International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Mucins, major components of mucus, are a family of high-molecular-weight proteins that are rich in proline, threonine and serine (PTS) residues and contain numerous O-glycosylation sites (1,2). Common mucins can be categorized into two types, secreted mucins and membrane-bound mucins, which form a selective protective barrier on epithelial surfaces and perform other physiological functions (3). In general, mucins consist of a core protein backbone and highly complex glycosylations (4). The glycan chains are linked to serine or threonine residues by O-glycosylation to form N-acetylgalactosamine (GalNAc) attached to the core polypeptide backbone (5). Additionally, mucins have numerous tandem repeats, especially around glycosylation sites, which allow mucins to carry massive glycan chains, resulting in their unique adhesive and protective functions (1,6,7). There are currently 22 known human genes encoding mucins, but there are likely additional mucin proteins waiting to be discovered (8-11).
Mucins are generally produced by epithelial cells to lubricate body cavities, where they serve as the first barrier against external attacks. Therefore, they are more abundant in the cavities of the respiratory tract, digestive system, eyes and lacrimal glands (12,13). Mucins also partly assume the role of maintaining the dynamic balance of mucus in terms of pH, oxygen concentration and hydration (14). Normally, mucin expression is relatively specific and restricted to specific tissues and organs at specific developmental stages. By contrast, aberrant expression of mucins is often considered to be associated with pathologic conditions such as inflammation and cancer (11,15,16).
The main challenges in treating cancer are its ability to invade surrounding tissues, metastasize remotely, and circumvent the surveillance of the host immune system through immune escape or suppression mechanisms (17-19). Since mucin glycosylation is diverse and most of its functions depend on its glycosylation and dimerization, changes in tumor-related glycosylation can greatly enhance tumor immune evasion or immunosuppression (20-22). Therefore, the abnormal expression of these mucins may be related to cancer, and they can even be used as tumor markers, as well as to assess patient prognosis and guide treatment. For example, researchers have combined toll-like receptor 7 (TLR7) agonists with abnormally glycosylated MUC1, which can produce a non-specific immune response and can enhance the body's cellular and humoral immunity against MUC1, thereby exerting an antitumor effect, which is used clinically for the treatment of recurrent breast cancer (23,24). Among mucin family members, MUC1 and MUC2 have been extensively studied in various cancers; by contrast, MUC6 remains relatively under-investigated despite its unique structural features, highly tissue-specific expression, and reported associations with several gastrointestinal (GI) and gynecologic tumors. Importantly, recent findings suggest that MUC6 polymorphisms and aberrant glycosylation patterns may affect tumor risk, immune interactions and prognosis. These characteristics point to MUC6 as a biologically and clinically relevant but underappreciated mucin, warranting a comprehensive review of its mechanisms, expression dynamics and translational potential.
The research prospects of the relationship between MUC and cancer are very broad. MUC1 is the most widely studied mucin in breast cancer, and the role of Mucin 6 (MUC6) has also emerged in recent studies (25,26). MUC6 belongs to the gel-forming mucin family and is the main component of gastric mucus. It is also widely expressed in other tissues, mainly in the hollow organs of the digestive system (including the stomach, gallbladder, duodenum, pancreas, bronchi and endocervix) (27-29). A recent large-scale tissue microarray analysis further demonstrated the expression profile of MUC6 across 119 tumor types, involving 15,412 tumor samples, highlighting its variable yet widespread presence beyond the GI tract (30). Studies have shown that the pyloric glands of the gastric antrum and mucopeptic cells of the neck zone also express MUC6 to a certain extent (24,31,32). Numerous studies have reported that aberrant expression of MUC6 is generally associated with GI diseases and cancer. For example, Lee et al (33) showed that the number of tandem repeat sequences of MUC6 can be used to predict gastric cancer risk. In addition, MUC6-TN (highlighted below as a cancer-associated mucin that has been considered a potential target for immunotherapy) can be used as a target for tumor vaccines, which may constitute a new direction for cancer therapy (34-37). However, the specific mechanisms of MUC6 in the tumor microenvironment (TME), for example, how MUC6 is involved in cancer development, invasion and metastasis, have not been fully elucidated. It remains unknown whether MUC6 acts synergistically or antagonistically with other mucins (for example, MUC1 and MUC5AC). These questions suggest that although the clinical promise of MUC6 is very high, numerous aspects still need to be explored. The clinical significance of MUC6 and its mechanism in human tumor development, especially in GI tumors, were reviewed with the aim of providing clinical diagnosis and targeted therapy for a variety of tumors.
The composition of mucins differs from that of most proteins in that their molecular weight is usually very large because of the presence of a variable number tandem repeat (VNTR) region, also known as minisatellite RNA (38). These repeats are rich in PTS; some are extremely similar, and some are identical (39-41). The repetitive sequences are arranged densely in an ordered array and are relatively concentrated (42). After translation, further folding and modification are needed, and glycosylation is the primary modification of mucins. Glycoproteins are transported to the Golgi apparatus via COP II to accomplish more complex biochemical reactions, such as N-glycosylation (43-46). Normal N-glycosylated proteins subsequently undergo further O-glycosylation and dimerization (43,47,48). O-glycosylation depends on the length and number of PTS repeats, and owing to gene polymorphisms, O-glycosylation is different for each mucin and even the same mucin in different contexts (43,49).
MUC6 belongs to the family of gel-forming secreted mucins (including MUC2, MUC5AC and MUC5B), which have high sequence similarity (50,51). These genes are concentrated at p15.5 in the recombination enrichment region of ~400 kb in the sub-telomeric region of chromosome 11, between H-ras and IGF2 (9). However, the transcription direction of MUC6 is opposite to that of other family members (52) (Fig. 1). Its gene is ~28 kb in length and includes 35 exons and 34 introns (41). A recent study using long-read sequencing confirmed that the exon 31 tandem repeat region of MUC6 encodes highly glycosylated PTS sequences, with significant copy number variation among individuals-explaining observed polymorphisms in gene length and protein isoforms (42). Furthermore, structural comparisons revealed that MUC6 lacks conserved domains (D4, B and C) found in other gel-forming mucins such as MUC2, causing it to adopt a distinct polymerization behavior (53). These genomic characteristics likely underpin MUC6's unique functional role in mucosal defense.
The core length of the tandem repeat sequence is ~169 amino acids, and the corresponding gene length is between 8 and 13.5 kb. The uncertainty of the tandem repeat sequence in exon 31 causes changes in the length of the MUC6 gene (54,55). The center of MUC6 is rich in serine and threonine residues, followed by the CK region, and the highly glycosylated central domain is encoded by tandem repeats. The N-terminal domain of MUC6 is similar to that of von Willebrand factor (vWF). Both have a D-like domain and high cysteine content. vWF is highly similar to MUC2, but unlike other homologous MUC genes, some exons encoding the D4, B and C regions were lost during the evolution of MUC6 (53,56-58). There is a cystine structural motif in its short C-segment region, and a class D domain or a cystine structural motif is very important for the dimerization of mucins (52,56,59).
Initially, MUC6 was discovered in a gastric cDNA library, and it is most abundantly expressed in the stomach (52,60). Together with MUC5AC, it participates in the formation of the mucus barrier on the gastric surface (31,61). Its side chain can interact with bicarbonate to resist external gastric acid and pepsin. It also helps protect the gallbladder and bile duct from being corroded by hydrophobic bile acids. The pancreatic tubules need to resist the action of a variety of digestive enzymes and trypsin, and the seminal vesicles are also rich in pepsin (31). The side chain structures of MUC6 and other mucins in the cervical and endometrial glands can interact with certain bacterial structures to prevent infection (31). These findings indicate that under normal physiological conditions, the expression of MUC6 in epithelial cells can protect the human body from invasion and damage caused by digestive enzymes and bacteria (29,31).
Previous studies have quantified the structural and rheological contributions of MUC6 to the mucus barrier. In humans, the gastric mucus layer-composed primarily of MUC5AC and MUC6-reaches thicknesses of 0.2-0.6 mm, forming a bi-layered gel essential for buffering gastric acid and limiting pathogen access (62). Additionally, the lectin trefoil factor family 2 (TFF2), co-secreted with MUC6, cross-links MUC6 via GlcNAc-specific interactions, enhancing mucus viscosity and barrier stability in vivo and in vitro (63). Mechanically, enhanced mucus viscoelastic behavior under shear stress underscores the importance of mucin glycoprotein architecture in determining barrier resilience and microenvironment homeostasis.
The role of mucin in cancer is becoming clearer, where it generally indicates an improved outcome. Its expression in numerous cancers, including gallbladder, cholangiocarcinoma, lung, colorectal, prostate, breast, gastric, pancreatic and endometrial cancer, is different from that in normal tissues, potentially improving the prognosis and reducing metastasis. Although the relationships between MUC6 and various clinical indicators of patients with cancer, such as sex, age, stage and depth of invasion, are not clear, the positive role of MUC6 in cancer is obvious (Table I). It is known that most of the functions of mucin are inseparable from glycosylation, and some mucin structures in tumors are produced by obstructed O-glycosylation, such as TN, sialyl-TN or TF antigens (2,34). Among them, MUC6-TN, which has different antigenicity than other mucins, is expressed in the MCF7 breast cancer cell line. Further research revealed that the formation of MUC6-TN requires the participation of polypeptide N-acetyl galactosamine transferases (ppGalNAc-Ts). Although the prepared MUC6-TN partially eliminated the ability of MUC6 to promote the proliferation of specific T cells, MUC6-TN can stimulate T cells to preferentially produce IL-17. MUC6-TN can be recognized by specific antibodies against MUC6 and TN, thereby inducing the production of corresponding IgG antigens in the body, and the specific TN-induced IgGs can recognize the TN antigen of tumor cells to activate antitumor immunity (34,35,37). Researchers have also reported that the participation of multiple ppGalNAc-Ts (-T1, -T2 and -T7) may act together to produce MUC6-TN with a relatively high TN concentration, thereby improving the quality of antitumor vaccines (35).
In tissues that normally express MUC6, the loss of MUC6 is often accompanied by increased tumor invasiveness. In the early stages of pancreatic intraepithelial neoplasms (PanINs), the level of MUC6, especially in gastric PanIN, is elevated, but MUC6 expression is lost during progression to invasive ductal adenocarcinoma, especially in the final stage (28,64,65). It has been revealed that there is almost no difference between the levels of the MUC6 N-terminus (MUC6-N) and the MUC6 C-terminus (MUC6-C) in breast cancer cells, which are inherently weakly invasive (56). In pancreatic cancer and colon cancer cell lines, expression of the MUC6 domain can effectively inhibit their invasiveness; however, its effect on poorly differentiated pancreatic cancer is less pronounced than that on moderately differentiated colon cancer. This may also be due to the faster adhesion of pancreatic cancer and its weaker invasiveness. It has been identified that the expression of MUC6-N and MUC6-C can significantly reduce the adhesion of pancreatic cancer cells to type I and IV collagen, but there is no significant difference in their adhesion to fibronectin and laminin (56). In addition, in colon cancer cells, the expression of MUC6-N more significantly affects the adhesion of cancer cells to the four aforementioned proteins, but the expression of MUC6-C has a lesser effect on adhesion. In gallbladder cancer, the downregulation of MUC6 may be necessary for cancer metastasis. Shortly, the loss of MUC6 often indicates an increase in tumor aggressiveness and cancer metastasis. This may explain why the expression of MUC6 in most cancers predicts an improved outcome (36,56,66). In the following chapters, the results of research on the roles of MUC6 in various tumors are separately introduced (part of the mechanism is schematically shown in Fig. 2).
Although the incidence and mortality of gastric cancer have decreased in recent years, it is still among the leading causes of cancer-related death worldwide, with a mortality rate of ~75% and an estimated 5-year survival rate of only 20% (67). According to GLOBOCAN cancer statistics, there were >1 million new gastric cancer cases and 769,000 gastric cancer-related deaths in 2020. It is the fifth most common cancer and the fourth leading cause of cancer-related death worldwide (68). As a global health problem that threatens human safety, all aspects of gastric cancer should be studied to promote an early clinical diagnosis, early treatment and improved prognosis (69,70). Helicobacter pylori (H. pylori) infection is a recognized risk factor for gastric cancer, and it has also been identified as a human carcinogen by the IARC (71). Studies have shown that shorter VNTRs in MUC6 promote infection by H. pylori. This is due to the presence of α-1,4-linked N-acetylglucosamine (α-1,4-GlcNAc) in the variable region of MUC6, which has antibacterial effects against H. pylori. Accordingly, shorter VNTRs lead to less α-1,4-GlcNAc, and the antibacterial effect against H. pylori is weaker, increasing the risk of infection and subsequent development of gastric cancer (72,73). This theory is also consistent with the distribution of H. pylori (74,75).
The relationships between MUC6 and patient sex, tumor location, lymphatic invasion, clinical stage, metastasis and histological classification in gastric cancer remain unclear, and a consensus has not yet been reached. Certain studies indicate that MUC6 has no significant relationship with these clinicopathological characteristics (76-83). It has been also indicated that the loss of mucin predicts cell degeneration, especially intestinal metaplasia (precancerous lesions preceding gastric cancer) (84). Therefore, MUC6 is involved in the development of malignant lesions from gastric epithelial cells. The downregulation of MUC6 is correlated with the progression of gastric cancer and a poor prognosis, supporting the aforementioned theory (85). However, a study by Oosterlinck et al (86) revealed that MUC6 abundance was associated with a more favorable prognostic outcome in patients with gastric adenocarcinoma. In addition, MUC6 is a promising diagnostic marker for endocervical gastric-type adenocarcinoma (GAS) (87). Furthermore, a population-based histopathological analysis revealed notable regional differences in MUC6 expression between European and Latin American patients with gastric cancer, suggesting potential ethnicity-related biological variability (88). These data collectively underscore the diagnostic and prognostic potential of MUC6, though the precise associations with clinical indicators remain to be definitively established.
Notably, a new breakthrough in the study of MUC6 gene polymorphisms was previously reported (89). Studies of the relationship between MUC6 gene polymorphisms and gastric cancer have concluded that smaller alleles increase the risk of gastric cancer, irrespective of its histological classification (89). Subsequent research revealed that shorter tandem repeats may inhibit the expression of MUC6, which is ultimately associated with an increased risk of gastric cancer (54). Overall, the length and genetic polymorphism of MUC6 can be used to predict the risk of gastric cancer, but whether the expression level of MUC6 in gastric cancer is related to multiple clinical indicators remains uncertain. More data are needed to verify the relationship between MUC6 and gastric cancer, and the relationship between polymorphisms of other minisatellites of MUC6 and the risk of gastric cancer also needs further verification.
Colorectal cancer is one of the most common cancers in the world, and its mortality is closely related to the failure of early screening (90,91). In 2020, a total of 1,931,590 cases of colorectal cancer were diagnosed worldwide, and 935,173 patients succumbed to the disease. The incidence of colon cancer varies greatly worldwide. Europe, Australia/New Zealand and North America have the highest incidences of colon cancer. The incidence of colon cancer in the most affected regions is 9-fold greater than that in the least affected regions (68,92).
MUC6 is generally not expressed in the colorectal mucosa, but some colorectal cancers are positive for MUC6 expression, and when most cancer cells in a colorectal tumor express MUC6, the prognosis of the patient tends to be improved. Therefore, MUC6 is recommended as a molecular marker for prospective risk assessment of patients with colorectal cancer (56). Studies have also shown that the expression of MUC6 is significantly correlated with a number of clinical indicators, such as whether the tumor is on the left or right, pathological type, tumor infiltrating lymphocytes, p53 expression, mismatch repair and MLH1 methylation, and other factors (93). In recent years, a new concept called the serrated pathway, which includes a series of molecular events, such as BRAF mutations, CpG island methylation, and microsatellite instability, has been proposed to explain the occurrence and development of colorectal cancer (10,14). This pathway starts with a variety of polyps, such as traditional serrated adenomas; sessile serrated adenomas, also termed sessile serrated polyps (SSAs/Ps); SSA/Ps with dysplasia (SSAs/PDs); and hyperplastic polyps (HPPs). Among them, HPPs are different from the other types and are considered benign. It has been noted that MUC6 may be involved in the serrated pathway of colorectal cancer (94). Compared with that of the HPPs, its level is significantly greater in SSA/P and SSA/PD. Although using this indicator to differentiate HPPs, SSA/P and SSA/PD is not ideal, it still supports the idea that HPPs may be a type of SSA. Although the relationship between MUC6 and various clinical indicators of colorectal cancer and polyps (including sex, age and size) is inconclusive (95,96), the results of these studies reached a consensus in some aspects. First, the expression of MUC6 is significantly increased in colon polyps, especially adenomatous polyps. Second, the physiological difference between the left colon and the right colon causes MUC6 expression in the serrated polyps of the right colon to be significantly greater than that in the serrated polyps of the left colon (95,97). As observed in gastric cancer, genetic polymorphisms of MUC6 may be associated with susceptibility to colon cancer (54). Similar to gastric cancer, having a shorter allele also indicates a higher probability of rectal cancer. However, unlike gastric cancer, MUC6 also has significant sex differences, whereby men generally have shorter alleles. However, that study was conducted in ethnic Asians, and this result is consistent with the fact that the prevalence of rectal cancer is higher in men than in women in Asians. Although the reason why the length of MUC6 is related to the incidence of cancer is not clear, there are more transcription factor-binding sites in the fifth tandem repeat of MUC6, and perhaps the underlying mechanism may be related to the regulation of gene expression (54,98). In general, genetic polymorphisms of MUC6 are also related to susceptibility to colon cancer, and MUC6 expression is significantly increased in adenoma, but its relationship with other clinical indicators is not clear. Research on the genetic polymorphisms of colon cancer is promising, and global multi-center big data studies are needed to verify the established conclusions.
Biliary tract cancer, including gallbladder cancer and cholangiocarcinoma, is caused by the malignant transformation of biliary epithelial cells. Biliary tract cancer is characterized by a poor prognosis and early dissemination. Moreover, most patients are diagnosed with late-stage disease, which results in a dismal 5-year survival rate of <10% (99). Therefore, it is particularly important to correctly evaluate its prognosis and study the factors affecting its early dissemination.
Studies have shown that MUC6 expression is correlated with reduced lymph node metastasis in cholangiocarcinoma, which may be related to the co-expression of TFF2. When TFF2 is co-expressed and combines with MUC6, they form a complex, increase the viscosity of the mucin around the tumor mass, and ultimately prevent the tumor from spreading. It has been revealed that the downregulation of MUC6 is necessary for the metastasis of cholangiocarcinoma (100) (Fig. 2). The expression of MUC6 in cholangiocarcinoma is related to the histological type and extent of metastasis (101,102). MUC6 is more abundantly expressed in the pyloric glands (31), and pseudopyloric metaplasia is closely related to the occurrence of gallbladder cancer. Therefore, MUC6 may help us study the occurrence and classification of gallbladder cancer (103-106). In moderately and well-differentiated gallbladder cancers, the upregulation of MUC6 indicates an improved prognosis, and after the cancer progresses further, the dedifferentiation of the cancer cells results in the disappearance of the antimetastatic effect of MUC6 (107). The level of MUC6 may be used as a new indicator for the classification of biliary tract cancer and may even be necessary for its metastasis. The expression level of MUC6 may be an indicator of the prognosis of patients with biliary tract cancer, but the mechanism is not clear, and further research is needed.
As a common malignant tumor of the digestive tract with the worst prognosis, pancreatic cancer usually exhibits symptoms only in the late stage (108). In 2018, 458,918 new cases of pancreatic cancer were diagnosed worldwide, and 432,242 patients succumbed to the disease (109). Despite the continuous advancements in pancreatic cancer detection and treatment technology, the five-year survival rate is still only ~9% (68,109).
Under normal physiological conditions, MUC6 is most abundantly expressed in the intercalated ducts of the pancreas, followed by occasional detection in centroacinar cells and almost no expression in the interlobular ducts (56). Studies have suggested that MUC6 can inhibit the growth and development of pancreatic cancer. This is mainly because long non-coding RNA (lncRNA) cancer susceptibility candidate 2 (CASC2) indirectly regulates the expression of MUC6 by regulating microRNA (miR)-24, thereby exerting a tumor suppressor effect (110-112). It was also reported that the NF-κB and Sp families of genes, which are related to the occurrence of pancreatic cancer, may regulate the expression of MUC6 (113-117). In addition, it was proposed that PDX1 can induce MUC6 transcription as an important factor in the foregut endoderm cell lineage. Therefore, the role of MUC6 in the early development of pancreatic cancer also has a certain physiological basis (113,118) (Fig. 3). Given the poor prognosis of patients with pancreatic tumors, early diagnosis is the only means to effectively improve their outcomes. A study confirmed the feasibility of semiquantitative immunohistochemical (IHC) evaluation of three MUC genes (MUC3, MUC5AC and MUC6) as diagnostic markers for pancreatic cancer, which can effectively distinguish pancreatic cancer, chronic pancreatitis and normal pancreas. However, more data and multicenter studies are needed to verify the feasibility of this approach (119). Interestingly, there is no correlation between MUC6 and pancreatic invasive ductal carcinoma, possibly because MUC6 is related only to adenocarcinoma (120). MUC6 cannot only play a clinical role but also be used to study the development of tumors. It has been shown that MUUC6 is expressed mainly in central glandular secretions and that MUC6 expression may indicate the progression of a pancreatic cystic tumor to the central gland (121). The CASC2 (lncRNA)-miR-24 axis and PDX1 can both regulate MUC6 levels, thereby inhibiting the occurrence and development of pancreatic cancer. Accordingly, MUC6 may be an effective marker for early screening of pancreatic cancer together with other mucins and may help to distinguish different pathological types of pancreatic cancer. Nevertheless, more data and samples from multiple centers are needed to verify the existing conclusions.
The incidence of breast cancer ranks first among female malignant tumors and has surpassed that of lung cancer to become the world's most common cancer, with 2,261,419 new cases of breast cancer diagnosed in 2020 (68).
MUC6 is not normally expressed in breast tissue but is expressed in non-cancerous tissues surrounding breast cancer (122). Some studies have shown that the expression of MUC6 is related to the grade and degree of lymphatic invasion of invasive breast cancer, which is related to prognosis (36,66,123-125), but other studies have reached different conclusions. Notably, the standard used to judge positive MUC6 expression has improved (>30% of cancer cells expressing MUC6 are considered positive), and subsequent studies have confirmed this theory. Perhaps the formation of a mucin barrier by MUC6 requires a sufficient amount of MUC6, which may also exert other biological effects. Therefore, the functional relationship between MUC6 and cancer is more pronounced at higher expression levels (66,123). There are numerous examples of mucin expression being regulated by steroid hormones, and MUC6 is also upregulated by hormones in vitro. The abnormal expression of MUC6 in breast cancer may therefore be related to abnormal levels of steroid hormones during the development of breast cancer (126). TN (α-GalNAc-O-Ser/Thr) is a specific tumor antigen that is found mainly on mucins, and MUC6 was found to be a very favorable substrate for ppGalNAc-ts (the enzyme related to the synthesis of the TN antigen) in breast cancer and a relatively bad substrate for 1β3Gal-T (the enzyme that converts the MUC6-TN glycopeptide into MUC6-TF). This leads to the preferential enrichment of MUC6-TN rather than MUC1-TN in breast cancer cells (36,37). The TN antigen has numerous positive effects on cancer; thus, the relationship between MUC6 and breast cancer is very promising.
Lung cancer is the cancer with the fastest increase in morbidity and mortality in recent years. As the second most common cancer in the world, there were 2,206,771 new cases of lung cancer in 2020, affecting twice as numerous men as women, and the number of deaths was 1,796,144 (68).
The expression of MUC6 has been increasingly recognized for its value in the classification and differential diagnosis of lung cancer subtypes. Specifically, subtype-specific expression patterns of MUC6 have been reported in lung adenocarcinoma, with immunoreactivity scores varying significantly between mucinous and non-mucinous forms (127). This aligns with earlier findings that MUC6 expression distinguishes bronchioloalveolar carcinoma from invasive mucinous adenocarcinoma and is often associated with a more favorable prognosis (119,128-131).
Moreover, MUC6 has shown diagnostic utility when combined with lineage-specific markers such as CDX2 and SATB2 in differentiating primary pulmonary mucinous adenocarcinoma from metastatic colorectal carcinoma (132). However, inconsistencies remain-some studies have found MUC6 expression to be positively correlated with lymph node metastasis, contradicting its previously reported protective role (133). These discrepancies underscore the need for larger, standardized studies to clarify the clinical relevance of MUC6 in lung cancer.
In 2020, there were 1,414,259 new cases of prostate cancer and 375,304 related deaths. It is the fourth most common cancer in the world and the second most common cancer in men, ranking fifth in terms of cancer-related deaths in men (68,134).
False-positive biopsy-based diagnosis of prostate cancer is often due to the presence of seminal vesicle-ejaculatory ducts. Studies have shown that MUC6 is not expressed in prostate cancer or normal prostate tissues but is strongly expressed in seminal vesicle-ejaculatory duct tissues. Therefore, MUC6 can provide strong evidence for distinguishing prostate cancer from seminal vesicles and ejaculatory ducts, greatly reducing the false-positive rate of prostate biopsy (60,135). Previous studies have indicated that MUC6 expression indicates an improved prognosis and is strongly correlated with the occurrence of prostate cancer (136,137). MUC6 expression may not directly lead to the occurrence of prostate cancer, but its presence may assist in increasing the detection rate of prostate cancer or have other indirect effects. More data are needed to verify the relationship between MUC6 and prostate cancer.
Endometrial cancer is a group of epithelial malignant tumors that occur in the endometria of perimenopausal and post-menopausal women (138,139). Endometrial cancer causes ~76,000 deaths among women worldwide annually (140,141). Cervical GAS, also known as minimally offset mucinous adenocarcinoma or malignant adenoma, is characterized by HIK1083 and/or MUC6 immunopositivity and is more aggressive than ordinary endometrial cancer. The expression of MUC6 is focal, thus the absence of MUC6 on a sub-biopsy section does not rule out a diagnosis (88,142). A 2023 meta-analysis further supported this diagnostic approach, confirming that MUC6, in combination with HIK1083, provides high specificity in identifying GAS, reinforcing its utility as a reliable IHC marker (143). The aforementioned steroid hormone regulation of mucin has also been mentioned in endometrial cancer, but the data are not complete (144). More in-depth research on endometrial cancer is needed since the current data are insufficient for existing conclusions to be translated into clinical applications, but there are broad research prospects.
Cancer immunotherapy focuses on the body's immune response to cancer cells without causing damage to normal cells. This requires cancer-specific antigens to induce a specific immune response (34), and only peptide modifications can be identified by the immune system (for example, mutations, glycosylation and phosphorylation) (145,146). MUC6, along with other mucins (for example, MUC1 and MUC5AC), all exhibit important biological functions, such as glycosylation, masking tumor antigens, and evading immune recognition (147-149). Glycosylations allow these mucins to form structural barriers that mask tumor antigens on the cell surface and reduce recognition by immune cells (for example, T cells and natural killer cells), thus facilitating tumor immune escape (20). MUC6, as a secreted mucin, impedes the penetration of immune cells, whereas MUC1, as a membrane-bound mucin, has a glycosylation that not only masks antigens but also participates in the process of immunosuppression through intracellular signaling (150,151). Moreover, the aberrant secretion of MUC5AC further aggravated the formation of the mucus barrier in the TME, limiting immune cell infiltration and antitumor activity (152). In terms of expression pattern, MUC6 is more commonly expressed in tissues such as the stomach and pancreas, whereas MUC1 and MUC5AC are more widely expressed and tend to be differentially expressed in different cancer types. Accordingly, highly glycosylated and relatively specific MUC6 is an attractive target. Abnormal mucin expression can expose core proteins (2). This makes MUC6 a new cancer immunotherapy target, and TN-related vaccines, such as α-N-acetylgalactosamine-O-serine/threonine conjugate vaccines, have shown promise (153). The ppGalNAc-T enzyme is a glycosyltransferase that can transfer GalNAc to a specific MUC6 residue and connect it with a serine or threonine to form a -TN antigen. There are numerous isozymes in the ppGalNAc-T family and choosing the one with the highest glycosylation efficiency has become a research hotspot. It was aforementioned that MUC6 is a favorable substrate for ppGalNAc-Ts and has a low reactivity to 1β3Gal-T, which leads to its enrichment in MCF7 breast cancer cells (36,153). The latter can also induce immune responses in mice, leading to the production of IgGs that can recognize TN antigens to attack tumor cells (36,153).
On this basis, Freire et al (35) glycosylated MUC6 with different combinations of ppGalNAc-Ts to obtain various MUC6: Tn glycoproteins and injected them into mice. Only MUC6: Tn(T2) and MUC6: Tn (T1 + T2 + T7) were found to induce specific antibodies capable of recognizing tumor cells, suggesting that the linkage of GalNAc to the MUC6 protein alters its immunogenicity and that there is no significant correlation between the number of GalNAc residues and the antigenicity of MUC6 or TN (35). Glycosylation may mask dominant T-cell epitopes, thereby reducing recognition, which may explain why MUC6 occasionally stimulates IL-2 to greater effect (35). In conclusion, the TN antigen of MUC6-TN affects its immunogenicity against B and T cells; thus, adjusting the combination of ppGalNAc-T is necessary when designing a vaccine to ensure its antigenicity (Table II).
Supporting this immunomodulatory role of MUC6 in vivo, it has been recently demonstrated that Muc6 upregulation in the mouse intestine after low-dose radiation exposure leads to mucosal immune alterations resembling inflammatory bowel disease (IBD) pathogenesis (154). Furthermore, MUC6 IHC staining has been proposed as a useful adjunct for histologic assessment in Crohn's disease, potentially identifying gastric-type metaplasia and related immune phenotypes (155). These observations collectively reinforce the concept that MUC6 is not only a structural mucin but also an active participant in regulating mucosal immunity-implications that extend to both autoimmunity and cancer immunotherapy.
In clinical oncology, MUC6 has emerged as a tissue-restricted marker with increasing relevance for both tumor classification and differential diagnosis, particularly in the GI tract (156,157). IHC detection remains the most widely used and practical method for assessing MUC6 expression in formalin-fixed tissues (158). Multiple tissue-based studies, including large-scale tumor microarray profiling, have validated MUC6 IHC in various cancers ranging from gastric and pancreatic to endometrial and breast carcinomas (159-161).
Recent experimental evidence suggests that altered glycosylation patterns of MUC6, particularly truncated Tn antigens, may offer enhanced specificity for identifying neoplastic transformation. A 2024 murine study demonstrated that loss of MUC6 leads to impaired O-glycosylation, enrichment of mannose-rich glycans, and subsequent tumor-promoting changes via MAPK signaling. These modifications were effectively detected by lectin-based assays and glycoform-sensitive IHC, providing a novel diagnostic approach (5).
Selection of wild-type vs. glycoform-specific MUC6 detection should be guided by tumor type and diagnostic goal. In pancreatic and biliary neoplasms, wild-type MUC6 is a useful surrogate marker for pyloric-type differentiation (63,162). By contrast, in breast, lung, or serrated colorectal lesions, MUC6 expression is ectopic and often associated with glycan alterations (163,164). In such settings, detection of Tn-modified MUC6 may better reflect tumor phenotype and immunogenicity (165).
Of particular interest is the potential co-expression of MUC6 with Claudin 18.2, a tight junction protein that has recently emerged as a promising therapeutic target across multiple GI cancers. A recent immunopathological study reported that over 57% of IBD-associated small bowel adenocarcinomas co-express MUC6 and Claudin 18.2, suggesting a shared gastric lineage phenotype (166). Similarly, Yang et al (87) demonstrated in GAS that dual staining for MUC6 and Claudin 18.2 significantly improves diagnostic sensitivity and subclassification.
Despite these advances, standardization of antibody clones, scoring criteria, and threshold definitions remains lacking. Furthermore, few large-scale prospective studies have evaluated MUC6 performance in clinical cohorts or in comparison with other epithelial markers such as MUC5AC, MUC1, or CDX2 (30). Integration of glycoform-specific detection tools-such as lectins or monoclonal antibodies targeting the Tn epitope-remains in early translational stages and requires further validation (167,168).
Nonetheless, the spatially restricted, differentiation-dependent nature of MUC6, combined with its potential to reflect glycosylation-linked immune signatures, positions it as a promising companion marker for tumor profiling. In selected cancers, especially those with gastric or pyloric features, MUC6 may also contribute to personalized immunotherapy or stratification for Claudin 18.2-targeted treatment.
The present study reviewed the current research status of MUC6, focusing on its biological roles in cancer, especially GI tumors, and the possibility of its clinical application. The expression of MUC6 has strong specificity. Under normal physiological conditions, it is expressed only in specific tissues and organs during a specific developmental stage. Therefore, abnormal MUC6 expression usually indicates disease (11,15,16). In most studies, the presence of MUC6 indicates an improved outcome. MUC6 can enhance the adhesion of tissues, therefore the erosion and spread of tumor cells become more difficult, thereby improving the prognosis (56,66). MUC6 can protect the cavities of the digestive system and other organs and tissues from attack by digestive enzymes and bacteria. Tumorigenesis can also be induced by repeated stimulation and inflammation due to damage to tissues and organs. Therefore, the correct expression of MUC6 is essential for preventing the occurrence of cancer. However, while the expression level of MUC6 does not appear to be a suitable choice for evaluating various clinical indicators of cancer, its genetic polymorphisms can be used as important factors in cancer risk assessment. The length of the tandem repeats in MUC6 is significantly related to susceptibility to gastric and colorectal cancer. Accordingly, the possible relationship between MUC6 gene polymorphisms and susceptibility to other cancers merits further investigation (54,98). MUC6 is also used to distinguish false positives in prostate cancer biopsies (60,135). Moreover, MUC6-TN may provide a new direction for cancer treatment, and its practicality may surpass that of the widely recognized MUC1 to become a new target for cancer vaccines (34,35,37).
In the future, genetic polymorphisms of MUC6, duplication of VNRT in small satellites and susceptibility to cancer have broad research prospects, and they may become important indicators for early cancer prevention (96,98). The relationship between colorectal serrated polyps and MUC6 also appears to be a direction worthy of further research. MUC6 is not expressed by colon tissue under normal physiological conditions, and its appearance is often accompanied by increased cancer risk or activation of the serrated pathway, which may serve as an effective indicator for colorectal cancer risk assessment (54,56). Because pancreatic disease itself is difficult to diagnose and distinguish, while pancreatic cancer has almost no symptoms in the early stage, effective indicators that can be used for early screening and differential diagnosis are urgently needed, and MUC6 has significant potential in this regard (119). The promising performance of MUC6 in cancer immunotherapy suggests that it has broad development prospects. Overall, the roles of MUC6 in all aspects of cancer biology and therapy require more in-depth research.
Not applicable.
PX contributed to the conception and design of the work, reviewed and revised the manuscript critically for important intellectual content, and approved the final version. XL drafted the initial version of the manuscript and participated in its revision. XF contributed to the interpretation of content and manuscript editing. YX and PX jointly reviewed and revised the manuscript for intellectual content. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
PTS |
proline, threonine and serine |
|
VNTR |
variable number tandem repeat region |
|
TLR7 |
Toll-like receptor 7 |
|
vWF |
von Willebrand factor |
|
ppGalNAc-Ts |
polypeptide N-acetylgalactosamine transferases |
|
α-1,4-GlcNAc |
α-1,4-linked N-acetylglucosamine |
|
SSA/P |
sessile serrated adenomas also termed sessile serrated polyps |
|
SSA/PD |
SSA/Ps with dysplasia |
|
HPPs |
hyperplastic polyps |
|
TFF2 |
trefoil factor family 2 |
|
lncRNA |
long non-coding RNA |
|
CASC2 |
cancer susceptibility candidate 2 |
Not applicable.
No funding was received.
|
Gendler SJ and Spicer AP: Epithelial mucin genes. Annu Rev Physiol. 57:607–634. 1995. View Article : Google Scholar : PubMed/NCBI | |
|
Hollingsworth MA and Swanson BJ: Mucins in cancer: Protection and control of the cell surface. Nat Rev Cancer. 4:45–60. 2004. View Article : Google Scholar | |
|
Demouveaux B, Gouyer V, Gottrand F, Narita T and Desseyn JL: Gel-forming mucin interactome drives mucus viscoelasticity. Adv Colloid Interface Sci. 252:69–82. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Kohout VR, Wardzala CL and Kramer JR: mirror image mucins and thio mucins with tunable biodegradation. J Am Chem Soc. 145:16573–16583. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Arai J, Hayakawa Y, Tateno H, Murakami K, Hayashi T, Hata M, Matsushita Y, Kinoshita H, Abe S, Kurokawa K, et al: Impaired glycosylation of gastric mucins drives gastric tumorigenesis and serves as a novel therapeutic target. Gastroenterology. 167:505–521 e19. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Perez-Vilar J and Hill RL: The structure and assembly of secreted mucins. J Biol Chem. 274:31751–31754. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Strous GJ and Dekker J: Mucin-type glycoproteins. Crit Rev Biochem Mol Biol. 27:57–92. 1992. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Y, Zhao YH, Kalaslavadi TB, Hamati E, Nehrke K, Le AD, Ann DK and Wu R: Genome-wide search and identification of a novel gel-forming mucin MUC19/Muc19 in glandular tissues. Am J Respir Cell Mol Biol. 30:155–165. 2004. View Article : Google Scholar | |
|
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. 279:1968–1979. 2004. View Article : Google Scholar | |
|
Marimuthu S, Rauth S, Ganguly K, Zhang C, Lakshmanan I, Batra SK and Ponnusamy MP: Mucins reprogram stemness, metabolism and promote chemoresistance during cancer progression. Cancer Metastasis Rev. 40:575–588. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Moniaux N, Escande F, Porchet N, Aubert JP and Batra SK: Structural organization and classification of the human mucin genes. Front Biosci. 6:D1192–D1206. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Chaturvedi P, Singh AP and Batra SK: Structure, evolution, and biology of the MUC4 mucin. FASEB J. 22:966–981. 2008. View Article : Google Scholar | |
|
Mukhopadhyay P, Chakraborty S, Ponnusamy MP, Lakshmanan I, Jain M and Batra SK: Mucins in the pathogenesis of breast cancer: Implications in diagnosis, prognosis and therapy. Biochim Biophys Acta. 1815:224–240. 2011.PubMed/NCBI | |
|
Breugelmans T, Oosterlinck B, Arras W, Ceuleers H, De Man J, Hold GL, De Winter BY and Smet A: The role of mucins in gastrointestinal barrier function during health and disease. Lancet Gastroenterol Hepatol. 7:455–471. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Devine PL and McKenzie IF: Mucins: Structure, function, and associations with malignancy. Bioessays. 14:619–625. 1992. View Article : Google Scholar : PubMed/NCBI | |
|
Gray T, Koo JS and Nettesheim P: Regulation of mucous differentiation and mucin gene expression in the tracheobronchial epithelium. Toxicology. 160:35–46. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Marzio A, Kurz E, Sahni JM, Di Feo G, Puccini J, Jiang S, Hirsch CA, Arbini AA, Wu WL, Pass HI, et al: EMSY inhibits homologous recombination repair and the interferon response, promoting lung cancer immune evasion. Cell. 185:169–183.e19. 2022. View Article : Google Scholar | |
|
Yang K, Wu J, Xu T, Zhou Y, Liu W and Yang L: Machine learning to predict distant metastasis and prognostic analysis of moderately differentiated gastric adenocarcinoma patients: A novel focus on lymph node indicators. Front Immunol. 15:13986852024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Lu Q, Zhou H, Liu J, Nadorp B, Lasry A, Sun Z, Lai B, Rona G, Zhang J, et al: A membrane-associated MHC-I inhibitory axis for cancer immune evasion. Cell. 186:3903–3920.e21. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Reily C, Stewart TJ, Renfrow MB and Novak J: Glycosylation in health and disease. Nat Rev Nephrol. 15:346–366. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Chen K, Gu Y, Cao Y, Fang H, Lv K, Liu X, He X, Wang J, Lin C, Liu H, et al: TIM3(+) cells in gastric cancer: Clinical correlates and association with immune context. Br J Cancer. 126:100–108. 2022. View Article : Google Scholar | |
|
Lai Y, Huang C, Wu J, Yang K and Yang L: Ferroptosis in cancer: A new perspective on T cells. Int Immunopharmacol. 143(Pt 3): 1135392024. View Article : Google Scholar : PubMed/NCBI | |
|
Ratan C, Cicily K D D, Nair B and Nath LR: MUC glycoproteins: Potential biomarkers and molecular targets for cancer therapy. Curr Cancer Drug Targets. 21:132–152. 2021. View Article : Google Scholar | |
|
Liu Y, Tang L, Gao N, Diao Y, Zhong J, Deng Y, Wang Z, Jin G and Wang X: Synthetic MUC1 breast cancer vaccine containing a Toll-like receptor 7 agonist exerts antitumor effects. Oncol Lett. 20:2369–2377. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Guillen-Poza PA, Sánchez-Fernández EM, Artigas G, García Fernández JM, Hinou H, Ortiz Mellet C, Nishimura SI and Garcia-Martin F: Amplified detection of breast cancer autoantibodies using MUC1-based Tn antigen mimics. J Med Chem. 63:8524–8533. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Xi X, Wang J, Qin Y, Huang W, You Y and Zhan J: Glycosylated modification of MUC1 maybe a new target to promote drug sensitivity and efficacy for breast cancer chemotherapy. Cell Death Dis. 13:7082022. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Yin L, Chen S, Li Z, Ding J, Wu J, Yang K and Xu J: The perspectives of NETosis on the progression of obesity and obesity-related diseases: mechanisms and applications. Front Cell Dev Biol. 11:12213612023. View Article : Google Scholar : PubMed/NCBI | |
|
Kim GE, Bae HI, Park HU, Kuan SF, Crawley SC, Ho JJ and Kim YS: Aberrant expression of MUC5AC and MUC6 gastric mucins and sialyl Tn antigen in intraepithelial neoplasms of the pancreas. Gastroenterology. 123:1052–1060. 2022. View Article : Google Scholar | |
|
Babu SD, Jayanthi V, Devaraj N, Reis CA and Devaraj H: Expression profile of mucins (MUC2, MUC5AC and MUC6) in Helicobacter pylori infected pre-neoplastic and neoplastic human gastric epithelium. Mol Cancer. 5:102006. View Article : Google Scholar : PubMed/NCBI | |
|
Dwertmann Rico S, Schliesser SJA, Gorbokon N, Dum D, Menz A, Buscheck F, Hinsch A, Lennartz M, von Bargen C, Bawahab AA, et al: Pattern of MUC6 expression across 119 different tumor types: A tissue microarray study on 15 412 tumors. Pathol Int. 73:281–296. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Bartman AE, Buisine MP, Aubert JP, Niehans GA, Toribara NW, Kim YS, Kelly EJ, Crabtree JE and Ho SB: The MUC6 secretory mucin gene is expressed in a wide variety of epithelial tissues. J Pathol. 186:398–405. 1998. View Article : Google Scholar | |
|
Fan H, Wu J, Yang K, Xiong C, Xiong S, Wu X, Fang Z, Zhu J and Huang J: Dietary regulation of intestinal stem cells in health and disease. Int J Food Sci Nutr. 74:730–745. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Lee HS, Lee HK, Kim HS, Yang HK, Kim YI and Kim WH: MUC1, MUC2, MUC5AC, and MUC6 expressions in gastric carcinomas: Their roles as prognostic indicators. Cancer. 92:1427–1434. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Freire T, Lo-Man R, Piller F, Piller V, Leclerc C and Bay S: Enzymatic large-scale synthesis of MUC6-Tn glycoconjugates for antitumor vaccination. Glycobiology. 16:390–401. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Freire T, Lo-Man R, Bay S and Leclerc C: Tn glycosylation of the MUC6 protein modulates its immunogenicity and promotes the induction of Th17-biased T cell responses. J Biol Chem. 286:7797–7811. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Freire T, Bay S, von Mensdorff-Pouilly S and Osinaga E: Molecular basis of incomplete O-glycan synthesis in MCF-7 breast cancer cells: Putative role of MUC6 in Tn antigen expression. Cancer Res. 65:7880–7887. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Tavares C, de Oliveira JT, Lopes C, Carvalheira J, Matos AJ, Rutteman G, Reis CA and Gärtner F: Mucin 6 and Tn antigen expression in canine mammary tumours: Correlation with pathological features. J Comp Pathol. 147:410–418. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Plender EG, Prodanov T, Hsieh P, Nizamis E, Harvey WT, Sulovari A, Munson KM, Kaufman EJ, O'Neal WK, Valdmanis PN, et al: Structural and genetic diversity in the secreted mucins MUC5AC and MUC5B. Am J Hum Genet. 111:1700–1716. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Gendler SJ, Burchell JM, Duhig T, Lamport D, White R, Parker M and Taylor-Papadimitriou J: Cloning of partial cDNA encoding differentiation and tumor-associated mucin glycoproteins expressed by human mammary epithelium. Proc Natl Acad Sci USA. 84:6060–6064. 1987. View Article : Google Scholar : PubMed/NCBI | |
|
Gum JR Jr, Hicks JW, Toribara NW, Rothe EM, Lagace RE and Kim YS: The human MUC2 intestinal mucin has cysteine-rich subdomains located both upstream and downstream of its central repetitive region. J Biol Chem. 267:21375–21383. 1992. View Article : Google Scholar : PubMed/NCBI | |
|
Toribara NW, Roberton AM, Ho SB, Kuo WL, Gum E, Hicks JW, Gum JR Jr, Byrd JC, Siddiki B and Kim YS: Human gastric mucin. Identification of a unique species by expression cloning. J Biol Chem. 268:5879–5885. 1993. View Article : Google Scholar : PubMed/NCBI | |
|
Lang T: Tandem repeats structure of gel-forming mucin domains could be revealed by SMRT sequencing data. Sci Rep. 12:206522022. View Article : Google Scholar : PubMed/NCBI | |
|
Karbhari DS, Panwalla NM, Mondkar AM and Motashaw ND: Eclampsia-the influence of antenatal care. J Postgrad Med. 18:175–180. 1972.PubMed/NCBI | |
|
Kornfeld R and Kornfeld S: Assembly of asparagine-linked oligosaccharides. Annu Rev Biochem. 54:631–664. 1985. View Article : Google Scholar : PubMed/NCBI | |
|
Schwarz F and Aebi M: Mechanisms and principles of N-linked protein glycosylation. Curr Opin Struct Biol. 21:576–582. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Ruddock LW and Molinari M: N-glycan processing in ER quality control. J Cell Sci. 119(Pt 21): 4373–4380. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Stanley P: Golgi glycosylation. Cold Spring Harb Perspect Biol. 3:a0051992011. View Article : Google Scholar : PubMed/NCBI | |
|
Marth JD: Complexity in O-linked oligosaccharide biosynthesis engendered by multiple polypeptide N-acetylgalactosaminyltransferases. Glycobiology. 6:701–705. 1996. View Article : Google Scholar : PubMed/NCBI | |
|
Rose MC and Voynow JA: Respiratory tract mucin genes and mucin glycoproteins in health and disease. Physiol Rev. 86:245–278. 2006. View Article : Google Scholar | |
|
Kasprzak A and Adamek A: Mucins: The old, the new and the promising factors in hepatobiliary carcinogenesis. Int J Mol Sci. 20:12882019. View Article : Google Scholar : PubMed/NCBI | |
|
Van Acker SI, Van den Bogerd B, Van Acker ZP, Vailionytė A, Haagdorens M, Koppen C, Ní Dhubhghaill S, Dartt DA and Pintelon I: Characterisation of gel-forming mucins produced in vivo and in ex vivo conjunctival explant cultures. Int J Mol Sci. 22:105282021. View Article : Google Scholar : PubMed/NCBI | |
|
Rousseau K, Byrne C, Kim YS, Gum JR, Swallow DM and Toribara NW: The complete genomic organization of the human MUC6 and MUC2 mucin genes. Genomics. 83:936–939. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Desseyn JL, Aubert JP, Porchet N and Laine A: Evolution of the large secreted gel-forming mucins. Mol Biol Evol. 17:1175–1184. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Kwon JA, Lee SY, Ahn EK, Seol SY, Kim MC, Kim SJ, Kim SI, Chu IS and Leem SH: Short rare MUC6 minisatellites-5 alleles influence susceptibility to gastric carcinoma by regulating gene. Hum Mutat. 31:942–949. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Han HS, Lee SY, Lee KY, Hong SN, Kim JH, Sung IK, Park HS, Jin CJ and Min YI: Unclassified mucin phenotype of gastric adenocarcinoma exhibits the highest invasiveness. J Gastroenterol Hepatol. 24:658–666. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Leir SH and Harris A: MUC6 mucin expression inhibits tumor cell invasion. Exp Cell Res. 317:2408–2419. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Lidell ME, Johansson ME and Hansson GC: An autocatalytic cleavage in the C terminus of the human MUC2 mucin occurs at the low pH of the late secretory pathway. J Biol Chem. 278:13944–13951. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Velcich A, Yang W, Heyer J, Fragale A, Nicholas C, Viani S, Kucherlapati R, Lipkin M, Yang K and Augenlicht L: Colorectal cancer in mice genetically deficient in the mucin Muc2. Science. 295:1726–1729. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Toribara NW, Ho SB, Gum E, Gum JR Jr, Lau P and Kim YS: The carboxyl-terminal sequence of the human secretory mucin, MUC6. Analysis Of the primary amino acid sequence. J Biol Chem. 272:16398–16403. 1997. View Article : Google Scholar : PubMed/NCBI | |
|
Cozzi PJ, Wang J, Delprado W, Perkins AC, Allen BJ, Russell PJ and Li Y: MUC1, MUC2, MUC4, MUC5AC and MUC6 expression in the progression of prostate cancer. Clin Exp Metastasis. 22:565–573. 2005. View Article : Google Scholar | |
|
Senapati S, Sharma P, Bafna S, Roy HK and Batra SK: The MUC gene family: Their role in the diagnosis and prognosis of gastric cancer. Histol Histopathol. 23:1541–1552. 2008.PubMed/NCBI | |
|
Boccellato F, Woelffling S, Imai-Matsushima A, Sanchez G, Goosmann C, Schmid M, Berger H, Morey P, Denecke C, Ordemann J and Meyer TF: Polarised epithelial monolayers of the gastric mucosa reveal insights into mucosal homeostasis and defence against infection. Gut. 68:400–413. 2019. View Article : Google Scholar | |
|
Yuki A, Fujii C, Yamanoi K, Matoba H, Harumiya S, Kawakubo M and Nakayama J: Glycosylation of MUC6 by α1,4-linked N-acetylglucosamine enhances suppression of pancreatic cancer malignancy. Cancer Sci. 113:576–586. 2022. View Article : Google Scholar | |
|
Nagata K, Horinouchi M, Saitou M, Higashi M, Nomoto M, Goto M and Yonezawa S: Mucin expression profile in pancreatic cancer and the precursor lesions. J Hepatobiliary Pancreat Surg. 14:243–254. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Prasad NB, Biankin AV, Fukushima N, Maitra A, Dhara S, Elkahloun AG, Hruban RH, Goggins M and Leach SD: Gene expression profiles in pancreatic intraepithelial neoplasia reflect the effects of Hedgehog signaling on pancreatic ductal epithelial cells. Cancer Res. 65:1619–1626. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Matsukita S, Nomoto M, Kitajima S, Tanaka S, Goto M, Irimura T, Kim YS, Sato E and Yonezawa S: Expression of mucins (MUC1, MUC2, MUC5AC and MUC6) in mucinous carcinoma of the breast: Comparison with invasive ductal carcinoma. Histopathology. 42:26–36. 2003. View Article : Google Scholar | |
|
Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015.PubMed/NCBI | |
|
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI | |
|
Ding JT, Yang KP, Zhou HN, Huang YF, Li H and Zong Z: Landscapes and mechanisms of CD8(+) T cell exhaustion in gastrointestinal cancer. Front Immunol. 14:11496222023. View Article : Google Scholar : PubMed/NCBI | |
|
Li Z, Qi H, Li Z, Bao Y, Yang K and Min Q: Research progress on the premature ovarian failure caused by cisplatin therapy. Front Oncol. 13:12763102023. View Article : Google Scholar : PubMed/NCBI | |
|
Schistosomes, liver flukes and Helicobacter pylori. IARC Monogr Eval Carcinog Risks Hum. 61:1–241. 1994.PubMed/NCBI | |
|
Nguyen TV, Janssen M Jr, Gritters P, te Morsche RH, Drenth JP, van Asten H, Laheij RJ and Jansen JB: Short mucin 6 alleles are associated with H pylori infection. World J Gastroenterol. 12:6021–6025. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Kawakubo M, Ito Y, Okimura Y, Kobayashi M, Sakura K, Kasama S, Fukuda MN, Fukuda M, Katsuyama T and Nakayama J: Natural antibiotic function of a human gastric mucin against Helicobacter pylori infection. Science. 305:1003–1006. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Byrd JC, Yan P, Sternberg L, Yunker CK, Scheiman JM and Bresalier RS: Aberrant expression of gland-type gastric mucin in the surface epithelium of Helicobacter pylori-infected patients. Gastroenterology. 113:455–464. 1997. View Article : Google Scholar : PubMed/NCBI | |
|
Van den Brink GR, Tytgat KM, Van der Hulst RW, Van der Loos CM, Einerhand AW, Buller HA and Dekker J: H pylori colocalises with MUC5AC in the human stomach. Gut. 46:601–607. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Jia Y, Persson C, Hou L, Zheng Z, Yeager M, Lissowska J, Chanock SJ, Chow WH and Ye W: A comprehensive analysis of common genetic variation in MUC1, MUC5AC, MUC6 genes and risk of stomach cancer. Cancer Causes Control. 21:313–321. 2010. | |
|
Khattab AZ, Nasif WA and Lotfy M: MUC2 and MUC6 apomucins expression in human gastric neoplasm: An immunohistochemical analysis. Med Oncol. 28(Suppl 1): S207–S213. 2011. View Article : Google Scholar | |
|
Marin F, Bonet C, Munoz X, Garcia N, Pardo ML, Ruiz-Liso JM, Alonso P, Capellà G, Sanz-Anquela JM, González CA and Sala N: Genetic variation in MUC1, MUC2 and MUC6 genes and evolution of gastric cancer precursor lesions in a long-term follow-up in a high-risk area in Spain. Carcinogenesis. 33:1072–1080. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Pinto-de-Sousa J, David L, Reis CA, Gomes R, Silva L and Pimenta A: Mucins MUC1, MUC2, MUC5AC and MUC6 expression in the evaluation of differentiation and clinico-biological behaviour of gastric carcinoma. Virchows Arch. 440:304–310. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Pinto-de-Sousa J, Reis CA, David L, Pimenta A and Cardoso-de-Oliveira M: MUC5B expression in gastric carcinoma: Relationship with clinico-pathological parameters and with expression of mucins MUC1, MUC2, MUC5AC and MUC6. Virchows Arch. 444:224–230. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Reis CA, David L, Carvalho F, Mandel U, de Bolos C, Mirgorodskaya E, Clausen H and Sobrinho-Simões M: Immunohistochemical study of the expression of MUC6 mucin and co-expression of other secreted mucins (MUC5AC and MUC2) in human gastric carcinomas. J Histochem Cytochem. 48:377–388. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Shiroshita H, Watanabe H, Ajioka Y, Watanabe G, Nishikura K and Kitano S: Re-evaluation of mucin phenotypes of gastric minute well-differentiated-type adenocarcinomas using a series of HGM, MUC5AC, MUC6, M-GGMC, MUC2 and CD10 stains. Pathol Int. 54:311–321. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Barua RR, Uozaki H, Chong JM, Ushiku T, Hino R, Chang MS, Nagai H and Fukayama M: Phenotype analysis by MUC2, MUC5AC, MUC6, and CD10 expression in Epstein-Barr virus-associated gastric carcinoma. J Gastroenterol. 41:733–739. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Tjandra D, Busuttil RA and Boussioutas A: Gastric intestinal metaplasia: Challenges and the opportunity for precision prevention. Cancers (Basel). 15:39132023. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng H, Takahashi H, Nakajima T, Murai Y, Cui Z, Nomoto K, Tsuneyama K and Takano Y: MUC6 down-regulation correlates with gastric carcinoma progression and a poor prognosis: An immunohistochemical study with tissue microarrays. J Cancer Res Clin Oncol. 132:817–823. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Oosterlinck B, Ceuleers H, Arras W, De Man JG, Geboes K, De Schepper H, Peeters M, Lebeer S, Skieceviciene J, Hold GL, et al: Mucin-microbiome signatures shape the tumor microenvironment in gastric cancer. Microbiome. 11:862023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang J, Peng Y, Ding Y, Liu Y, Wang Y, Liu Y and Liu C: The clinicopathological and molecular characteristics of endocervical gastric-type adenocarcinoma and the use of claudin18.2 as a potential therapeutic target. Mod Pathol. 37:1005692024. View Article : Google Scholar : PubMed/NCBI | |
|
Martinez-Ciarpaglini C, Barros R, Caballero C, Boggino H, Alarcon-Molero L, Peleteiro B, Ruiz-García E, Fernandez-Figueroa E, Herrera-Goepfert R, Díaz-Romero C, et al: Comprehensive histopathological analysis of gastric cancer in European and Latin America populations reveals differences in PDL1, HER2, p53 and MUC6 expression. Gastric Cancer. 28:160–173. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Garcia E, Carvalho F, Amorim A and David L: MUC6 gene polymorphism in healthy individuals and in gastric cancer patients from northern Portugal. Cancer Epidemiol Biomarkers Prev. 6:1071–1074. 1997. | |
|
Dashti F, Mirazimi SMA, Rabiei N, Fathazam R, Rabiei N, Piroozmand H, Vosough M, Rahimian N, Hamblin MR and Mirzaei H: The role of non-coding RNAs in chemotherapy for gastrointestinal cancers. Mol Ther Nucleic Acids. 26:892–926. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Hashemipour M, Boroumand H, Mollazadeh S, Tajiknia V, Nourollahzadeh Z, Rohani Borj M, Pourghadamyari H, Rahimian N, Hamblin MR and Mirzaei H: Exosomal microRNAs and exosomal long non-coding RNAs in gynecologic cancers. Gynecol Oncol. 161:314–327. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Aghdam AM, Amiri A, Salarinia R, Masoudifar A, Ghasemi F and Mirzaei H: MicroRNAs as diagnostic, prognostic, and therapeutic biomarkers in prostate cancer. Crit Rev Eukaryot Gene Expr. 29:127–139. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Walsh MD, Clendenning M, Williamson E, Pearson SA, Walters RJ, Nagler B, Packenas D, Win AK, Hopper JL, Jenkins MA, et al: Expression of MUC2, MUC5AC, MUC5B, and MUC6 mucins in colorectal cancers and their association with the CpG island methylator phenotype. Mod Pathol. 26:1642–1656. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Tanaka Y, Eizuka M, Uesugi N, Kawasaki K, Yamano H, Suzuki H, Matsumoto T and Sugai T: Traditional serrated adenoma has two distinct genetic pathways for molecular tumorigenesis with potential neoplastic progression. J Gastroenterol. 55:846–857. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Bartley AN, Thompson PA, Buckmeier JA, Kepler CY, Hsu CH, Snyder MS, Lance P, Bhattacharyya A and Hamilton SR: Expression of gastric pyloric mucin, MUC6, in colorectal serrated polyps. Mod Pathol. 23:169–176. 2010. View Article : Google Scholar | |
|
Gibson JA, Hahn HP, Shahsafaei A and Odze RD: MUC expression in hyperplastic and serrated colonic polyps: Lack of specificity of MUC6. Am J Surg Pathol. 35:742–749. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Owens SR, Chiosea SI and Kuan SF: Selective expression of gastric mucin MUC6 in colonic sessile serrated adenoma but not in hyperplastic polyp aids in morphological diagnosis of serrated polyps. Mod Pathol. 21:660–669. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Ahn MH, Bae KB, Kwon JA, Choi HJ, Lee SR, Kim SH, Jung TD, Kim SH, An MS, Hong KH, et al: Association of MUC6-minisatellite variants with susceptibility to rectal carcinoma. Mol Biol Rep. 40:303–308. 2013. View Article : Google Scholar | |
|
Abou-Alfa GK, Andersen JB, Chapman W, Choti M, Forbes SJ, Gores GJ, Hong TS, Harding JJ, Vander Heiden MG, Javle M, et al: Advances in cholangiocarcinoma research: Report from the third Cholangiocarcinoma Foundation Annual Conference. J Gastrointest Oncol. 7:819–827. 2016. View Article : Google Scholar | |
|
Thuwajit P, Chawengrattanachot W, Thuwajit C, Sripa B, Paupairoj A and Chau-In S: Enhanced expression of mucin 6 glycoprotein in cholangiocarcinoma tissue from patients in Thailand as a prognostic marker for survival. J Gastroenterol Hepatol. 23:771–778. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Aishima S, Kuroda Y, Nishihara Y, Taguchi K, Taketomi A, Maehara Y and Tsuneyoshi M: Gastric mucin phenotype defines tumour progression and prognosis of intrahepatic cholangiocarcinoma: gastric foveolar type is associated with aggressive tumour behaviour. Histopathology. 49:35–44. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Park SY, Roh SJ, Kim YN, Kim SZ, Park HS, Jang KY, Chung MJ, Kang MJ, Lee DG and Moon WS: Expression of MUC1, MUC2, MUC5AC and MUC6 in cholangiocarcinoma: Prognostic impact. Oncol Rep. 22:649–657. 2009.PubMed/NCBI | |
|
Albores-Saavedra J, Chable-Montero F, Gonzalez-Romo MA, Ramirez Jaramillo M and Henson DE: Adenomas of the gallbladder. Morphologic features, expression of gastric and intestinal mucins, and incidence of high-grade dysplasia/carcinoma in situ and invasive carcinoma. Hum Pathol. 43:1506–1513. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Nagata S, Ajioka Y, Nishikura K, Watanabe G, Inoue T, Yamaguchi K, Watanabe H, Tanaka M and Tsuneyoshi M: Co-expression of gastric and biliary phenotype in pyloric-gland type adenoma of the gallbladder: Immunohistochemical analysis of mucin profile and CD10. Oncol Rep. 17:721–729. 2007.PubMed/NCBI | |
|
Sasaki M, Yamato T, Nakanuma Y, Ho SB and Kim YS: Expression of MUC2, MUC5AC and MUC6 apomucins in carcinoma, dysplasia and non-dysplastic epithelia of the gallbladder. Pathol Int. 49:38–44. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Sato K, Ito E, Masaki Y and Ogawa M: A case report of gallbladder cancer and pancreas cystic neoplasm associated with pancreaticobiliary maljunction. Int J Surg Case Rep. 85:1061702021. View Article : Google Scholar : PubMed/NCBI | |
|
Carrasco C, Tittarelli A, Paillaleve N, Pozo MD, Rojas-Sepulveda D, Barria O, Fluxá P, Hott M, Martin C, Quezada C and Salazar-Onfray F: The evaluation of 17 gastrointestinal tumor markers reveals prognosis value for MUC6, CK17, and CD10 in gallbladder-cancer patients. Diagnostics (Basel). 11:1532021. View Article : Google Scholar : PubMed/NCBI | |
|
Kamisawa T, Wood LD, Itoi T and Takaori K: Pancreatic cancer. Lancet. 388:73–85. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Rawla P, Sunkara T and Gaduputi V: Epidemiology of pancreatic cancer: Global trends, etiology and risk factors. World J Oncol. 10:10–27. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Xu DF, Wang LS and Zhou JH: Long noncoding RNA CASC2 suppresses pancreatic cancer cell growth and progression by regulating the miR24/MUC6 axis. Int J Oncol. 56:494–507. 2020.PubMed/NCBI | |
|
Lahdaoui F, Delpu Y, Vincent A, Renaud F, Messager M, Duchene B, Leteurtre E, Mariette C, Torrisani J, Jonckheere N and Van Seuningen I: miR-219-1-3p is a negative regulator of the mucin MUC4 expression and is a tumor suppressor in pancreatic cancer. Oncogene. 34:780–788. 2015. View Article : Google Scholar | |
|
Mohr AM, Bailey JM, Lewallen ME, Liu X, Radhakrishnan P, Yu F, Tapprich W and Hollingsworth MA: MUC1 regulates expression of multiple microRNAs involved in pancreatic tumor progression, including the miR-200c/141 cluster. PLoS One. 8:e733062013. View Article : Google Scholar : PubMed/NCBI | |
|
Basturk O, Khayyata S, Klimstra DS, Hruban RH, Zamboni G, Coban I and Adsay NV: Preferential expression of MUC6 in oncocytic and pancreatobiliary types of intraductal papillary neoplasms highlights a pyloropancreatic pathway, distinct from the intestinal pathway, in pancreatic carcinogenesis. Am J Surg Pathol. 34:364–370. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Li XL, Eishi Y, Bai YQ, Sakai H, Akiyama Y, Tani M, Takizawa T, Koike M and Yuasa Y: Expression of the SRY-related HMG box protein SOX2 in human gastric carcinoma. Int J Oncol. 24:257–263. 2004.PubMed/NCBI | |
|
Sakai H, Jinawath A, Yamaoka S and Yuasa Y: Upregulation of MUC6 mucin gene expression by NFkappaB and Sp factors. Biochem Biophys Res Commun. 333:1254–1260. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Banerjee S, Li Y, Rahman KM, Zhang Y and Sarkar FH: Down-regulation of notch-1 inhibits invasion by inactivation of nuclear factor-kappaB, vascular endothelial growth factor, and matrix metalloproteinase-9 in pancreatic cancer cells. Cancer Res. 66:2778–2784. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Banerjee S, Wang Z, Xu H, Zhang L, Mohammad R, Aboukameel A, Adsay NV, Che M, Abbruzzese JL, et al: Antitumor activity of epidermal growth factor receptor-related protein is mediated by inactivation of ErbB receptors and nuclear factor-kappaB in pancreatic cancer. Cancer Res. 66:1025–1032. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Lee CS, Sund NJ, Vatamaniuk MZ, Matschinsky FM, Stoffers DA and Kaestner KH: Foxa2 controls Pdx1 gene expression in pancreatic beta-cells in vivo. Diabetes. 51:2546–2551. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Sierzega M, Mlynarski D, Tomaszewska R and Kulig J: Semiquantitative immunohistochemistry for mucin (MUC1, MUC2, MUC3, MUC4, MUC5AC, and MUC6) profiling of pancreatic ductal cell adenocarcinoma improves diagnostic and prognostic performance. Histopathology. 69:582–591. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Jinfeng M, Kimura W, Hirai I, Sakurai F, Moriya T and Mizutani M: Expression of MUC5AC and MUC6 in invasive ductal carcinoma of the pancreas and relationship with prognosis. Int J Gastrointest Cancer. 34:9–18. 2003. View Article : Google Scholar | |
|
Awaya H, Takeshima Y, Yamasaki M and Inai K: Expression of MUC1, MUC2, MUC5AC, and MUC6 in atypical adenomatous hyperplasia, bronchioloalveolar carcinoma, adenocarcinoma with mixed subtypes, and mucinous bronchioloalveolar carcinoma of the lung. Am J Clin Pathol. 121:644–653. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Pereira MB, Dias AJ, Reis CA and Schmitt FC: Immunohistochemical study of the expression of MUC5AC and MUC6 in breast carcinomas and adjacent breast tissues. J Clin Pathol. 54:210–213. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Do SI, Kim K, Kim DH, Chae SW, Park YL, Park CH and Sohn JH: Associations between the Expression of Mucins (MUC1, MUC2, MUC5AC, and MUC6) and clinicopathologic parameters of human breast ductal carcinomas. J Breast Cancer. 16:152–158. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Oral O, Unverdi H, Kumcu E, Turkbey D, Dogan S and Hucumenoglu S: Associations between the expression of mucins (MUC1, MUC2, MUC5AC and MUC6) and clinicopathologic parameters of human breast carcinomas. Indian J Pathol Microbiol. 63:551–558. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Rakha EA, Boyce RW, Abd El-Rehim D, Kurien T, Green AR, Paish EC, Robertson JF and Ellis IO: Expression of mucins (MUC1, MUC2, MUC3, MUC4, MUC5AC and MUC6) and their prognostic significance in human breast cancer. Mod Pathol. 18:1295–1304. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
de Bolos C, Guma M, Barranco C, Garrido M, Kim YS and Real FX: MUC6 expression in breast tissues and cultured cells: abnormal expression in tumors and regulation by steroid hormones. Int J Cancer. 77:193–199. 1998. View Article : Google Scholar : PubMed/NCBI | |
|
Buyuk M, Ozluk Y, Bakkaloglu DV, Ozkan B, Firat P and Yilmazbayhan D: Evaluation of MUC1, MUC2, MUC5AC, and MUC6 expression differences in lung adenocarcinoma subtypes by using a final immunoreactivity score (FIRS). Turk Patoloji Derg. 39:64–74. 2023. | |
|
Copin MC, Buisine MP, Leteurtre E, Marquette CH, Porte H, Aubert JP, Gosselin B and Porchet N: Mucinous bronchioloalveolar carcinomas display a specific pattern of mucin gene expression among primary lung adenocarcinomas. Hum Pathol. 32:274–281. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Hamamoto A, Abe Y, Nishi M, Fujimori S, Ohnishi Y, Yamazaki H, Oida Y, Miyazaki N, Inada K, Ueyama Y, et al: Aberrant expression of the gastric mucin MUC6 in human pulmonary adenocarcinoma xenografts. Int J Oncol. 26:891–896. 2005.PubMed/NCBI | |
|
Kishikawa S, Hayashi T, Saito T, Takamochi K, Kohsaka S, Sano K, Sasahara N, Sasa K, Kurihara T, Hara K, et al: Diffuse expression of MUC6 defines a distinct clinicopathological subset of pulmonary invasive mucinous adenocarcinoma. Mod Pathol. 34:786–797. 2021. View Article : Google Scholar | |
|
Lee HK, Kwon MJ, Seo J, Kim JW, Hong M, Park HR, Min SK, Choe JY, Ra YJ, Jang SH, et al: Expression of mucins (MUC1, MUC2, MUC5AC and MUC6) in ALK-positive lung cancer: Comparison with EGFR-mutated lung cancer. Pathol Res Pract. 215:459–465. 2019. View Article : Google Scholar | |
|
Rerkpichaisuth V, Lau RP, Meyerson C and Fishbein GA: The utility of the lineage specific immunohistochemical stains SATB2, CDX2, and villin, and the mucin glycoproteins MUC2, MUC5AC, and MUC6 to distinguish pulmonary invasive mucinous adenocarcinoma from metastatic colorectal carcinoma. Hum Pathol. 151:1056272024. View Article : Google Scholar : PubMed/NCBI | |
|
Nishiumi N, Abe Y, Inoue Y, Hatanaka H, Inada K, Kijima H, Yamazaki H, Tatematsu M, Ueyama Y, Iwasaki M, et al: Use of 11p15 mucins as prognostic factors in small adenocarcinoma of the lung. Clin Cancer Res. 9:5616–5619. 2003.PubMed/NCBI | |
|
Rahimian N, Razavi ZS, Aslanbeigi F, Mirkhabbaz AM, Piroozmand H, Shahrzad MK, Hamblin MR and Mirzaei H: Non-coding RNAs related to angiogenesis in gynecological cancer. Gynecol Oncol. 161:896–912. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Leroy X, Ballereau C, Villers A, Saint F, Aubert S, Gosselin B, Porchet N and Copin MC: MUC6 is a marker of seminal vesicle-ejaculatory duct epithelium and is useful for the differential diagnosis with prostate adenocarcinoma. Am J Surg Pathol. 27:519–521. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Kudryavtseva AV, Lukyanova EN, Kharitonov SL, Nyushko KM, Krasheninnikov AA, Pudova EA, Guvatova ZG, Alekseev BY, Kiseleva MV, Kaprin AD, et al: Bioinformatic identification of differentially expressed genes associated with prognosis of locally advanced lymph node-positive prostate cancer. J Bioinform Comput Biol. 17:19500032019. View Article : Google Scholar : PubMed/NCBI | |
|
Schaid DJ, McDonnell SK, FitzGerald LM, DeRycke L, Fogarty Z, Giles GG, MacInnis RJ, Southey MC, Nguyen-Dumont T, Cancel-Tassin G, et al: Two-stage study of familial prostate cancer by whole-exome sequencing and custom capture identifies 10 novel genes associated with the risk of prostate cancer. Eur Urol. 79:353–361. 2021. View Article : Google Scholar : | |
|
Sarvizadeh M, Hasanpour O, Naderi Ghale-Noie Z, Mollazadeh S, Rezaei M, Pourghadamyari H, Masoud Khooy M, Aschner M, Khan H, Rezaei N, et al: Allicin and digestive system cancers: From chemical structure to its therapeutic opportunities. Front Oncol. 11:6502562021. View Article : Google Scholar : PubMed/NCBI | |
|
Savardashtaki A, Shabaninejad Z, Movahedpour A, Sahebnasagh R, Mirzaei H and Hamblin MR: miRNAs derived from cancer-associated fibroblasts in colorectal cancer. Epigenomics. 11:1627–1645. 2016. View Article : Google Scholar | |
|
Urick ME and Bell DW: Clinical actionability of molecular targets in endometrial cancer. Nat Rev Cancer. 19:510–521. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Razavi ZS, Tajiknia V, Majidi S, Ghandali M, Mirzaei HR, Rahimian N, Hamblin MR and Mirzaei H: Gynecologic cancers and non-coding RNAs: Epigenetic regulators with emerging roles. Crit Rev Oncol Hematol. 157:1031922021. View Article : Google Scholar | |
|
Hodgson A, Parra-Herran C and Mirkovic J: Immunohistochemical expression of HIK1083 and MUC6 in endometrial carcinomas. Histopathology. 75:552–558. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Fulgione C, Raffone A, Travaglino A, Arciuolo D, Santoro A, Cianfrini F, Russo D, Varricchio S, Raimondo I, Inzani F, et al: Diagnostic accuracy of HIK1083 and MUC6 as immunohistochemical markers of endocervical gastric-type adenocarcinoma: A systematic review and meta-analysis. Pathol Res Pract. 241:1542612023. View Article : Google Scholar | |
|
Alameda F, Mejias-Luque R, Garrido M and de Bolos C: Mucin genes (MUC2, MUC4, MUC5AC, and MUC6) detection in normal and pathological endometrial tissues. Int J Gynecol Pathol. 26:61–65. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Carbone FR and Gleeson PA: Carbohydrates and antigen recognition by T cells. Glycobiology. 7:725–730. 1997. View Article : Google Scholar : PubMed/NCBI | |
|
Cloos PA and Christgau S: Post-translational modifications of proteins: implications for aging, antigen recognition, and autoimmunity. Biogerontology. 5:139–158. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Kontani K, Taguchi O, Narita T, Izawa M, Hiraiwa N, Zenita K, Takeuchi T, Murai H, Miura S and Kannagi R: Modulation of MUC1 mucin as an escape mechanism of breast cancer cells from autologous cytotoxic T-lymphocytes. Br J Cancer. 84:1258–1264. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Hoshi H, Sawada T, Uchida M, Iijima H, Kimura K, Hirakawa K and Wanibuchi H: MUC5AC protects pancreatic cancer cells from TRAIL-induced death pathways. Int J Oncol. 42:887–893. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Thomas A and Giaccone G: Why has active immunotherapy not worked in lung cancer? Ann Oncol. 26:2213–2220. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Xu X, Padilla MT, Li B, Wells A, Kato K, Tellez C, Belinsky SA, Kim KC and Lin Y: MUC1 in macrophage: Contributions to cigarette smoke-induced lung cancer. Cancer Res. 74:460–470. 2014. View Article : Google Scholar | |
|
Pelosi G and Sonzogni A: MUC6 distribution in the spectrum of pulmonary mucinous adenocarcinoma. Mod Pathol. 35:2025–2026. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Hoshi H, Sawada T, Uchida M, Saito H, Iijima H, Toda-Agetsuma M, Wada T, Yamazoe S, Tanaka H, Kimura K, et al: Tumor-associated MUC5AC stimulates in vivo tumorigenicity of human pancreatic cancer. Int J Oncol. 38:619–627. 2011.PubMed/NCBI | |
|
Slovin SF, Ragupathi G, Musselli C, Olkiewicz K, Verbel D, Kuduk SD, Schwarz JB, Sames D, Danishefsky S, Livingston PO and Scher HI: Fully synthetic carbohydrate-based vaccines in biochemically relapsed prostate cancer: Clinical trial results with alpha-N-acetylgalactosamine-O-serine/threonine conjugate vaccine. J Clin Oncol. 21:4292–4298. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Kang S, Bae MJ, Kang MK, Kim H, Kang YR, Jo WS, Lee CG, Jung B, Lee J, Moon C, et al: Possible association of G6PC2 and MUC6 induced by low-dose-rate irradiation in mouse intestine with inflammatory bowel disease. Mol Med Rep. 30:1272024. View Article : Google Scholar | |
|
Cin M, Gundogar O, Yarikkaya E and Cin S: The importance of MUC6 immunohistochemistry staining in the histopathologic examination of Crohn's disease: Can we enhance our diagnostic power? Ann Diagn Pathol. 78:1524822025. View Article : Google Scholar : PubMed/NCBI | |
|
Wong N, Beavers S, Gill P, Heryet A and Linares J: Calponin and MUC6 complement inhibin as diagnostic immunomarkers of serous cystadenoma in endoscopic ultrasound-guided aspiration/biopsy specimens. Histopathology. 79:252–259. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yamada S, Yamanoi K, Sato Y and Nakayama J: Diffuse MIST1 expression and decreased alpha1,4-linked N-acetylglucosamine (alphaGlcNAc) glycosylation on MUC6 are distinct hallmarks for gastric neoplasms showing oxyntic gland differentiation. Histopathology. 77:413–422. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhong F, Ren Z, Li W, Jones T, Zeng X, Wang S, Tao X and Zhao C: The sensitivity and specificity of Claudin18.2 and MUC6 in the differential diagnosis of endocervical gastric-type glandular lesions. Hum Pathol. 160:1058372025. View Article : Google Scholar : PubMed/NCBI | |
|
Shi H, Zhang W, Hu B, Wang Y, Zhang Z, Sun Y, Mao G, Li C and Lu S: Whole-exome sequencing identifies a set of genes as markers of hepatocellular carcinoma early recurrence. Hepatol Int. 17:393–405. 2023. View Article : Google Scholar | |
|
Kim GH, Lee JS, Lee JH and Park YS: Oxyntic gland neoplasms - from adenoma to advanced gastric cancer: A review of 29 cases. J Gastric Cancer. 24:378–390. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Kurosawa T, Murakami T, Yamashiro Y, Terukina H, Hayashi T, Saito T, Nojiri S, Sakamoto K, Nagahara A and Yao T: Mucin phenotypes and clinicopathological features of colorectal adenocarcinomas: Correlation with colorectal adenocarcinoma with enteroblastic differentiation. Pathol Res Pract. 232:1538402022. View Article : Google Scholar : PubMed/NCBI | |
|
Tanabe H, Uehara T and Ota H: Cell lineage-specific immunohistochemical markers in biliary intraepithelial neoplasia: Implications for subclassification and grading. Pathol Res Pract. 269:1558962025. View Article : Google Scholar : PubMed/NCBI | |
|
Riley NM, Wen RM, Bertozzi CR, Brooks JD and Pitteri SJ: Measuring the multifaceted roles of mucin-domain glycoproteins in cancer. Adv Cancer Res. 157:83–121. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Gupta R, Ponangi R and Indresh KG: Role of glycosylation in breast cancer progression and metastasis: Implications for miRNA, EMT and multidrug resistance. Glycobiology. 33:545–555. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Costa VD, Marino KV, Rodriguez-Zraquia SA, Festari MF, Lores P, Costa M, Landeira M, Rabinovich GA, van Vliet SJ and Freire T: Lung tumor cells with different Tn antigen expression present distinctive immunomodulatory properties. Int J Mol Sci. 23:120472022. View Article : Google Scholar : PubMed/NCBI | |
|
Iwaya M, Kodama M, Abe K, Maeda K, Nakajima T, Uehara T, Nishio R, Yamana T, Riddell R, Ota H, et al: Variability in morphology and immunohistochemistry of Crohn's disease-associated small bowel neoplasms: Implications of Claudin 18 and Cadherin 17 expression for tumor-targeted immunotherapies. Virchows Arch. 486:595–603. 2025. View Article : Google Scholar | |
|
Yang Y, Akashi Y, Shimomura O, Tateno H, Saito S, Hiemori K, Miyazaki Y, Furuta T, Kitaguchi D, Kuroda Y, et al: Glycan expression profile of signet ring cell gastric cancer cells and potential applicability of rBC2LCN-targeted lectin drug conjugate therapy. Gastric Cancer. 25:896–905. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ideo H, Tsuchida A and Takada Y: Lectin-based approaches to analyze the role of glycans and their clinical application in disease. Int J Mol Sci. 25:102312024. View Article : Google Scholar : PubMed/NCBI |