Next Article in Journal
Nitric Oxide Regulates Multiple Signal Pathways in Plants via Protein S-Nitrosylation
Previous Article in Journal
Bioinformatic RNA-Seq Functional Profiling of the Tumor Suppressor Gene OPCML in Ovarian Cancers: The Multifunctional, Pleiotropic Impacts of Having Three Ig Domains
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Mucins in Cancer and Cancer Progression: A Comprehensive Review

1
Department of Internal Medicine, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
2
Department of Surgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
3
Departments of Health Science Education and Pathology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
4
Health Care Engineering Systems Center, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2025, 47(6), 406; https://doi.org/10.3390/cimb47060406
Submission received: 15 April 2025 / Revised: 21 May 2025 / Accepted: 28 May 2025 / Published: 29 May 2025
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)

Abstract

:
Mucin, a heavily glycosylated glycoprotein, serves an important function in forming protective and immune defense barriers against the exterior environment on epithelial surfaces. While secreted-type mucins are involved in mucous production, transmembrane mucins, which contain O-glycosylated tandem repeats, play a pivotal role in cellular signaling, especially in immune modulation and mediating inflammatory response. However, dysregulation in mucin expressions, such as MUC1, MUC2, MUC4, MUC5AC, and MUC16, have been observed in many cancer cells. More specifically, alterations in the expression and glycosylation of MUC1 have been associated with the upregulation of pathways involving the cell proliferation, angiogenesis, migration, and invasion of cancer cells. With mucin’s extensive involvement in cancer biology, several mucin biomarkers, such as CA125, CA19-9, and CEA, have been utilized as diagnostic and prognostic monitoring biomarkers in ovarian, pancreatic, and colon cancer. Vaccines and antibody therapy against abnormal mucin glycosylation have also been investigated for potential therapy for mucin-related cancers that are resistant to traditional chemotherapy agents. Despite the lack of specificity in mucin biomarkers and challenges in efficient drug delivery systems, the current advancement in mucin-targeted immunotherapy highlighted the pivotal potential in developing therapeutic targets to improve cancer prognosis.

1. Introduction to Mucins and Cancer

Mucus, secreted by goblet cells, is a viscous secretion that covers epithelial surfaces throughout the body. Its main components are water, salts, immunoglobulins, proteins, and mucins [1,2]. Mucin’s high molecular weight and heavily O-linked glycoproteins provide stable structure and protective function to mucosal surfaces [2]. As a primary component of mucus, mucins form a gel-like apical coat that lines the interface between the epithelium and the external environment [3]. They contribute to the viscoelasticity and hydration of the mucus barrier, acting as bio-adhesives and molecular sieves.
Over the past five decades, studies have demonstrated the dysregulation of mucins in the progression of human malignancies, highlighting their roles in inflammation, cancer recognition, and therapy resistance [4]. Elevated mucin levels are observed in the mucus-secreting cells of gastric, colorectal, and pancreatic adenocarcinomas, affecting the progression of cancer cells in these tissues [5,6,7].
For instance, MUC1 binds to E-cadherin and alters its function, acting as a sequestration factor for β-catenin, a key protein in the Wnt signaling pathway. The 20 μm mucin-rich pericellular halo around cancer cells provides a stealth cloak against the immune system and a slippery barrier to drug penetration [2,8]. Within the cancer microenvironment, tumor cells are encapsulated in mucus [3]. Cleared mucus, including recombinant mucins, can elicit varying immune responses, from strong activation to negative regulation [9,10]. Consequently, clearing infiltrated mucin families is exploited in various pathophysiological conditions [4,11,12].

1.1. Definition and Classification of Mucins

Mucins are highly glycosylated proteins commonly associated with the apical cell surface [9,10]. These gel-forming glycoproteins protect epithelial surfaces from solute particles, chemical irritants, pathogens, and mechanical stress [10]. Mucins are broadly classified into two main categories: secreted and transmembrane. Secreted mucins form a physical protective barrier with a viscous gel at the epithelium, most commonly in the gastrointestinal and urinary systems. Transmembrane mucins contribute to this protective gel through their O-glycosylated tandem repeats that extend above the apical cell surface [4]. Secreted mucins, such as MUC2, MUC5AC, MUC5B, and MUC6, are encoded by a cluster of genes on chromosome 11q15 [11]. Due to their shared similarity in dimerization, domain organization, pH-dependent assembly, and protective function, secreted mucins are proposed to share a common ancestor with the von Willebrand factor (vWF) [12]. Secreted mucins can be divided into monomeric and gel-forming mucins [13]. Monomeric mucins, such as MUC7, found in saliva [14], and MUC20, present in the kidney and urinary tract [15], serve specialized functions in their respective locations. Gel-forming mucins, including MUC2, MUC5AC, and MUC5B, are primarily involved in mucus production in the respiratory and gastrointestinal tracts [16]. These mucins adhere to the cell surface to protect the epithelium from pathogenic and mechanical injuries [17,18]. Their gel-like properties enable them to trap pathogens and particulate matter, facilitating their removal from the body.
Transmembrane mucins, such as MUC1, MUC13, and MUC16, are hydrophobic and attached to the cell surface [19]. The highly glycosylated extracellular and intracellular domains also enable them to participate in signaling pathways [20]. MUC1, for example, interacts with various intracellular signaling molecules, influencing cellular responses to external stimuli and playing a role in cell adhesion and immune response modulation [20,21]. This multifunctionality makes transmembrane mucins critical in maintaining epithelial integrity and responding to environmental changes.

1.2. Overview of Cancer and Cancer Progression

Epithelial cells line the surface of the epithelium, playing essential roles in protection, cell signaling, inflammation, and infection [22]. These cells are single-layered, exhibit apical-basal polarity, and have high turnover rates [23,24]. Under normal conditions, mucins are located at the epithelium apical surface, facing the external environment, while receptor tyrosine kinases (RTKs) are situated on the basolateral side. Chronic stress and inflammation can cause epithelial cells to lose their polarity, leading to the repositioning and interaction of apical mucins with basolateral proteins [25]. This interaction between mucins and RTKs promotes the survival of inflamed tissue and causes goblet cell hyperplasia, resulting in the hypersecretion of mucins [26]. The loss of polarity, along with the disruption of tight junctions and adherens junctions, triggers an epithelial–mesenchymal transition (EMT). EMT endows cells with characteristics of cancer cells, further activating cellular proliferation, differentiation, and survival mechanisms [25,26,27,28,29,30,31]. Recent research has extensively studied the role of mucin glycoproteins (MUC) in cancer. For example, the activation of ERBB2 RTK by transmembrane MUC1 following the loss of cellular polarity has been linked to the disruption of tight and adherens junctions mediated by E-cadherin [25,31,32]. These alterations in mucin expression and interactions contribute significantly to cancer formation and progression. The transformation from normal to neoplasia occurs incrementally, advancing stepwise through stages of hyperproliferation, dysplasia, loss of differentiation, transformation, and metastasis [33,34]. These changes lead to the development of various subtypes of colonic tumors, including hyperplastic/serrated adenomas, tubular adenomas, tubulovillous adenomas, villous adenomas, and adenocarcinomas [35]. Understanding the mechanisms underlying these transformations, particularly the roles of mucins and epithelial cell polarity, provides valuable insights into the development and progression of cancer, which is crucial for developing targeted therapies and diagnostic tools to combat cancer effectively.

2. Structure and Function of Mucins

Mucins are structurally characterized as large, viscous glycoproteins primarily composed of carbohydrates and amino acids linked via O-glycosylation [36]. At the molecular level, mucins are elongated, rod-shaped molecules with a linear polypeptide core [37]. The glycosylated regions, which often contain simple disaccharides like sialic acid, maintain the mucin molecule’s configuration, bind to water, promote hydration, and contribute to gel formation [36,37]. Proline residues play a crucial role in ensuring proper unfolding during the process of O-glycosylation. In contrast, non-glycosylated regions are rich in cysteine, which is vital for polymerization [3]. This polymerization is critical for the aggregation of intestinal [38] and tracheobronchial mucins [39].
Both intramolecular and intermolecular interactions are essential for mucin’s structural organization, including the cross-linking of mucin polymers leading to gel formation. Mucin secretions possess significant protective and lubricative properties. In the upper gastrointestinal (GI) tract, such as the stomach, mucins protect epithelial cells from the corrosive effects of digestive acids [40]. In the lower GI tract, particularly in the colon, mucins form a protective interface between host cells and the assemblage of gut microbiota and bacterial antigens [41,42]. The extensive mucin glycosylation serves in host defense by maintaining gut homeostasis and preventing pathogen invasion [13]. More specifically, the carbohydrate chains on mucins act as decoy receptors for pathogens, preventing their adherence to epithelial cells and subsequent infection [43,44]. Additionally, the highly hydrated nature of mucins allows the trapping of microorganisms, facilitating their clearance from the epithelial surface [45]. Furthermore, the regulation of mucin secretion and composition is finely tuned to respond to various physiological and pathological stimuli. For example, in response to inflammation or infection, there is often an upregulation of mucin production to reinforce the protective barrier [46]. The dysregulation of mucin expression patterns, on the other hand, can lead to diseases such as cystic fibrosis, inflammatory bowel disease, and certain cancers [45,47,48].

2.1. General Features of Transmembrane Mucins

2.1.1. Extracellular Domain

Transmembrane mucins are distinguished by their extracellular domains, which are rich in tandem repeat sequences composed predominantly of serine (Ser), threonine (Thr), and proline (Pro) residues [13,49]. These sequences are heavily O-glycosylated, which serves to shield the protein backbone from proteolytic degradation by bacteria and host proteases, thereby contributing to the formation of a robust barrier [50]. The glycan composition of mucins can vary significantly due to the differential expression of glycosyltransferases, resulting in a wide array of oligosaccharide structures. The terminal glycan residues, such as Gal, GlcNAc, Fuc, GalNAc, and sialic acid, can define blood group antigens and histo-blood group antigens, resulting in the A, B, and H as well as Lewis a, b, x, and y epitopes on mucins, within cells, and on erythrocytes [51,52]. This extensive process confers mucins with their protective and lubricative properties. In the upper gastrointestinal (GI) tract, mucins form a protective barrier against the harsh, acidic environment of the stomach, preventing damage to the epithelial cells [40]. In the lower GI tract, particularly the colon, mucins create a physical barrier that separates the epithelial cells from the gut microbiota, protecting the host from bacterial invasion and antigenic stimulation [41,42].

2.1.2. SEA Domain

The extracellular domains of most transmembrane mucins, such as MUC1 and MUC16, include a sea urchin sperm protein, enterokinase, and agrin (SEA) domain [53]. This domain is located between the O-glycosylated tandem repeats and the transmembrane domain [54]. They undergo autoproteolytic cleavage in the endoplasmic reticulum [55], leading to the formation of an extracellular α-chain and a β-chain that remain noncovalently associated [56]. The SEA domain protects epithelial cells from mechanical stress by breaking before the apical membrane ruptures [56,57]. Additionally, it facilitates the release of the mucin extracellular domain, which is often detected in biological fluids such as serum and the lumen of the intestinal tract. This shedding process is frequently observed in metastatic carcinoma, inflammatory bowel disease, and cystic fibrosis, indicating a pathological relevance [58,59,60,61] (Table 1).

2.1.3. EGF-like Domains

Many transmembrane mucins contain epidermal growth factor (EGF)-like domains, which are characterized by six cysteine residues that form three disulfide bonds [67,68]. Through interaction with EGF receptors and the activation of receptor signaling pathways, EGF-like domains are crucial for mucosal repair and mucin expression regulation [69]. For example, MUC4, which lacks a SEA domain, contains three predicted EGF domains that facilitate repair and regeneration processes in the gastrointestinal and respiratory tracts [70]. The release of the extracellular domain may enable these EGF domains to interact with their ligands more effectively, potentially playing a role in distant signaling like cytokines.

2.1.4. Intracellular Domain

The intracellular domains, which are often referred to as the cytoplasmic tail of transmembrane mucins, are integral to cellular signaling pathways [20]. They contain PDZ-binding motifs that facilitate the trafficking and anchoring of receptor proteins, thereby organizing signaling complexes at cellular membranes [71,72]. In the gastrointestinal tract, the PDZ-binding motifs of MUC3, MUC12, and MUC17 compete with those of cystic fibrosis transmembrane conductance regulator (CFTR) chloride channels for adaptor proteins that regulate lysosomal degradation [73]. Furthermore, these cytoplasmic tails are subject to phosphorylation, mediating interactions with other proteins [20]. For instance, the phosphorylated cytoplasmic tail of MUC1 competes with E-cadherin for β-catenin binding, disrupting cell–cell adhesion and promoting cell detachment and growth [74]. Moreover, transmembrane mucins such as MUC1, MUC13, and MUC16 can localize to the nucleus, where their cytoplasmic tails influence transcription factors and regulatory proteins, contributing to cellular processes such as differentiation and proliferation [20] (Table 1).

2.1.5. Glycosylation

In humans, eight core mucin structures have been identified, with cores 1 to 4 being most prevalent in the intestinal tract [13,75]. These core structures can be elongated with various combinations of Gal-GlcNAc residues, forming either ß1-3 (type 1 chain) or ß1-4 (type 2 chain linkages [50,51]. This elongation process leads to the formation of a protective glycan coat that shields the mucin protein core from enzymatic degradation and maintains its hydration and viscosity [76] (Figure 1).
The O-glycosylation process, which occurs in the Golgi apparatus, begins with the addition of N-acetylgalactosamine (GalNAc) to the hydroxyl groups of Ser and Thr residues. This is followed by the sequential addition of other sugars, including galactose (Gal), N-acetylglucosamine (GlcNAc), fucose (Fuc), and sialic acid [1,77,78]. Proline residues help stabilize the mucin polypeptide in an extended conformation, facilitating O-glycosylation. This process results in the formation of diverse glycan structures, which protect the protein core from proteolytic degradation, enhance solubility and hydration, and contribute to the gel-forming properties of mucins [76,79,80] (Table 1).

2.1.6. Signaling Roles

In addition to the protective functions, transmembrane mucins also play critical roles in transducing signaling pathways [81]. With numerous potential sites of phosphorylation and through various interactions with RTKs, such as hepatocyte growth factor receptor (Met), epidermal growth factor receptor (EGFR), and platelet-derived growth factor receptor β (PDGFR β), the cytoplasmic tail of MUC1 executes a wide array of phosphorylation signaling patterns [82,83,84]. Though the precise function of these phosphorylation remains unknown, it has been discovered that MUC1 has the potential to translocate to the nucleus and regulate the expression of transcription factors, such as p53, Wnt/β-catenin, the signal transducer and activator of transcription (STAT), and nuclear factor (NF)- κB RelA pathways [85,86,87,88]. Through interaction with EGFR, ErbB2, and RTK at the cell membrane, MUC1 is involved in the activation of PI3K-AKT and MEK-ERK pathways [88,89,90,91] (Figure 2).
Though MUC4 lacks the SEA domain, it contains EGF-like domains with tyrosine and serine residues in its cytoplasmic tail that serve as potential sites of amino acid phosphorylation [92,93]. MUC4 was found to interact with ErbB family receptors and initiate signaling cascades involving RTK ErbB-2 (ErbB2/HER2) and ErbB3/HER3 [94,95], which leads to the cellular differentiation, proliferation, and inhibition of apoptosis. These MUC4-induced signaling pathways were found to protect tumor cells from HER2-targeted therapy in studies of cancer treatments [95,96]. In addition to HER2, MUC4 also interacts with HER3, which activates PI3K-ERK and focal adhesion kinase (FAK)-associated pathways, resulting in proliferation, metastasis, and angiogenesis in pancreatic cancer cells [94,97,98].
Similarly to MUC1, MUC16 is another transmembrane mucin with SEA domain, the largest core protein in size and longest mucin-type tandem repeats [99,100]. They also contain cytoplasmic tails that enable several potential phosphorylation sites [9]. MUC16 participates in signaling pathways at the cell surface and the nucleus via translocation [101]. It has been found that the cytoplasmic tail of MUC16 interacts with Janus kinase 2 (JAK2) and increases the activities of STAT3. These modulations induce cellular proliferation, the inhibition of apoptosis, and promote the metastasis of breast and pancreatic cancer cells [65,102]. MUC16 also interacts with Src and tyrosine protein kinases, which results in the phosphorylation of tyrosine in the MUC16 cytoplasmic tail, the shedding of the extracellular domain, and the dysregulation of β-catenin and E-cadherin [103,104].

2.1.7. Cleavage and Shedding

In MUC1, the transmembrane domain is proteolytically cleaved after synthesis, creating an α and β subunit that binds in a noncovalent fashion [105]. This cleavage occurs at the SEA domain, which is vital for the proper maturation and function of mucins [106]. The EGF-like domain of MUC4 is also proteolytically cleaved in a manner essential for its complex formation [70,92,107]. The shedding of mucin extracellular domains can occur via proteolytic cleavage near the plasma membrane by sheddases or metalloproteases such as TNF-α converting enzyme/A Disintegrin and Metalloprotease-like (TACE/ADAM17) and membrane-type 1 matrix metalloproteases (MT1-MMP) [108]. These cleavages of the SEA domain, which can be triggered by mechanical force, microbial interactions, alterations in pH, ionic concentration, hydration, or inflammatory stimuli, also require serine, threonine, or cysteine at the proteolytic site [106]. As a result, cleaved mucin extracellular domains function as decoy receptors for pathogens at the mucosal surface, preventing pathogen adhesion to epithelial cells (Table 1).

2.2. Role of Mucins in Normal Physiology

Biophysically, the heavily glycosylated mucins contribute to the viscoelastic properties of the mucus gel [9]. In general, mucins lubricate, moisturize, and protect the surface of mucus from pathogens, prevent the rupture of the epithelium barrier, and remove invaders [13]. The heavily glycosylated mucins are commonly found in the gastrointestinal tract, lungs, salivary glands, sweat glands, and breast [3]. Typically, the structural pattern of O-glycosylation determines mucin classification and function. For example, secretory mucins, such as MUC2, are cysteine-rich and secreted by goblet cells in the airway and intestine. These cysteine-rich regions allow the formation of a complex covalent structure that protects the intestinal epithelium [109]. On the other hand, membrane-bound mucins, such as MUC1 and MUC3, are synthesized by epithelial cells and integrated into the apical plasma membrane. The C-terminal trans-membrane anchor on membrane-bound mucins is important for cellular signaling, allowing the coordination of cellular responses, such as cell proliferation, differentiation, apoptosis, or product secretion [110].
The outer mucin layer provides a physical barrier against microorganisms and irritants while maintaining the local microenvironment. For example, the sulfate-rich mucins are also more acidic and contribute to the protective function against mucin-degrading bacteria [111]. Furthermore, MUC5AC and MUC6 in the stomach protect its lining from gastric acid [112,113] by holding the secreted bicarbonate ion in their mucin layers [114]. By increasing mucin sialylation, the duodenal mucin prevents the diffusion of gastric acid [115] and maintains the optimal alkaline environment in the intestine.
In addition to the formation of physical barriers, mucins also release active molecules to initiate immune defense and mediate processes of inflammation and repair [116,117,118]. The release of trefoil factors (TFFs) increases mucus viscosity [119], promotes wound healing, and reconstitutes damaged mucus epithelium [120,121]. Besides restorations, the TFFs also inhibit apoptosis, promote cellular motility, and promote differentiation [122]. By secreting cells with IgG Fc binding proteins and IgA, mucin also plays a role in immune surveillance along the GI tract [123,124].

3. Altered Mucin Expression in Cancer

There are characteristic differences between healthy and cancerous tissues, especially in the localization, glycosylation, electrophoresis, and transcriptional expression profiles of mucin in the apical surface [4]. The overexpression of mucin provides oncogenic signals that favor the initiation of tumor progression [125]. For example, MUC1 stabilizes β-catenin and promotes nuclear translocation to active Wnt genes that drive the proliferation, invasion, and metastasis of intrahepatic cholangiocarcinoma [126]. It has been speculated that altered MUC2 expression, in combination with genetic factors, may increase colorectal cancer (CRC) risk by creating a microenvironment suitable for tumor growth and metastasis [127]. The decrease in expression of MUC5AC mucin is also correlated with a poorer prognosis of gastric cancer [128]. Alterations in MUC4 and MUC1 are noted in bladder cancer, pancreatic ductal adenocarcinoma, and oral squamous cell carcinoma [129,130,131,132]. Mucin can also cause abnormalities in the mucin network and disturb signaling pathways, leading to changes in the biomechanical properties of pancreatic tumors [133]. For instance, MUC1 sterically disrupts the PAR3-PAR-6-aPKC complex, resulting in the loss of tight junction and cellular polarity. As a result, mucins interact with growth factors and perpetuate oncogenic signaling [27,74]. In this section, we will delve into the role of mucin in cancer development.

3.1. Upregulated Mucins in Cancer

Upon the examination of the cancer cell membrane, several mucins are overexpressed and secreted to the cell surface [4,46]. In addition to the protective barrier, some membrane-spanning mucins also serve as cell surface receptors that aid in the survival, differentiation, and metastasis of cancer cells [4,95,134,135,136].
Current studies have found a profound association of MUC1 overexpression in numerous cancer types. In the study of mucin expression in mouse models, MUC1 overexpression was associated with the metastasis of pancreatic adenocarcinoma to the lung, liver, and peritoneal organs [137], as well as increased multidrug resistance in pancreatic cancer cells [138]. In mammary gland tumors, the upregulation of MUC1 also leads to tumorigenesis via its interaction with EGFR and the downstream activation of MAP kinase (MAPK) [83]. By phosphorylating the cytoplasmic tail of MUC1 at the YEKV motif, EGFR facilitates the binding of MUC1 to β-catenin, which enhances tumorigenesis [139]. In intrahepatic cholangiocarcinoma (ICC), the upregulation of myeloid leukemia factor 1 (MLF1) is significantly correlated with the upregulation of MUC1 expression involving the EMT of cancer progression [140]. The increased cell migration and evasion results in MUC1-induced EMT through the increased expression of vimentin and β-catenin [141]. In ovarian cancer, the upregulation of MUC16 activates the PI3K/Akt signaling pathway and promotes the expression of mesenchymal markers, such as N-cadherin, vimentin, and Snail, leading to increased proliferation, invasion, and metastasis [142]. By inducing DNA methyltransferase (DNMT) 1 and 3b, MUC1 also contributes to the suppression of tumor-suppressor genes via promoter-specific DNA methylation [143]. A high level of VEGF expression was also found in tumors with increased MUC1 via the AKT pathway [144]. As MUC1 is overexpressed, the phosphorylated Ser/Thy at the cytoplasmic portion of MUC1 conducts signals through ß-catenin and MAPK pathways [145]. The overexpression of MUC4 and MUC5AC at mRNA and protein levels was also observed during the progression of pancreatic intraepithelial neoplasia (PanIN) [146,147].

3.2. Downregulated Mucins in Cancer

Acting as a tumor suppressor, the MUC2 knockout mice models showed early development of GI tumor and invasion of carcinoma, along with a reduced number of goblet cells, decreased apoptosis, and the increased migration of intestinal epithelial cells [148]. In salivary gland tumors, reduced MUC-4 expression has been associated with higher-graded tumors [64]. Studies on the immunohistochemistry of colon cancer also revealed that the low expression of MUC2 is associated with colon tumor and less favorable disease outcome, and lack of expression of MUC5AC and MUC6 were associated with worsened stages of CRC [149]. It is worth noting that there were conflicting results regarding the prognostic values of MUC5AC in gastric cancer. Some studies revealed that a lower MUC5AC expression results in better survival, but some revealed that decreased MUC5AC expression results in poor prognosis [150,151]. In a meta-analysis performed by Zhang et al. in 2015, they concluded that decreased MUC5AC expression has a significant correlation with lymph node metastasis, aggressive histopathological patterns, and poor survival in gastric cancer patients [128].

4. Mucins in Cancer Metastatic Cascade

To date, mucin expressions have been well documented in many cancer types, where they are known to have key roles in host cell growth, differentiation, mobility, immune response, and apoptotic escape [4,95,134,135,136]. The role of mucin in cancer metastasis has been characterized in MUC1 and MUC16. MUC1 modulates the transcription of cell adhesion molecules and signaling pathways, promoting cancer cell migration, invasion, and immune evasion [87]. The cell surface-associated glycopeptides and extracellular signals derived from the proteolytic processing of MUC1 on the cancer cells alter the cytotoxicity and migration of natural killer (NK) cells and T-cells by modulating NK group 2D (NKG2D) ligands and β-catenin, respectively [74,152,153]. The downregulation of NKG2D ligands on tumor cells reduces NK cell recognition and cytotoxicity in hepatocellular carcinoma [154]. In colorectal and pancreatic cancer, the recruitment of regulatory T cells suppresses cytotoxic CD8+ T cells and promotes immune tolerance, providing an opportunity for uncontrolled cell growth and migration [155,156]. The role of MUC16 in metastasis involves regulating the expression levels of E-cadherin and vimentin [103,104], inhibiting the release of apoptotic ligands from breast cancer cells [102], promoting the nuclear translocation of JAK2 in pancreatic cancer cells [65], and modulating immune evasion [157]. Hallmarks of cancer metastasis include the growth of neoplastic cells, angiogenesis, intravasation into circulation, extravasation, and colonization at a secondary site. These series of processes are termed “metastatic cascade” [158] (Figure 3). In this section, the role of mucin glycoproteins in the progression and metastasis of cancer and their functions, as well as their therapeutic implications, will be discussed in depth [159] (Figure 3).

4.1. Cancer Cell Invasion and Migration

MUC4, MUC1, and MUC16 are involved in the regional invasion and migration of cancer cells through various signaling pathways. In the study of pancreatic cancer, the inhibition of MUC4 expression resulted in reduced tumor metastasis by enhancing cell proliferation and reducing apoptosis [134,160]. MUC1-C, the C-terminal domain of MUC1, functions as an oncoprotein, promoting the upregulation of genes and proteins involved in signaling pathways that are dysregulated in advanced cancers. In studies of mammary tumors, MUC1 augments the dimerization of Her 2 and EGFR, which has been associated with invasive papillary mucinous neoplasm [161,162]. Additionally, MUC16’s interaction with JAK2, Src, and FAK was found to facilitate tumor growth and metastasis [102,103,163]. Lakshmanan et al. also identified that MUC16 enhanced the endothelial cell and p-selectin binding of pancreatic cancer cells to favor the metastatic spread of pancreatic ductal adenocarcinoma [164].

4.2. Cancer Cell Adhesion and Colonization

In the study of metastatic breast cancer, the under-glycosylated MUC1 expresses sialyl Lewis x and a, which slows extravascular rolling and enhances the adhesion to distant sites [165]. Galectin-3 (Gal-3), an endogenous lectin, helps stabilize MUC4 mRNA in the cytoplasm, which is highly expressed in pancreatic cancer cells [166]. In addition to MUC4, Gal-3 is also a ligand for MUC1 in epithelial cells, which induces MUC1 cell surface polarization, increased cancer cell aggregation, and adhesion to vascular endothelium [167,168,169].

4.3. Immune Evasion and Tumor Microenvironment

The phenomenon of immune modulation and evasion by cancer cells is fundamental to cancer development. Tumors exploit the immune cells’ ability to suppress responses through an adaptive process that is known as immunoediting [170]. By remaining dormant under immunosurveillance, coexisting with the immune system, bypassing checkpoints, and escaping from immune attacks, tumors began to proliferate and migrate in an unregulated fashion [170]. In the context of mucin, the expression of sialosyl-Tn (STn) antigen expressed by mucin significantly inhibits the cytotoxicity of natural killer (NK) and lymphocytic cells [171,172]. Additionally, MUC1 disrupts the interaction between tumor cell antigen and MHC-I receptors in innate immune cells. Besides MUC1, MUC16 also reduces T effector cell function by binding to toll-like receptors on dendritic cells [173].
The tumor microenvironment (TME) is composed of non-cancerous cellular and structural components, including cancer-associated fibroblasts (CAFs), adipocytes, vascular endothelial cells, and immune cells, along with the extracellular matrix (ECM) and extracellular vesicles these cells secrete. This three-dimensional scaffold not only supports cancer cell survival and proliferation but also plays a pivotal role in the transition from normal to malignant tissue [174,175]. Within the TME, mucins serve as critical ligands and signaling molecules for immune cells. They effectively modulate mechanisms of immune surveillance and immune escape. For example, MUC1 modulates TME through its interaction with lectin Siglec-9 on myeloid cells, inducing monocytes to secrete Il6, macrophage colony-stimulating factors (M-CSF), and plasminogen activator inhibitor (PAI-1) [58]. The altered expression of mucins in various cancers impacts their utility as potential biomarkers for early diagnosis and therapeutic monitoring [173,176]. These therapies include immune checkpoint inhibitors and chimeric antigen receptor (CAR) T-cell therapies, which are at the forefront of personalized cancer treatment strategies. The intricate nature of the TME can influence the effectiveness of therapeutic interventions, including the viability of surgical resection, which remains the primary curative treatment in many cases.

4.4. Mucins and Angiogenesis

By forming new blood vessels, angiogenesis provides a route for nutrient delivery, waste removal, and immune surveillance [177]. To support the growth and enhance the invasive potential of tumors, malignant cells overexpressed angiogenic factors, such as VEGF and their receptors (VEGFRs), to promote the neovascularization and remodeling of tumor blood vessels [178]. As many cancer cells thrive in a hypoxic environment, the increase in MUC1 expression helps induce the increase in VEGF, connective tissue growth factor (CTGF), and platelet-derived growth factor (PDGF-β) to support tumor angiogenesis [179]. More specifically, the overexpression of MUC1 in pancreatic and breast cancer cells interacts with signaling pathways, such as the MAPK, JAK/STAT, and the PI3K/AKT/mTOR pathways, and VEGF co-receptor, neuropilin-1, to promote the generation of endothelial cell and ectopic blood vessels [144,180]. Hypoxia-inducible factor-1 alpha (HIF-1α) is a transcription factor that is normally deactivated in an oxygen-rich environment but activated in an oxygen-poor environment and upregulated gene expression related to cancer progression, such as angiogenesis and metabolism reprogramming [181]. The upregulation of MUC1 expression is one of the downstream effects of HIF-1α activation, which is consistent with MUC-1-induced cancer cell migration and invasion [182].

5. Mucins in Different Types of Cancer

In normal physiological contexts, mucin’s long glycan chains create a water-rich antiadhesive mucus that protects tissues from dehydration, infections, and physical damage [37,46]. However, the deregulation of mucin glycosylation and altered mucin cellular localization are observed in numerous cancer types [129,130,131,132]. Cancer-associated mucins, especially those expressed at the cell surface, imped the cellular clearance of low-sialylated cellular proteins, deregulate the expression of immune activation markers, induce the secretion of VEGF, CTGF, metastatic markers, and inhibit cytotoxic lymphocytes [171,172,178,179]. The current chapter shows the importance of various mucins (MUC1, MUC3A, MUC4, MUC4β, MUC5AC, MUC5B, MUC7, and MUC16) and their mucin-bound carrier proteins in different cancers (Table 2).

5.1. Breast Cancer

In normal breast epithelial cells, MUC1 is expressed at the apical surface at low levels [183], in which their high molecular weight glycoproteins with complex O-glycans function as cytoprotective and lubricating agents [20]. In studies of breast cancer cells, the increased activity in sialyltransferase (STGal-I) was observed in the exposure of otherwise cryptic peptide epitopes expressed by MUC1 [184]. Moreover, the mucin expressed by breast cancer cell lines has a shorter sidechain that consists of Galβ1-3f, which is also associated with the addition of sialic acid by STGal-I that terminates the glycan chain extension [185]. Among the various N-acetylglucosaminyltransferases (GalNAcTs), GalNAcT6 has been identified to stabilize MUC1 [186], and GalNAcT14 is highly expressed in breast cancers [181,187]. The overexpression of ST3Gal-I also leads to the increased expression of sialyate core 1 in mucins and is associated with the tumorigenesis of breast cancer [188]. MUC1-ST, a tumor glycoform of MUC1, interacts with siglec-9 expressed by tumor-associated macrophages [189], which activates MEK-ERK and contributes to the poorer prognosis of breast cancer [58]. With the extensive involvement of MUC1 in breast cancer, recent studies have focused on the role of mucin in immunotherapy [190], which will be further discussed in a later chapter (Table 2).

5.2. Colorectal Cancer

In normal colon mucosa, mucins are equipped with Core 1-4 of O-glycans [191]. These 4 O-glycans can be synthesized and modified by different families of glycosyltransferases [192]. The increased expression of Tn and STn antigens correlate with the progression of colonic malignancy and are used as markers for poorly differentiated mucinous carcinomas [193,194]. More specifically, the expression of the sialyl Lewis x epitope in colon cancer is associated with poor survival [195]. Similarly to MUC1 discussed in breast cancer, MUC16 has also been identified to bind to Siglec-9 [196], attenuating T and NK cell functions to help the survival of cancer cells.
Aside from the alteration in the mucin glycosylation, the downregulation of MUC2 is also associated with metastasis and poor prognosis in colon cancer [197,198]. With abundant and variable O-glycan, MUC2 is a major mucin in the colon mucus that serves as an essential protective physical barrier against bacteria and inflammation [199]. The suppression of MUC2 expression is found to be associated with IL-6 overexpression, which leads to inflammation and tumor growth [200]. In addition to MUC2, the abnormal expression of MUC5AC is also observed in colorectal cancer [201]. In histological studies of the colorectal adenocarcinoma differentiation, the MUC2 level was found to decrease, and MUC5AC tends to increase when CRC progresses from moderately to poorly differentiated adenocarcinoma [202]. The overexpression of MUC5AC is associated with BRAF mutation [203] and the activation of the CD44/β-catenin/p53/p21 signaling pathway that contributes to tumorigenesis and chemoresistance [204]. The increased MUC5 and decreased MUC2 expression in CRC is also associated with the increased prevalence of lymph node metastasis and advanced tumor stages [201] (Table 2).

5.3. Pancreatic Cancer

Pancreatic cancer (PC) is characterized by a decline in 5-year survival rate and ranks as the seventh leading cause of death among all cancers in America [205]. In the search for biomarkers, current research has illustrated the significance of glycoproteins, such as CA19-9 and CA125, in PC diagnosis and prognosis markers [205]. Produced as an aberrant version of disialyl Lewis a sialic acid residual, CA19-9 has been studied extensively as a promising tumor marker for PC diagnosis and staging [206,207]. CA125 is another high molecular weight glycoprotein that is part of the MUC16 extracellular domain, of which studies of anti-MUC16 monoclonal antibodies have shown to reduce the actives of pancreatic tumors [208]. Though the mechanism remained unknown, a meta-analysis by Huang et al. has identified a significant association between high MUC4 expression and poor prognosis of metastatic pancreatic cancer [209]. In studies of PC in mice models, MUC4 knockout cells had less tumorigenicity and were more sensitive to gemcitabine treatment [63].
Though expressed in the apical membrane of the intralobular duct, glycoforms of MUC1 are undetectable in the main pancreatic duct [210]. However, in studies of pancreatic cancer, high MUC1 expression has been strongly associated with the invasiveness of pancreatic cancer [211,212]. The cytoplasmic tail of MUC1 is known to interact with β-catenin and EGFR to potentiate cell proliferation, motility, and chemoresistance in subtypes of PC via AKT and BCL-2 pathways [213,214]. Aside from cancer progression, MUC1 is also involved in chemoresistance in PC cells through the inhibition of BRCA1 to enhance glucose utilization [211] (Table 2).
Table 2. Summary of mucin deregulation in cancer.
Table 2. Summary of mucin deregulation in cancer.
Cancer Mucin and Mucin-Associated DeregulationProposed Mechanisms
BreastMUC1: GalNAcT14 [187], GALNT6 [186] MEK/ERK [58], MAPK, JAK/STAT, and PI3K/AKT/mTOR pathways [144,180], neuropilin-1 [187,215]
MUC16: Sialyl Lewis x and [165], and JAK2 [65,102]
Stabilization of MUC1 [186,187].
Increase tumorigenesis [58,187], inhibit apoptosis [65], angiogenesis [215].
Enhance cellular adhesion for distal metastasis [165].
Increase G2/M transition for cellular proliferation [65].
ColorectalMUC2: IL-6 overexpression [201]
MUC5AC: CD44/β-catenin/p53/p21 signaling pathway [201,202]
MUC16 [196]: Sialyl Lewis x epitope [195]
Inflammation and tumor growth [200]
Tumorigenesis and chemoresistance [204]
Invasion and metastasis [195], attenuation of NK and T cell [196]
PancreaticMUC1: β-catenin and EGFR, AKT and BCL-2, BRCA1, MAPK, JAK/STAT and the PI3K/AKT/mTOR pathways, and neuropilin-1 [210,213,214]
MUC4: integrin-mediated cell adhesion [134], HER2/neu [160],
MUC16: PI3K/AKT/mTOR pathways [180], Treg [216]
Enhance cell proliferation, motility, chemoresistance, glucose utilization, and angiogenesis. [210,213,214]
Inhibit integrin-mediated cell adhesion [134], increased cellular proliferation and metastasis [160].
Increase tumor survival [180], promote immunosuppressive tumor microenvironment [216]

6. Experimental Models and Techniques in Mucin Research

To investigate the role of mucin in cancer development, cell culture models are commonly used to study mucin expression, glycosylation, and secretion in cancer progression, metastasis and treatment resistance. Some commonly used adenocarcinoma cell lines include those coming from gastrointestinal, pancreatic, breast, and lung cancers [136,217,218,219]. For example, CAPAN-1 and CAPAN-2 cell lines, derived from pancreatic ductal adenocarcinoma, expressed high levels of MUC1 and MUC4, which were used in studies of chemotherapist-resistant pancreatic cancer [218]. In addition to traditional 2D cell models, recent studies have investigated the use of 3D multicellular spheroid cell models in cancer research [220]. With the ability to develop metabolic gradients in spheroid models, this technique enabled researchers to mimic in vitro environments under in vivo conditions, which could also resemble avascular stages of solid tumors [221,222,223]. Three-dimensional spheroids with a combination of siRNA transfections, lentiviral transductions, and quantitative PCR are valuable tools in gene functional and mechanistic assays. Taking this into account, spheroids can serve as a unique form of in vitro models to improve the relevance of in vitro research for human disease in an immunocompetent and controlled condition [39,114,117].

7. Mucins as Cancer Biomarkers

Through genetic, epigenetic, proteomic, glycomic, and microbial changes, the expression of mucin is altered in different disease stages [224,225,226]. By understanding the complex interplay of protein structure and mucin function in different pathophysiological conditions, such as colorectal cancer, we hope to elude markers that enable the early diagnosis of serious illness [125].

7.1. Current Mucin Biomarker

The search for more specific markers for the early detection of tumorigenesis has led to an interest in altered mucin protein glycosylation. The developments of novel detection methods for mutated O-glycan epitopes and the currently available commercial assays enable the assessment of mucin-based glyco-markers for cancer diagnosis, prognosis, and therapy monitoring [227]. For instance, MUC1, which is overexpressed in the colon, breast, lung, pancreatic, bladder, ovarian, bladder, gastric, and esophageal cancer [228,229,230,231,232,233,234], has been reported as a critical protein for the development of pluripotency, platform proteins secreted by tumor-associated surface proteases [58,235]. This evidence supports that MUC1 can be considered as an early diagnostic biomarker (lung adenocarcinoma [236], pancreatic [237,238], or ovarian [239] cancer) or as a prognostic biomarker (bladder carcinoma [240], node-negative breast [241], lung squamous cell [242], and esophageal cancer [243].
Numerous studies have shown that patterns of mucin expression may have a role in tracking pathologies or phenotyping when a detailed histological tumor definition is not pervasive. For example, recent evidence has suggested that mucin expression may be indispensable in cancer biology as a diagnostic marker for genitourinary squamous cell carcinomas and adenocarcinomas [240,244]. More specifically, endocervical and endometrial mucin markers demonstrate widely varying and potential diagnostic accuracy [245,246]. In fact, by adjusting immunophenotypes, all adenocarcinomas (i.e., carcinomas with mucus-producing epithelia) of the uteri are split into two groups: papillary endocervical and endometrial adenocarcinomas (EMCA) [247]. Regarding ovarian carcinoma and cysts, MSA (UMA1) and MUC16 (CA125) can present equivocal test scenarios [248,249]. Tests are often negative in invasive serous carcinomas for ovarian cysts, endometrioid cysts, and borderline lesions [250]. The usual differences in disease biology are confusing players in cancer diagnostics and cancer cell propagation.
The two carbohydrate antigens (CAs), MUC5AC and carcinoembryonic antigen (CEA), are more frequently detected in esophageal than in gastric cancers, according to both immunohistochemical and biochemical data [251]. In most cases, gastric MUC5AC is reduced or absent [128], while CEA usually demonstrates a higher positivity in esophageal cancer than in gastric cancer [252,253]. The prognostic significance of these mucins is also partially different in the two tumor types, with MUC5AC being more helpful in gastric [128] and CEA in esophageal cancers [254].

7.2. Limitation of Mucin as Biomarker

Though the altered expression of mucin is detected in various cancer cells, there are several limitations of mucin as biomarkers for cancer diagnosis. Mucin expression varies across cancer types. The increased expression of MUC5AC in pancreatic and lung cancer is correlated with poor prognosis [255,256,257], while in gastric carcinoma, decreased MUC5AC expression correlates with poor prognosis [258]. The variability in the O-glycosylation profile in mucin post-translational modification and the lack of tissue specificity also pose challenges in utilizing mucin proteins as cancer biomarkers [259]. The lack of sensitivity in early cancer detection is another notable limitation. For example, while CA125, a MUC16 marker, has been widely used as a serum biomarker for ovarian cancer, the limited sensitivity makes it difficult to screen early-stage ovarian cancer [260,261]. While the detection of mucin protein backbone or O-glycans may yield low sensitivity and specificity, some studies have combined mucin and its O-glycan feature to increase specificity as cancer biomarkers [259]. Though mucin shows promise as a cancer biomarker, the variable expression across cancer types, limited specificity, and sensitivity make it challenging to definite diagnostic tools.

8. Therapeutic Targeting of Mucins in Cancer

The aberrant glycosylation of mucin, including increased N-glycan branching, sialic acid glycans, and shortened O-glycans, is the core signature of mucin-associated cancer cells [262]. Though the exact mechanisms are under investigation, this abnormality has been associated with proliferation [263], the loss of adhesion [264], and the evasion of immune surveillance [62] in cancer cells. With its unique patterns and cancer-associated epitopes, MUC1 has been studied as a potential therapeutic target for mucin-associated cancer cells [265].

8.1. Current Strategies and Challenges

Widely expressed in many cancer types, MUC1 is heavily studied in cancer biology research. With the interaction between the MUC1 C-terminal subunit (MUC1-C) and receptor tyrosine kinases at the cell membrane, the MUC1-C inhibitor has been studied as a potential therapeutic intervention for breast cancer [88]. Recent studies have examined the use of monoclonal antibodies (mAbs) and anti-mucin vaccines in treating cancers with overexpressed mucin proteins. For example, AS1402, an IgG mAbs that binds to aberrant extracellular MUC1 peptide that is not normally exposed in normal breast, has demonstrated its efficacy in Phase I and II clinical trials [266,267]. In treating pancreatic cancer, mAb and TAB004, which target tumors associated with MUC1 (tMUC1), have shown a reduction in colony formation in the PDAC cell line [268]. In addition to mAbs, vaccines against cancer have also been developed. Examples of MUC1-based cancer vaccines include subunit vaccines, DNA vaccines, viral vector vaccines, dendritic cell vaccines, and glycoprotein vaccines [269]. Julien et al. also observed a significant delay in tumor growth when treating tumor cells with a coupled sialyl-Tn vaccine [270]. Preclinical studies on adjuvanted MUC1 subunit vaccines have shown their antitumor activities by inducing Th1 and NK cell response to suppress tumor cells in mice models [271,272,273]. Several subunit vaccines, such as L-BLP25, have undergone trials and shown improved survival outcomes in patients with non-small cell lung cancer and prostate cancer [274,275].
Current challenges in mucin-target cancer therapy include the immunosuppressive tumor microenvironment, the negative charge of the mucin barrier, and non-specific drug distribution systems. The immunosuppressive microenvironment has posed a challenge in the efficient delivery of MUC-1-based vaccines, which activate immune cells and exert cytotoxic pathways [276]. The negatively charged mucin also creates a diffusion barrier for positively charged drugs, leading to chemoresistance in cancer cells [277,278,279]. By disrupting mucin synthesis through the glucosaminyl transferase 3 inhibitor (GCNT3), it is hoped to improve drug delivery and delay PanIN progression [279]. However, its therapeutic potential requires further investigation. To overcome challenges in tumor penetration of antitumor mAbs, radiopharmaceuticals act as carriers to improve the stability and immune reactivity of antibody treatments [280,281]. Other shortcomings of cancer therapy include high toxicity and systemic side effects due to non-specific drug distribution. To overcome these challenges, recent studies have shown an improvement in the sensitivity of chemotherapy drugs by coupling mAbs with nanoparticles and chemokine receptor CXR4 antagonists [282].

8.2. Novel Approaches and Future Directions

With protein glycosylation playing a role in malignant transformation and immune tolerance, current studies have explored the therapeutic potential of chimeric antigen receptor T cells (CAR-T) in solid tumors [283,284,285]. Posey et al. demonstrated target-specific cytotoxicity activity of anit-Tn-MUC1 CAR-T cells in pancreatic xenograft models [283]. By transducing CAR-T cells with TAB004, Zhou et al. demonstrated a novel therapeutic approach to target tMUC1 in breast cancer by inducing the production of cytokines and chemokines to suppress tMUC1-positive tumors without disrupting normal breast epithelial cells [286]. The Tn-MUC1 targeted CAR-T cells also showed effectiveness in targeting and eliminating Tn-MUC1-positive intrahepatic cholangiocarcinoma in both in vivo and in vitro models [287]. Similarly to the mAbs and vaccine therapy mentioned above, the tumor-associated macrophages (TAM) in the tumor microenvironment inhibit the efficacy of CAR-T cells [288]. To target the TAMs and increase phagocytosis of tumor cells, CAR-macrophages (CAR-M) have been studied to enhance tumor antigen presentation and T-cell activation [289,290]. By adjuvating CAR-T and CAR-M therapy, it is hoped to improve the efficiency of cancer immunotherapy. Though in vitro models have shown promising results in the novel antitumor activities in targeting mucin-associated cancer cells, further investigation and clinical trials are warranted to evaluate the safety and efficacy of cancer treatments.

9. Conclusions and Future Perspectives

The complexity and versatility of mucin function and signaling pathways are essential for biological survival. Extensive glycosylation and glycopeptides mucins serve an essential role as a protective barrier and cellular signaling. Disruption in mucin expression, such as the upregulation, downregulation, and exposure of normally hidden mucin antigen on cell surfaces, leads to the malignant transformation of epithelial cells in breast, pancreatic, and colon cancer. While MUC1, MUC4, MUC5, and MUC16 are all involved in cancer progression, MUC1 has been extensively studied in its ability to modulate the innate immune system, develop tumor microenvironment, and promote tumor angiogenesis that contributes to the proliferation, invasion, and metastasis of cancer cells. While most mucins are upregulated in their association with cancer, the downregulation of mucin, such as MUC2, expression is also involved in pro-inflammatory behavior that favors cancer progression.
With mucin’s extensive involvement in cancer formation, many studies have investigated mucin’s potential to serve as a biomarker. Comparing mucin’s pattern of expression, immunophenotypes, histological presentation, and serum antigen levels are potential diagnostic and disease monitoring tools for clinicians. However, the variability in mucin glycosylation and lack of specificity in mucin expression pose challenges in early cancer detection. Despite current challenges in searching for highly sensitive and specific diagnostic mucin markers, recent discoveries and investigations into mucin-targeted immunotherapy through autoimmune antibodies and vaccines have yielded new strategies to improve diagnostics and therapeutics in mucin-associated cancers.
The involvement of mucin in cancer is extremely intricate, especially with its diverse glycosylation and cleavage patterns and involvement with numerous signaling pathways. The irregular dysregulation of mucin and the overlapping expression of mucin across different cancer types have made it challenging to characterize their diagnostic and therapeutic potential. Yet, recent discoveries in mucin’s role as biomarkers and therapeutic targets inspire further innovative approaches to cancer research. To encompass our current understanding of mucins, our study provided a comprehensive review of mucins and their association with cancer. Through a thorough discussion of the biochemical structure of mucin, we highlighted its significance in cellular communication within physiological, pathological, and oncogenic pathways. This review emphasized the role of mucin, especially MUC1, in modulating the tumor microenvironment and the immune system in favor of cancer proliferation. By discussing novel therapeutic approaches that target mucin-associated cancer and illustrating the current barriers in diagnostic and therapeutic modalities, we provide novel insights into the possible future directions of mucin cancer research.

Funding

Internal funding for this research was provided by the Department of Internal Medicine University of Illinois Collage of Medicine Peoria, Peoria, IL, USA.

Acknowledgments

The figures were made using Biorender (www.biorender.com, accessed on 18 July 2024).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Corfield, A.P. Mucins: A biologically relevant glycan barrier in mucosal protection. Biochim. Biophys. Acta 2015, 1850, 236–252. [Google Scholar] [CrossRef] [PubMed]
  2. Webber, S.E. Receptors mediating the effects of substance P and neurokinin A on mucus secretion and smooth muscle tone of the ferret trachea: Potentiation by an enkephalinase inhibitor. Br. J. Pharmacol. 1989, 98, 1197–1206. [Google Scholar] [CrossRef]
  3. Bansil, R.; Stanley, E.; LaMont, J.T. Mucin biophysics. Annu. Rev. Physiol. 1995, 57, 635–657. [Google Scholar] [CrossRef] [PubMed]
  4. Kufe, D.W. Mucins in cancer: Function, prognosis and therapy. Nat. Rev. Cancer 2009, 9, 874–885. [Google Scholar] [CrossRef]
  5. Pai, P.; Rachagani, S.; Dhawan, P.; Batra, S.K. Mucins and Wnt/β-catenin signaling in gastrointestinal cancers: An unholy nexus. Carcinogenesis 2016, 37, 223–232. [Google Scholar] [CrossRef] [PubMed]
  6. Pothuraju, R.; Krishn, S.R.; Gautam, S.K.; Pai, P.; Ganguly, K.; Chaudhary, S.; Rachagani, S.; Kaur, S.; Batra, S.K. Mechanistic and Functional Shades of Mucins and Associated Glycans in Colon Cancer. Cancers 2020, 12, 649. [Google Scholar] [CrossRef]
  7. Cox, K.E.; Liu, S.; Lwin, T.M.; Hoffman, R.M.; Batra, S.K.; Bouvet, M. The Mucin Family of Proteins: Candidates as Potential Biomarkers for Colon Cancer. Cancers 2023, 15, 1491. [Google Scholar] [CrossRef]
  8. Strous, G.J.; Dekker, J. Mucin-type glycoproteins. Crit. Rev. Biochem. Mol. Biol. 1992, 27, 57–92. [Google Scholar] [CrossRef]
  9. Hattrup, C.L.; Gendler, S.J. Structure and function of the cell surface (tethered) mucins. Annu. Rev. Physiol. 2008, 70, 431–457. [Google Scholar] [CrossRef]
  10. Demouveaux, B.; Gouyer, V.; Gottrand, F.; Narita, T.; Desseyn, J. Gel-forming mucin interactome drives mucus viscoelasticity. Adv. Colloid Interface Sci. 2018, 252, 69–82. [Google Scholar] [CrossRef]
  11. Desseyn, J.L.; Aubert, J.P.; Porchet, N.; Laine, A. Evolution of the large secreted gel-forming mucins. Mol. Biol. Evol. 2000, 17, 1175–1184. [Google Scholar] [CrossRef] [PubMed]
  12. Javitt, G.; Khmelnitsky, L.; Albert, L.; Bigman, L.S.; Elad, N.; Morgenstern, D.; Ilani, T.; Levy, Y.; Diskin, R.; Fass, D. Assembly Mechanism of Mucin and von Willebrand Factor Polymers. Cell 2020, 183, 717–729.e16. [Google Scholar] [CrossRef] [PubMed]
  13. Hansson, G.C. Mucins and the Microbiome. Annu. Rev. Biochem. 2020, 89, 769–793. [Google Scholar] [CrossRef] [PubMed]
  14. Bobek, L.A.; Liu, J.; Sait, S.N.; Shows, T.B.; Bobek, Y.A.; Levine, M.J. Structure and chromosomal localization of the human salivary mucin gene, MUC7. Genomics 1996, 31, 277–282. [Google Scholar] [CrossRef]
  15. Higuchi, T.; Orita, T.; Nakanishi, S.; Katsuya, K.; Watanabe, H.; Yamasaki, Y.; Waga, I.; Nanayama, T.; Yamamoto, Y.; Munger, W.; et al. Molecular cloning, genomic structure, and expression analysis of MUC20, a novel mucin protein, up-regulated in injured kidney. J. Biol. Chem. 2004, 279, 1968–1979. [Google Scholar] [CrossRef]
  16. Johansson, M.E.V.; Hansson, G.C. Immunological aspects of intestinal mucus and mucins. Nat. Rev. Immunol. 2016, 16, 639–649. [Google Scholar] [CrossRef]
  17. Fekete, E.; Buret, A.G. The role of mucin O-glycans in microbiota dysbiosis, intestinal homeostasis, and host-pathogen interactions. Am. J. Physiol. Gastrointest. Liver Physiol. 2023, 324, G452–G465. [Google Scholar] [CrossRef]
  18. McKenzie, C.; Thim, L.; Parsons, M.E. Topical and intravenous administration of trefoil factors protect the gastric mucosa from ethanol-induced injury in the rat. Aliment. Pharmacol. Ther. 2000, 14, 1033–1040. [Google Scholar] [CrossRef]
  19. Bruce, J.N.; Oldfield, E.H. Method for sequential sampling of cerebrospinal fluid in humans. Neurosurgery 1988, 23, 788–790. [Google Scholar] [CrossRef]
  20. van Putten, J.P.M.; Strijbis, K. Transmembrane Mucins: Signaling Receptors at the Intersection of Inflammation and Cancer. J. Innate Immun. 2017, 9, 281–299. [Google Scholar] [CrossRef]
  21. Du, J.; Zhou, S.; Cheng, Z.; Xu, W.; Zhang, R.; Wang, L.; Guo, J. MUC1 Specific Immune Responses Enhanced by Coadministration of Liposomal DDA/MPLA and Lipoglycopeptide. Front. Chem. 2022, 10, 814880. [Google Scholar] [CrossRef] [PubMed]
  22. Powell, D.W. Barrier function of epithelia. Am. J. Physiol. 1981, 241, 275. [Google Scholar] [CrossRef]
  23. Cavey, M.; Lecuit, T. Molecular bases of cell-cell junctions stability and dynamics. Cold Spring Harb. Perspect. Biol. 2009, 1, a002998. [Google Scholar] [CrossRef] [PubMed]
  24. Tepass, U.; Tanentzapf, G.; Ward, R.; Fehon, R. Epithelial cell polarity and cell junctions in Drosophila. Annu. Rev. Genet. 2001, 35, 747–784. [Google Scholar] [CrossRef]
  25. Vermeer, P.D.; Einwalter, L.A.; Moninger, T.O.; Rokhlina, T.; Kern, J.A.; Zabner, J.; Welsh, M.J. Segregation of receptor and ligand regulates activation of epithelial growth factor receptor. Nature 2003, 422, 322–326. [Google Scholar] [CrossRef] [PubMed]
  26. Polyak, K.; Weinberg, R.A. Transitions between epithelial and mesenchymal states: Acquisition of malignant and stem cell traits. Nat. Rev. Cancer 2009, 9, 265–273. [Google Scholar] [CrossRef]
  27. Ganguly, K.; Rauth, S.; Marimuthu, S.; Kumar, S.; Batra, S.K. Unraveling mucin domains in cancer and metastasis: When protectors become predators. Cancer Metastasis Rev. 2020, 39, 647–659. [Google Scholar] [CrossRef]
  28. Thiery, J.P. Epithelial-mesenchymal transitions in tumour progression. Nat. Rev. Cancer 2002, 2, 442–454. [Google Scholar] [CrossRef]
  29. Shin, K.; Fogg, V.C.; Margolis, B. Tight junctions and cell polarity. Annu. Rev. Cell Dev. Biol. 2006, 22, 207–235. [Google Scholar] [CrossRef]
  30. Gumbiner, B.M. Regulation of cadherin-mediated adhesion in morphogenesis. Nat. Rev. Mol. Cell Biol. 2005, 6, 622–634. [Google Scholar] [CrossRef]
  31. Yamamoto, M.; Bharti, A.; Li, Y.; Kufe, D. Interaction of the DF3/MUC1 breast carcinoma-associated antigen and beta-catenin in cell adhesion. J. Biol. Chem. 1997, 272, 12492–12494. [Google Scholar] [CrossRef] [PubMed]
  32. Li, Y.; Yu, W.; Ren, J.; Chen, W.; Huang, L.; Kharbanda, S.; Loda, M.; Kufe, D. Heregulin targets gamma-catenin to the nucleolus by a mechanism dependent on the DF3/MUC1 oncoprotein. Mol. Cancer Res. 2003, 1, 765–775. [Google Scholar]
  33. Hassan, C.; Zullo, A.; Risio, M.; Rossini, F.P.; Morini, S. Histologic risk factors and clinical outcome in colorectal malignant polyp: A pooled-data analysis. Dis. Colon Rectum 2005, 48, 1588–1596. [Google Scholar] [CrossRef]
  34. Deschner, E.E. Cell turnover and colon tumor development. Prev. Med. 1987, 16, 580–585. [Google Scholar] [CrossRef]
  35. Cappell, M.S. Pathophysiology, clinical presentation, and management of colon cancer. Gastroenterol. Clin. N. Am. 2008, 37, 1–24. [Google Scholar] [CrossRef]
  36. Gerken, T.A.; Butenhof, K.J.; Shogren, R. Effects of glycosylation on the conformation and dynamics of O-linked glycoproteins: Carbon-13 NMR studies of ovine submaxillary mucin. Biochemistry 1989, 28, 5536–5543. [Google Scholar] [CrossRef]
  37. Shogren, R.; Gerken, T.A.; Jentoft, N. Role of glycosylation on the conformation and chain dimensions of O-linked glycoproteins: Light-scattering studies of ovine submaxillary mucin. Biochemistry 1989, 28, 5525–5536. [Google Scholar] [CrossRef] [PubMed]
  38. Gum, J.R.J.; Hicks, J.W.; Toribara, N.W.; Rothe, E.M.; Lagace, R.E.; Kim, Y.S. The human MUC2 intestinal mucin has cysteine-rich subdomains located both upstream and downstream of its central repetitive region. J. Biol. Chem. 1992, 267, 21375–21383. [Google Scholar] [CrossRef] [PubMed]
  39. Meezaman, D.; Charles, P.; Daskal, E.; Polymeropoulos, M.H.; Martin, B.M.; Rose, M.C. Cloning and analysis of cDNA encoding a major airway glycoprotein, human tracheobronchial mucin (MUC5). J. Biol. Chem. 1994, 269, 12932–12939. [Google Scholar] [CrossRef]
  40. Jin, C.; Kenny, D.T.; Skoog, E.C.; Padra, M.; Adamczyk, B.; Vitizeva, V.; Thorell, A.; Venkatakrishnan, V.; Lindén, S.K.; Karlsson, N.G. Structural Diversity of Human Gastric Mucin Glycans. Mol. Cell Proteom. 2017, 16, 743–758. [Google Scholar] [CrossRef]
  41. Linden, S.K.; Sutton, P.; Karlsson, N.G.; Korolik, V.; McGuckin, M.A. Mucins in the mucosal barrier to infection. Mucosal Immunol. 2008, 1, 183–197. [Google Scholar] [CrossRef] [PubMed]
  42. Lu, J.; Teh, C.; Kishore, U.; Reid, K.B.M. Collectins and ficolins: Sugar pattern recognition molecules of the mammalian innate immune system. Biochim. Biophys. Acta 2002, 1572, 387–400. [Google Scholar] [CrossRef] [PubMed]
  43. Thornton, D.J.; Rousseau, K.; McGuckin, M.A. Structure and function of the polymeric mucins in airways mucus. Annu. Rev. Physiol. 2008, 70, 459–486. [Google Scholar] [CrossRef]
  44. Fernández-Blanco, J.A.; Fakih, D.; Arike, L.; Rodríguez-Piñeiro, A.M.; Martínez-Abad, B.; Skansebo, E.; Jackson, S.; Root, J.; Singh, D.; McCrae, C.; et al. Attached stratified mucus separates bacteria from the epithelial cells in COPD lungs. J. Clin. Investig. 2018, 3, e120994. [Google Scholar] [CrossRef]
  45. Ma, J.; Rubin, B.K.; Voynow, J.A. Mucins, Mucus, and Goblet Cells. Chest 2018, 154, 169–176. [Google Scholar] [CrossRef]
  46. Joshi, S.; Kumar, S.; Bafna, S.; Rachagani, S.; Wagner, K.; Jain, M.; Batra, S.K. Genetically engineered mucin mouse models for inflammation and cancer. Cancer Metastasis Rev. 2015, 34, 593–609. [Google Scholar] [CrossRef]
  47. Zhang, Y.; Wang, L.; Ocansey, D.K.W.; Wang, B.; Wang, L.; Xu, Z. Mucin-Type O-Glycans: Barrier, Microbiota, and Immune Anchors in Inflammatory Bowel Disease. J. Inflamm. Res. 2021, 14, 5939–5953. [Google Scholar] [CrossRef]
  48. Schwerbrock, N.M.J.; Makkink, M.K.; van der Sluis, M.; Büller, H.A.; Einerhand, A.W.C.; Sartor, R.B.; Dekker, J. Interleukin 10-deficient mice exhibit defective colonic Muc2 synthesis before and after induction of colitis by commensal bacteria. Inflamm. Bowel Dis. 2004, 10, 811–823. [Google Scholar] [CrossRef] [PubMed]
  49. Johansson, M.E.V.; Ambort, D.; Pelaseyed, T.; Schütte, A.; Gustafsson, J.K.; Ermund, A.; Subramani, D.B.; Holmén-Larsson, J.M.; Thomsson, K.A.; Bergström, J.H.; et al. Composition and functional role of the mucus layers in the intestine. Cell Mol. Life Sci. 2011, 68, 3635–3641. [Google Scholar] [CrossRef]
  50. Bergstrom, K.; Fu, J.; Johansson, M.E.V.; Liu, X.; Gao, N.; Wu, Q.; Song, J.; McDaniel, J.; McGee, S.; Chen, W.; et al. Core 1- and 3-derived O-glycans collectively maintain the colonic mucus barrier and protect against spontaneous colitis in mice. Mucosal Immunol. 2017, 10, 91–103. [Google Scholar] [CrossRef]
  51. Yamaguchi, M.; Yamamoto, K. Mucin glycans and their degradation by gut microbiota. Glycoconj. J. 2023, 40, 493–512. [Google Scholar] [CrossRef] [PubMed]
  52. Schauer, R. Sialic acids as regulators of molecular and cellular interactions. Curr. Opin. Struct. Biol. 2009, 19, 507–514. [Google Scholar] [CrossRef] [PubMed]
  53. Palmai-Pallag, T.; Khodabukus, N.; Kinarsky, L.; Leir, S.; Sherman, S.; Hollingsworth, M.A.; Harris, A. The role of the SEA (sea urchin sperm protein, enterokinase and agrin) module in cleavage of membrane-tethered mucins. FEBS J. 2005, 272, 2901–2911. [Google Scholar] [CrossRef] [PubMed]
  54. Bork, P.; Patthy, L. The SEA module: A new extracellular domain associated with O-glycosylation. Protein Sci. 1995, 4, 1421–1425. [Google Scholar] [CrossRef] [PubMed]
  55. Wreschner, D.H.; McGuckin, M.A.; Williams, S.J.; Baruch, A.; Yoeli, M.; Ziv, R.; Okun, L.; Zaretsky, J.; Smorodinsky, N.; Keydar, I.; et al. Generation of ligand-receptor alliances by “SEA” module-mediated cleavage of membrane-associated mucin proteins. Protein Sci. 2002, 11, 698–706. [Google Scholar] [CrossRef]
  56. Macao, B.; Johansson, D.G.A.; Hansson, G.C.; Härd, T. Autoproteolysis coupled to protein folding in the SEA domain of the membrane-bound MUC1 mucin. Nat. Struct. Mol. Biol. 2006, 13, 71–76. [Google Scholar] [CrossRef]
  57. Pelaseyed, T.; Zäch, M.; Petersson, A.C.; Svensson, F.; Johansson, D.G.A.; Hansson, G.C. Unfolding dynamics of the mucin SEA domain probed by force spectroscopy suggest that it acts as a cell-protective device. FEBS J. 2013, 280, 1491–1501. [Google Scholar] [CrossRef]
  58. Beatson, R.; Tajadura-Ortega, V.; Achkova, D.; Picco, G.; Tsourouktsoglou, T.-D.; Klausing, S.; Hillier, M.; Maher, D.A.J.; Noll, S.K.T.; Crocker, P.R.; et al. The mucin MUC1 modulates the tumor immunological microenvironment through engagement of the lectin Siglec-9. Nat. Immunol. 2016, 17, 1273–1281. [Google Scholar] [CrossRef]
  59. Yin, B.W.T.; Dnistrian, A.; Lloyd, K.O. Ovarian cancer antigen CA125 is encoded by the MUC16 mucin gene. Int. J. Cancer 2002, 98, 737–740. [Google Scholar] [CrossRef]
  60. Morrison, C.B.; Markovetz, M.R.; Ehre, C. Mucus, mucins, and cystic fibrosis. Pediatr. Pulmonol. 2019, 54 (Suppl. S3), S84–S96. [Google Scholar] [CrossRef]
  61. Kudelka, M.R.; Stowell, S.R.; Cummings, R.D.; Neish, A.S. Intestinal epithelial glycosylation in homeostasis and gut microbiota interactions in IBD. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 597–617. [Google Scholar] [CrossRef] [PubMed]
  62. Madsen, C.B.; Lavrsen, K.; Steentoft, C.; Vester-Christensen, M.B.; Clausen, H.; Wandall, H.H.; Pedersen, A.E. Glycan elongation beyond the mucin associated Tn antigen protects tumor cells from immune-mediated killing. PLoS ONE 2013, 8, e72413. [Google Scholar] [CrossRef]
  63. Sagar, S.; Leiphrakpam, P.D.; Thomas, D.; McAndrews, K.L.; Caffrey, T.C.; Swanson, B.J.; Clausen, H.; Wandall, H.H.; Hollingsworth, M.A.; Radhakrishnan, P. MUC4 enhances gemcitabine resistance and malignant behaviour in pancreatic cancer cells expressing cancer-associated short O-glycans. Cancer Lett. 2021, 503, 91–102. [Google Scholar] [CrossRef] [PubMed]
  64. Alos, L.; Lujan, B.; Castillo, M.; Nadal, A.; Carreras, M.; Caballero, M.; de Bolos, C.; Cardesa, A. Expression of membrane-bound mucins (MUC1 and MUC4) and secreted mucins (MUC2, MUC5AC, MUC5B, MUC6 and MUC7) in mucoepidermoid carcinomas of salivary glands. Am. J. Surg. Pathol. 2005, 29, 806–813. [Google Scholar] [CrossRef]
  65. Das, S.; Rachagani, S.; Torres-Gonzalez, M.P.; Lakshmanan, I.; Majhi, P.D.; Smith, L.M.; Wagner, K.; Batra, S.K. Carboxyl-terminal domain of MUC16 imparts tumorigenic and metastatic functions through nuclear translocation of JAK2 to pancreatic cancer cells. Oncotarget 2015, 6, 5772–5787. [Google Scholar] [CrossRef] [PubMed]
  66. Carraway, K.L.; Ramsauer, V.P.; Haq, B.; Carothers Carraway, C.A. Cell signaling through membrane mucins. Bioessays 2003, 25, 66–71. [Google Scholar] [CrossRef]
  67. Savage, C.R.J.; Hash, J.H.; Cohen, S. Epidermal growth factor. Location of disulfide bonds. J. Biol. Chem. 1973, 248, 7669–7672. [Google Scholar] [CrossRef]
  68. Winkler, M.E.; Bringman, T.; Marks, B.J. The purification of fully active recombinant transforming growth factor alpha produced in Escherichia coli. J. Biol. Chem. 1986, 261, 13838–13843. [Google Scholar] [CrossRef]
  69. Nadel, J.A. Role of epidermal growth factor receptor activation in regulating mucin synthesis. Respir. Res. 2001, 2, 85–89. [Google Scholar] [CrossRef]
  70. Moniaux, N.; Nollet, S.; Porchet, N.; Degand, P.; Laine, A.; Aubert, J.P. Complete sequence of the human mucin MUC4: A putative cell membrane-associated mucin. Biochem. J. 1999, 338 Pt 2, 325–333. [Google Scholar] [CrossRef]
  71. Malmberg, E.K.; Pelaseyed, T.; Petersson, A.C.; Seidler, U.E.; De Jonge, H.; Riordan, J.R.; Hansson, G.C. The C-terminus of the transmembrane mucin MUC17 binds to the scaffold protein PDZK1 that stably localizes it to the enterocyte apical membrane in the small intestine. Biochem. J. 2008, 410, 283–289. [Google Scholar] [CrossRef] [PubMed]
  72. Lamprecht, G.; Seidler, U. The emerging role of PDZ adapter proteins for regulation of intestinal ion transport. Am. J. Physiol. Gastrointest. Liver Physiol. 2006, 291, 766. [Google Scholar] [CrossRef]
  73. Pelaseyed, T.; Hansson, G.C. CFTR anion channel modulates expression of human transmembrane mucin MUC3 through the PDZ protein GOPC. J. Cell Sci. 2011, 124, 3074–3083. [Google Scholar] [CrossRef]
  74. Huang, L.; Chen, D.; Liu, D.; Yin, L.; Kharbanda, S.; Kufe, D. MUC1 oncoprotein blocks glycogen synthase kinase 3beta-mediated phosphorylation and degradation of beta-catenin. Cancer Res. 2005, 65, 10413–10422. [Google Scholar] [CrossRef]
  75. Tailford, L.E.; Crost, E.H.; Kavanaugh, D.; Juge, N. Mucin glycan foraging in the human gut microbiome. Front. Genet. 2015, 6, 81. [Google Scholar] [CrossRef]
  76. Kato, K.; Jeanneau, C.; Tarp, M.A.; Benet-Pagès, A.; Lorenz-Depiereux, B.; Bennett, E.P.; Mandel, U.; Strom, T.M.; Clausen, H. Polypeptide GalNAc-transferase T3 and familial tumoral calcinosis. Secretion of fibroblast growth factor 23 requires O-glycosylation. J. Biol. Chem. 2006, 281, 18370–18377. [Google Scholar] [CrossRef]
  77. Ringot-Destrez, B.; Kalach, N.; Mihalache, A.; Gosset, P.; Michalski, J.; Léonard, R.; Robbe-Masselot, C. How do they stick together? Bacterial adhesins implicated in the binding of bacteria to the human gastrointestinal mucins. Biochem. Soc. Trans. 2017, 45, 389–399. [Google Scholar] [CrossRef] [PubMed]
  78. Thompson, N.; Wakarchuk, W. O-glycosylation and its role in therapeutic proteins. Biosci. Rep. 2022, 42, BSR20220094. [Google Scholar] [CrossRef] [PubMed]
  79. Znamenskaya, Y.; Sotres, J.; Gavryushov, S.; Engblom, J.; Arnebrant, T.; Kocherbitov, V. Water sorption and glass transition of pig gastric mucin studied by QCM-D. J. Phys. Chem. B 2013, 117, 2554–2563. [Google Scholar] [CrossRef]
  80. Svensson, O.; Lindh, L.; Cárdenas, M.; Arnebrant, T. Layer-by-layer assembly of mucin and chitosan—Influence of surface properties, concentration and type of mucin. J. Colloid Interface Sci. 2006, 299, 608–616. [Google Scholar] [CrossRef]
  81. Hanson, R.L.; Hollingsworth, M.A. Functional Consequences of Differential O-glycosylation of MUC1, MUC4, and MUC16 (Downstream Effects on Signaling). Biomolecules 2016, 6, 34. [Google Scholar] [CrossRef] [PubMed]
  82. Singh, P.K.; Wen, Y.; Swanson, B.J.; Shanmugam, K.; Kazlauskas, A.; Cerny, R.L.; Gendler, S.J.; Hollingsworth, M.A. Platelet-derived growth factor receptor beta-mediated phosphorylation of MUC1 enhances invasiveness in pancreatic adenocarcinoma cells. Cancer Res. 2007, 67, 5201–5210. [Google Scholar] [CrossRef]
  83. Schroeder, J.A.; Thompson, M.C.; Gardner, M.M.; Gendler, S.J. Transgenic MUC1 interacts with epidermal growth factor receptor and correlates with mitogen-activated protein kinase activation in the mouse mammary gland. J. Biol. Chem. 2001, 276, 13057–13064. [Google Scholar] [CrossRef] [PubMed]
  84. Singh, P.K.; Behrens, M.E.; Eggers, J.P.; Cerny, R.L.; Bailey, J.M.; Shanmugam, K.; Gendler, S.J.; Bennett, E.P.; Hollingsworth, M.A. Phosphorylation of MUC1 by Met modulates interaction with p53 and MMP1 expression. J. Biol. Chem. 2008, 283, 26985–26995. [Google Scholar] [CrossRef]
  85. Wei, X.; Xu, H.; Kufe, D. Human MUC1 oncoprotein regulates p53-responsive gene transcription in the genotoxic stress response. Cancer Cell 2005, 7, 167–178. [Google Scholar] [CrossRef]
  86. Li, Y.; Kufe, D. The Human DF3/MUC1 carcinoma-associated antigen signals nuclear localization of the catenin p120(ctn). Biochem. Biophys. Res. Commun. 2001, 281, 440–443. [Google Scholar] [CrossRef] [PubMed]
  87. Liu, X.; Yi, C.; Wen, Y.; Radhakrishnan, P.; Tremayne, J.R.; Dao, T.; Johnson, K.R.; Hollingsworth, M.A. Interactions between MUC1 and p120 catenin regulate dynamic features of cell adhesion, motility, and metastasis. Cancer Res. 2014, 74, 1609–1620. [Google Scholar] [CrossRef]
  88. Kufe, D.W. MUC1-C oncoprotein as a target in breast cancer: Activation of signaling pathways and therapeutic approaches. Oncogene 2013, 32, 1073–1081. [Google Scholar] [CrossRef]
  89. Khodarev, N.; Ahmad, R.; Rajabi, H.; Pitroda, S.; Kufe, T.; McClary, C.; Joshi, M.D.; MacDermed, D.; Weichselbaum, R.; Kufe, D. Cooperativity of the MUC1 oncoprotein and STAT1 pathway in poor prognosis human breast cancer. Oncogene 2010, 29, 920–929. [Google Scholar] [CrossRef]
  90. Ahmad, R.; Raina, D.; Joshi, M.D.; Kawano, T.; Ren, J.; Kharbanda, S.; Kufe, D. MUC1-C oncoprotein functions as a direct activator of the nuclear factor-kappaB p65 transcription factor. Cancer Res. 2009, 69, 7013–7021. [Google Scholar] [CrossRef]
  91. Ahmad, R.; Rajabi, H.; Kosugi, M.; Joshi, M.D.; Alam, M.; Vasir, B.; Kawano, T.; Kharbanda, S.; Kufe, D. MUC1-C oncoprotein promotes STAT3 activation in an autoinductive regulatory loop. Sci. Signal 2011, 4, ra9. [Google Scholar] [CrossRef] [PubMed]
  92. Duraisamy, S.; Ramasamy, S.; Kharbanda, S.; Kufe, D. Distinct evolution of the human carcinoma-associated transmembrane mucins, MUC1, MUC4 and MUC16. Gene 2006, 373, 28–34. [Google Scholar] [CrossRef]
  93. Chaturvedi, P.; Singh, A.P.; Batra, S.K. Structure, evolution, and biology of the MUC4 mucin. FASEB J. 2008, 22, 966–981. [Google Scholar] [CrossRef] [PubMed]
  94. Lakshmanan, I.; Seshacharyulu, P.; Haridas, D.; Rachagani, S.; Gupta, S.; Joshi, S.; Guda, C.; Yan, Y.; Jain, M.; Ganti, A.K.; et al. Novel HER3/MUC4 oncogenic signaling aggravates the tumorigenic phenotypes of pancreatic cancer cells. Oncotarget 2015, 6, 21085–21099. [Google Scholar] [CrossRef] [PubMed]
  95. Chaturvedi, P.; Singh, A.P.; Chakraborty, S.; Chauhan, S.C.; Bafna, S.; Meza, J.L.; Singh, P.K.; Hollingsworth, M.A.; Mehta, P.P.; Batra, S.K. MUC4 mucin interacts with and stabilizes the HER2 oncoprotein in human pancreatic cancer cells. Cancer Res. 2008, 68, 2065–2070. [Google Scholar] [CrossRef]
  96. Nagy, P.; Friedländer, E.; Tanner, M.; Kapanen, A.I.; Carraway, K.L.; Isola, J.; Jovin, T.M. Decreased accessibility and lack of activation of ErbB2 in JIMT-1, a herceptin-resistant, MUC4-expressing breast cancer cell line. Cancer Res. 2005, 65, 473–482. [Google Scholar] [CrossRef]
  97. Zhi, X.; Tao, J.; Xie, K.; Zhu, Y.; Li, Z.; Tang, J.; Wang, W.; Xu, H.; Zhang, J.; Xu, Z. MUC4-induced nuclear translocation of β-catenin: A novel mechanism for growth, metastasis and angiogenesis in pancreatic cancer. Cancer Lett. 2014, 346, 104–113. [Google Scholar] [CrossRef]
  98. Tang, J.; Zhu, Y.; Xie, K.; Zhang, X.; Zhi, X.; Wang, W.; Li, Z.; Zhang, Q.; Wang, L.; Wang, J.; et al. The role of the AMOP domain in MUC4/Y-promoted tumour angiogenesis and metastasis in pancreatic cancer. J. Exp. Clin. Cancer Res. 2016, 35, 91. [Google Scholar] [CrossRef]
  99. O’Brien, T.J.; Beard, J.B.; Underwood, L.J.; Dennis, R.A.; Santin, A.D.; York, L. The CA 125 gene: An extracellular superstructure dominated by repeat sequences. Tumour Biol. 2001, 22, 348–366. [Google Scholar] [CrossRef]
  100. Haridas, D.; Ponnusamy, M.P.; Chugh, S.; Lakshmanan, I.; Seshacharyulu, P.; Batra, S.K. MUC16: Molecular analysis and its functional implications in benign and malignant conditions. FASEB J. 2014, 28, 4183–4199. [Google Scholar] [CrossRef]
  101. Das, S.; Majhi, P.D.; Al-Mugotir, M.H.; Rachagani, S.; Sorgen, P.; Batra, S.K. Membrane proximal ectodomain cleavage of MUC16 occurs in the acidifying Golgi/post-Golgi compartments. Sci. Rep. 2015, 5, 9759. [Google Scholar] [CrossRef]
  102. Lakshmanan, I.; Ponnusamy, M.P.; Das, S.; Chakraborty, S.; Haridas, D.; Mukhopadhyay, P.; Lele, S.M.; Batra, S.K. MUC16 induced rapid G2/M transition via interactions with JAK2 for increased proliferation and anti-apoptosis in breast cancer cells. Oncogene 2012, 31, 805–817. [Google Scholar] [CrossRef] [PubMed]
  103. Akita, K.; Tanaka, M.; Tanida, S.; Mori, Y.; Toda, M.; Nakada, H. CA125/MUC16 interacts with Src family kinases, and over-expression of its C-terminal fragment in human epithelial cancer cells reduces cell-cell adhesion. Eur. J. Cell Biol. 2013, 92, 257–263. [Google Scholar] [CrossRef]
  104. Thériault, C.; Pinard, M.; Comamala, M.; Migneault, M.; Beaudin, J.; Matte, I.; Boivin, M.; Piché, A.; Rancourt, C. MUC16 (CA125) regulates epithelial ovarian cancer cell growth, tumorigenesis and metastasis. Gynecol. Oncol. 2011, 121, 434–443. [Google Scholar] [CrossRef]
  105. Ligtenberg, M.J.; Kruijshaar, L.; Buijs, F.; van Meijer, M.; Litvinov, S.V.; Hilkens, J. Cell-associated episialin is a complex containing two proteins derived from a common precursor. J. Biol. Chem. 1992, 267, 6171–6177. [Google Scholar] [CrossRef] [PubMed]
  106. Levitin, F.; Stern, O.; Weiss, M.; Gil-Henn, C.; Ziv, R.; Prokocimer, Z.; Smorodinsky, N.I.; Rubinstein, D.B.; Wreschner, D.H. The MUC1 SEA module is a self-cleaving domain. J. Biol. Chem. 2005, 280, 33374–33386. [Google Scholar] [CrossRef]
  107. Escande, F.; Lemaitre, L.; Moniaux, N.; Batra, S.K.; Aubert, J.; Buisine, M. Genomic organization of MUC4 mucin gene. Towards the characterization of splice variants. Eur. J. Biochem. 2002, 269, 3637–3644. [Google Scholar] [CrossRef] [PubMed]
  108. Thathiah, A.; Carson, D.D. MT1-MMP mediates MUC1 shedding independent of TACE/ADAM17. Biochem J. 2004, 382, 363–373. [Google Scholar] [CrossRef]
  109. Zhang, L.; Muirhead, K.J.; Syed, Z.A.; Dimitriadis, E.K.; Ten Hagen, K.G. A novel cysteine-rich adaptor protein is required for mucin packaging and secretory granule stability in vivo. Proc. Natl. Acad. Sci. USA 2024, 121, e2314309121. [Google Scholar] [CrossRef]
  110. Hollingsworth, M.A.; Swanson, B.J. Mucins in cancer: Protection and control of the cell surface. Nat. Rev. Cancer 2004, 4, 45–60. [Google Scholar] [CrossRef]
  111. Roberton, A.M.; McKenzie, C.G.; Sharfe, N.; Stubbs, L.B. A glycosulphatase that removes sulphate from mucus glycoprotein. Biochem. J. 1993, 293 Pt 3, 683–689. [Google Scholar] [CrossRef] [PubMed]
  112. Nordman, H.; Davies, J.R.; Lindell, G.; Carlstedt, I. Human gastric mucins--a major population identified as MUC5. Biochem Soc. Trans. 1995, 23, 533S. [Google Scholar] [CrossRef] [PubMed]
  113. Ho, S.B.; Roberton, A.M.; Shekels, L.L.; Lyftogt, C.T.; Niehans, G.A.; Toribara, N.W. Expression cloning of gastric mucin complementary DNA and localization of mucin gene expression. Gastroenterology 1995, 109, 735–747. [Google Scholar] [CrossRef]
  114. Bhaskar, K.R.; Garik, P.; Turner, B.S.; Bradley, J.D.; Bansil, R.; Stanley, H.E.; LaMont, J.T. Viscous fingering of HCl through gastric mucin. Nature 1992, 360, 458–461. [Google Scholar] [CrossRef]
  115. Cunningham, M.; Beachey, E.H. Immunochemical properties of streptococcal M protein purified by isoelectric focusing. J. Immunol. 1975, 115, 1002–1006. [Google Scholar] [CrossRef]
  116. Cebo, C.; Dambrouck, T.; Maes, E.; Laden, C.; Strecker, G.; Michalski, J.C.; Zanetta, J.P. Recombinant human interleukins IL-1alpha, IL-1beta, IL-4, IL-6, and IL-7 show different and specific calcium-independent carbohydrate-binding properties. J. Biol. Chem. 2001, 276, 5685–5691. [Google Scholar] [CrossRef]
  117. Dabbagh, K.; Takeyama, K.; Lee, H.M.; Ueki, I.F.; Lausier, J.A.; Nadel, J.A. IL-4 induces mucin gene expression and goblet cell metaplasia in vitro and in vivo. J. Immunol. 1999, 162, 6233–6237. [Google Scholar] [CrossRef] [PubMed]
  118. Enss, M.L.; Cornberg, M.; Wagner, S.; Gebert, A.; Henrichs, M.; Eisenblätter, R.; Beil, W.; Kownatzki, R.; Hedrich, H.J. Proinflammatory cytokines trigger MUC gene expression and mucin release in the intestinal cancer cell line LS180. Inflamm Res. 2000, 49, 162–169. [Google Scholar] [CrossRef]
  119. Thim, L.; Madsen, F.; Poulsen, S.S. Effect of trefoil factors on the viscoelastic properties of mucus gels. Eur. J. Clin. Investig. 2002, 32, 519–527. [Google Scholar] [CrossRef]
  120. Tran, C.P.; Cook, G.A.; Yeomans, N.D.; Thim, L.; Giraud, A.S. Trefoil peptide TFF2 (spasmolytic polypeptide) potently accelerates healing and reduces inflammation in a rat model of colitis. Gut 1999, 44, 636–642. [Google Scholar] [CrossRef]
  121. Taupin, D.; Wu, D.C.; Jeon, W.K.; Devaney, K.; Wang, T.C.; Podolsky, D.K. The trefoil gene family are coordinately expressed immediate-early genes: EGF receptor- and MAP kinase-dependent interregulation. J. Clin. Investig. 1999, 103, 31. [Google Scholar] [CrossRef] [PubMed]
  122. Wright, N.A.; Hoffmann, W.; Otto, W.R.; Rio, M.C.; Thim, L. Rolling in the clover: Trefoil factor family (TFF)-domain peptides, cell migration and cancer. FEBS Lett. 1997, 408, 121–123. [Google Scholar] [CrossRef]
  123. Kobayashi, K.; Ogata, H.; Morikawa, M.; Iijima, S.; Harada, N.; Yoshida, T.; Brown, W.R.; Inoue, N.; Hamada, Y.; Ishii, H.; et al. Distribution and partial characterisation of IgG Fc binding protein in various mucin producing cells and body fluids. Gut 2002, 51, 169–176. [Google Scholar] [CrossRef]
  124. Phalipon, A.; Cardona, A.; Kraehenbuhl, J.P.; Edelman, L.; Sansonetti, P.J.; Corthésy, B. Secretory component: A new role in secretory IgA-mediated immune exclusion in vivo. Immunity 2002, 17, 107–115. [Google Scholar] [CrossRef] [PubMed]
  125. Niv, Y.; Rokkas, T. Mucin Expression in Colorectal Cancer (CRC): Systematic Review and Meta-Analysis. J. Clin. Gastroenterol. 2019, 53, 434–440. [Google Scholar] [CrossRef] [PubMed]
  126. Song, F.; Chen, F.-Y.; Wu, S.-Y.; Hu, B.; Liang, X.-L.; Yang, H.-Q.; Cheng, J.-W.; Wang, P.-X.; Guo, W.; Zhou, J.; et al. Mucin 1 promotes tumor progression through activating WNT/β-catenin signaling pathway in intrahepatic cholangiocarcinoma. J. Cancer 2021, 12, 6937–6947. [Google Scholar] [CrossRef] [PubMed]
  127. Gundamaraju, R.; Chong, W.C. Consequence of distinctive expression of MUC2 in colorectal cancers: How much is actually bad? Biochim. Biophys. Acta Rev. Cancer 2021, 1876, 188579. [Google Scholar] [CrossRef]
  128. Zhang, C.; He, K.; Pan, F.; Li, Y.; Wu, J. Prognostic value of Muc5AC in gastric cancer: A meta-analysis. World J. Gastroenterol. 2015, 21, 10453–10460. [Google Scholar] [CrossRef]
  129. Rathee, R.; Devi, A.; Narwal, A.; Kamboj, M.; Singh, S. Immunohistochemical Coexpression of MUC1 and MUC4 in Oral Leukoplakia and Oral Squamous Cell Carcinoma. Head Neck Pathol. 2021, 15, 831–842. [Google Scholar] [CrossRef]
  130. Albrecht, H.; Carraway, K.L. MUC1 and MUC4: Switching the emphasis from large to small. Cancer Biother. Radiopharm. 2011, 26, 261–271. [Google Scholar] [CrossRef]
  131. Kaur, S.; Momi, N.; Chakraborty, S.; Wagner, D.G.; Horn, A.J.; Lele, S.M.; Theodorescu, D.; Batra, S.K. Altered expression of transmembrane mucins, MUC1 and MUC4, in bladder cancer: Pathological implications in diagnosis. PLoS ONE 2014, 9, e92742. [Google Scholar] [CrossRef]
  132. Yokoyama, S.; Higashi, M.; Kitamoto, S.; Oeldorf, M.; Knippschild, U.; Kornmann, M.; Maemura, K.; Kurahara, H.; Wiest, E.; Hamada, T.; et al. Aberrant methylation of MUC1 and MUC4 promoters are potential prognostic biomarkers for pancreatic ductal adenocarcinomas. Oncotarget 2016, 7, 42553–42565. [Google Scholar] [CrossRef] [PubMed]
  133. Kaur, S.; Kumar, S.; Momi, N.; Sasson, A.R.; Batra, S.K. Mucins in pancreatic cancer and its microenvironment. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 607–620. [Google Scholar] [CrossRef] [PubMed]
  134. Chaturvedi, P.; Singh, A.P.; Moniaux, N.; Senapati, S.; Chakraborty, S.; Meza, J.L.; Batra, S.K. MUC4 mucin potentiates pancreatic tumor cell proliferation, survival, and invasive properties and interferes with its interaction to extracellular matrix proteins. Mol. Cancer Res. 2007, 5, 309–320. [Google Scholar] [CrossRef]
  135. Ponnusamy, M.P.; Singh, A.P.; Jain, M.; Chakraborty, S.; Moniaux, N.; Batra, S.K. MUC4 activates HER2 signalling and enhances the motility of human ovarian cancer cells. Br. J. Cancer 2008, 99, 520–526. [Google Scholar] [CrossRef]
  136. Singh, P.K.; Hollingsworth, M.A. Cell surface-associated mucins in signal transduction. Trends Cell Biol. 2006, 16, 467–476. [Google Scholar] [CrossRef] [PubMed]
  137. Besmer, D.M.; Curry, J.M.; Roy, L.D.; Tinder, T.L.; Sahraei, M.; Schettini, J.; Hwang, S.; Lee, Y.Y.; Gendler, S.J.; Mukherjee, P. Pancreatic ductal adenocarcinoma mice lacking mucin 1 have a profound defect in tumor growth and metastasis. Cancer Res. 2011, 71, 4432–4442. [Google Scholar] [CrossRef]
  138. Nath, S.; Daneshvar, K.; Roy, L.D.; Grover, P.; Kidiyoor, A.; Mosley, L.; Sahraei, M.; Mukherjee, P. MUC1 induces drug resistance in pancreatic cancer cells via upregulation of multidrug resistance genes. Oncogenesis 2013, 2, e51. [Google Scholar] [CrossRef]
  139. Li, Y.; Ren, J.; Yu, W.; Li, Q.; Kuwahara, H.; Yin, L.; Carraway, K.L. 3.; Kufe, D. The epidermal growth factor receptor regulates interaction of the human DF3/MUC1 carcinoma antigen with c-Src and beta-catenin. J. Biol. Chem. 2001, 276, 35239–35242. [Google Scholar] [CrossRef]
  140. Tang, Z.; Yang, Y.; Chen, W.; Liang, T. Epigenetic deregulation of MLF1 drives intrahepatic cholangiocarcinoma progression through EGFR/AKT and Wnt/β-catenin signaling. Hepatol. Commun. 2023, 7, e0204. [Google Scholar] [CrossRef]
  141. Rajabi, H.; Alam, M.; Takahashi, H.; Kharbanda, A.; Guha, M.; Ahmad, R.; Kufe, D. MUC1-C oncoprotein activates the ZEB1/miR-200c regulatory loop and epithelial-mesenchymal transition. Oncogene 2014, 33, 1680–1689. [Google Scholar] [CrossRef]
  142. Zhai, Y.; Lu, Q.; Lou, T.; Cao, G.; Wang, S.; Zhang, Z. MUC16 affects the biological functions of ovarian cancer cells and induces an antitumor immune response by activating dendritic cells. Ann. Transl. Med. 2020, 8, 1494–6388. [Google Scholar] [CrossRef]
  143. Rajabi, H.; Tagde, A.; Alam, M.; Bouillez, A.; Pitroda, S.; Suzuki, Y.; Kufe, D. DNA methylation by DNMT1 and DNMT3b methyltransferases is driven by the MUC1-C oncoprotein in human carcinoma cells. Oncogene 2016, 35, 6439–6445. [Google Scholar] [CrossRef]
  144. Woo, J.K.; Choi, Y.; Oh, S.; Jeong, J.; Choi, D.; Seo, H.; Kim, C. Mucin 1 enhances the tumor angiogenic response by activation of the AKT signaling pathway. Oncogene 2012, 31, 2187–2198. [Google Scholar] [CrossRef] [PubMed]
  145. Zrihan-Licht, S.; Baruch, A.; Elroy-Stein, O.; Keydar, I.; Wreschner, D.H. Tyrosine phosphorylation of the MUC1 breast cancer membrane proteins. Cytokine receptor-like molecules. FEBS Lett. 1994, 356, 130–136. [Google Scholar] [CrossRef]
  146. Rachagani, S.; Torres, M.P.; Kumar, S.; Haridas, D.; Baine, M.; A Macha, M.; Kaur, S.; Ponnusamy, M.P.; Dey, P.; Seshacharyulu, P.; et al. Mucin (Muc) expression during pancreatic cancer progression in spontaneous mouse model: Potential implications for diagnosis and therapy. J. Hematol. Oncol. 2012, 5, 68. [Google Scholar] [CrossRef] [PubMed]
  147. Andrianifahanana, M.; Moniaux, N.; Schmied, B.M.; Ringel, J.; Friess, H.; Hollingsworth, M.A.; Büchler, M.W.; Aubert, J.P.; Batra, S.K. Mucin (MUC) gene expression in human pancreatic adenocarcinoma and chronic pancreatitis: A potential role of MUC4 as a tumor marker of diagnostic significance. Clin. Cancer Res. 2001, 7, 4033–4040. [Google Scholar] [PubMed]
  148. Velcich, A.; Yang, W.; Heyer, J.; Fragale, A.; Nicholas, C.; Viani, S.; Kucherlapati, R.; Lipkin, M.; Yang, K.; Augenlicht, L. Colorectal cancer in mice genetically deficient in the mucin Muc2. Science 2002, 295, 1726–1729. [Google Scholar] [CrossRef]
  149. Betge, J.; Schneider, N.I.; Harbaum, L.; Pollheimer, M.J.; Lindtner, R.A.; Kornprat, P.; Ebert, M.P.; Langner, C. MUC1, MUC2, MUC5AC, and MUC6 in colorectal cancer: Expression profiles and clinical significance. Virchows Arch. 2016, 469, 255–265. [Google Scholar] [CrossRef]
  150. Wang, J.; Chang, C.; Hsieh, J.; Lee, L.; Huang, T.; Chai, C.; Lin, S. Role of MUC1 and MUC5AC expressions as prognostic indicators in gastric carcinomas. J. Surg. Oncol. 2003, 83, 253–260. [Google Scholar] [CrossRef]
  151. Kocer, B.; Soran, A.; Kiyak, G.; Erdogan, S.; Eroglu, A.; Bozkurt, B.; Solak, C.; Cengiz, O. Prognostic significance of mucin expression in gastric carcinoma. Dig. Dis. Sci. 2004, 49, 954–964. [Google Scholar] [CrossRef] [PubMed]
  152. Morimoto, Y.; Yamashita, N.; Daimon, T.; Hirose, H.; Yamano, S.; Haratake, N.; Ishikawa, S.; Bhattacharya, A.; Fushimi, A.; Ahmad, R.; et al. MUC1-C is a master regulator of MICA/B NKG2D ligand and exosome secretion in human cancer cells. J. Immunother. Cancer 2023, 11, e006238. [Google Scholar] [CrossRef] [PubMed]
  153. Kufe, D.W. Emergence of MUC1 in Mammals for Adaptation of Barrier Epithelia. Cancers 2022, 14, 4805. [Google Scholar] [CrossRef]
  154. Cadoux, M.; Caruso, S.; Pham, S.; Gougelet, A.; Pophillat, C.; Riou, R.; Loesch, R.; Colnot, S.; Nguyen, C.T.; Calderaro, J.; et al. Expression of NKG2D ligands is downregulated by β-catenin signalling and associates with HCC aggressiveness. J. Hepatol. 2021, 74, 1386–1397. [Google Scholar] [CrossRef] [PubMed]
  155. Wang, D.; Yang, L.; Yu, W.; Wu, Q.; Lian, J.; Li, F.; Liu, S.; Li, A.; He, Z.; Liu, J.; et al. Colorectal cancer cell-derived CCL20 recruits regulatory T cells to promote chemoresistance via FOXO1/CEBPB/NF-κB signaling. J. Immunother. Cancer 2019, 7, 215. [Google Scholar] [CrossRef]
  156. Ganguly, K.; Shah, A.; Atri, P.; Rauth, S.; Ponnusamy, M.P.; Kumar, S.; Batra, S.K. Chemokine-mucinome interplay in shaping the heterogeneous tumor microenvironment of pancreatic cancer. Semin. Cancer Biol. 2022, 86, 511–520. [Google Scholar] [CrossRef]
  157. Shimizu, A.; Hirono, S.; Tani, M.; Kawai, M.; Okada, K.; Miyazawa, M.; Kitahata, Y.; Nakamura, Y.; Noda, T.; Yokoyama, S.; et al. Coexpression of MUC16 and mesothelin is related to the invasion process in pancreatic ductal adenocarcinoma. Cancer Sci. 2012, 103, 739–746. [Google Scholar] [CrossRef]
  158. Welch, D.R.; Hurst, D.R. Defining the Hallmarks of Metastasis. Cancer Res. 2019, 79, 3011–3027. [Google Scholar] [CrossRef]
  159. Dhanisha, S.S.; Guruvayoorappan, C. Pathological Implications of Mucin Signaling in Metastasis. Curr. Cancer Drug Targets 2023, 23, 585–602. [Google Scholar] [CrossRef]
  160. Singh, A.P.; Moniaux, N.; Chauhan, S.C.; Meza, J.L.; Batra, S.K. Inhibition of MUC4 expression suppresses pancreatic tumor cell growth and metastasis. Cancer Res. 2004, 64, 622–630. [Google Scholar] [CrossRef]
  161. Ohira, G.; Kimura, K.; Yamada, N.; Amano, R.; Nakata, B.; Doi, Y.; Murata, A.; Yashiro, M.; Tanaka, S.; Ohsawa, M.; et al. MUC1 and HER2 might be associated with invasive phenotype of intraductal papillary mucinous neoplasm. Hepatogastroenterology 2013, 60, 1067–1072. [Google Scholar]
  162. Pang, Z.; Dong, X.; Deng, H.; Wang, C.; Liao, X.; Liao, C.; Liao, Y.; Tian, W.; Cheng, J.; Chen, G.; et al. MUC1 triggers lineage plasticity of Her2 positive mammary tumors. Oncogene 2022, 41, 3064–3078. [Google Scholar] [CrossRef] [PubMed]
  163. Muniyan, S.; Haridas, D.; Chugh, S.; Rachagani, S.; Lakshmanan, I.; Gupta, S.; Seshacharyulu, P.; Smith, L.M.; Ponnusamy, M.P.; Batra, S.K. MUC16 contributes to the metastasis of pancreatic ductal adenocarcinoma through focal adhesion mediated signaling mechanism. Genes. Cancer 2016, 7, 110–124. [Google Scholar] [CrossRef]
  164. Lakshmanan, I.; Marimuthu, S.; Chaudhary, S.; Seshacharyulu, P.; Rachagani, S.; Muniyan, S.; Chirravuri-Venkata, R.; Atri, P.; Rauth, S.; Nimmakayala, R.K.; et al. Muc16 depletion diminishes KRAS-induced tumorigenesis and metastasis by altering tumor microenvironment factors in pancreatic ductal adenocarcinoma. Oncogene 2022, 41, 5147–5159. [Google Scholar] [CrossRef]
  165. Geng, Y.; Yeh, K.; Takatani, T.; King, M.R. Three to Tango: MUC1 as a Ligand for Both E-Selectin and ICAM-1 in the Breast Cancer Metastatic Cascade. Front. Oncol. 2012, 2, 76. [Google Scholar] [CrossRef] [PubMed]
  166. Coppin, L.; Vincent, A.; Frénois, F.; Duchêne, B.; Lahdaoui, F.; Stechly, L.; Renaud, F.; Villenet, C.; Van Seuningen, I.; Leteurtre, E.; et al. Galectin-3 is a non-classic RNA binding protein that stabilizes the mucin MUC4 mRNA in the cytoplasm of cancer cells. Sci. Rep. 2017, 7, 43927. [Google Scholar] [CrossRef]
  167. Yu, L.-G.; Andrews, N.; Zhao, Q.; McKean, D.; Williams, J.F.; Connor, L.J.; Gerasimenko, O.V.; Hilkens, J.; Hirabayashi, J.; Kasai, K.; et al. Galectin-3 interaction with Thomsen-Friedenreich disaccharide on cancer-associated MUC1 causes increased cancer cell endothelial adhesion. J. Biol. Chem. 2007, 282, 773–781. [Google Scholar] [CrossRef]
  168. Zhao, Q.; Barclay, M.; Hilkens, J.; Guo, X.; Barrow, H.; Rhodes, J.M.; Yu, L. Interaction between circulating galectin-3 and cancer-associated MUC1 enhances tumour cell homotypic aggregation and prevents anoikis. Mol. Cancer 2010, 9, 154. [Google Scholar] [CrossRef] [PubMed]
  169. Zhao, Q.; Guo, X.; Nash, G.B.; Stone, P.C.; Hilkens, J.; Rhodes, J.M.; Yu, L. Circulating galectin-3 promotes metastasis by modifying MUC1 localization on cancer cell surface. Cancer Res. 2009, 69, 6799–6806. [Google Scholar] [CrossRef]
  170. Abbott, M.; Ustoyev, Y. Cancer and the Immune System: The History and Background of Immunotherapy. Semin. Oncol. Nurs. 2019, 35, 150923. [Google Scholar] [CrossRef]
  171. Ogata, S.; Maimonis, P.J.; Itzkowitz, S.H. Mucins bearing the cancer-associated sialosyl-Tn antigen mediate inhibition of natural killer cell cytotoxicity. Cancer Res. 1992, 52, 4741–4746. [Google Scholar]
  172. van de Wiel-van Kemenade, E.; Ligtenberg, M.J.; de Boer, A.J.; Buijs, F.; Vos, H.L.; Melief, C.J.; Hilkens, J.; Figdor, C.G. Episialin (MUC1) inhibits cytotoxic lymphocyte-target cell interaction. J. Immunol. 1993, 151, 767–776. [Google Scholar] [CrossRef]
  173. Bhatia, R.; Gautam, S.K.; Cannon, A.; Thompson, C.; Hall, B.R.; Aithal, A.; Banerjee, K.; Jain, M.; Solheim, J.C.; Kumar, S.; et al. Cancer-associated mucins: Role in immune modulation and metastasis. Cancer Metastasis Rev. 2019, 38, 223–236. [Google Scholar] [CrossRef] [PubMed]
  174. Maman, S.; Witz, I.P. A history of exploring cancer in context. Nat. Rev. Cancer 2018, 18, 359–376. [Google Scholar] [CrossRef]
  175. Anderson, N.M.; Simon, M.C. The tumor microenvironment. Curr. Biol. 2020, 30, R921–R925. [Google Scholar] [CrossRef]
  176. Lei, X.; Lei, Y.; Li, J.; Du, W.; Li, R.; Yang, J.; Li, J.; Li, F.; Tan, H. Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy. Cancer Lett. 2020, 470, 126–133. [Google Scholar] [CrossRef]
  177. Lyssiotis, C.A.; Kimmelman, A.C. Metabolic Interactions in the Tumor Microenvironment. Trends Cell Biol. 2017, 27, 863–875. [Google Scholar] [CrossRef]
  178. Wang, M.; Zhao, J.; Zhang, L.; Wei, F.; Lian, Y.; Wu, Y.; Gong, Z.; Zhang, S.; Zhou, J.; Cao, K.; et al. Role of tumor microenvironment in tumorigenesis. J. Cancer 2017, 8, 761–773. [Google Scholar] [CrossRef]
  179. Kitamoto, S.; Yokoyama, S.; Higashi, M.; Yamada, N.; Takao, S.; Yonezawa, S. MUC1 enhances hypoxia-driven angiogenesis through the regulation of multiple proangiogenic factors. Oncogene 2013, 32, 4614–4621. [Google Scholar] [CrossRef] [PubMed]
  180. Roy, L.D.; Sahraei, M.; Subramani, D.B.; Besmer, D.; Nath, S.; Tinder, T.L.; Bajaj, E.; Shanmugam, K.; Lee, Y.Y.; Hwang, S.I.L.; et al. MUC1 enhances invasiveness of pancreatic cancer cells by inducing epithelial to mesenchymal transition. Oncogene 2011, 30, 1449–1459. [Google Scholar] [CrossRef] [PubMed]
  181. Rashid, M.; Zadeh, L.R.; Baradaran, B.; Molavi, O.; Ghesmati, Z.; Sabzichi, M.; Ramezani, F. Up-down regulation of HIF-1α in cancer progression. Gene 2021, 798, 145796. [Google Scholar] [CrossRef] [PubMed]
  182. Aubert, S.; Fauquette, V.; Hémon, B.; Lepoivre, R.; Briez, N.; Bernard, D.; Van Seuningen, I.; Leroy, X.; Perrais, M. MUC1, a new hypoxia inducible factor target gene, is an actor in clear renal cell carcinoma tumor progression. Cancer Res. 2009, 69, 5707–5715. [Google Scholar] [CrossRef]
  183. Jing, X.; Liang, H.; Hao, C.; Yang, X.; Cui, X. Overexpression of MUC1 predicts poor prognosis in patients with breast cancer. Oncol. Rep. 2019, 41, 801–810. [Google Scholar] [CrossRef] [PubMed]
  184. Brockhausen, I.; Yang, J.M.; Burchell, J.; Whitehouse, C.; Taylor-Papadimitriou, J. Mechanisms underlying aberrant glycosylation of MUC1 mucin in breast cancer cells. Eur. J. Biochem. 1995, 233, 607–617. [Google Scholar] [CrossRef] [PubMed]
  185. Burchell, J.; Poulsom, R.; Hanby, A.; Whitehouse, C.; Cooper, L.; Clausen, H.; Miles, D.; Taylor-Papadimitriou, J. An alpha2,3 sialyltransferase (ST3Gal I) is elevated in primary breast carcinomas. Glycobiology 1999, 9, 1307–1311. [Google Scholar] [CrossRef]
  186. Park, J.; Nishidate, T.; Kijima, K.; Ohashi, T.; Takegawa, K.; Fujikane, T.; Hirata, K.; Nakamura, Y.; Katagiri, T. Critical roles of mucin 1 glycosylation by transactivated polypeptide N-acetylgalactosaminyltransferase 6 in mammary carcinogenesis. Cancer Res. 2010, 70, 2759–2769. [Google Scholar] [CrossRef]
  187. Wu, C.; Guo, X.; Wang, W.; Wang, Y.; Shan, Y.; Zhang, B.; Song, W.; Ma, S.; Ge, J.; Deng, H.; et al. N-Acetylgalactosaminyltransferase-14 as a potential biomarker for breast cancer by immunohistochemistry. BMC Cancer 2010, 10, 123. [Google Scholar] [CrossRef]
  188. Picco, G.; Julien, S.; Brockhausen, I.; Beatson, R.; Antonopoulos, A.; Haslam, S.; Mandel, U.; Dell, A.; Pinder, S.; Taylor-Papadimitriou, J.; et al. Over-expression of ST3Gal-I promotes mammary tumorigenesis. Glycobiology 2010, 20, 1241–1250. [Google Scholar] [CrossRef]
  189. Tiainen, S.; Tumelius, R.; Rilla, K.; Hämäläinen, K.; Tammi, M.; Tammi, R.; Kosma, V.; Oikari, S.; Auvinen, P. High numbers of macrophages, especially M2-like (CD163-positive), correlate with hyaluronan accumulation and poor outcome in breast cancer. Histopathology 2015, 66, 873–883. [Google Scholar] [CrossRef]
  190. Smarr, K.L.; Parker, J.C.; Wright, G.E.; Stucky-Ropp, R.C.; Buckelew, S.P.; Hoffman, R.W.; O’Sullivan, F.X.; Hewett, J.E. The importance of enhancing self-efficacy in rheumatoid arthritis. Arthritis Care Res. 1997, 10, 18–26. [Google Scholar] [CrossRef]
  191. Brockhausen, I. Mucin-type O-glycans in human colon and breast cancer: Glycodynamics and functions. EMBO Rep. 2006, 7, 599–604. [Google Scholar] [CrossRef] [PubMed]
  192. Brockhausen, I. Pathways of O-glycan biosynthesis in cancer cells. Biochim. Biophys. Acta 1999, 1473, 67–95. [Google Scholar] [CrossRef] [PubMed]
  193. Shibao, K.; Izumi, H.; Nakayama, Y.; Ohta, R.; Nagata, N.; Nomoto, M.; Matsuo, K.; Yamada, Y.; Kitazato, K.; Itoh, H.; et al. Expression of UDP-N-acetyl-alpha-D-galactosamine-polypeptide galNAc N-acetylgalactosaminyl transferase-3 in relation to differentiation and prognosis in patients with colorectal carcinoma. Cancer 2002, 94, 1939–1946. [Google Scholar] [CrossRef] [PubMed]
  194. Itzkowitz, S.H.; Bloom, E.J.; Kokal, W.A.; Modin, G.; Hakomori, S.; Kim, Y.S. Sialosyl-Tn. A novel mucin antigen associated with prognosis in colorectal cancer patients. Cancer 1990, 66, 1960–1966. [Google Scholar] [CrossRef]
  195. Nakamori, S.; Kameyama, M.; Imaoka, S.; Furukawa, H.; Ishikawa, O.; Sasaki, Y.; Kabuto, T.; Iwanaga, T.; Matsushita, Y.; Irimura, T. Increased expression of sialyl Lewis x antigen correlates with poor survival in patients with colorectal carcinoma: Clinicopathological and immunohistochemical study. Cancer Res. 1993, 53, 3632–3637. [Google Scholar]
  196. Belisle, J.A.; Horibata, S.; Jennifer, G.A.; Petrie, S.; Kapur, A.; André, S.; Gabius, H.-J.; Rancourt, C.; Connor, J.; Paulson, J.C.; et al. Identification of Siglec-9 as the receptor for MUC16 on human NK cells, B cells, and monocytes. Mol. Cancer 2010, 9, 118. [Google Scholar] [CrossRef]
  197. Kang, H.; Min, B.S.; Lee, K.Y.; Kim, N.K.; Kim, S.N.; Choi, J.; Kim, H. Loss of E-cadherin and MUC2 expressions correlated with poor survival in patients with stages II and III colorectal carcinoma. Ann. Surg. Oncol. 2011, 18, 711–719. [Google Scholar] [CrossRef]
  198. Ajioka, Y.; Allison, L.J.; Jass, J.R. Significance of MUC1 and MUC2 mucin expression in colorectal cancer. J. Clin. Pathol. 1996, 49, 560–564. [Google Scholar] [CrossRef]
  199. Johansson, M.E.V.; Phillipson, M.; Petersson, J.; Velcich, A.; Holm, L.; Hansson, G.C. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc. Natl. Acad. Sci. USA 2008, 105, 15064–15069. [Google Scholar] [CrossRef]
  200. Shan, Y.; Hsu, H.; Lai, M.; Yen, M.; Fang, J.; Weng, T.; Chen, Y. Suppression of mucin 2 promotes interleukin-6 secretion and tumor growth in an orthotopic immune-competent colon cancer animal model. Oncol. Rep. 2014, 32, 2335–2342. [Google Scholar] [CrossRef]
  201. Wang, H.; Jin, S.; Lu, H.; Mi, S.; Shao, W.; Zuo, X.; Yin, H.; Zeng, S.; Shimamoto, F.; Qi, G. Expression of survivin, MUC2 and MUC5 in colorectal cancer and their association with clinicopathological characteristics. Oncol. Lett. 2017, 14, 1011–1016. [Google Scholar] [CrossRef] [PubMed]
  202. Imai, Y.; Yamagishi, H.; Fukuda, K.; Ono, Y.; Inoue, T.; Ueda, Y. Differential mucin phenotypes and their significance in a variation of colorectal carcinoma. World J. Gastroenterol. 2013, 19, 3957–3968. [Google Scholar] [CrossRef] [PubMed]
  203. Walsh, M.D.; Clendenning, M.; Williamson, E.; Pearson, S.-A.; Walters, R.J.; Nagler, B.; Packenas, D.; Win, A.K.; Hopper, J.L.; Jenkins, M.A.; et al. Expression of MUC2, MUC5AC, MUC5B, and MUC6 mucins in colorectal cancers and their association with the CpG island methylator phenotype. Mod. Pathol. 2013, 26, 1642–1656. [Google Scholar] [CrossRef]
  204. Pothuraju, R.; Rachagani, S.; Krishn, S.R.; Chaudhary, S.; Nimmakayala, R.K.; Siddiqui, J.A.; Ganguly, K.; Lakshmanan, I.; Cox, J.L.; Mallya, K.; et al. Molecular implications of MUC5AC-CD44 axis in colorectal cancer progression and chemoresistance. Mol. Cancer 2020, 19, 37. [Google Scholar] [CrossRef]
  205. McGuigan, A.; Kelly, P.; Turkington, R.C.; Jones, C.; Coleman, H.G.; McCain, R.S. Pancreatic cancer: A review of clinical diagnosis, epidemiology, treatment and outcomes. World J. Gastroenterol. 2018, 24, 4846–4861. [Google Scholar] [CrossRef]
  206. Kannagi, R. Carbohydrate antigen sialyl Lewis a—Its pathophysiological significance and induction mechanism in cancer progression. Chang Gung Med. J. 2007, 30, 189–209. [Google Scholar]
  207. Ballehaninna, U.K.; Chamberlain, R.S. Serum CA 19-9 as a Biomarker for Pancreatic Cancer—A Comprehensive Review. Indian J. Surg. Oncol. 2011, 2, 88–100. [Google Scholar] [CrossRef]
  208. Liu, J.F.; Moore, K.N.; Birrer, M.J.; Berlin, S.; Matulonis, U.A.; Infante, J.R.; Wolpin, B.; Poon, K.A.; Firestein, R.; Xu, J.; et al. Phase I study of safety and pharmacokinetics of the anti-MUC16 antibody-drug conjugate DMUC5754A in patients with platinum-resistant ovarian cancer or unresectable pancreatic cancer. Ann. Oncol. 2016, 27, 2124–2130. [Google Scholar] [CrossRef] [PubMed]
  209. Huang, X.; Wang, X.; Lu, S.; Chen, C.; Wang, J.; Zheng, Y.; Ren, B.; Xu, L. Clinicopathological and prognostic significance of MUC4 expression in cancers: Evidence from meta-analysis. Int. J. Clin. Exp. Med. 2015, 8, 10274–10283. [Google Scholar]
  210. Klöppel, G.; Kosmahl, M. Is the intraductal papillary mucinous neoplasia of the biliary tract a counterpart of pancreatic papillary mucinous neoplasm? J. Hepatol. 2006, 44, 249–250. [Google Scholar] [CrossRef]
  211. Fu, X.; Tang, N.; Xie, W.; Mao, L.; Qiu, Y. MUC1 promotes glycolysis through inhibiting BRCA1 expression in pancreatic cancer. Chin. J. Nat. Med. 2020, 18, 178–185. [Google Scholar] [CrossRef] [PubMed]
  212. Levi, E.; Klimstra, D.S.; Andea, A.; Basturk, O.; Adsay, N.V. MUC1 and MUC2 in pancreatic neoplasia. J. Clin. Pathol. 2004, 57, 456–462. [Google Scholar] [CrossRef] [PubMed]
  213. Nath, S.; Roy, L.D.; Grover, P.; Rao, S.; Mukherjee, P. Mucin 1 Regulates Cox-2 Gene in Pancreatic Cancer. Pancreas 2015, 44, 909–917. [Google Scholar] [CrossRef]
  214. Trehoux, S.; Duchene, B.; Jonckheere, N.; Van Seuningen, I. The MUC1 oncomucin regulates pancreatic cancer cell biological properties and chemoresistance. Implication of p42-44 MAPK, Akt, Bcl-2 and MMP13 pathways. Biochem. Biophys. Res. Commun. 2015, 456, 757–762. [Google Scholar] [CrossRef]
  215. Zhou, R.; Curry, J.M.; Roy, L.D.; Grover, P.; Haider, J.; Moore, L.J.; Wu, S.-T.; Kamesh, A.; Yazdanifar, M.; A Ahrens, W.; et al. A novel association of neuropilin-1 and MUC1 in pancreatic ductal adenocarcinoma: Role in induction of VEGF signaling and angiogenesis. Oncogene 2016, 35, 5608–5618. [Google Scholar] [CrossRef]
  216. Fan, K.; Yang, C.; Fan, Z.; Huang, Q.; Zhang, Y.; Cheng, H.; Jin, K.; Lu, Y.; Wang, Z.; Luo, G.; et al. MUC16 C terminal-induced secretion of tumor-derived IL-6 contributes to tumor-associated Treg enrichment in pancreatic cancer. Cancer Lett. 2018, 418, 167–175. [Google Scholar] [CrossRef]
  217. Gagnon, M.; Zihler Berner, A.; Chervet, N.; Chassard, C.; Lacroix, C. Comparison of the Caco-2, HT-29 and the mucus-secreting HT29-MTX intestinal cell models to investigate Salmonella adhesion and invasion. J. Microbiol. Methods 2013, 94, 274–279. [Google Scholar] [CrossRef] [PubMed]
  218. Andrianifahanana, M.; Moniaux, N.; Batra, S.K. Regulation of mucin expression: Mechanistic aspects and implications for cancer and inflammatory diseases. Biochim. Biophys. Acta 2006, 1765, 189–222. [Google Scholar] [CrossRef]
  219. Ehre, C.; Ridley, C.; Thornton, D.J. Cystic fibrosis: An inherited disease affecting mucin-producing organs. Int. J. Biochem. Cell Biol. 2014, 52, 136–145. [Google Scholar] [CrossRef]
  220. Leong, D.T.; Ng, K.W. Probing the relevance of 3D cancer models in nanomedicine research. Adv. Drug Deliv. Rev. 2014, 79-80, 95–106. [Google Scholar] [CrossRef]
  221. Nederman, T.; Norling, B.; Glimelius, B.; Carlsson, J.; Brunk, U. Demonstration of an extracellular matrix in multicellular tumor spheroids. Cancer Res. 1984, 44, 3090–3097. [Google Scholar] [PubMed]
  222. Rotin, D.; Robinson, B.; Tannock, I.F. Influence of hypoxia and an acidic environment on the metabolism and viability of cultured cells: Potential implications for cell death in tumors. Cancer Res. 1986, 46, 2821–2826. [Google Scholar] [PubMed]
  223. Kelm, J.M.; Timmins, N.E.; Brown, C.J.; Fussenegger, M.; Nielsen, L.K. Method for generation of homogeneous multicellular tumor spheroids applicable to a wide variety of cell types. Biotechnol. Bioeng. 2003, 83, 173–180. [Google Scholar] [CrossRef]
  224. Agrawal, B.; Krantz, M.J.; Parker, J.; Longenecker, B.M. Expression of MUC1 mucin on activated human T cells: Implications for a role of MUC1 in normal immune regulation. Cancer Res. 1998, 58, 4079–4081. [Google Scholar]
  225. Treon, S.P.; Maimonis, P.; Bua, D.; Young, G.; Raje, N.; Mollick, J.; Chauhan, D.; Tai, Y.T.; Hideshima, T.; Shima, Y.; et al. Elevated soluble MUC1 levels and decreased anti-MUC1 antibody levels in patients with multiple myeloma. Blood 2000, 96, 3147–3153. [Google Scholar] [CrossRef] [PubMed]
  226. Fremd, C.; Stefanovic, S.; Beckhove, P.; Pritsch, M.; Lim, H.; Wallwiener, M.; Heil, J.; Golatta, M.; Rom, J.; Sohn, C.; et al. Mucin 1-specific B cell immune responses and their impact on overall survival in breast cancer patients. Oncoimmunology 2015, 5, e1057387. [Google Scholar] [CrossRef]
  227. Matsuda, A.; Kuno, A.; Matsuzaki, H.; Kawamoto, T.; Shikanai, T.; Nakanuma, Y.; Yamamoto, M.; Ohkohchi, N.; Ikehara, Y.; Shoda, J.; et al. Glycoproteomics-based cancer marker discovery adopting dual enrichment with Wisteria floribunda agglutinin for high specific glyco-diagnosis of cholangiocarcinoma. J. Proteom. 2013, 85, 1–11. [Google Scholar] [CrossRef]
  228. Kim, Y.; Pecha, R.L.; Keihanian, T.; Mercado, M.; Pena-Munoz, S.V.; Lang, K.; Van Buren, G.; Dhingra, S.; Othman, M.O. MUC1 Expressions and Its Prognostic Values in US Gastric Cancer Patients. Cancers 2023, 15, 998. [Google Scholar] [CrossRef]
  229. Strawbridge, R.J.; Nistér, M.; Brismar, K.; Grönberg, H.; Li, C. MUC1 as a Putative Prognostic Marker for Prostate Cancer. Biomark. Insights 2008, 3, 303–315. [Google Scholar] [CrossRef]
  230. Li, C.; Liu, T.; Yin, L.; Zuo, D.; Lin, Y.; Wang, L. Prognostic and clinicopathological value of MUC1 expression in colorectal cancer: A meta-analysis. Medicine 2019, 98, e14659. [Google Scholar] [CrossRef]
  231. Yin, Y.; Yang, C.; Xu, J.; Luo, Y.; Xia, Q.; He, K. MUC1 promotes lung metastases of liver cancer by impairing anti-tumor immunity. Discov. Oncol. 2023, 14, 18. [Google Scholar] [CrossRef] [PubMed]
  232. Duffy, M.J.; Shering, S.; Sherry, F.; McDermott, E.; O’Higgins, N. CA 15-3: A prognostic marker in breast cancer. Int. J. Biol. Markers 2000, 15, 330–333. [Google Scholar] [CrossRef] [PubMed]
  233. Qing, L.; Li, Q.; Yang, Y.; Xu, W.; Dong, Z. A prognosis marker MUC1 correlates with metabolism and drug resistance in bladder cancer: A bioinformatics research. BMC Urol. 2022, 22, 114–118. [Google Scholar] [CrossRef] [PubMed]
  234. Yamashita, N.; Long, M.; Fushimi, A.; Yamamoto, M.; Hata, T.; Hagiwara, M.; Bhattacharya, A.; Hu, Q.; Wong, K.-K.; Liu, S.; et al. MUC1-C integrates activation of the IFN-γ pathway with suppression of the tumor immune microenvironment in triple-negative breast cancer. J. Immunother. Cancer 2021, 9, e002115. [Google Scholar] [CrossRef]
  235. Beckwith, D.M.; Cudic, M. Tumor-associated O-glycans of MUC1: Carriers of the glyco-code and targets for cancer vaccine design. Semin. Immunol. 2020, 47, 101389. [Google Scholar] [CrossRef]
  236. Kato, T.; Ujiie, H.; Hatanaka, K.C.; Nange, A.; Okumura, A.; Tsubame, K.; Naruchi, K.; Sato, M.; Kaga, K.; Matsuno, Y.; et al. A novel Tn antigen epitope-recognizing antibody for MUC1 predicts clinical outcome in patients with primary lung adenocarcinoma. Oncol. Lett. 2021, 21, 202. [Google Scholar] [CrossRef]
  237. Lüttges, J.; Zamboni, G.; Longnecker, D.; Klöppel, G. The immunohistochemical mucin expression pattern distinguishes different types of intraductal papillary mucinous neoplasms of the pancreas and determines their relationship to mucinous noncystic carcinoma and ductal adenocarcinoma. Am. J. Surg. Pathol. 2001, 25, 942–948. [Google Scholar] [CrossRef]
  238. Banerjee, S.; Mujumdar, N.; Dudeja, V.; Mackenzie, T.; Krosch, T.K.; Sangwan, V.; Vickers, S.M.; Saluja, A.K. MUC1c regulates cell survival in pancreatic cancer by preventing lysosomal permeabilization. PLoS ONE 2012, 7, e43020. [Google Scholar] [CrossRef]
  239. Scholz, C.; Heublein, S.; Lenhard, M.; Friese, K.; Mayr, D.; Jeschke, U. Glycodelin A is a prognostic marker to predict poor outcome in advanced stage ovarian cancer patients. BMC Res. Notes 2012, 5, 551. [Google Scholar] [CrossRef]
  240. Stojnev, S.; Ristic-Petrovic, A.; Velickovic, L.J.; Krstic, M.; Bogdanovic, D.; Khanh, D.T.; Ristic, A.; Conic, I.; Stefanovic, V. Prognostic significance of mucin expression in urothelial bladder cancer. Int. J. Clin. Exp. Pathol. 2014, 7, 4945–4958. [Google Scholar]
  241. Mirili, C.; Paydas, S.; Kilic, E.B.; Seydaoglu, G.; Ogul, A.; Gokcay, S.; Buyuksimsek, M.; Yetisir, A.E.; Karaalioglu, B.; Tohumcuoglu, M.; et al. Prognostic significance of EGFR, MUC1 and PD-L1 expressions in cases with triple negative breast cancer. J. BUON 2020, 25, 159–167. [Google Scholar] [PubMed]
  242. Wang, Q.; Zou, H.; Wang, Y.; Shang, J.; Yang, L.; Shen, J. CCR7-CCL21 axis promotes the cervical lymph node metastasis of tongue squamous cell carcinoma by up-regulating MUC1. J Craniomaxillofac Surg. 2021, 49, 562–569. [Google Scholar] [CrossRef]
  243. Kahkhaie, K.R.; Moaven, O.; Abbaszadegan, M.R.; Montazer, M.; Gholamin, M. Specific MUC1 splice variants are correlated with tumor progression in esophageal cancer. World J. Surg. 2014, 38, 2052–2057. [Google Scholar] [CrossRef] [PubMed]
  244. Ro, J.Y.; Grignon, D.J.; Troncoso, P.; Ayala, A.G. Mucin in prostatic adenocarcinoma. Semin. Diagn. Pathol. 1988, 5, 273–283. [Google Scholar] [PubMed]
  245. Castrillon, D.H.; Lee, K.R.; Nucci, M.R. Distinction between endometrial and endocervical adenocarcinoma: An immunohistochemical study. Int. J. Gynecol. Pathol. 2002, 21, 4–10. [Google Scholar] [CrossRef]
  246. Kong, C.S.; Beck, A.H.; Longacre, T.A. A panel of 3 markers including p16, ProExC, or HPV ISH is optimal for distinguishing between primary endometrial and endocervical adenocarcinomas. Am. J. Surg. Pathol. 2010, 34, 915–926. [Google Scholar] [CrossRef]
  247. Ronnett, B.M. Endocervical adenocarcinoma: Selected diagnostic challenges. Mod. Pathol. 2016, 29 (Suppl. S1), 12. [Google Scholar] [CrossRef]
  248. Fawzy, A.; Mohamed, M.R.; Ali, M.A.M.; Abd El-Magied, M.H.; Helal, A.M. Tissue CA125 and HE4 Gene Expression Levels Offer Superior Accuracy in Discriminating Benign from Malignant Pelvic Masses. Asian Pac. J. Cancer Prev. 2016, 17, 323–333. [Google Scholar] [CrossRef]
  249. McGuckin, M.A.; Ramm, L.E.; Joy, G.J.; Free, K.E.; Ward, B.C. Preoperative discrimination between ovarian carcinoma, non-ovarian gynecological malignancy and benign adnexal masses using serum levels of CA125 and the polymorphic epithelial mucin antigens CASA, OSA and MSA. Int. J. Gynecol. Cancer 1992, 2, 119–128. [Google Scholar] [CrossRef]
  250. Chen, X.; Zhou, H.; Chen, R.; He, J.; Wang, Y.; Huang, L.; Sun, L.; Duan, C.; Luo, X.; Yan, H. Development of a multimarker assay for differential diagnosis of benign and malignant pelvic masses. Clin. Chim. Acta 2015, 440, 57–63. [Google Scholar] [CrossRef]
  251. Xu, Y.; Zhang, L.; Hu, G. Potential application of alternatively glycosylated serum MUC1 and MUC5AC in gastric cancer diagnosis. Biologicals 2009, 37, 18–25. [Google Scholar] [CrossRef]
  252. Roșu, M.C.; Mihnea, P.D.; Ardelean, A.; Moldovan, S.D.; Popețiu, R.O.; Totolici, B.D. Clinical significance of tumor necrosis factor-alpha and carcinoembryonic antigen in gastric cancer. J. Med. Life 2022, 15, 4–6. [Google Scholar] [CrossRef]
  253. Bagaria, B.; Sood, S.; Sharma, R.; Lalwani, S. Comparative study of CEA and CA19-9 in esophageal, gastric and colon cancers individually and in combination (ROC curve analysis). Cancer Biol. Med. 2013, 10, 148–157. [Google Scholar] [CrossRef] [PubMed]
  254. Scarpa, M.; Noaro, G.; Saadeh, L.; Cavallin, F.; Cagol, M.; Alfieri, R.; Plebani, M.; Castoro, C. Esophageal cancer management: Preoperative CA19.9 and CEA serum levels may identify occult advanced adenocarcinoma. World J. Surg. 2015, 39, 424–432. [Google Scholar] [CrossRef] [PubMed]
  255. Manne, A.; Esnakula, A.; Abushahin, L.; Tsung, A. Understanding the Clinical Impact of MUC5AC Expression on Pancreatic Ductal Adenocarcinoma. Cancers 2021, 13, 3059. [Google Scholar] [CrossRef] [PubMed]
  256. Dong, Y.; Zhou, L.; Zhao, D.; Li, K.; Liu, Z.; Che, N.; Liu, H. MUC5AC enhances tumor heterogeneity in lung adenocarcinoma with mucin production and is associated with poor prognosis. Jpn. J. Clin. Oncol. 2020, 50, 701–711. [Google Scholar] [CrossRef]
  257. Liu, D.; Chang, C.; Gold, D.V.; Goldenberg, D.M. Identification of PAM4 (clivatuzumab)-reactive epitope on MUC5AC: A promising biomarker and therapeutic target for pancreatic cancer. Oncotarget 2015, 6, 4274–4285. [Google Scholar] [CrossRef]
  258. Javanbakht, M.; Akhavanmoghadam, J.; Talaei, A.J.; Aghyani, M.; Mozafari, M.; Khedmat, L.; Mohebbi, M. Differential expression of two genes Oct-4 and MUC5AC associates with poor outcome in patients with gastric cancer. Clin. Exp. Pharmacol. Physiol. 2017, 44, 1099–1105. [Google Scholar] [CrossRef]
  259. Pinto, R.; Carvalho, A.S.; Conze, T.; Magalhães, A.; Picco, G.; Burchell, J.M.; Taylor-Papadimitriou, J.; Reis, C.A.; Almeida, R.; Mandel, U.; et al. Identification of new cancer biomarkers based on aberrant mucin glycoforms by in situ proximity ligation. J. Cell Mol. Med. 2012, 16, 1474–1484. [Google Scholar] [CrossRef]
  260. Dikmen, Z.G.; Colak, A.; Dogan, P.; Tuncer, S.; Akbiyik, F. Diagnostic performances of CA125, HE4, and ROMA index in ovarian cancer. Eur. J. Gynaecol. Oncol. 2015, 36, 457–462. [Google Scholar]
  261. Ferraro, S.; Braga, F.; Lanzoni, M.; Boracchi, P.; Biganzoli, E.M.; Panteghini, M. Serum human epididymis protein 4 vs carbohydrate antigen 125 for ovarian cancer diagnosis: A systematic review. J. Clin. Pathol. 2013, 66, 273–281. [Google Scholar] [CrossRef] [PubMed]
  262. Pinho, S.S.; Reis, C.A. Glycosylation in cancer: Mechanisms and clinical implications. Nat. Rev. Cancer 2015, 15, 540–555. [Google Scholar] [CrossRef] [PubMed]
  263. Mungul, A.; Cooper, L.; Brockhausen, I.; Ryder, K.; Mandel, U.; Clausen, H.; Rughetti, A.; Miles, D.W.; Taylor-Papadimitriou, J.; Burchell, J.M. Sialylated core 1 based O-linked glycans enhance the growth rate of mammary carcinoma cells in MUC1 transgenic mice. Int. J. Oncol. 2004, 25, 937–943. [Google Scholar] [CrossRef]
  264. Paschos, K.A.; Canovas, D.; Bird, N.C. The role of cell adhesion molecules in the progression of colorectal cancer and the development of liver metastasis. Cell Signal 2009, 21, 665–674. [Google Scholar] [CrossRef]
  265. Sun, L.; Zhang, Y.; Li, W.; Zhang, J.; Zhang, Y. Mucin Glycans: A Target for Cancer Therapy. Molecules 2023, 28, 7033. [Google Scholar] [CrossRef]
  266. Pegram, M.D.; Borges, V.F.; Ibrahim, N.; Fuloria, J.; Shapiro, C.; Perez, S.; Wang, K.; Schaedli Stark, F.; Courtenay Luck, N. Phase I dose escalation pharmacokinetic assessment of intravenous humanized anti-MUC1 antibody AS1402 in patients with advanced breast cancer. Breast Cancer Res. 2009, 11, R73. [Google Scholar] [CrossRef] [PubMed]
  267. Ibrahim, N.K.; Yariz, K.O.; Bondarenko, I.; Manikhas, A.; Semiglazov, V.; Alyasova, A.; Komisarenko, V.; Shparyk, Y.; Murray, J.L.; Jones, D.; et al. Randomized phase II trial of letrozole plus anti-MUC1 antibody AS1402 in hormone receptor-positive locally advanced or metastatic breast cancer. Clin. Cancer Res. 2011, 17, 6822–6830. [Google Scholar] [CrossRef]
  268. Bose, M.; Sanders, A.; De, C.; Zhou, R.; Lala, P.; Shwartz, S.; Mitra, B.; Brouwer, C.; Mukherjee, P. Targeting tumor-associated MUC1 overcomes anoikis-resistance in pancreatic cancer. Transl. Res. 2023, 253, 41–56. [Google Scholar] [CrossRef]
  269. Gao, T.; Cen, Q.; Lei, H. A review on development of MUC1-based cancer vaccine. Biomed. Pharmacother. 2020, 132, 110888. [Google Scholar] [CrossRef]
  270. Julien, S.; Picco, G.; Sewell, R.; Vercoutter-Edouart, A.; Tarp, M.; Miles, D.; Clausen, H.; Taylor-Papadimitriou, J.; Burchell, J.M. Sialyl-Tn vaccine induces antibody-mediated tumour protection in a relevant murine model. Br. J. Cancer 2009, 100, 1746–1754. [Google Scholar] [CrossRef]
  271. Fang, F.; Ma, J.; NI, W.; Wang, F.; Sun, X.; Li, Y.; Li, Q.; Xie, F.; Wang, J.; Zhai, R.; et al. MUC1 and maltose-binding protein recombinant fusion protein combined with Bacillus Calmette-Guerin induces MUC1-specific and nonspecific anti-tumor immunity in mice. Mol. Med. Rep. 2014, 10, 1056–1064. [Google Scholar] [CrossRef] [PubMed]
  272. Zhang, Q.; Ni, W.; Zhao, X.; Wang, F.; Gao, Z.; Tai, G. Synergistic antitumor effects of Escherichia coli maltose binding protein and Bacillus Calmette-Guerin in a mouse lung carcinoma model. Immunol. Lett. 2011, 136, 108–113. [Google Scholar] [CrossRef] [PubMed]
  273. Li, D.; Li, H.; Zhang, P.; Wu, X.; Wei, H.; Wang, L.; Wan, M.; Deng, P.; Zhang, Y.; Wang, J.; et al. Heat shock fusion protein induces both specific and nonspecific anti-tumor immunity. Eur. J. Immunol. 2006, 36, 1324–1336. [Google Scholar] [CrossRef] [PubMed]
  274. Palmer, M.; Parker, J.; Modi, S.; Butts, C.; Smylie, M.; Meikle, A.; Kehoe, M.; MacLean, G.; Longenecker, M. Phase I study of the BLP25 (MUC1 peptide) liposomal vaccine for active specific immunotherapy in stage IIIB/IV non-small-cell lung cancer. Clin. Lung Cancer 2001, 3, 49–57. [Google Scholar] [CrossRef]
  275. North, S.A.; Graham, K.; Bodnar, D.; Venner, P. A pilot study of the liposomal MUC1 vaccine BLP25 in prostate specific antigen failures after radical prostatectomy. J. Urol. 2006, 176, 91–95. [Google Scholar] [CrossRef]
  276. Beatty, P.L.; van der Geest, R.; Hashash, J.G.; Kimura, T.; Gutkin, D.; Brand, R.E.; Finn, O.J. Immunobiology and immunosurveillance in patients with intraductal papillary mucinous neoplasms (IPMNs), premalignant precursors of pancreatic adenocarcinomas. Cancer Immunol. Immunother. 2016, 65, 771–778. [Google Scholar] [CrossRef]
  277. Messager, M.; Lefevre, J.H.; Pichot-Delahaye, V.; Souadka, A.; Piessen, G.; Mariette, C.; FREGAT working group—FRENCH. The impact of perioperative chemotherapy on survival in patients with gastric signet ring cell adenocarcinoma: A multicenter comparative study. Ann. Surg. 2011, 254, 684–693; discussion 693. [Google Scholar] [CrossRef]
  278. Khanvilkar, K.; Donovan, M.D.; Flanagan, D.R. Drug transfer through mucus. Adv. Drug Deliv. Rev. 2001, 48, 173–193. [Google Scholar] [CrossRef]
  279. Rao, C.V.; Janakiram, N.B.; Mohammed, A. Molecular Pathways: Mucins and Drug Delivery in Cancer. Clin. Cancer Res. 2017, 23, 1373–1378. [Google Scholar] [CrossRef]
  280. Okarvi, S.M. Peptide-based radiopharmaceuticals and cytotoxic conjugates: Potential tools against cancer. Cancer Treat. Rev. 2008, 34, 13–26. [Google Scholar] [CrossRef]
  281. Salouti, M.; Rajabi, H.; Babaei, M.H.; Rasaee, M.J. Breast tumor targeting with (99m)Tc-HYNIC-PR81 complex as a new biologic radiopharmaceutical. Nucl. Med. Biol. 2008, 35, 763–768. [Google Scholar] [CrossRef] [PubMed]
  282. Xie, Y.; Wang, Y.; Li, J.; Hang, Y.; Jaramillo, L.; Wehrkamp, C.J.; Phillippi, M.A.; Mohr, A.M.; Chen, Y.; Talmon, G.A.; et al. Cholangiocarcinoma therapy with nanoparticles that combine downregulation of MicroRNA-210 with inhibition of cancer cell invasiveness. Theranostics 2018, 8, 4305–4320. [Google Scholar] [CrossRef]
  283. Posey, A.D., Jr.; Schwab, R.D.; Boesteanu, A.C.; Steentoft, C.; Mandel, U.; Engels, B.; Stone, J.D.; Madsen, T.D.; Schreiber, K.; Haines, K.M.; et al. Engineered CAR T Cells Targeting the Cancer-Associated Tn-Glycoform of the Membrane Mucin MUC1 Control Adenocarcinoma. Immunity 2016, 44, 1444–1454. [Google Scholar] [CrossRef] [PubMed]
  284. Rabinovich, G.A.; Croci, D.O. Regulatory circuits mediated by lectin-glycan interactions in autoimmunity and cancer. Immunity 2012, 36, 322–335. [Google Scholar] [CrossRef] [PubMed]
  285. Ren, D.; Jia, L.; Li, Y.; Gong, Y.; Liu, C.; Zhang, X.; Wang, N.; Zhao, Y. ST6GalNAcII mediates the invasive properties of breast carcinoma through PI3K/Akt/NF-κB signaling pathway. IUBMB Life 2014, 66, 300–308. [Google Scholar] [CrossRef]
  286. Zhou, R.; Yazdanifar, M.; Roy, L.D.; Whilding, L.M.; Gavrill, A.; Maher, J.; Mukherjee, P. CAR T Cells Targeting the Tumor MUC1 Glycoprotein Reduce Triple-Negative Breast Cancer Growth. Front. Immunol. 2019, 10, 1149. [Google Scholar] [CrossRef]
  287. Mao, L.; Su, S.; Li, J.; Yu, S.; Gong, Y.; Chen, C.; Hu, Z.; Huang, X. Development of Engineered CAR T Cells Targeting Tumor-Associated Glycoforms of MUC1 for the Treatment of Intrahepatic Cholangiocarcinoma. J. Immunother. 2023, 46, 89–95. [Google Scholar] [CrossRef]
  288. Wang, S.; Yang, Y.; Ma, P.; Zha, Y.; Zhang, J.; Lei, A.; Li, N. CAR-macrophage: An extensive immune enhancer to fight cancer. EBioMedicine 2022, 76, 103873. [Google Scholar] [CrossRef]
  289. Niu, Z.; Chen, G.; Chang, W.; Sun, P.; Luo, Z.; Zhang, H.; Zhi, L.; Guo, C.; Chen, H.; Yin, M.; et al. Chimeric antigen receptor-modified macrophages trigger systemic anti-tumour immunity. J. Pathol. 2021, 253, 247–257. [Google Scholar] [CrossRef]
  290. Zhang, W.; Liu, L.; Su, H.; Liu, Q.; Shen, J.; Dai, H.; Zheng, W.; Lu, Y.; Zhang, W.; Bei, Y.; et al. Chimeric antigen receptor macrophage therapy for breast tumours mediated by targeting the tumour extracellular matrix. Br. J. Cancer 2019, 121, 837–845. [Google Scholar] [CrossRef]
Figure 1. Four types of mucin core in human intestinal tract. The mucin core is composed of N-acetylgalactosamine (GalNAc) with different variations in galactose (Gal) and N-acetylglucosamine (GlcNAc) linkage via ß1-3 or ß1-6 [13].
Figure 1. Four types of mucin core in human intestinal tract. The mucin core is composed of N-acetylgalactosamine (GalNAc) with different variations in galactose (Gal) and N-acetylglucosamine (GlcNAc) linkage via ß1-3 or ß1-6 [13].
Cimb 47 00406 g001
Figure 2. Downstream signaling of MUC1. The cytoplasmic tail of MUC1 interacts with various RTKs to activate PI3K/AKT/mTOR pathway, as well as MEK-REK pathways to promote cancer cell progression. The C-terminal of MUC1 also regulates the transcription of p53, β-catenin, STAT, and NF-kB to promote cancer cell migration and invasion. The red arrow indicates inhibition of transcription, while the blue arrow indicates promotion of transcription.
Figure 2. Downstream signaling of MUC1. The cytoplasmic tail of MUC1 interacts with various RTKs to activate PI3K/AKT/mTOR pathway, as well as MEK-REK pathways to promote cancer cell progression. The C-terminal of MUC1 also regulates the transcription of p53, β-catenin, STAT, and NF-kB to promote cancer cell migration and invasion. The red arrow indicates inhibition of transcription, while the blue arrow indicates promotion of transcription.
Cimb 47 00406 g002
Figure 3. A schematic representation of the hematogenous metastatic cascade. Neoplastic cell growth and angiogenesis supply blood and nutrients that create an optimal tumor microenvironment for cancer cell growth, intravasation, migration, extravasation, and colonization at distal organs [158].
Figure 3. A schematic representation of the hematogenous metastatic cascade. Neoplastic cell growth and angiogenesis supply blood and nutrients that create an optimal tumor microenvironment for cancer cell growth, intravasation, migration, extravasation, and colonization at distal organs [158].
Cimb 47 00406 g003
Table 1. The general features of transmembrane mucins.
Table 1. The general features of transmembrane mucins.
FeatureDescriptionMucin Examples
Structural CompositionExtracellular Domain: Rich in serine, threonine, and proline residues, heavily O-glycosylated. Forms a protective barrier against proteolytic enzymes, binds water, and forms gels.
SEA Domain: Undergoes autoproteolytic cleavage, resulting in an extracellular α-chain and a transmembrane/intracellular β-chain.
Extracellular Domain (membrane-bound):
MUC 1, 4, 16 [62,63,64,65]
Extracellular Domain (secreted): MUC 2, 6, 19 [1,9]
SEA Domain:
MUC 1, 12, 13, [1]
Intracellular Domain:
MUC 1, 3, 12, 17, [20]
ECF-like Domain:
MUC 4, 12, 13, 17, [1]
GlycosylationExtensively glycosylated to shield the protein backbone and maintain hydration. Glycosylation patterns vary, leading to different functional properties.MUC 1, 4, 16 [1]
Signaling RolesIntracellular tails participate in signaling pathways, influencing cellular responses. For example, the MUC1 tail can be phosphorylated, affecting cell adhesion and proliferation.MUC 1 and 4 [66]
Shedding and CleavageExtracellular domains can be shed from the cell surface, especially during inflammatory responses or in cancer. Shedding is mediated by proteases and modulates signaling and interactions.MUC 1, 4, 16 [20]
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

Chen, C.; Patel, A.; Demirkhanyan, L.; Gondi, C.S. The Role of Mucins in Cancer and Cancer Progression: A Comprehensive Review. Curr. Issues Mol. Biol. 2025, 47, 406. https://doi.org/10.3390/cimb47060406

AMA Style

Chen C, Patel A, Demirkhanyan L, Gondi CS. The Role of Mucins in Cancer and Cancer Progression: A Comprehensive Review. Current Issues in Molecular Biology. 2025; 47(6):406. https://doi.org/10.3390/cimb47060406

Chicago/Turabian Style

Chen, Clare, Ameena Patel, Lusine Demirkhanyan, and Christopher S. Gondi. 2025. "The Role of Mucins in Cancer and Cancer Progression: A Comprehensive Review" Current Issues in Molecular Biology 47, no. 6: 406. https://doi.org/10.3390/cimb47060406

APA Style

Chen, C., Patel, A., Demirkhanyan, L., & Gondi, C. S. (2025). The Role of Mucins in Cancer and Cancer Progression: A Comprehensive Review. Current Issues in Molecular Biology, 47(6), 406. https://doi.org/10.3390/cimb47060406

Article Metrics

Back to TopTop