuan, X.; Liu, Y.; Wu, W. Notch3 Signaling Promotes Colorectal Tumor Growth by Enhancing Immunosuppressive Cells Infiltration in the Microenvironment. BMC Cancer 2023, 23, 55. [Google Scholar] [CrossRef]
  188. George, D.C.; Bertrand, F.E.; Sigounas, G. Notch-3 Affects Chemoresistance in Colorectal Cancer via DNA Base Excision Repair Enzymes. Adv. Biol. Regul. 2024, 91, 101013. [Google Scholar] [CrossRef]
  189. Sharma, A.K.; Nimisha; Apurva; Kumar, A.; Ali, A.; Singh Saluja, S.; Prasad, B. Elevated Expression of Notch 2 & Notch 3 Is Associated with Disease Progression in Colorectal Cancer. Int. J. Appl. Biol. Pharm. 2022, 13, 33–50. [Google Scholar] [CrossRef]
  190. Wang, F.; Long, J.; Li, L.; Wu, Z.X.; Da, T.T.; Wang, X.Q.; Huang, C.; Jiang, Y.H.; Yao, X.Q.; Ma, H.Q.; et al. Single-Cell and Spatial Transcriptome Analysis Reveals the Cellular Heterogeneity of Liver Metastatic Colorectal Cancer. Sci. Adv. 2023, 9, eadf5464. [Google Scholar] [CrossRef]
  191. Gowrikumar, S.; Ahmad, R.; Uppada, S.B.; Washington, M.K.; Shi, C.; Singh, A.B.; Dhawan, P. Upregulated Claudin-1 Expression Promotes Colitis-Associated Cancer by Promoting β-Catenin Phosphorylation and Activation in Notch/p-AKT-Dependent Manner. Oncogene 2019, 38, 5321–5337. [Google Scholar] [CrossRef]
  192. Meng, R.D.; Shelton, C.C.; Li, Y.M.; Qin, L.X.; Notterman, D.; Paty, P.B.; Schwartz, G.K. γ-Secretase Inhibitors Abrogate Oxaliplatin-Induced Activation of the Notch-1 Signaling Pathway in Colon Cancer Cells Resulting in Enhanced Chemosensitivity. Cancer Res. 2009, 69, 573–582. [Google Scholar] [CrossRef]
  193. Liu, H.; Yin, Y.; Hu, Y.; Feng, Y.; Bian, Z.; Yao, S.; Li, M.; You, Q.; Huang, Z. MiR-139-5p Sensitizes Colorectal Cancer Cells to 5-Fluorouracil by Targeting NOTCH-1. Pathol. Res. Pr. 2016, 212, 643–649. [Google Scholar] [CrossRef]
  194. Riccio, O.; van Gijn, M.E.; Bezdek, A.C.; Pellegrinet, L.; van Es, J.H.; Zimber-Strobl, U.; Strobl, L.J.; Honjo, T.; Clevers, H.; Radtke, F. Loss of Intestinal Crypt Progenitor Cells Owing to Inactivation of Both Notch1 and Notch2 Is Accompanied by Derepression of CDK Inhibitors P27Kip1 and P57Kip2. EMBO Rep. 2008, 9, 377–383. [Google Scholar] [CrossRef]
  195. Van Es, J.H.; Van Gijn, M.E.; Riccio, O.; Van Den Born, M.; Vooijs, M.; Begthel, H.; Cozijnsen, M.; Robine, S.; Winton, D.J.; Radtke, F.; et al. Notch/γ-Secretase Inhibition Turns Proliferative Cells in Intestinal Crypts and Adenomas into Goblet Cells. Nature 2005, 435, 959–963. [Google Scholar] [CrossRef]
  196. Wang, R.; Ye, X.; Bhattacharya, R.; Boulbes, D.R.; Fan, F.; Xia, L.; Ellis, L.M. A Disintegrin and Metalloproteinase Domain 17 Regulates Colorectal Cancer Stem Cells and Chemosensitivity Via Notch1 Signaling. Stem Cells Transl. Med. 2016, 5, 331–338. [Google Scholar] [CrossRef]
  197. Li, D.D.; Zhao, C.H.; Ding, H.W.; Wu, Q.; Ren, T.S.; Wang, J.; Chen, C.Q.; Zhao, Q.C. A Novel Inhibitor of ADAM17 Sensitizes Colorectal Cancer Cells to 5-Fluorouracil by Reversing Notch and Epithelial-Mesenchymal Transition in Vitro and in Vivo. Cell Prolif. 2018, 51, e12480. [Google Scholar] [CrossRef]
  198. Dosch, J.; Ziemke, E.; Wan, S.; Luker, K.; Welling, T.; Hardiman, K.; Fearon, E.; Thomas, S.; Flynn, M.; Rios-Doria, J.; et al. Targeting ADAM17 Inhibits Human Colorectal Adenocarcinoma Progression and Tumor-Initiating Cell Frequency. Oncotarget 2017, 8, 65090–65099. [Google Scholar] [CrossRef] [PubMed]
  199. Jimenez-Luna, C.; González-Flores, E.; Ortiz, R.; Martínez-González, L.J.; Antúnez-Rodríguez, A.; Expósito-Ruiz, M.; Melguizo, C.; Caba, O.; Prados, J. Circulating Ptgs2, Jag1, Gucy2c and Pgf Mrna in Peripheral Blood and Serum as Potential Biomarkers for Patients with Metastatic Colon Cancer. J. Clin. Med. 2021, 10, 2248. [Google Scholar] [CrossRef]
  200. Nakata, T.; Shimizu, H.; Nagata, S.; Ito, G.; Fujii, S.; Suzuki, K.; Kawamoto, A.; Ishibashi, F.; Kuno, R.; Anzai, S.; et al. Indispensable Role of Notch Ligand-Dependent Signaling in the Proliferation and Stem Cell Niche Maintenance of APC-Deficient Intestinal Tumors. Biochem. Biophys. Res. Commun. 2017, 482, 1296–1303. [Google Scholar] [CrossRef]
  201. Yamamoto, S.; Tateishi, K.; Kudo, Y.; Yamamoto, K.; Isagawa, T.; Nagae, G.; Nakatsuka, T.; Asaoka, Y.; Ijichi, H.; Hirata, Y.; et al. Histone Demethylase KDM4C Regulates Sphere Formation by Mediating the Cross Talk between Wnt and Notch Pathways in Colonic Cancer Cells. Carcinogenesis 2013, 34, 2380–2388. [Google Scholar] [CrossRef] [PubMed]
  202. Pannequin, J.; Bonnans, C.; Delaunay, N.; Ryan, J.; Bourgaux, J.-F.; Joubert, D.; Hollande, F. The Wnt Target Jagged-1 Mediates the Activation of Notch Signaling by Progastrin in Human Colorectal Cancer Cells. Cancer Res. 2009, 69, 6065–6073. [Google Scholar] [CrossRef] [PubMed]
  203. López-Arribillaga, E.; Rodilla, V.; Colomer, C.; Vert, A.; Shelton, A.; Cheng, J.H.; Yan, B.; Gonzalez-Perez, A.; Junttila, M.R.; Iglesias, M.; et al. Manic Fringe Deficiency Imposes Jagged1 Addiction to Intestinal Tumor Cells. Nat. Commun. 2018, 9, 2992. [Google Scholar] [CrossRef]
  204. Kuintzle, R.; Santat, L.A.; Elowitz, M.B. Diversity in Notch Ligand-Receptor Signaling Interactions. Elife 2025, 12, RP91422. [Google Scholar] [CrossRef]
  205. Stanley, P.; Okajima, T. Roles of Glycosylation in Notch Signaling. In Current Topics in Developmental Biology; Elsevier: Amsterdam, The Netherlands, 2010; Volume 92. [Google Scholar]
  206. Haltiwanger, R.S.; Stanley, P. Modulation of Receptor Signaling by Glycosylation: Fringe Is an O-Fucose-Β1,3-N-Acetylglucosaminyltransferase. Biochim. Biophys. Acta Gen. Subj. 2002, 1573, 328–335. [Google Scholar] [CrossRef]
  207. Tan, J.B.; Xu, K.; Cretegny, K.; Visan, I.; Yuan, J.S.; Egan, S.E.; Guidos, C.J. Lunatic and Manic Fringe Cooperatively Enhance Marginal Zone B Cell Precursor Competition for Delta-like 1 in Splenic Endothelial Niches. Immunity 2009, 30, 254–263. [Google Scholar] [CrossRef]
  208. Kim, M.-H.; Kim, H.-B.; Yoon, S.P.; Lim, S.-C.; Cha, M.J.; Jeon, Y.J.; Park, S.G.; Chang, I.-Y.; You, H.J. Colon Cancer Progression Is Driven by APEX1-Mediated Upregulation of Jagged. J. Clin. Investig. 2013, 123, 3211–3230. [Google Scholar] [CrossRef]
  209. Kim, H.B.; Lim, H.J.; Lee, H.J.; Park, J.H.; Park, S.G. Evaluation and Clinical Significance of Jagged-1-Activated Notch Signaling by APEX1 in Colorectal Cancer. Anticancer. Res. 2019, 39, 6097–6105. [Google Scholar] [CrossRef] [PubMed]
  210. Van Schaeybroeck, S.; Kalimutho, M.; Dunne, P.D.; Carson, R.; Allen, W.; Jithesh, P.V.; Redmond, K.L.; Sasazuki, T.; Shirasawa, S.; Blayney, J.; et al. ADAM17-Dependent c-MET-STAT3 Signaling Mediates Resistance to MEK Inhibitors in KRAS Mutant Colorectal Cancer. Cell Rep. 2014, 7, 1940–1955. [Google Scholar] [CrossRef]
  211. Saad, M.I.; Alhayyani, S.; McLeod, L.; Yu, L.; Alanazi, M.; Deswaerte, V.; Tang, K.; Jarde, T.; Smith, J.A.; Prodanovic, Z.; et al. ADAM 17 Selectively Activates the IL -6 Trans-signaling/ERK MAPK Axis in KRAS -addicted Lung Cancer. EMBO Mol. Med. 2019, 11, e9976. [Google Scholar] [CrossRef] [PubMed]
  212. Díaz-Rodríguez, E.; Montero, J.C.; Esparís-Ogando, A.; Yuste, L.; Pandiella, A. Extracellular Signal-Regulated Kinase Phosphorylates Tumor Necrosis Factor Alpha-Converting Enzyme at Threonine 735: A Potential Role in Regulated Shedding. Mol. Biol. Cell 2002, 13, 2031–2044. [Google Scholar] [CrossRef]
  213. Lu, J.; Ye, X.; Fan, F.; Xia, L.; Bhattacharya, R.; Bellister, S.; Tozzi, F.; Sceusi, E.; Zhou, Y.; Tachibana, I.; et al. Endothelial Cells Promote the Colorectal Cancer Stem Cell Phenotype through a Soluble Form of Jagged-1. Cancer Cell 2013, 23, 171–185. [Google Scholar] [CrossRef] [PubMed]
  214. Martinez-Balibrea, E.; Martínez-Cardus, A.; Gines, A.; Ruiz De Porras, V.; Moutinho, C.; Layos, L.; Manzano, J.L.; Buges, C.; Bystrup, S.; Esteller, M.; et al. Tumor-Related Molecular Mechanisms of Oxaliplatin Resistance. Mol. Cancer Ther. 2015, 14, 1767–1776. [Google Scholar] [CrossRef]
  215. Briffa, R.; Langdon, S.P.; Grech, G.; Harrison, D.J. Acquired and Intrinsic Resistance to Colorectal Cancer Treatment. In Colorectal Cancer—Diagnosis, Screening and Management; InTechOpen: London, UK, 2018. [Google Scholar]
  216. Curry, C.L.; Reed, L.L.; Golde, T.E.; Miele, L.; Nickoloff, B.J.; Foreman, K.E. Gamma Secretase Inhibitor Blocks Notch Activation and Induces Apoptosis in Kaposi’s Sarcoma Tumor Cells. Oncogene 2005, 24, 6333–6344. [Google Scholar] [CrossRef]
  217. Farnie, G.; Clarke, R.B.; Spence, K.; Pinnock, N.; Brennan, K.; Anderson, N.G.; Bundred, N.J. Novel Cell Culture Technique for Primary Ductal Carcinoma in Situ: Role of Notch and Epidermal Growth Factor Receptor Signaling Pathways. J. Natl. Cancer Inst. 2007, 99, 616–627. [Google Scholar] [CrossRef]
  218. Gounder, M.; Ratan, R.; Alcindor, T.; Schöffski, P.; van der Graaf, W.T.; Wilky, B.A.; Riedel, R.F.; Lim, A.; Smith, L.M.; Moody, S.; et al. Nirogacestat, a γ-Secretase Inhibitor for Desmoid Tumors. N. Engl. J. Med. 2023, 388, 898–912. [Google Scholar] [CrossRef]
  219. Sanchez-Martin, M.; Ambesi-Impiombato, A.; Qin, Y.; Herranz, D.; Bansal, M.; Girardi, T.; Paietta, E.; Tallman, M.S.; Rowe, J.M.; De Keersmaecker, K.; et al. Synergistic Antileukemic Therapies in NOTCH1-Induced T-ALL. Proc. Natl. Acad. Sci. USA 2017, 114, 2006–2011. [Google Scholar] [CrossRef]
  220. Govaerts, I.; Prieto, C.; Vandersmissen, C.; Gielen, O.; Jacobs, K.; Provost, S.; Nittner, D.; Maertens, J.; Boeckx, N.; De Keersmaecker, K.; et al. PSEN1-Selective Gamma-Secretase Inhibition in Combination with Kinase or XPO-1 Inhibitors Effectively Targets T Cell Acute Lymphoblastic Leukemia. J. Hematol. Oncol. 2021, 14, 97. [Google Scholar] [CrossRef]
  221. Kukcinaviciute, E.; Jonusiene, V.; Sasnauskiene, A.; Dabkeviciene, D.; Eidenaite, E.; Laurinavicius, A. Significance of Notch and Wnt Signaling for Chemoresistance of Colorectal Cancer Cells HCT116. J. Cell Biochem. 2018, 119, 5913–5920. [Google Scholar] [CrossRef]
  222. Aleksic, T.; Feller, S.M. Gamma-Secretase Inhibition Combined with Platinum Compounds Enhances Cell Death in a Large Subset of Colorectal Cancer Cells. Cell Commun. Signal. 2008, 6, 8. [Google Scholar] [CrossRef]
  223. Timme, C.R.; Gruidl, M.; Yeatman, T.J. Gamma-Secretase Inhibition Attenuates Oxaliplatin-Induced Apoptosis through Increased Mcl-1 and/or Bcl-XL in Human Colon Cancer Cells. Apoptosis 2013, 18, 1163–1174. [Google Scholar] [CrossRef]
  224. Strosberg, J.R.; Yeatman, T.; Weber, J.; Coppola, D.; Schell, M.J.; Han, G.; Almhanna, K.; Kim, R.; Valone, T.; Jump, H.; et al. A Phase II Study of RO4929097 in Metastatic Colorectal Cancer. Eur. J. Cancer 2012, 48, 997–1003. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of Jagged1 signaling mechanisms. Jagged1 can activate Notch receptors on adjacent cells through canonical trans-activation, or initiate a non-canonical, reverse signaling cascade within ligand-expressing cells following sequential proteolytic cleavages that generate the Jagged1 intracellular domain (Jag1-ICD). In the figure, solid arrows indicate activation events, nuclear translocation or proteolytic processing steps within the signaling cascade. Dashed arrows represent endocytosis or degradation processes. Bar-headed lines denote inhibitory effects. In the color scheme, light blue reflects the role of Jagged1 as a Notch ligand, pink represents the canonical Notch signal transduction pathway occurring in the signal-receiving cell and green depicts the non-canonical Jagged1 reverse signaling. The figure is created in https://BioRender.com. URL accessed on 14 November 2025.
Figure 1. Schematic representation of Jagged1 signaling mechanisms. Jagged1 can activate Notch receptors on adjacent cells through canonical trans-activation, or initiate a non-canonical, reverse signaling cascade within ligand-expressing cells following sequential proteolytic cleavages that generate the Jagged1 intracellular domain (Jag1-ICD). In the figure, solid arrows indicate activation events, nuclear translocation or proteolytic processing steps within the signaling cascade. Dashed arrows represent endocytosis or degradation processes. Bar-headed lines denote inhibitory effects. In the color scheme, light blue reflects the role of Jagged1 as a Notch ligand, pink represents the canonical Notch signal transduction pathway occurring in the signal-receiving cell and green depicts the non-canonical Jagged1 reverse signaling. The figure is created in https://BioRender.com. URL accessed on 14 November 2025.
Cells 14 01815 g001
Figure 2. Context-specific oncogenic functions of Jagged1 signaling. Jagged1 exerts a distinct oncogenic role depending on the cellular and molecular context, acting as either a Notch-co-activator or as an autonomous nuclear effector. In T-ALL (left panel), nuclear Jag1-ICD cooperates with Notch3-ICD and RBP-Jκ transcription factor within a transcriptional complex that amplifies JAG1 expression, establishing a positive feedback loop that reinforces Notch activation in both ligand and receptor-expressing cells, thereby sustaining leukemic cell proliferation and survival. In CLL (central panel), IL-4 promotes JAG1 expression and proteolytic processing through the PI3K/AKT signaling, concomitant with increased Notch activation, that correlates with enhanced cell survival. Conversely, in Glioma/Glioblastoma (right panel), Jag1-ICD exerts Notch-independent transcriptional activity by integrating into a DDX17/SMAD3/TGIF2 complex that induces SOX2 and TWIST1 expression to sustain self-renewal and EMT-related invasiveness. In prostate cancer (right panel), Jag1-ICD potentiates AR signaling and upregulates CD133 and NANOG expression, reinforcing cancer stem-like properties and malignant progression. Red solid arrows indicate activation event and nuclear translocation. In the figure, black solid arrows denote transcriptional induction. Yellow circles mark oncogenic nodes, highlighting factors that exert tumor-promoting effects in the depicted pathways. Background colors distinguish tumor types: green for T-ALL, pink for CLL and light blue for Glioma/Glioblastoma and prostate cancer. The figure is created in https://BioRender.com. URL accessed on 14 November 2025.
Figure 2. Context-specific oncogenic functions of Jagged1 signaling. Jagged1 exerts a distinct oncogenic role depending on the cellular and molecular context, acting as either a Notch-co-activator or as an autonomous nuclear effector. In T-ALL (left panel), nuclear Jag1-ICD cooperates with Notch3-ICD and RBP-Jκ transcription factor within a transcriptional complex that amplifies JAG1 expression, establishing a positive feedback loop that reinforces Notch activation in both ligand and receptor-expressing cells, thereby sustaining leukemic cell proliferation and survival. In CLL (central panel), IL-4 promotes JAG1 expression and proteolytic processing through the PI3K/AKT signaling, concomitant with increased Notch activation, that correlates with enhanced cell survival. Conversely, in Glioma/Glioblastoma (right panel), Jag1-ICD exerts Notch-independent transcriptional activity by integrating into a DDX17/SMAD3/TGIF2 complex that induces SOX2 and TWIST1 expression to sustain self-renewal and EMT-related invasiveness. In prostate cancer (right panel), Jag1-ICD potentiates AR signaling and upregulates CD133 and NANOG expression, reinforcing cancer stem-like properties and malignant progression. Red solid arrows indicate activation event and nuclear translocation. In the figure, black solid arrows denote transcriptional induction. Yellow circles mark oncogenic nodes, highlighting factors that exert tumor-promoting effects in the depicted pathways. Background colors distinguish tumor types: green for T-ALL, pink for CLL and light blue for Glioma/Glioblastoma and prostate cancer. The figure is created in https://BioRender.com. URL accessed on 14 November 2025.
Cells 14 01815 g002
Figure 3. The two-way role of Jagged in CRC. In CRC, sequential mutations in APC and KRAS drive tumor progression from dysplasia to adenocarcinoma. Loss of APC function leads to β-Catenin stabilization and nuclear accumulation, promoting JAG1 transcription and overexpression (steps 12). In the canonical signaling, membrane-bound Jagged1 acts as a ligand for Notch receptors on adjacent signal-receiving cells, activating downstream transcriptional programs that sustain EMT, self-renewal and anti-apoptotic signaling. In addition, oncogenic KRAS mutations activate the RAF/MEK/ERK cascade (step 3), inducing ADAM17 activation and Jagged1 proteolytic cleavage. The released Jag1-ICD translocates into the nucleus (step 4), functioning as a transcriptional co-effector independent of RPB-Jκ, which activates pro-survival and EMT-related genes. In the figure, solid arrows represent activation events, nuclear translocation, transcriptional induction or proteolytic processing steps within the signaling cascade, while yellow circles mark key oncogenic nodes that contribute to CRC progression. The tumor cell harboring APC and KRAS mutations is shown in light blue and the adjacent Notch receptor-expressing cell is depicted in pink. The figure is created in https://BioRender.com. URL accessed on 14 November 2025.
Figure 3. The two-way role of Jagged in CRC. In CRC, sequential mutations in APC and KRAS drive tumor progression from dysplasia to adenocarcinoma. Loss of APC function leads to β-Catenin stabilization and nuclear accumulation, promoting JAG1 transcription and overexpression (steps 12). In the canonical signaling, membrane-bound Jagged1 acts as a ligand for Notch receptors on adjacent signal-receiving cells, activating downstream transcriptional programs that sustain EMT, self-renewal and anti-apoptotic signaling. In addition, oncogenic KRAS mutations activate the RAF/MEK/ERK cascade (step 3), inducing ADAM17 activation and Jagged1 proteolytic cleavage. The released Jag1-ICD translocates into the nucleus (step 4), functioning as a transcriptional co-effector independent of RPB-Jκ, which activates pro-survival and EMT-related genes. In the figure, solid arrows represent activation events, nuclear translocation, transcriptional induction or proteolytic processing steps within the signaling cascade, while yellow circles mark key oncogenic nodes that contribute to CRC progression. The tumor cell harboring APC and KRAS mutations is shown in light blue and the adjacent Notch receptor-expressing cell is depicted in pink. The figure is created in https://BioRender.com. URL accessed on 14 November 2025.
Cells 14 01815 g003
Table 1. Taxonomy of CRC represented as four consensus molecular subtypes (CMS 1–4) [64].
Table 1. Taxonomy of CRC represented as four consensus molecular subtypes (CMS 1–4) [64].
CMS SubtypeSubtype NameMolecular and Genomic FeaturesPathway Activation/Biological CharacteristicsClinical/Prognostic Features
CMS1MSI ImmuneHypermutated DNA
MSI
CIMP-High
Frequent BRAF mutations
Strong immune activation and infiltrationPoor overall survival and relapse outcomes
CMS2CanonicalChromosomal instability (CIN)
High somatic copy number alterations (SCNA)
Activation of Wnt and Myc pathwaysTypical “classical” subtype with intermediate prognosis
CMS3MetabolicMixed MSI status
Low SCNA and low CIMP
KRAS mutations enriched
Epithelial features with metabolic dysregulationVariable prognosis; often linked to altered metabolism
CMS4MesenchymalHigh SCNA
Stromal infiltration and angiogenesis
Activation of TGFβ pathwayWorst prognosis, associated with poor relapse-free and overall survival
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zema, S.; Di Fazio, F.; Palermo, R.; Talora, C.; Bellavia, D. The Two-Way Role of Jagged1 in Cancer: A Focus on CRC. Cells 2025, 14, 1815. https://doi.org/10.3390/cells14221815

AMA Style

Zema S, Di Fazio F, Palermo R, Talora C, Bellavia D. The Two-Way Role of Jagged1 in Cancer: A Focus on CRC. Cells. 2025; 14(22):1815. https://doi.org/10.3390/cells14221815

Chicago/Turabian Style

Zema, Sabrina, Francesca Di Fazio, Rocco Palermo, Claudio Talora, and Diana Bellavia. 2025. "The Two-Way Role of Jagged1 in Cancer: A Focus on CRC" Cells 14, no. 22: 1815. https://doi.org/10.3390/cells14221815

APA Style

Zema, S., Di Fazio, F., Palermo, R., Talora, C., & Bellavia, D. (2025). The Two-Way Role of Jagged1 in Cancer: A Focus on CRC. Cells, 14(22), 1815. https://doi.org/10.3390/cells14221815

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