Open Access

Roles of long non‑coding RNA SNHG16 in human digestive system cancer (Review)

  • Authors:
    • Lujie Zhao
    • Yuling Kan
    • Lu Wang
    • Jiquan Pan
    • Yun Li
    • Haiyan Zhu
    • Zhongfa Yang
    • Lin Xiao
    • Xinhua Fu
    • Fujun Peng
    • Haipeng Ren
  • View Affiliations

  • Published online on: June 28, 2024     https://doi.org/10.3892/or.2024.8765
  • Article Number: 106
  • Copyright: © Zhao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The incidence of tumors in the human digestive system is relatively high, including esophageal cancer, liver cancer, pancreatic cancer, gastric cancer and colorectal cancer. These malignancies arise from a complex interplay of environmental and genetic factors. Among them, long non‑coding RNAs (lncRNAs), which cannot be translated into proteins, serve an important role in the development, progression, migration and prognosis of tumors. Small nucleolar RNA host gene 16 (SNHG16) is a typical lncRNA, and its relationship with digestive system tumors has been widely explored. The prevailing hypothesis suggests that the principal molecular mechanism of SNHG16 in digestive system tumors involves it functioning as a competitive endogenous RNA that interacts with other proteins, regulates various genes and influences a downstream target molecule. The present review summarizes recent research on the relationship between SNHG16 and numerous types of digestive system cancer, encompassing its biological functions, underlying mechanisms and potential clinical implications. Furthermore, it outlines the association between SNHG16 expression and pertinent risk factors, such as smoking, infection and diet. The present review indicated the promise of SNHG16 as a potential biomarker and therapeutic target in human digestive system cancer.

Introduction

In 2020, human digestive system tumors, mainly esophageal cancer (EC), hepatocellular carcinoma (HCC), pancreatic cancer (PC), gastric cancer (GC) and colorectal cancer (CRC), resulted in >5 million new cases and ~4 million cancer deaths worldwide. These malignancies are associated with personal suffering, and impose a substantial economic burden on patients, families and society (1,2). For example, the median medical expenditure per patient with EC in China increased from 6,851 to 57,554 CNY during 1996–2013, with an average growth rate of 11.89% (3). From a societal perspective, the costs associated with CRC in Europe reached ~19 billion EUR in 2018 (4). In the past few decades, although modern cancer treatments, including surgical treatment, chemotherapy, radiotherapy, immunotherapy, targeted therapy and traditional Chinese medicine, have markedly improved the quality of life of patients, limited effects have been achieved on patients with advanced or metastatic cancer (57). It is well known that the development processes of cancer are influenced by genetic and environmental factors, including environmental agents; lifestyle habits, such as a poor diet; and social behavior, such as immoderate consumption of alcohol. Various clinical trials have been conducted on targeted therapy for digestive tract cancer. Since gene mutations, such as those in the BRCA2 gene, are prevalent in PC and CRC, the development of corresponding targeted drugs has been initiated (810). However, these drugs still face the challenge of acquired resistance, and the efficacy of targeted therapy remains less pronounced than traditional therapy, such as chemotherapy, in some tumors. There is therefore a pressing need to identify new therapeutic targets to provide theoretical support for the clinical treatment of digestive system cancer.

Long non-coding RNAs (lncRNAs) are single-strand RNA molecules >200 nucleotides long, which are transcribed by RNA polymerase II and lack protein-coding ability (11). Extensive research has underscored the substantial impact of lncRNAs on the development, proliferation, migration and prognosis of various types of cancer. These lncRNAs interact with target genes at the transcriptional level, regulating a series of biological processes, such as histone modification and chromatin remodeling. They also function as competing endogenous RNAs (ceRNAs) that interact with microRNAs (miRNAs), which are ~22 nucleotides long (1113). For example, lncRNA BC069792 acts as a ceRNA sponge to interact with miR-658 and miR-4739, and increases the expression of the target gene KCNQ4, leading to AKT phosphorylation, and subsequently to inhibition of the proliferation and invasion of breast cancer in vitro and in vivo (14).

Small nucleolar RNA host gene 16 (SNHG16) is a member of the SNHG family that is located on human chromosome 17q25.1 and consists of four exons. SNHG16 was initially identified as a potent oncogenic factor and has been reported to promote the progression of neuroblastoma (15). In addition, SNHG16 has been recognized as non-coding RNA that is expressed in aggressive neuroblastoma (15). Numerous studies have further revealed that SNHG16 is extensively involved in the complex molecular regulatory network of various types of human cancer (1618). For example, the knockdown of SNHG16 has been reported to suppress the proliferation and radioresistance of nasopharyngeal carcinoma cells by regulating the miR-31-5p/SFN axis (19). Furthermore, SNHG16 has been implicated as an oncogene, capable of promoting the proliferation and reducing the apoptosis of bladder cancer cells. This effect was shown to be achieved by binding and recruiting the enhancer of zeste homolog 2 (EZH2) to p21 promoter and silencing the expression of p21 (20). SNHG16 has also been shown to serve a key role in the staging, distant metastasis and poor prognosis of ovarian cancer by increasing the expression of MMP9 (21). In oral squamous cell carcinoma, the expression of SNHG16 is regulated by the transcription factor c-Myc, which recruits histone acetyltransferase and induces RNA polymerase II clearance (22). These findings suggested that SNHG16 has an important role in the progression, invasion and carcinogenesis of human cancer through upstream regulatory and downstream molecular mechanisms.

The present review begins with a concise summary of the relationship between risk factors, such as smoking, and human digestive system tumors. Then, it delves into an examination of research on the expression, biological function, related mechanisms and potential clinical significance of SNHG16 for digestive tumors, indicating a connection between SNHG16 and digestive cancers. Additionally, it outlines the association of these risk factors with SNHG16 in all reported publications. These findings collectively underscore the potential of SNHG16 as both a potential biomarker and therapeutic target in human digestive system tumors.

Risk factors and human digestive system cancer

Several factors have been reported to have a significant import on cancer etiology, including smoking, excessive alcohol consumption, physical activity, infection, radiation, living environment, family history, diet, disease and genomic characteristics. In the subsequent sections, the association of these risk factors with human digestive system cancers is described.

Smoking

Smoking is the leading cause of cancer, and smokers are at a higher risk of developing digestive system disorders, including digestive tract cancers (23,24). In 2019, tobacco smoking was responsible for ~203,000 deaths of patients with EC worldwide (25). Increasing evidence has demonstrated that smoking is closely associated with the development and progression of HCC, with 13% of HCC cases reported to be caused by smoking worldwide (26,27). In a statistical analysis of HCC, patients were categorized as non-smokers, current smokers and ex-smokers, according to smoking status; notably, non-smokers had higher late survival rates than current smokers and ex-smokers (28). For PC, tobacco smoking is considered a major risk factor, with former or current smokers exhibiting a higher odds ratio of 1.42–1.74 than non-smokers (24,29). A number of causative factors have been epidemiologically confirmed to have an association with PC, among which smoking shows the most positive correlation with the risk of PC and is a recognized risk factor (3033). Furthermore, smoking has been shown to affect the prognosis of patients with PC (34). Similarly, smoking constitutes an established risk factor for CRC. Compared with 6,866 healthy individuals, 6,264 patients with CRC had a higher smoking status, suggesting a strong association between smoking and CRC evident across early- and late-stage CRC (35).

Notably, research in esophageal squamous cell carcinoma (ESCC), bladder cancer, non-small lung cancer and CRC has demonstrated that there is no correlation between SNHG16 expression and smoking (20,3638). Specifically, in lung cancer, SNHG16 expression was not related to the clinical data of patients, including age and smoking history (39). Furthermore, a meta-analysis study indicated that SNHG16 expression was not associated with smoking (40). By contrast, in a study on CRC, Zhou et al revealed that smoking influenced the combination of rs7353, rs8038 and rs15278 sites located in the SNHG16 gene. In addition, through multifactor dimensionality reduction analysis, changes in the expression levels of SNHG16 were revealed to increase or decrease the risk of CRC susceptibility (41). In the future, the association between smoking and SNHG16 expression in other types of cancer requires further research.

Excessive alcohol consumption

Similar to smoking, excessive alcohol consumption is associated with an increased risk of all digestive system tumors, including EC (24,42), HCC (43), PC (44), GC (45) and CRC (46). Nevertheless, moderate alcohol consumption has shown no association with certain types of digestive cancer, such as HCC, PC, GC and CRC (24,47,48). An Australian study revealed that the risk of ESCC was significantly increased with combined tobacco and alcohol use, surpassing >20-fold higher risk compared with non-smokers and non-drinkers (49).

In addition to smoking, Zhou et al (41) also revealed that drinking was a factor affecting SNHG16 single nucleotide polymorphisms and expression, thus affecting CRC susceptibility. To the best of our knowledge, the association between excessive alcohol consumption and SNHG16 expression has not been reported in other diseases.

Physical activity

Physical activity involves the use of skeletal muscles and requires energy expenditure. Numerous studies have demonstrated the association between physical activity and cancers (5053). Physical activity can decrease the risk of EC by 19–51%, GC by 15–19% and colon cancer by 21–27% (50). In particular, high levels of physical activity, such as running and jumping rope, may decrease the risk of PC by 9–25% (50). Moore et al reported that high levels of physical activity decreased the risk of EC, HCC and GC by >20% (51). Kasvis and Kilgour (53) suggested that physical activity interventions may alleviate malnutrition and muscle wasting, which are common in PC. In China, a decade-long prospective study showed that CRC risk was 25% lower in the highest-level-of-activity group compared with in the lowest-level-of-activity group (52). In addition, a meta-analysis showed that a moderate-to-high physical activity level serves as a common protective factor that can significantly reduce the overall risk of digestive system cancer (54). To date, there is an absence of evidence to indicate the relationship between physical activity and SNHG16 expression, which should be explored in the future.

Infection

Evidence has suggested that bacterial infection serves a key role in tumor progression, such as in EC (55). Porphyromonas gingivalis is an important periodontal disease pathogen that has been detected in 61% of ESCC tissues (56). It has been suggested that EC is moderately positively associated with chronic hepatitis C virus (HCV) infection, with a combined relative risk of 1.61 (95% CI, 1.19–2.17) (57). For HCC, the most common risk factor is chronic hepatitis caused by hepatitis B virus (HBV) and HCV infection, and long-term chronic hepatitis can lead to cirrhosis and eventually develop into HCC (58). Furthermore, HBV products and HBV mutations may disrupt normal cell signaling pathways, leading to HBV-induced HCC (59). Fusobacterium has been identified as a potential prognostic biomarker for PC (60). A prospective study reported that patients with high concentrations of P. gingivalis have a higher risk of PC (61). Helicobacter pylori, which is found only in the human stomach, has been shown to be closely associated with GC as a separate risk factor (62,63). Substantial evidence has suggested that H. pylori carrying CagA and VacA virulence factors is highly associated with distal GC by promoting GC epithelial-mesenchymal transition (EMT) through disruption of the gastric tissue microenvironment (6467). The Epstein-Barr virus infection can also cause GC, accounting for ~10% of patients with GC (68). In patients with CRC, Escherichia coli has been reported to contain a polyketide synthase gene that not only induces inflammation, epithelial cell damage and cell proliferation, but also encodes colibactin, which destroys DNA and ultimately leads to the formation of CRC (69). Fusobacterium has also been reported to be associated with the occurrence of CRC (70).

In some clinical samples of HCC, HBV infection showed no correlation with the expression of SNHG16 (7175). In addition, in a meta-analysis by Liu et al, there was no association detected between SNHG16 expression and HBV infection (76). To the best of our knowledge, in other human digestive system tumors, the association between infection and SNHG16 expression has not been reported. In some cells, SNHG16 expression was upregulated in Cryptococcus-treated dendritic cells compared with in wild-type dendritic cells (77). In addition, Mycobacterium tuberculosis infection can increase the expression levels of SNHG16 in a dose- and time-dependent manner in macrophages (78).

Radiation

Radiation is a common tool in modern medicine, including ionizing radiation and radiotherapy, and is one of the main treatments for cancer (7981). However, the disadvantages of radiation cannot be overlooked. In patients with head and neck tumors, the risk of ESCC is associated with the dose of radiotherapy (80). In addition, α-radiation emitted by plutonium is strongly associated with genetic mutations in HCC (82). Dores et al (83) proposed that both radiotherapy and chemotherapy can substantially increase the risk of PC in Hodgkin lymphoma survivors treated previously. In addition, Yusefi et al (84) showed that ionizing radiation is a possible risk factor for GC. Low-dose radiation exposure among uranium miners has been reported to be positively associated with GC (85). Computerized tomography radiation slightly increases the risk of CRC, whereas the benefits of computerized tomography (CT) radiation far outweigh the risks (86). Notably, to the best of our knowledge, the association between radiation and SNHG16 expression has not been reported in disease.

Living environment

A study in China revealed that drinking from untreated water sources can increase the risk of ESCC by 2-fold (87). The use of polluted water containing nitrate is considered an essential risk factor for HCC (88), PC (89), GC (90) and CRC (91). Exposure to external airborne agents, such as fine particulate matter, may also increase the risk of digestive system cancer, especially EC and GC (92,93). Tsai et al (93) demonstrated that PM2.5 was strongly associated with the mortality of HCC, which agrees with the results of another study, where a strong association was detected between PM2.5 and HCC and CRC (94). A previous study reported that SNHG16 expression presented no significant differences between Intensive Care Unit (ICU)-hospitalized and non-ICU hospitalized patients (95). To the best of our knowledge, in digestive system cancer, the effect of living environment on SNHG16 expression is not known.

Family history

A number of studies have shown that a family history of cancer is strongly associated with the incidence rate of certain types of cancer, such as EC (96), HCC (97), PC (98), GC (99) and CRC (100). Parents and siblings of a person with EC and HCC have been reported to exhibit a higher risk of developing EC and HCC. Research has found that there is a clear ‘dose-response’ relationship with the number of first-degree relatives of EC (97,101). Furthermore, familial inheritance is a known cause of GC (102). A positive first-degree family history of CRC and GC can reduce the risk of cancer recurrence and death compared with patients without a family history, based on a well-defined cohort enrolled in a clinical trial (103,104). Su et al (105) reported that people with a family history of EC had a 2-fold higher risk of developing the disease with a poorer prognosis. However, in another study, the HCC survival rate was higher in the familial cancer group than in the sporadic cancer group (106). Furthermore, individuals with a family history of CRC have been shown to have a slightly increased risk of getting PC (107).

In a study on CRC, Zhou et al indicated that family history affected the combination of rs7353, rs8038 and rs15278 sites of the SNHG16 gene, which increased or decreased SNHG16 expression and influenced CRC susceptibility (41). Whereas in other diseases, this relationship has not been presented.

Diet

There is a consensus on the strong association between diet and digestive system tumors. Long-term unbalanced diets, such as high-calorie hot beverages or food, can lead to esophageal epithelial cell damage and exacerbate the risk of EC (108). A number of studies have demonstrated that reducing vegetable and fruit intake, and low-fiber diets, may increase the risk of HCC and PC, and increasing the intake of salted and preserved foods and meat could increase the risk of HCC and GC (109114). Similarly, diet also influences CRC. For example, calcium, fiber, milk, wholegrains and 2′5-hydroxyvitamin D have been shown to inhibit the development of CRC; however, consumption of a large amount of red or culinary meat can increase the risk of CRC (115,116). Furthermore, patients with CRC have been reported to be deficient in vitamin C, vitamin E and folate (117). To the best of our knowledge, the association between diet and SNHG16 expression has not been published.

Disease

Existing studies have shown that both infectious diseases and chronic inflammation may account for ~25% of carcinogenic factors (118). Reactive oxygen/nitrogen species are produced under the condition of chronic inflammation, which can cause DNA damage in various organs, thereby inducing cellular carcinogenesis (118,119). Furthermore, prolonged acid reflux can cause reflux esophagitis in the proximal esophagus and expedite esophageal carcinogenesis (120). Non-alcoholic fatty liver disease can result in a series of diseases, including steatosis accumulation, non-alcoholic steatohepatitis, inflammation, liver fibrosis and cirrhosis, which may eventually lead to HCC (121123). Chronic pancreatitis is also a causative factor in the development of PC. Patients who have had this disease for >2 years face a 2.71-fold higher risk of developing PC (124). Chronic gastritis, one of the most common types of chronic inflammation, is considered a precursor of GC (125,126). Chronic enteritis and dysbiosis of the intestinal microflora can increase the risk of CRC (127). Notably, diabetes mellitus (DM) and obesity are risk factors for digestive system cancer, enhancing the development of EC (128), HCC (29), PC (129,130), GC (131,132) and CRC (133). For example, type 2 DM (T2DM) is often recognized as an independent risk factor for HCC, with a 2- to 4-fold increased risk in patients with T2DM compared with the general population (134).

In HCC, liver cirrhosis has been reported to not necessarily be associated with the expression of SNHG16 (71,74,75,135). By contrast, investigations into the association between portal vein tumor thrombus (PVTT) and SNHG16 expression have yielded dissimilar results. Guo et al (135) revealed a positive correlation between SNHG16 expression and PVTT, while another study indicated that high SNHG16 expression was independent of PVTT (136). Patients with sepsis and acute respiratory distress syndrome (ARDS) have been shown to exhibit a decline SNHG16 expression compared with those without ARDS, indicating that SNHG16 may possess a certain ability to discriminate patients with sepsis and ARDS from those without ARDS, according to the area under curve (137). Moreover, SNHG16 in patients with sepsis has been discovered to have a negative correlation with diabetes and chronic obstructive pulmonary disease history, rather than other medical history, such as hypertension (137). In addition, some studies have found that SNHG16 serves an important role in sepsis-induced acute lung injury and inflammation via an involvement in the pathogenesis of ARDS (138140). In patients with acute ischemic stroke, SNHG16 expression was revealed to be negatively related to comorbidities, such as hyperlipidemia and disease severity (141). SNHG16 has been shown to be upregulated in unilateral ureteral obstruction-induced renal fibrotic tissues of mice (142).

Genomic characteristics

Cancer is a multi-stage process disease and its occurrence is not only disturbed by external factors, but also by intrinsic genetic mutations. The occurrence of genetic mutations serves an important role in the development of digestive system tumors. KRAS, a proto-oncogene, affects the cellular proliferation and differentiation in digestive system cancer, and can influence the prognosis of these patients (143,144). KRAS mutations have been found in ~85% of patients with PC and ~45% of patients with CRC (145). Similar results have been reported regarding tumor suppressor genes. Mutations in the TP53 gene usually occur in the early stages of GC and can accelerate the progression of GC (146). In HCC, TP53 mutations have been detected in circulating exosomal DNA and are associated with the prognosis of patients (147). APC mutations are associated with tumorigenesis, affecting the overall survival of patients with CRC (148,149). A previous meta-analysis showed the neutral function of PIK3CA mutations on the overall survival and progression-free survival of patients with CRC (144). Specifically, individuals who have both genetic and lifestyle-related risks have a ~190 times higher risk of ESCC than those without these risks (150). To the best of our knowledge, the association between genomic characteristics and SNHG16 expression has not yet been presented.

Epigenetic influences, such as DNA methylation, histone modifications and non-coding RNA regulation, are also important factors (151). The role of lncRNA in the process of cancer development and progression cannot be overlooked. A number of studies have shown that SNHG16 is a key factor in the process of digestive system cancer, including promoting the proliferation of cancer cells, resisting cancer therapeutic drugs and enhancing cancer cell invasiveness. The possible mechanisms and functional characterization of SNHG16 in human digestive system cancer are presented in Fig. 1 and Table I, respectively. Furthermore, the association between SNHG16 expression and clinicopathological characteristics is summarized in Table II.

Table I.

Functional characterization of SNHG16 in human digestive system cancer.

Table I.

Functional characterization of SNHG16 in human digestive system cancer.

A, EC

Clinical sampleCellular levelFunctional roleAnimal level




First author, yearSample sizeExpression in tissueCell linesExpression in cancer cellsRelated genesProliferationApoptosis Invasion/Migration ChemoresistanceName of cell line injected into the animalsIn vivo modelEffect on tumors(Refs.)
Han, 2018128UpHEEC, EC-1,Up β-catenin,PromoteInhibitPromoteNANANANA(36)
ECa-109, TE-13, cyclin-D1,
TE-1 c-Myc
Zhang, 201868UpHEEC, kyse-30,UpmiR-140-5p,PromoteInhibitPromoteNAkyse-70BALB/cInhibit(152)
kyse-70, Eca109, ZEB1 (kd-SNHG16)athymic
EC9706, TE1 nude mice
Ren, 202255UpHET-1A, Eca109,UpEIF4A3,PromoteInhibitPromoteNAEca-109BALB/cPromote(154)
KYSE30, RhoU (oe-SNHG16nude mice(oe);
KYSE140, and kd- inhibit
KYSE410 SNHG16) (kd)
Zhang, 2022NANAEC9706,UpmiR-802,PromoteNANANAEC9706BALB/cInhibit(153)
KYSE150 PTCH1 (kd-SNHG16)nude mice

B, HCC

Clinical sampleCellular levelFunctional roleAnimal level




First author, yearSample sizeExpression in tissueCell linesExpression in cancer cellsRelated genesProliferationApoptosis Invasion/Migration ChemoresistanceName of cell line injected into the animalsIn vivo modelEffect on tumors(Refs.)

Xie, 201940UpLO2, HepG2,UpmiR-195PromoteNAPromoteNAHepG2BALB/cInhibit(155)
Huh7, SMMC7721, (kd-SNHG16)nude mice
Hep3B, Bel7402
Lin, 201988aUpLO2, Huh7,UpmiR-4500,PromoteInhibitPromoteNANANANA(72)
SMMC721, HepG2, E-cadherin,
MHCC-97H STAT3,
N-cadherin
Ye, 2019103UpLO2, HepG2,UpmiR-140-5pNANANAPromoteHepG2Nude miceInhibit(74)
SK-hep1, Huh7, (kd-SNHG16)
HCCLM3
Li, 202123UpLO2, Huh7, HepG2,UpmiR-4500,PromoteNAPromoteNAHuh7BALB/cInhibit(156)
MHCC97H, GALNT1 (kd-SNHG16)nude mice
MHCC97L
Zhong, 2020108UpLO2, Huh7, HepG2,UpmiR-17-5p,PromoteInhibitPromoteNAHepG2 (oe-AthymicPromote(71)
SMCC7721, p62 SNHG16 andnude mice(oe);
QGY-7703 kd-SNHG16) inhibit
(kd)
Chen, 201950UpLO2, Hep-3B,UpmiR-186,PromoteNAPromoteNAHep-3BBALB/cPromote(73)
Huh7, PLC, ROCK1 (oe-SNHG16),nude mice(oe);
Sk-hep-1, SMMC- Sk-hep-1 inhibit
7721 (kd- SNHG16) (kd)
Li, 201934UpLO2, Huh7, HepG2,UpmiR-302a-PromoteNANANANANANA(157)
SMMC7721, 3p, FGF19
SK-Hep1, Hep3B
Li, 202047UpLO2, MHCC97H,Uplet-7b-5p,PromoteInhibitPromotePromoteHuh7BALB/cInhibit(158)
Huh7, SMMC7721, CDC25B, (kd-SNHG16)nude mice
Hep3B, HepG2 CDK1,
HMGA2
Guo, 201961UpLO2, SK-Hep-1,upNAPromoteInhibitPromotePromoteNANANA(135)
Huh7, Hep3B,
HepG2
Jing, 202040UpHep3BUpmiR-23b-3p,PromoteInhibitNAPromoteHep3BNOD/Inhibit(159)
EGR1 (kd-SNHG16)SCID
male mice
Xu, 202388UpLO2, MHCC97,UpmiR-942-3p,NANAPromoteNAMHCC97Nude miceInhibit(160)
HCCLM3 MMP9 (kd-SNHG16)
Hu, 202078UpLO2, HCCLM3,UpmiR-605-3p,NANAPromoteNAHCCLM3Nude miceInhibit(75)
MHCC97L, TRAF6, (kdSNHG16)
MHCC-97H, NF-κB HepG2
Hep3B, HepG2 (kd-SNHG16)
Zhang, 2022158UpLO2, SMMC-7721,UpECM-PromoteNAPromoteNANANANA(136)
HepG2, Hep3B, receptor
MHCC-97L,
MHCC-97H,
HCCLM3, Huh7,
BEL-7402, PLC/
PRF/5, SK-Hep1
Xu, 201843DownTHLE2, THLE3,DownmiR-93InhibitNANAInhibitHuh7FemaleInhibit(165)
Hep3B, Huh7, (oe-SNHG16)athymic
SNU398, SNU423, nude mice
SNU429, Hep3G2,
SK-HEP-1, PLC/
PRF/5

C, PC

Clinical sampleCellular levelFunctional roleAnimal level




First author, yearSample sizeExpression in tissueCell linesExpression in cancer cellsRelated genesProliferationApoptosis Invasion/Migration ChemoresistanceName of cell line injected into the animalsIn vivo modelEffect on tumors(Refs.)

Yu, 2019NANAHPDE6-C7,UpmiR-195,PromoteNAPromoteNANANANA(168)
AsPC-1, Panc-1, SREBP2
BxPC-3, SW1990
Xu, 202130UpHPY-Y5, BxPC3,UpmiR-302b-3p,PromoteInhibitPromoteNANANANA(166)
Panc-1 SLC2A4
Yu, 2022350UpHPDE, SW1990,UpEZH2,PromoteInhibitNAPromoteNANANA(167)
PANC-1, ASPC-1, Smad4
BxPC3

D, GC

Clinical sampleCellular levelFunctional roleAnimal level




First author, yearSample sizeExpression in tissueCell linesExpression in cancer cellsRelated genesProliferationApoptosis Invasion/Migration ChemoresistanceName of cell line injected into the animalsIn vivo modelEffect on tumors(Refs.)

Ding, 202255UpGES-1, AGS,UpmiR-506-3p,PromoteInhibitNAPromoteAGSNude miceNA(169)
BGC-823, MKN-45, PTBP1
MGC-803, SGC-
7901
Lian, 2017122UpGES-1, AGS,UpNAPromoteInhibitPromoteNAMGC-803BALB/cInhibit(170)
BGC-823, (kd-SNHG16)nude mice
MGC-803,
SGC-7901, MKN45
Zhao, 202260UpGES-1, AGS,Upc-Myc, P53,PromoteInhibitPromoteNANANANA(171)
SGC-7901, P21, P27,
BGC-823 cyclin-E1/A2/
D1, cdk2/6
Zhou, 201920UpAGS, HGC27NADKK3PromoteNAPromoteNANANANA(172)
Yang, 202260UpRGM-1, GIST882,UpmiR-128-3p,PromoteInhibitPromoteNAGIST882BALB/cInhibit(173)
GIST48, GIST430, CASC3 (kd-SNHG16)nude mice
GIST-T1
Wang, 201932UpGES-1, BGC823,UpmiR-135a,PromoteInhibitPromoteNANANANA(174)
MGC803, MKN45, JAK2,
SGC7901 STAT3

E, CRC

Clinical sampleCellular levelFunctional roleAnimal level




First author, yearSample sizeExpression in tissueCell linesExpression in cancer cellsRelated genesProliferationApoptosis Invasion/Migration ChemoresistanceName of cell line injected into the animalsIn vivo modelEffect on tumors(Refs.)

Li, 201956UpCCC-HIE-2,UpmiR-PromoteNAPromoteNALoVoMalePromote(38)
CaCO-2, SW480, 200a-3p (oe-SNHG16)BALB/c
HCT116, LoVo nude mice
Chen, 2022120UpFHC, SW480,UpmiR-124-3p,PromoteInhibitPromoteNASW480Nude miceInhibit(176)
HCT116, DLD-1, MCP-1 (kd-SNHG16)
LoVo
Tan, 202220UpNCM460, HCT116,UpmiR-214-3p,PromoteInhibitNANAHCT116MaleInhibit(177)
HT29, SW480, ABCB1 (kd-SNHG16)BALB/c
SW620, CaCo2 nude mice
He, 202050UpCCD841 CoN,UpmiR-132-3p,PromoteInhibitPromoteNASW620MaleInhibit(178)
SW480, SW620 USP22 (kd-SNHG16)BALB/c
nude mice
Ke, 2020NANAHCT116, CaCo-2NAmiR-302a-PromoteNANANANANANA(179)
3p, AKT
Christensen,606UpHT29, HCT116,NAASCL2,PromoteInhibitPromoteNANANANA(182)
2016 HCT15, Colo205, ETS2,
DLD1, LS174T, c-Myc
SW620, SW480,
CaCo2
Xiang, 2022111UpNCM-460, DLD-1,UpmiR-195-5p,PromoteNAPromoteNAHCT116Nude miceInhibit(183)
HCT-116, HT-29, YAP1, (kd-SNHG16)
SW620, SW480, TEAD1
LoVo, CaCo2
Zhou, 2020721UpNANANANANANANANANANA(41)

{ label (or @symbol) needed for fn[@id='tfn1-or-52-2-08765'] } CRC, colorectal cancer; EC, esophageal carcinoma; GC, gastric cancer; HCC, hepatocellular carcinoma; kd, knockdown; NA, not applicable; oe, overexpression; PC, pancreatic cancer; PVTT, portal vein tumor thrombus; SNHG16, small nucleolar RNA host gene 16; miRNA/miR, microRNA

a 10 samples from serum, remaining samples from tissue.

Table II.

Association between SNHG16 expression and clinicopathological characteristics in human digestive system cancer.

Table II.

Association between SNHG16 expression and clinicopathological characteristics in human digestive system cancer.

A, EC

Expression of SNHG16 and clinical features

First author, yearSexAgeSmoking statusDegree of tumor differentiationTumor sizeOverall survivalInvasion depthLymph nodes metastasisTumor stageHistological gradeClinical stageTumor locationHBV infectionPVTTLiver cirrhosisMacrovascular invasion(Refs.)
Han, 2018/++++++/////(36)
Ren, 2022+/+////////(154)

B, HCC

Expression of SNHG16 and clinical features

First author, yearSexAgeSmoking statusDegree of tumor differentiationTumor sizeOverall survivalInvasion depthLymph nodes metastasisTumor stageHistological gradeClinical stageTumor locationHBV infectionPVTTLiver cirrhosisMacrovascular invasion(Refs.)

Xie, 2019////++///////(155)
Lin, 2019//++/++++//////(72)
Ye, 2019/+/+/++//////(74)
Zhong, 2020//++//////+(71)
Chen, 2019//+///++///++///(73)
Guo, 2019//+++/++////++/(135)
Hu, 2020/++///////(75)
Zhang, 2022/++////////(136)

C, PC

Expression of SNHG16 and clinical features

First author, yearSexAgeSmoking statusDegree of tumor differentiationTumor sizeOverall survivalInvasion depthLymph nodes metastasisTumor stageHistological gradeClinical stageTumor locationHBV infectionPVTTLiver cirrhosisMacrovascular invasion(Refs.)

Xu, 2021//+++/+///////(166)

D, GC

Expression of SNHG16 and clinical features

First author, yearSexAgeSmoking statusDegree of tumor differentiationTumor sizeOverall survivalInvasion depthLymph nodes metastasisTumor stageHistological gradeClinical stageTumor locationHBV infectionPVTTLiver cirrhosisMacrovascular invasion(Refs.)

Lian, 2017/++/++++++//////(170)
Zhao, 2022///+///////(171)
Wang, 2019//++++//+///////(174)

E, CRC

Expression of SNHG16 and clinical features

First author, yearSexAgeSmoking statusDegree of tumor differentiationTumor sizeOverall survivalInvasion depthLymph nodes metastasisTumor stageHistological gradeClinical stageTumor locationHBV infectionPVTTLiver cirrhosisMacrovascular invasion(Refs.)

Chen, 2022/////++//++///////(176)
Tan, 2022/////+++///////(177)
Xiang, 2022/+++++//////(183)
Zhou, 2020////////+//////(41)

[i] CRC, colorectal cancer; EC, esophageal carcinoma; GC, gastric cancer; HBV, hepatitis B virus; HCC, hepatocellular carcinoma; PC, pancreatic cancer; PTVV, portal vein tumor thrombus. ++, P<0.01; +, P<0.05; -, P>0.05; /, not applicable. P<0.05 was considered statistically significant.

SNHG16 and human digestive system cancer

SNHG16 and EC

EC is one of the major cancer types worldwide, ranking 7th (3.1%, 604,100 new cases) and 6th (5.5%, 544, 076 deaths) among all types of cancer in terms of incidence and mortality rate, respectively (2). Its incidence and mortality rates vary between geographic regions (2). For example, due to economic underdevelopment and dietary habits, the burden of EC is higher in East Asia with a predominance of patients with ESCC (2). Studies have shown that SNHG16 expression is upregulated in EC, and is closely associated with tumor stage, lymph node metastasis and clinical stage (36,152154). The knockdown of SNHG16 has been reported to suppress the proliferation and invasion, and promote apoptosis by reducing the expression of β-catenin, cyclin D1 and c-Myc protein in EC-1 and Eca-109 cells (36). In addition, Zhang et al verified by reverse transcription-quantitative PCR (RT-qPCR) that the expression levels of SNHG16 were upregulated in EC tissues or cells compared with those in normal tissues or cells (P<0.01). This previous study also observed that the disruption of SNHG16 expression suppressed proliferation, promoted apoptosis and inhibited EMT through the miR-140-5p/ZEB1 axis in vivo and in vitro (152). In another study on ESCC, the expression of SNHG16 was revealed to be associated with tumor differentiation and T stage, and increased expression of SNHG16 could promote ESCC growth and metastasis. The underlying mechanism may be that SNHG16 binds to and recruits EIF4A3 to modulate RhoU expression, thereby enhancing the stability of RhoU mRNA (154). Zhang et al (153) demonstrated that SNHG16 acts as a sponge of miR-802 to upregulate PTCH1 and activate the Hedgehog pathway, thus facilitating EC proliferation and self-renewal.

These results show that the upregulation of SNHG16 may be strongly associated with the development of ESCC, suggesting the potential utility of SNHG16 as a marker for ESCC. These insights offer novel avenues for the clinical management of ESCC.

SNHG16 and liver cancer

HCC is one of the most common malignancies worldwide, accounting for 4.7% (906,000 new cases) of all new cancer cases and 8.3% (830,000 deaths) of all cancer-related mortalities. HCC is ranked as the 6th most commonly diagnosed cancer and the 3rd leading cause of cancer-related deaths (2). In most studies, SNHG16 has been considered a proto-oncogene of HCC, and RT-qPCR has been used to detect the expression of SNHG16 in HCC tissues and corresponding non-tumor tissues. The results showed that the expression levels of SNHG16 in HCC samples were much higher than those in matched non-tumor samples, and upregulation of SNHG16 expression was highly associated with poor prognosis and tumor stage of HCC. Furthermore, the patients with advanced-stage HCC exhibited a significantly higher SNHG16 expression level than the patients with early-stage HCC (72,155). Moreover, the high expression of SNHG16 has been shown to be associated with the tumor size, TNM stage and vascular infiltration of patients with HCC (74). SNHG16, as a ceRNA, can target STAT3 and GALNT1 through sponging miR-4500 in Huh7 cells and human umbilical vein endothelial cells (HUVECs), respectively, to promote proliferation, metastasis and invasion of Huh7 cells, and enhance angiogenesis of HUVECs (72,156). In addition, through regulating miR-195, miR-17-5p/P62, miR-302a-3p/FGF19 and miR-186 expression, SNHG16 can inhibit the proliferation, migration and invasion of HepG2 and Hep3B cells (71,73,155,157). Overexpression of SNHG16 may also affect the G2/M transition of HCC cells by regulating CDC25B expression through sponging miR-let-7b-5p (158). A previous study reported that SNHG16 is upregulated in sorafenib-resistant tumor tissues and cells, and that the overexpression of SNHG16 can enhance sorafenib resistance in HCC (74). By contrast, when the expression of SNHG16 is suppressed, sorafenib resistance disappears (135). Jing et al (159) also suggested that SNHG16 may enhance HCC autophagy via the miR-23b-3p/EGR1 axis and protect HCC from sorafenib resistance. In addition, it has been reported that SNHG16 can be phagocytized by telocytes and can mediate telocytes to promote HCC cell metastasis by regulating the miR-942-3p/MMP9 axis (160). Furthermore, Hu et al (75) demonstrated that the overexpression of SNHG16 promotes TRAF6 expression by sponging miR-605-3p, activates NF-κB and exacerbates the development of HCC. Specifically, the activated NF-κB can enhance SNHG16 promoter activity, forming a positive SNHG16/NF-κB feedback loop that further worsens HCC (75). The overexpression of SNHG16 has also been shown to be associated with tumor recurrence and poor prognosis after surgery, and mechanistic analyses suggested that SNHG16 markedly activates the extracellular matrix-receptor interaction pathway (136).

Studies have demonstrated that SNHG16 regulates a large lncRNA-miRNA-mRNA network in HCC, and is closely associated with the infiltration of immune cells, the release of immunomodulatory factors and the expression of chemokines in tumor tissues (161163). Notably, UBE4B and SEMA3F may promote HCC progression regulated by their upstream SNHG16/miR-22-3p and SNHG16/let-7c-5p axes, respectively (163,164). Liu et al (76) revealed that SNHG16 can be used as a potential biomarker for patients with HCC with a poor prognoses. In summary, SNHG16 may be upregulated in HCC and can promote HCC development. Notably, a previous study presented the opposite argument, suggesting that SNHG16 expression may be reduced in HCC tissue compared with in normal liver tissue, and that overexpression of SNHG16 could decrease the proliferation of Hep3B and Huh7 cells, and inhibit HCC development and chemoresistance via sponging miR-93 (165). This discrepancy in findings may stem from the diverse dysregulation patterns of SNHG16 in human cancer, with its expression being either upregulated or downregulated. Such variations could be influenced by the specific cancer types, their anatomical locations and the microenvironments involved (165). This discrepancy prompts further research into the role of SNHG16 in HCC.

SNHG16 and PC

PC is one of the most serious malignancies of the digestive system. Due to its poor prognosis, PC accounts for almost as many deaths (466,000) as cases (496,000) in the world, and is the 7th leading cause of cancer death in both men and women (2). Similar to EC, the incidence rate of PC is 4-fold and 5-fold higher in high human development index (HDI) countries compared with in low HDI countries (2). Studies have shown that the expression levels of SNHG16 are upregulated in PC tissue compared with those in normal tissue (166,167). Altering the expression of SNHG16 may inhibit the adipogenesis of AsPC-1 and PANC-1 cells through the miR-195/SREBP2 axis (168). Inhibition of SNHG16 expression can result in the release of miR-302b-3p, which inhibits SLC2A4 expression and promotes apoptosis in PC cells (166). Overexpression of SNHG16 has also been reported to be closely associated with gemcitabine resistance in PC cells. SNHG16 can interact with EZH2, suppressing SMAD4 expression via EZH2 binding to the SMAD4 promoter (167). Downregulation of SMAD4 has a reduced ability to inhibit AKT phosphorylation, thereby promoting gemcitabine resistance in PC cells (167). These findings suggested that SNHG16 may serve a critical role in the development of PC and that it could be regarded as a marker of poor prognosis in PC.

SNHG16 and GC

GC remains an important type of cancer worldwide, and is considered the 5th most frequent malignancy (5.6%, >1,000,000 new cases) and the 4th most common cause of death (7.7%, ~769,000 deaths) among oncological patients (2). The region with the highest age-standardized incidence rate is Eastern Asia, followed by Central and Eastern Europe (2). The expression of SNHG16 is significantly associated with the depth of infiltration, lymph node metastasis, TNM stage, histological differentiation and PTBP1 expression of GC (169,170). Knockdown of SNHG16 can significantly suppress the migration, invasion and arrest of cells in the G1 phase, and can decrease c-Myc expression, and affect the formation of the p27/cyclin D1/CDK6, p53/cyclin E1 and cyclin A2/CDK2 complexes (169171). A number of patients with GC develop 5-fluorouracil (5-FU) resistance, showing a higher vulnerability than parental GC cells. Notably, blocking the SNHG16/miR-506-3p/PTBP1 axis may effectively limit 5-FU-resistant GC cell originated-xenograft tumor growth under 5-FU treatment. Specifically, PTBP1 stabilizes the mRNA expression of glycolysis enzymes by directly binding to 3′UTR regions (169). In addition, it has been shown that SNHG16 can promote EMT by downregulating the WNT signaling pathway and inhibiting DKK3 expression, and can regulate β-catenin protein expression without participating in the β-catenin translocation between the cytoplasm and nucleus (172). In particular, SNHG16 activated by CCCTC binding factor (CTCF) can modulate gastrointestinal stromal tumor cell proliferation, migration, invasion and apoptosis through the miR-128-3p/CASC3 axis (173). In another study, SNHG16 was also demonstrated to be able to mediate the upregulation of JAK2 and STAT3 by sponging miR-135a to influence the proliferation, invasion and apoptosis of GC cells, with SNHG16 being regulated by phosphorylated-STAT3 directly or indirectly (174). In summary, SNHG16 may be closely related to the occurrence and development of GC, and could be a potential marker of poor GC prognosis.

SNHG16 and CRC

Notably, >1,900,000 new cases of CRC and 935,000 CRC-related deaths occurred worldwide in 2020; during this year, it was the third most common cancer, after female breast cancer and lung cancer, and it exhibited a close mortality rate to lung cancer (2). Growing evidence has suggested that the expression levels of SNHG16 are positively associated with advanced TNM stage, distant metastasis and shorter overall survival time in CRC (38,175177). SNHG16 is mainly present in the cytoplasm, functioning as a ceRNA to regulate multiple miRNAs and target genes. Li et al (38) revealed that SNHG16 was associated with malignancy and poor prognosis in patients with CRC by sponging miR-200a-3p. Tan et al (177) indicated that SNHG16 could promote CRC proliferation by upregulating its target gene ABCB1 through interacting with miR-214-3p. He et al (178) concluded that SNHG16 could activate USP22 expression to promote CRC progression via absorbing miR-132-3p. Ke et al (179) demonstrated that SNHG16 supported colon cancer cell proliferation by targeting the miR-302a-3p/AKT axis. Chen et al (176) revealed that the expression of SNHG16 was higher in cancer tissues from patients than in the matched normal tissues, and was positively related to CRC grade. It was also revealed that SNHG16 may serve a contributory role in the proliferation, migration and EMT of CRC cells through the miR-124-3p/MCP-1 axis (176). Some bioinformatics analyses also reached a similar conclusion, in that SNHG16 may have an important role in CRC (175,180). In particular, SNHG16 has been reported to be closely associated with autophagy in CRC (175,181).

The expression of SNHG16 may be activated by other proteins, such as c-Myc. Christensen et al reported that the expression of SNHG16 is determined by Wnt-regulated transcription factors such as c-Myc in CRC (182). Specifically, knockdown of β-catenin could reduce the expression of SNHG16 and c-Myc, whereas c-Myc knockdown or overexpression could decrease or increase the SNHG16 expression, respectively (182). In a study by Xiang et al, the SNHG16/YAP1/TEA domain transcription factor 1 (TEAD1) positive feedback loop was detected in CRC cells (183). SNHG16 was shown to act as a ceRNA that can physically bind miR-195-5p, further regulating YAP1 expression and facilitating tumor progression. YAP1 binds to TEAD1 to form a YAP1/TEAD1 complex, which in turn binds to two sites in the promoter of SNHG16 and activates SNHG16 transcription (183).

In addition, SNHG16 polymorphisms have been shown to be significantly associated with CRC susceptibility. Research has revealed that the rs7353 site A>G of the SNHG16 gene is associated with decreased susceptibility of CRC; however, the rs8038 site G>A, rs15278 site A>G and rs15278 site G>A variations may increase CRC susceptibility (41).

SNHG16 and other types of cancer

SNHG16 has also been studied in other gastrointestinal tumors. For example, SNHG16 expression has been shown to be upregulated in cholangiocarcinoma tissues and cell lines. When SNHG16 expression was suppressed, the proliferation rate of RBE and HuCCT1 cells was reduced, whereas apoptosis was activated (184). Wu et al (184) also revealed that there was a potential binding site for miR-146a-5p at the 3′UTR end of GATA6 and that SNHG16 could sponge miR-146a-5p. Interfering with the expression of miR-146a-5p reversed the SNHG16 knockdown-induced apoptosis in RBE and HuCCT1 cells, whereas overexpression of GATA6 also achieved the same effect (184). These findings suggested that SNHG16 is important for the development of cholangiocarcinoma and it could be a potential target for future drug development against cholangiocarcinoma.

Neuroendocrine tumors account for a very small portion of tumors at each site; for example, the incidence of pancreatic neuroendocrine tumors was <1 case per 100,000 people/year worldwide (1), and the incidence of gastric neuroendocrine tumors was ~0.4 per 100,000 individuals in America in 2017 (185). However, as the number of patients with cancer increases, the proportion of neuroendocrine tumors has also increased, thus highlighting the need for attention to be paid to neuroendocrine tumors. Although, to the best of our knowledge, the role of SNHG16 in neuroendocrine tumors in the digestive system has not been reported, it is a valuable direction for improving the management of neuroendocrine tumors in the future.

Discussion and conclusion

A growing number of studies have demonstrated that tumorigenesis is caused by a combination of genetics and environmental factors. At present, environmental factors, such as diet, require attention to prevent their effects on personal health. The present review briefly summarized the relationship between risk factors and human digestive system cancer, identifying risk factors, such as smoking and diet, which may severely affect tumorigenesis. Subsequently, this review focused on outlining the role of SNHG16 in the formation and progression of digestive system cancer (Fig. 2). Available data have suggested that SNHG16 is strongly associated with proliferation, migration, invasion, apoptosis and prognosis in EC, HCC, PC, GC and CRC.

Upregulation of SNHG16 is clinically important, and is associated with tumor stage, lymph node metastasis and tumor size. It may be used as a novel diagnostic or prognostic biomarker for human digestive system tumors (Table II). For example, SNHG16 was revealed to be more highly expressed in EC tissues compared with in normal EC tissues, and similar results were observed in EC cell lines (36,152,154). In addition, overexpression of SNHG16 is strongly associated with the tumor stage, lymph node metastasis and clinical stage of EC (36). A Kaplan-Meier assay demonstrated that patients with EC and high SNHG16 expression had poorer overall survival rates than those with low SNHG16 expression (36). Notably, univariate and multivariate analyses have revealed that SNHG16 expression may be considered an independent predictor for the overall survival of patients with EC (36). In a number of studies, SNHG16 expression has been reported to be higher in HCC tissues or cells than in normal HCC tissues or cells (7175,135,156160); however, a study by Xu et al demonstrated that SNHG16 was significantly downregulated in both HCC tissues and cells (165). Clinical data have revealed that SNHG16 expression may be closely associated with tumor size, lymph node status and TNM stage, and enhanced SNHG16 expression could be related to advanced stages and short overall survival of patients with HCC (72,74,135). Multivariate analysis indicated that SNHG16 expression was an independent prognostic factor of HCC. Notably, some studies have reported that tumor size is not affected by SNHG16 expression, which may be due to individual differences or the small size of patients (74,75,136,155). However, reduced expression of SNHG16 has been reported in mice with smaller tumor sizes (71,155). In addition to the aforementioned cancer types, similar results were also reported in GC and CRC (170,183). Generally, the aforementioned findings suggested that SNHG16 expression is strongly associated with poor diagnosis or prognosis of human digestive system tumors.

In recent years, gene therapy has emerged as a promising avenue for significantly improving the survival rate of patients with cancer (186). As aforementioned, the clinical significance and biological functions of SNHG16 may serve as a promising therapeutic target for human digestive system cancer (Table I). However, Xu et al (165) reported contradictory results, which may be attributed to the sites and microenvironments specific to certain cancer types. The present review of publications on SNHG16 and cancer has resulted in a consistent conclusion that SNHG16 upregulation may promote proliferation, enhance migration and invasion, inhibit apoptosis, and affect lipid metabolism and chemoresistance. For example, silencing the expression of SNHG16 could attenuate the proliferation, activate the apoptosis, and inhibit the migratory, invasive and malignant phenotype in GC cell lines. In addition, the knockdown of SNHG16 could reduce tumor volume and weight in a nude mouse human-GC xenograft model (173).

The chemoresistance of various types of cancer may also be associated with the expression of SNHG16. A series of studies on HCC have demonstrated that downregulation of SNHG16 expression can reverse sorafenib and cisplatin resistance in HCC cell lines (such as Hep3B) or xenograft models, by acting as an endogenous sponge for miR-140-5p, miR-23b-3p and let-7b-5p (74,135,158,159). However, contrasting results reported by Xu et al showed that upregulation of SNHG16 inhibited 5-FU chemoresistance through competitive linking to miR-93 in Hep3B and Huh7 cells (165). In PC, SNHG16 may reduce SMAD4 to induce gemcitabine resistance via EZH2-mediated epigenetic modifications (167). In GC, the SNHG16-mediated miR-506-3p/PTBP1 axis effectively led to 5-FU resistance (169). These findings suggested that decreasing SNHG16 expression may be a promising therapy for suppressing GC progression. However, to the best of our knowledge, there are not currently any clinical trials focused on SNHG16; the development of inhibitors of SNHG16 have only been researched in cancer cell lines and animal models. In addition, the association between SNHG16 expression and chemoresistance in EC and CRC has not been researched. Therefore, whether SNHG16 will become a potential target of therapy, requires further study.

In this review, we described the association between risk factors and SNHG16 expression in ESCC, HCC and CRC. Notably, smoking may not influence the expression of SNHG16 in patients with ESCC (36). Furthermore, both HBV infection and liver cirrhosis were reported to not affect the expression of SNHG16 in HCC (7176,135). The association between PVTT and SNHG16 expression in HCC also remains controversial and is still under exploration (135,136). In CRC, the expression of SNHG16 could be regulated by risk factors, such as smoking and family history (41). To date, although smoking, excessive alcohol consumption, physical activity, infection, radiation, living environment, family history, diet, disease and genomic characteristics, are risk factors that can influence human digestive system cancer, the association between a number of risk factors and SNHG16 expression remains unclear. Therefore, further research using large-scale data will be necessary to thoroughly investigate the relationship between risk factors and SNHG16 expression.

Thus far, the upstream regulatory and downstream molecular mechanisms of SNHG16 in human digestive system cancer encompass four primary aspects (Fig. 2). i) Numerous transcription factors, including CTCF, c-Myc, NF-κB, STAT3 and TEAD1, are positively associated with SNHG16. ii) SNHG16 directly regulates the expression of downstream target genes, such as DKK3. iii) SNHG16 can bind to and recruit EIF4A3 to regulate RhoU expression and enhance RhoU mRNA stability. Meanwhile, SNHG16 also binds to EZH2 and recruits EZH2 to the SMAD4 promoter, subsequently suppressing SMAD4 expression. iv) SNHG16 can compete with miRNAs to mediate the expression of downstream target genes and activate different signaling pathways.

In conclusion, the expression of SNHG16 is associated with the clinical and pathological characteristics of patients with cancer, and regulates cell proliferation, apoptosis, invasion and metastasis through various potential mechanisms. These findings suggested that SNHG16 may serve an oncogenic role in human cancer, making it a promising target for cancer diagnosis and treatment, as well as a potential biomarker for cancer prognosis.

Acknowledgements

Not applicable.

Funding

This study was supported by the Shandong Youth Natural Science Foundation of China (grant no. ZR2021QH367); the Scientific and Technological Innovation Program for Medical Workers in Shandong Province (grant no. SDYWZGKCJHLH202212); and the Medical and Health Science and Technology Development Program of Shandong Province (grant nos. 202103100485 and 202102080115).

Availability of data and materials

Not applicable.

Authors' contributions

HR, FP and LX conceptualized the study. YL, YK and LW wrote the manuscript. LW, JP, LZ and HZ searched the literatures and collected the information from the literature. YL, YK, ZY and FP designed the figures and tables. LZ and XF revised and submitted the manuscript. Data authentication is not applicable. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

ARDS

acute respiratory distress syndrome

ceRNAs

competing endogenous RNAs

CRC

colorectal cancer

CT

computerized tomography

CTCF

CCCTC binding factor

DM

diabetes mellitus

EC

esophageal cancer

EMT

epithelial-mesenchymal transition

ESCC

esophageal squamous cell carcinoma

EZH2

enhancer of zeste homolog 2

GC

gastric cancer

HBV

hepatitis B virus

HCC

hepatocellular carcinoma

HCV

hepatitis C virus

HDI

human development index

lncRNA

long non-coding RNA

miRNAs

microRNAs

PC

pancreatic cancer

SNHG16

small nucleolar RNA host gene 16

T2DM

type 2 diabetes mellitus

TEAD1

TEA domain transcription factor 1

References

1 

Assarzadegan N and Montgomery E: What is New in the 2019 World Health Organization (WHO) classification of tumors of the digestive system: Review of selected updates on neuroendocrine neoplasms, appendiceal tumors, and molecular testing. Arch Pathol Lab Med. 145:664–677. 2021. View Article : Google Scholar : PubMed/NCBI

2 

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. View Article : Google Scholar : PubMed/NCBI

3 

Guo LW, Shi CL, Huang HY, Wang L, Yue XP, Liu SZ, Li J, Su K, Dai M, Sun XB and Shi JF: Economic burden of esophageal cancer in China from 1996 to 2015: A systematic review. Zhonghua Liu Xing Bing Xue Za Zhi. 38:102–109. 2017.(In Chinese). PubMed/NCBI

4 

Klimeck L, Heisser T, Hoffmeister M and Brenner H: Colorectal cancer: A health and economic problem. Best Pract Res Clin Gastroenterol. 66:1018392023. View Article : Google Scholar : PubMed/NCBI

5 

Hojman P, Gehl J, Christensen JF and Pedersen BK: Molecular mechanisms linking exercise to cancer prevention and treatment. Cell Metab. 27:10–21. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Xu W, Li B, Xu M, Yang T and Hao X: Traditional Chinese medicine for precancerous lesions of gastric cancer: A review. Biomed Pharmacother. 146:1125422022. View Article : Google Scholar : PubMed/NCBI

7 

Zhang X, Qiu H, Li C, Cai P and Qi F: The positive role of traditional Chinese medicine as an adjunctive therapy for cancer. Biosci Trends. 15:283–298. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Halbrook CJ, Lyssiotis CA, Pasca di Magliano M and Maitra A: Pancreatic cancer: Advances and challenges. Cell. 186:1729–1754. 2023. View Article : Google Scholar : PubMed/NCBI

9 

Vincent A, Herman J, Schulick R, Hruban RH and Goggins M: Pancreatic cancer. Lancet. 378:607–620. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Xie YH, Chen YX and Fang JY: Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct Target Ther. 5:222020. View Article : Google Scholar : PubMed/NCBI

11 

Bhan A, Soleimani M and Mandal SS: Long noncoding RNA and cancer: A new paradigm. Cancer Res. 77:3965–3981. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Lorenzi L, Avila Cobos F, Decock A, Everaert C, Helsmoortel H, Lefever S, Verboom K, Volders PJ, Speleman F, Vandesompele J and Mestdagh P: Long noncoding RNA expression profiling in cancer: Challenges and opportunities. Genes Chromosomes Cancer. 58:191–199. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Salmena L, Poliseno L, Tay Y, Kats L and Pandolfi PP: A ceRNA hypothesis: The Rosetta Stone of a hidden RNA language? Cell. 146:353–358. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Zhang Y, Dong X, Guo X, Li C, Fan Y, Liu P, Yuan D, Ma X, Wang J, Zheng J, et al: LncRNA-BC069792 suppresses tumor progression by targeting KCNQ4 in breast cancer. Mol Cancer. 22:412023. View Article : Google Scholar : PubMed/NCBI

15 

Yu M, Ohira M, Li Y, Niizuma H, Oo ML, Zhu Y, Ozaki T, Isogai E, Nakamura Y, Koda T, et al: High expression of ncRAN, a novel non-coding RNA mapped to chromosome 17q25.1, is associated with poor prognosis in neuroblastoma. Int J Oncol. 34:931–938. 2009.PubMed/NCBI

16 

Ghafouri-Fard S, Khoshbakht T, Taheri M and Shojaei S: A review on the role of small nucleolar RNA host gene 6 long non-coding RNAs in the carcinogenic processes. Front Cell Dev Biol. 9:7416842021. View Article : Google Scholar : PubMed/NCBI

17 

Gong CY, Tang R, Nan W, Zhou KS and Zhang HH: Role of SNHG16 in human cancer. Clin Chim Acta. 503:175–180. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Yang M and Wei W: SNHG16: A novel long-non coding RNA in human cancers. Onco Targets Ther. 12:11679–11690. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Zhang W, Zhou X, Tang Z, Fu L, Zou S and Tang S: Knockdown of lncRNA SNHG16 attenuates the proliferation and radioresistance of nasopharyngeal carcinoma cells by mediating miR-31-5p/SFN axis. Radiat Res. 199:124–131. 2023.PubMed/NCBI

20 

Cao X, Xu J and Yue D: LncRNA-SNHG16 predicts poor prognosis and promotes tumor proliferation through epigenetically silencing p21 in bladder cancer. Cancer Gene Ther. 25:10–17. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Yang XS, Wang GX and Luo L: Long non-coding RNA SNHG16 promotes cell growth and metastasis in ovarian cancer. Eur Rev Med Pharmacol Sci. 22:616–622. 2018.PubMed/NCBI

22 

Li S, Zhang S and Chen J: c-Myc induced upregulation of long non-coding RNA SNHG16 enhances progression and carcinogenesis in oral squamous cell carcinoma. Cancer Gene Ther. 26:400–410. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Sealock T and Sharma S: Smoking Cessation. StatPearls [Internet]. StatPearls Publishing; Treasure Island, FL: 2024

24 

Yuan S, Chen J, Ruan X, Sun Y, Zhang K, Wang X, Li X, Gill D, Burgess S, Giovannucci E and Larsson SC: Smoking, alcohol consumption, and 24 gastrointestinal diseases: Mendelian randomization analysis. Elife. 12:e840512023. View Article : Google Scholar : PubMed/NCBI

25 

GBD 2019 Cancer Risk Factors Collaborators, . The global burden of cancer attributable to risk factors, 2010-19: A systematic analysis for the global burden of disease study 2019. Lancet. 400:563–591. 2022. View Article : Google Scholar : PubMed/NCBI

26 

Marti-Aguado D, Clemente-Sanchez A and Bataller R: Cigarette smoking and liver diseases. J Hepatol. 77:191–205. 2022. View Article : Google Scholar : PubMed/NCBI

27 

Baecker A, Liu X, La Vecchia C and Zhang ZF: Worldwide incidence of hepatocellular carcinoma cases attributable to major risk factors. Eur J Cancer Prev. 27:205–212. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Cho WR, Wang CC, Tsai MJ, Lin CC, Yen YH, Chen CH, Kuo YH, Yao CC, Hung CH, Huang PY, et al: Smoking as a risk factor for very late recurrence in surgically resected early-stage primary hepatocellular carcinoma. Clin Med Insights Oncol. 18:117955492412282322024. View Article : Google Scholar : PubMed/NCBI

29 

Klein AP: Pancreatic cancer epidemiology: Understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol. 18:493–502. 2021. View Article : Google Scholar : PubMed/NCBI

30 

Lynch SM, Vrieling A, Lubin JH, Kraft P, Mendelsohn JB, Hartge P, Canzian F, Steplowski E, Arslan AA, Gross M, et al: Cigarette smoking and pancreatic cancer: A pooled analysis from the pancreatic cancer cohort consortium. Am J Epidemiol. 170:403–413. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Bosetti C, Lucenteforte E, Silverman DT, Petersen G, Bracci PM, Ji BT, Negri E, Li D, Risch HA, Olson SH, et al: Cigarette smoking and pancreatic cancer: An analysis from the international pancreatic cancer case-control consortium (Panc4). Ann Oncol. 23:1880–1888. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Ghadirian P, Lynch HT and Krewski D: Epidemiology of pancreatic cancer: An overview. Cancer Detect Prev. 27:87–93. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Iodice S, Gandini S, Maisonneuve P and Lowenfels AB: Tobacco and the risk of pancreatic cancer: A review and meta-analysis. Langenbecks Arch Surg. 393:535–545. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Weissman S, Takakura K, Eibl G, Pandol SJ and Saruta M: The diverse involvement of cigarette smoking in pancreatic cancer development and prognosis. Pancreas. 49:612–620. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Li H, Chen X, Hoffmeister M and Brenner H: Associations of smoking with early- and late-onset colorectal cancer. JNCI Cancer Spectr. 7:pkad0042023. View Article : Google Scholar : PubMed/NCBI

36 

Han GH, Lu KJ, Wang P, Ye J, Ye YY and Huang JX: LncRNA SNHG16 predicts poor prognosis in ESCC and promotes cell proliferation and invasion by regulating Wnt/β-catenin signaling pathway. Eur Rev Med Pharmacol Sci. 22:3795–3803. 2018.PubMed/NCBI

37 

Han W, Du X, Liu M, Wang J, Sun L and Li Y: Increased expression of long non-coding RNA SNHG16 correlates with tumor progression and poor prognosis in non-small cell lung cancer. Int J Biol Macromol. 121:270–278. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Li Y, Lu Y and Chen Y: Long non-coding RNA SNHG16 affects cell proliferation and predicts a poor prognosis in patients with colorectal cancer via sponging miR-200a-3p. Biosci Rep. 39:BSR201824982019. View Article : Google Scholar : PubMed/NCBI

39 

Gheliji T, Oskooei VK, Ashrafi Hafez A, Taheri M and Ghafouri-Fard S: Evaluation of expression of vitamin D receptor related lncRNAs in lung cancer. Noncoding RNA Res. 5:83–87. 2020. View Article : Google Scholar : PubMed/NCBI

40 

Jiao R, Jiang W, Wei X, Zhang M, Zhao S and Huang G: Clinicopathological significance and prognosis of long noncoding RNA SNHG16 expression in human cancers: A meta-analysis. BMC Cancer. 20:6622020. View Article : Google Scholar : PubMed/NCBI

41 

Zhou L, Zhang Y, Jin J and Gu X: Correlation between lncRNA SNHG16 gene polymorphism and its interaction with environmental factors and susceptibility to colorectal cancer. Medicine (Baltimore). 99:e233722020. View Article : Google Scholar : PubMed/NCBI

42 

Uhlenhopp DJ, Then EO, Sunkara T and Gaduputi V: Epidemiology of esophageal cancer: Update in global trends, etiology and risk factors. Clin J Gastroenterol. 13:1010–1021. 2020. View Article : Google Scholar : PubMed/NCBI

43 

McGee EE, Jackson SS, Petrick JL, Van Dyke AL, Adami HO, Albanes D, Andreotti G, Beane-Freeman LE, Berrington de Gonzalez A, Buring JE, et al: Smoking, alcohol, and biliary tract cancer risk: A pooling project of 26 prospective studies. J Natl Cancer Inst. 111:1263–1278. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Wood LD, Canto MI, Jaffee EM and Simeone DM: Pancreatic cancer: Pathogenesis, screening, diagnosis, and treatment. Gastroenterology. 163:386–402.e1. 2022. View Article : Google Scholar : PubMed/NCBI

45 

Laszkowska M, Rodriguez S, Kim J and Hur C: Heavy alcohol use is associated with gastric cancer: Analysis of the national health and nutrition examination survey from 1999 to 2010. Am J Gastroenterol. 116:1083–1086. 2021. View Article : Google Scholar : PubMed/NCBI

46 

McNabb S, Harrison TA, Albanes D, Berndt SI, Brenner H, Caan BJ, Campbell PT, Cao Y, Chang-Claude J, Chan A, et al: Meta-analysis of 16 studies of the association of alcohol with colorectal cancer. Int J Cancer. 146:861–873. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Sheikh M, Roshandel G, McCormack V and Malekzadeh R: Current status and future prospects for esophageal cancer. Cancers (Basel). 15:7652023. View Article : Google Scholar : PubMed/NCBI

48 

Larsson SC, Carter P, Kar S, Vithayathil M, Mason AM, Michaëlsson K and Burgess S: Smoking, alcohol consumption, and cancer: A mendelian randomisation study in UK Biobank and international genetic consortia participants. PLoS Med. 17:e10031782020. View Article : Google Scholar : PubMed/NCBI

49 

Dong J and Thrift AP: Alcohol, smoking and risk of oesophago-gastric cancer. Best Pract Res Clin Gastroenterol. 31:509–517. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Friedenreich CM, Ryder-Burbidge C and McNeil J: Physical activity, obesity and sedentary behavior in cancer etiology: Epidemiologic evidence and biologic mechanisms. Mol Oncol. 15:790–800. 2021. View Article : Google Scholar : PubMed/NCBI

51 

Moore SC, Lee IM, Weiderpass E, Campbell PT, Sampson JN, Kitahara CM, Keadle SK, Arem H, Berrington de Gonzalez A, Hartge P, et al: Association of leisure-time physical activity with risk of 26 types of cancer in 1.44 million adults. JAMA Intern Med. 176:816–825. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Su J, Jiang Y, Fan X, Tao R, Wu M, Lu Y, Hua Y, Jin J, Guo Y, Lv J, et al: Association between physical activity and cancer risk among Chinese adults: A 10-year prospective study. Int J Behav Nutr Phys Act. 19:1502022. View Article : Google Scholar : PubMed/NCBI

53 

Kasvis P and Kilgour RD: Diet and exercise interventions in patients with pancreatic cancer: A scoping review. Pancreas. 50:657–666. 2021. View Article : Google Scholar : PubMed/NCBI

54 

Xie F, You Y, Huang J, Guan C, Chen Z, Fang M, Yao F and Han J: Association between physical activity and digestive-system cancer: An updated systematic review and meta-analysis. J Sport Health Sci. 10:4–13. 2021. View Article : Google Scholar : PubMed/NCBI

55 

Garrett WS: Cancer and the microbiota. Science. 348:80–86. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Nasrollahzadeh D, Malekzadeh R, Ploner A, Shakeri R, Sotoudeh M, Fahimi S, Nasseri-Moghaddam S, Kamangar F, Abnet CC, Winckler B, et al: Variations of gastric corpus microbiota are associated with early esophageal squamous cell carcinoma and squamous dysplasia. Sci Rep. 5:88202015. View Article : Google Scholar : PubMed/NCBI

57 

Ponvilawan B, Rittiphairoj T, Charoenngam N, Rujirachun P, Wattanachayakul P, Tornsatitkul S and Ungprasert P: Association between chronic hepatitis C virus infection and esophageal cancer: A systematic review and meta-analysis. J Clin Gastroenterol. 56:55–63. 2022. View Article : Google Scholar : PubMed/NCBI

58 

Chidambaranathan-Reghupaty S, Fisher PB and Sarkar D: Hepatocellular carcinoma (HCC): Epidemiology, etiology and molecular classification. Adv Cancer Res. 149:1–61. 2021. View Article : Google Scholar : PubMed/NCBI

59 

Singh AK, Kumar R and Pandey AK: Hepatocellular carcinoma: Causes, mechanism of progression and biomarkers. Curr Chem Genom Transl Med. 12:9–26. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Mitsuhashi K, Nosho K, Sukawa Y, Matsunaga Y, Ito M, Kurihara H, Kanno S, Igarashi H, Naito T, Adachi Y, et al: Association of Fusobacterium species in pancreatic cancer tissues with molecular features and prognosis. Oncotarget. 6:7209–7220. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Fan X, Alekseyenko AV, Wu J, Peters BA, Jacobs EJ, Gapstur SM, Purdue MP, Abnet CC, Stolzenberg-Solomon R, Miller G, et al: Human oral microbiome and prospective risk for pancreatic cancer: A population-based nested case-control study. Gut. 67:120–127. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Wang YC: Medicinal plant activity on Helicobacter pylori related diseases. World J Gastroenterol. 20:10368–10382. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Ailloud F, Didelot X, Woltemate S, Pfaffinger G, Overmann J, Bader RC, Schulz C, Malfertheiner P and Suerbaum S: Within-host evolution of Helicobacter pylori shaped by niche-specific adaptation, intragastric migrations and selective sweeps. Nat Commun. 10:22732019. View Article : Google Scholar : PubMed/NCBI

64 

Espinoza JL, Matsumoto A, Tanaka H and Matsumura I: Gastric microbiota: An emerging player in Helicobacter pylori-induced gastric malignancies. Cancer Lett. 414:147–152. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Baj J, Korona-Glowniak I, Forma A, Maani A, Sitarz E, Rahnama-Hezavah M, Radzikowska E and Portincasa P: Mechanisms of the epithelial-mesenchymal transition and tumor microenvironment in Helicobacter pylori-induced gastric cancer. Cells. 9:10552020. View Article : Google Scholar : PubMed/NCBI

66 

Xia R, Zhang B, Wang X and Jia Q: Pathogenic interactions between Helicobacter pylori adhesion protein HopQ and human cell surface adhesion molecules CEACAMs in gastric epithelial cells. Iran J Basic Med Sci. 22:710–715. 2019.PubMed/NCBI

67 

Roesler BM, Rabelo-Gonçalves EMA and Zeitune JMR: Virulence factors of Helicobacter pylori: A review. Clin Med Insights Gastroenterol. 7:9–17. 2014. View Article : Google Scholar : PubMed/NCBI

68 

Sasaki S, Nishikawa J, Sakai K, Iizasa H, Yoshiyama H, Yanagihara M, Shuto T, Shimokuri K, Kanda T, Suehiro Y, et al: EBV-associated gastric cancer evades T-cell immunity by PD-1/PD-L1 interactions. Gastric Cancer. 22:486–496. 2019. View Article : Google Scholar : PubMed/NCBI

69 

Cuevas-Ramos G, Petit CR, Marcq I, Boury M, Oswald E and Nougayrède JP: Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells. Proc Natl Acad Sci USA. 107:11537–11542. 2010. View Article : Google Scholar : PubMed/NCBI

70 

Flemer B, Warren RD, Barrett MP, Cisek K, Das A, Jeffery IB, Hurley E, O'Riordain M, Shanahan F and O'Toole PW: The oral microbiota in colorectal cancer is distinctive and predictive. Gut. 67:1454–1463. 2018. View Article : Google Scholar : PubMed/NCBI

71 

Zhong JH, Xiang X, Wang YY, Liu X, Qi LN, Luo CP, Wei WE, You XM, Ma L, Xiang BD and Li LQ: The lncRNA SNHG16 affects prognosis in hepatocellular carcinoma by regulating p62 expression. J Cell Physiol. 235:1090–1102. 2020. View Article : Google Scholar : PubMed/NCBI

72 

Lin Q, Zheng H, Xu J, Zhang F and Pan H: LncRNA SNHG16 aggravates tumorigenesis and development of hepatocellular carcinoma by sponging miR-4500 and targeting STAT3. J Cell Biochem. 120:11604–11615. 2019. View Article : Google Scholar : PubMed/NCBI

73 

Chen H, Li M and Huang P: LncRNA SNHG16 promotes hepatocellular carcinoma proliferation, migration and invasion by regulating miR-186 expression. J Cancer. 10:3571–3581. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Ye J, Zhang R, Du X, Chai W and Zhou Q: Long noncoding RNA SNHG16 induces sorafenib resistance in hepatocellular carcinoma cells through sponging miR-140-5p. Onco Targets Ther. 12:415–422. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Hu YL, Feng Y, Chen YY, Liu JZ, Su Y, Li P, Huang H, Mao QS and Xue WJ: SNHG16/miR-605-3p/TRAF6/NF-κB feedback loop regulates hepatocellular carcinoma metastasis. J Cell Mol Med. 24:7637–7651. 2020. View Article : Google Scholar : PubMed/NCBI

76 

Liu Q, Gao P, Li Q, Xu C, Qu K and Zhang J: Long non-coding RNA SNHG16 as a potential biomarker in hepatocellular carcinoma: A meta-analysis. Medicine (Baltimore). 100:e271782021. View Article : Google Scholar : PubMed/NCBI

77 

Teng Y, Li M, Tao X, Huang Y, Ding X, Xu D, Fan Y and Shen Z: Cryptococcosis inhibits the immune response of dendritic cells through the snhg1-miR-145a-3p-Bcl2 axis. Exp Clin Transplant. 21:441–450. 2023. View Article : Google Scholar : PubMed/NCBI

78 

Sun W, Zhang X, He X, Zhang J, Wang X, Lin W, Wang X and Wu X: Long non-coding RNA SNHG16 silencing inhibits proliferation and inflammation in Mycobacterium tuberculosis-infected macrophages by targeting miR-140-5p expression. Infect Genet Evol. 103:1053252022. View Article : Google Scholar : PubMed/NCBI

79 

Allen C, Her S and Jaffray DA: Radiotherapy for cancer: Present and future. Adv Drug Deliv Rev. 109:1–2. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Nobel TB, Barbetta A, Hsu M, Tan KS, Pinchinat T, Schlottmann F, Bains MS, Ku GY, Wu AJ, Patti MG, et al: Outcomes of radiation-associated esophageal squamous cell carcinoma: The MSKCC experience. J Gastrointest Surg. 23:11–22. 2019. View Article : Google Scholar : PubMed/NCBI

81 

Thierry-Chef I, Cardis E, Damilakis J, Frija GA, Hierath M and Hoeschen C: Medical applications of ionizing radiation and radiation protection for European patients, population and environment. EPJ Nuclear Sci Technol. 8:442022. View Article : Google Scholar

82 

Goerlitz DS, Blancato J, Ramesh A, Islam M, Graham GT, Revina V, Kallakury B, Zeck J, Kirillova E and Loffredo CA: Somatic mutation signatures in primary liver tumors of workers exposed to ionizing radiation. Sci Rep. 9:181992019. View Article : Google Scholar : PubMed/NCBI

83 

Dores GM, Curtis RE, van Leeuwen FE, Stovall M, Hall P, Lynch CF, Smith SA, Weathers RE, Storm HH, Hodgson DC, et al: Pancreatic cancer risk after treatment of Hodgkin lymphoma. Ann Oncol. 25:2073–2079. 2014. View Article : Google Scholar : PubMed/NCBI

84 

Yusefi AR, Bagheri Lankarani K, Bastani P, Radinmanesh M and Kavosi Z: Risk factors for gastric cancer: A systematic review. Asian Pac J Cancer Prev. 19:591–603. 2018.PubMed/NCBI

85 

Kreuzer M, Straif K, Marsh JW, Dufey F, Grosche B, Nosske D and Sogl M: Occupational dust and radiation exposure and mortality from stomach cancer among German uranium miners, 1946–2003. Occup Environ Med. 69:217–223. 2012. View Article : Google Scholar : PubMed/NCBI

86 

Albert JM: Radiation risk from CT: Implications for cancer screening. AJR Am J Roentgenol. 201:W81–W87. 2013. View Article : Google Scholar : PubMed/NCBI

87 

Sun Y, Zhang T, Wu W, Zhao D, Zhang N, Cui Y, Liu Y, Gu J, Lu P, Xue F, et al: Risk factors associated with precancerous lesions of esophageal squamous cell carcinoma: A screening study in a high risk Chinese population. J Cancer. 10:3284–3290. 2019. View Article : Google Scholar : PubMed/NCBI

88 

Zhang L, Wan X, Shi R, Gong P and Si Y: Comparing spatial patterns of 11 common cancers in Mainland China. BMC Public Health. 22:15512022. View Article : Google Scholar : PubMed/NCBI

89 

Yang CY, Tsai SS and Chiu HF: Nitrate in drinking water and risk of death from pancreatic cancer in Taiwan. J Toxicol Environ Health A. 72:397–401. 2009. View Article : Google Scholar : PubMed/NCBI

90 

Picetti R, Deeney M, Pastorino S, Miller MR, Shah A, Leon DA, Dangour AD and Green R: Nitrate and nitrite contamination in drinking water and cancer risk: A systematic review with meta-analysis. Environ Res. 210:1129882022. View Article : Google Scholar : PubMed/NCBI

91 

Nasseri Maleki G, Bayati Khatibi M, Khamnian Z, Jalali Z, Dastgiri S and Ghodrati Aroogh H: Association between nitrate concentration in drinking water and rate of colorectal cancer: A case study in northwestern Iran. Int J Environ Health Res. 32:1791–1800. 2022. View Article : Google Scholar : PubMed/NCBI

92 

Lee W, Kim J, Lim SS, Kim Y, Ahn YS and Yoon JH: External airborne-agent exposure increase risk of digestive tract cancer. Sci Rep. 10:86172020. View Article : Google Scholar : PubMed/NCBI

93 

Tsai SS, Hsu CT and Yang C: Risk of death from liver cancer in relation to long-term exposure to fine particulate air pollution in Taiwan. J Toxicol Environ Health A. 86:135–143. 2023. View Article : Google Scholar : PubMed/NCBI

94 

Pritchett N, Spangler EC, Gray GM, Livinski AA, Sampson JN, Dawsey SM and Jones RR: Exposure to outdoor particulate matter air pollution and risk of gastrointestinal cancers in adults: A systematic review and meta-analysis of epidemiologic evidence. Environ Health Perspect. 130:360012022. View Article : Google Scholar : PubMed/NCBI

95 

Taheri M, Rad LM, Hussen BM, Nicknafs F, Sayad A and Ghafouri-Fard S: Evaluation of expression of VDR-associated lncRNAs in COVID-19 patients. BMC Infect Dis. 21:5882021. View Article : Google Scholar : PubMed/NCBI

96 

Zhang S, Chen J, Li B, Cai X, Wang K, Tan Z, Zheng Y and Liu Q: Family history of cancer is a prognostic factor for better survival in operable esophageal squamous cell carcinoma: A propensity score matching analysis. Front Oncol. 12:9459372022. View Article : Google Scholar : PubMed/NCBI

97 

Yang Y, Wu QJ, Xie L, Chow WH, Rothman N, Li HL, Gao YT, Zheng W, Shu XO and Xiang YB: Prospective cohort studies of association between family history of liver cancer and risk of liver cancer. Int J Cancer. 135:1605–1614. 2014. View Article : Google Scholar : PubMed/NCBI

98 

Abe K, Kitago M, Kitagawa Y and Hirasawa A: Hereditary pancreatic cancer. Int J Clin Oncol. 26:1784–1792. 2021. View Article : Google Scholar : PubMed/NCBI

99 

Choi IJ, Kim CG, Lee JY, Kim YI, Kook MC, Park B and Joo J: Family history of gastric cancer and Helicobacter pylori treatment. N Engl J Med. 382:427–436. 2020. View Article : Google Scholar : PubMed/NCBI

100 

Sninsky JA, Shore BM, Lupu GV and Crockett SD: Risk factors for colorectal polyps and cancer. Gastrointest Endosc Clin N Am. 32:195–213. 2022. View Article : Google Scholar : PubMed/NCBI

101 

Chen T, Cheng H, Chen X, Yuan Z, Yang X, Zhuang M, Lu M, Jin L and Ye W: Family history of esophageal cancer increases the risk of esophageal squamous cell carcinoma. Sci Rep. 5:160382015. View Article : Google Scholar : PubMed/NCBI

102 

Setia N, Clark JW, Duda DG, Hong TS, Kwak EL, Mullen JT and Lauwers GY: Familial gastric cancers. Oncologist. 20:1365–1377. 2015. View Article : Google Scholar : PubMed/NCBI

103 

Chan JA, Meyerhardt JA, Niedzwiecki D, Hollis D, Saltz LB, Mayer RJ, Thomas J, Schaefer P, Whittom R, Hantel A, et al: Association of family history with cancer recurrence and survival among patients with stage III colon cancer. JAMA. 299:2515–2523. 2008. View Article : Google Scholar : PubMed/NCBI

104 

Han MA, Oh MG, Choi IJ, Park SR, Ryu KW, Nam BH, Cho SJ, Kim CG, Lee JH and Kim YW: Association of family history with cancer recurrence and survival in patients with gastric cancer. J Clin Oncol. 30:701–708. 2012. View Article : Google Scholar : PubMed/NCBI

105 

Su Z, Zou GR, Mao YP, OuYang PY, Cao XL, Xie FY and Li Q: Prognostic impact of family history of cancer in Southern Chinese patients with esophageal squamous cell cancer. J Cancer. 10:1349–1357. 2019. View Article : Google Scholar : PubMed/NCBI

106 

Dai WC, Fan ST, Cheung TT, Chok KS, Chan AC, Tsang SH, Poon RT and Lo CM: The impact of family history of hepatocellular carcinoma on its patients' survival. Hepatobiliary Pancreat Dis Int. 11:160–164. 2012. View Article : Google Scholar : PubMed/NCBI

107 

Hamada T, Yuan C, Yurgelun MB, Perez K, Khalaf N, Morales-Oyarvide V, Babic A, Nowak JA, Rubinson DA, Giannakis M, et al: Family history of cancer, Ashkenazi Jewish ancestry, and pancreatic cancer risk. Br J Cancer. 120:848–854. 2019. View Article : Google Scholar : PubMed/NCBI

108 

Chen Y, Tong Y, Yang C, Gan Y, Sun H, Bi H, Cao S, Yin X and Lu Z: Consumption of hot beverages and foods and the risk of esophageal cancer: A meta-analysis of observational studies. BMC Cancer. 15:4492015. View Article : Google Scholar : PubMed/NCBI

109 

Keszei AP, Goldbohm RA, Schouten LJ, Jakszyn P and van den Brandt PA: Dietary N-nitroso compounds, endogenous nitrosation, and the risk of esophageal and gastric cancer subtypes in the Netherlands cohort study. Am J Clin Nutr. 97:135–146. 2013. View Article : Google Scholar : PubMed/NCBI

110 

Ibiebele TI, Hughes MC, Whiteman DC and Webb PM; Australian Cancer Study, : Dietary patterns and risk of oesophageal cancers: A population-based case-control study. Br J Nutr. 107:1207–1216. 2012. View Article : Google Scholar : PubMed/NCBI

111 

Kim J, Cho YA, Choi WJ and Jeong SH: Gene-diet interactions in gastric cancer risk: A systematic review. World J Gastroenterol. 20:9600–9610. 2014. View Article : Google Scholar : PubMed/NCBI

112 

Cheng YK, Yao SM, Xu YR and Niu RG: Life-style habits in a high-risk area for upper gastrointestinal cancers: A population-based study from Shanxi, China. Asian Pac J Cancer Prev. 17:4301–4306. 2016.PubMed/NCBI

113 

Berretta M, Lleshi A, Fisichella R, Berretta S, Basile F, Li Volti G, Bolognese A, Biondi A, De Paoli P, Tirelli U and Cappellani A: The role of nutrition in the development of esophageal cancer: What do we know? Front Biosci (Elite Ed). 4:351–357. 2012. View Article : Google Scholar : PubMed/NCBI

114 

Zhang Z and Zhang X: Salt taste preference, sodium intake and gastric cancer in China. Asian Pac J Cancer Prev. 12:1207–1210. 2011.PubMed/NCBI

115 

Thanikachalam K and Khan G: Colorectal cancer and nutrition. Nutrients. 11:1642019. View Article : Google Scholar : PubMed/NCBI

116 

Song M, Garrett WS and Chan AT: Nutrients, foods, and colorectal cancer prevention. Gastroenterology. 148:1244–1260.e16. 2015. View Article : Google Scholar : PubMed/NCBI

117 

Rosato V, Bosetti C, Levi F, Polesel J, Zucchetto A, Negri E and La Vecchia C: Risk factors for young-onset colorectal cancer. Cancer Causes Control. 24:335–341. 2013. View Article : Google Scholar : PubMed/NCBI

118 

Murata M: Inflammation and cancer. Environ Health Prev Med. 23:502018. View Article : Google Scholar : PubMed/NCBI

119 

Ohnishi S, Ma N, Thanan R, Pinlaor S, Hammam O, Murata M and Kawanishi S: DNA damage in inflammation-related carcinogenesis and cancer stem cells. Oxid Med Cell Longev. 2013:3870142013. View Article : Google Scholar : PubMed/NCBI

120 

Wang RH: From reflux esophagitis to Barrett's esophagus and esophageal adenocarcinoma. World J Gastroenterol. 21:5210–5219. 2015. View Article : Google Scholar : PubMed/NCBI

121 

Ioannou GN: Epidemiology and risk-stratification of NAFLD-associated HCC. J Hepatol. 75:1476–1484. 2021. View Article : Google Scholar : PubMed/NCBI

122 

Powell EE, Wong VW and Rinella M: Non-alcoholic fatty liver disease. Lancet. 397:2212–2224. 2021. View Article : Google Scholar : PubMed/NCBI

123 

Pouwels S, Sakran N, Graham Y, Leal A, Pintar T, Yang W, Kassir R, Singhal R, Mahawar K and Ramnarain D: Non-alcoholic fatty liver disease (NAFLD): A review of pathophysiology, clinical management and effects of weight loss. BMC Endocr Disord. 22:632022. View Article : Google Scholar : PubMed/NCBI

124 

Duell EJ, Lucenteforte E, Olson SH, Bracci PM, Li D, Risch HA, Silverman DT, Ji BT, Gallinger S, Holly EA, et al: Pancreatitis and pancreatic cancer risk: A pooled analysis in the international pancreatic cancer case-control consortium (PanC4). Ann Oncol. 23:2964–2970. 2012. View Article : Google Scholar : PubMed/NCBI

125 

Sipponen P and Maaroos HI: Chronic gastritis. Scand J Gastroenterol. 50:657–667. 2015. View Article : Google Scholar : PubMed/NCBI

126 

Tempera PJ, Michael M, Tageldin O and Hasak S: Gastric cancer due to chronic H. pylori infection: What we know and where we are going. Diseases. 10:572022. View Article : Google Scholar : PubMed/NCBI

127 

Kim J and Lee HK: Potential role of the gut microbiome in colorectal cancer progression. Front Immunol. 12:8076482022. View Article : Google Scholar : PubMed/NCBI

128 

Xu B, Zhou X, Li X, Liu C and Yang C: Diabetes mellitus carries a risk of esophageal cancer: A meta-analysis. Medicine (Baltimore). 96:e79442017. View Article : Google Scholar : PubMed/NCBI

129 

Li M, Park JY, Sheikh M, Kayamba V, Rumgay H, Jenab M, Narh CT, Abedi-Ardekani B, Morgan E, de Martel C, et al: Population-based investigation of common and deviating patterns of gastric cancer and oesophageal cancer incidence across populations and time. Gut. 72:846–854. 2023. View Article : Google Scholar : PubMed/NCBI

130 

Chari ST, Leibson CL, Rabe KG, Ransom J, de Andrade M and Petersen GM: Probability of pancreatic cancer following diabetes: A population-based study. Gastroenterology. 129:504–511. 2005. View Article : Google Scholar : PubMed/NCBI

131 

Lim JH, Shin CM, Han KD, Lee SW, Jin EH, Choi YJ, Yoon H, Park YS, Kim N and Lee DH: Association between the persistence of obesity and the risk of gastric cancer: A nationwide population-based study. Cancer Res Treat. 54:199–207. 2022. View Article : Google Scholar : PubMed/NCBI

132 

Guo J, Liu C, Pan J and Yang J: Relationship between diabetes and risk of gastric cancer: A systematic review and meta-analysis of cohort studies. Diabetes Res Clin Pract. 187:1098662022. View Article : Google Scholar : PubMed/NCBI

133 

Soltani G, Poursheikhani A, Yassi M, Hayatbakhsh A, Kerachian M and Kerachian MA: Obesity, diabetes and the risk of colorectal adenoma and cancer. BMC Endocr Disord. 19:1132019. View Article : Google Scholar : PubMed/NCBI

134 

Plaz Torres MC, Jaffe A, Perry R, Marabotto E, Strazzabosco M and Giannini EG: Diabetes medications and risk of HCC. Hepatology. 76:1880–1897. 2022. View Article : Google Scholar : PubMed/NCBI

135 

Guo Z, Zhang J, Fan L, Liu J, Yu H, Li X and Sun G: Long noncoding RNA (lncRNA) small nucleolar RNA host gene 16 (SNHG16) predicts poor prognosis and sorafenib resistance in hepatocellular carcinoma. Med Sci Monit. 25:2079–2086. 2019. View Article : Google Scholar : PubMed/NCBI

136 

Zhang QJ, Li DZ, Lin BY, Geng L, Yang Z and Zheng SS: SNHG16 promotes hepatocellular carcinoma development via activating ECM receptor interaction pathway. Hepatobiliary Pancreat Dis Int. 21:41–49. 2022. View Article : Google Scholar : PubMed/NCBI

137 

Zhang C, Huang Q and He F: Correlation of small nucleolar RNA host gene 16 with acute respiratory distress syndrome occurrence and prognosis in sepsis patients. J Clin Lab Anal. 36:e245162022. View Article : Google Scholar : PubMed/NCBI

138 

Sun J, Xin K, Leng C and Ge J: Down-regulation of SNHG16 alleviates the acute lung injury in sepsis rats through miR-128-3p/HMGB3 axis. BMC Pulm Med. 21:1912021. View Article : Google Scholar : PubMed/NCBI

139 

Xia L, Zhu G, Huang H, He Y and Liu X: LncRNA small nucleolar RNA host gene 16 (SNHG16) silencing protects lipopolysaccharide (LPS)-induced cell injury in human lung fibroblasts WI-38 through acting as miR-141-3p sponge. Biosci Biotechnol Biochem. 85:1077–1087. 2021. View Article : Google Scholar : PubMed/NCBI

140 

Zhou Z, Zhu Y, Gao G and Zhang Y: Long noncoding RNA SNHG16 targets miR-146a-5p/CCL5 to regulate LPS-induced WI-38 cell apoptosis and inflammation in acute pneumonia. Life Sci. 228:189–197. 2019. View Article : Google Scholar : PubMed/NCBI

141 

Xie C, Zhu B, Gu J and Sun M: The correlation of lncRNA SNHG16 with inflammatory cytokines, adhesion molecules, disease severity, and prognosis in acute ischemic stroke patients. J Clin Lab Anal. 36:e244392022. View Article : Google Scholar : PubMed/NCBI

142 

Zhao Y, Wang H, Tang Y, Wang J, Wu X, He Z, He Y and Tang Z: SNHG16/miR-205/HDAC5 is involved in the progression of renal fibrosis. J Biochem Mol Toxicol. 38:e236172024. View Article : Google Scholar : PubMed/NCBI

143 

Luo J: KRAS mutation in pancreatic cancer. Semin Oncol. 48:10–18. 2021. View Article : Google Scholar : PubMed/NCBI

144 

Mei ZB, Duan CY, Li CB, Cui L and Ogino S: Prognostic role of tumor PIK3CA mutation in colorectal cancer: A systematic review and meta-analysis. Ann Oncol. 27:1836–1848. 2016. View Article : Google Scholar : PubMed/NCBI

145 

Cox AD, Fesik SW, Kimmelman AC, Luo J and Der CJ: Drugging the undruggable RAS: Mission possible? Nat Rev Drug Discov. 13:828–851. 2014. View Article : Google Scholar : PubMed/NCBI

146 

Busuttil RA, Zapparoli GV, Haupt S, Fennell C, Wong SQ, Pang JM, Takeno EA, Mitchell C, Di Costanzo N, Fox S, et al: Role of p53 in the progression of gastric cancer. Oncotarget. 5:12016–12026. 2014. View Article : Google Scholar : PubMed/NCBI

147 

Li Y, Wu J, Li E, Xiao Z, Lei J, Zhou F, Yin X, Hu D, Mao Y, Wu L and Wenjun L: TP53 mutation detected in circulating exosomal DNA is associated with prognosis of patients with hepatocellular carcinoma. Cancer Biol Ther. 23:439–445. 2022. View Article : Google Scholar : PubMed/NCBI

148 

Wang W, Shao F, Yang X, Wang J, Zhu R, Yang Y, Zhao G, Guo D, Sun Y, Wang J, et al: METTL3 promotes tumour development by decreasing APC expression mediated by APC mRNA N(6)-methyladenosine-dependent YTHDF binding. Nat Commun. 12:38032021. View Article : Google Scholar : PubMed/NCBI

149 

Zeineldin M and Neufeld KL: Understanding phenotypic variation in rodent models with germline Apc mutations. Cancer Res. 73:2389–2399. 2013. View Article : Google Scholar : PubMed/NCBI

150 

Cui R, Kamatani Y, Takahashi A, Usami M, Hosono N, Kawaguchi T, Tsunoda T, Kamatani N, Kubo M, Nakamura Y and Matsuda K: Functional variants in ADH1B and ALDH2 coupled with alcohol and smoking synergistically enhance esophageal cancer risk. Gastroenterology. 137:1768–1775. 2009. View Article : Google Scholar : PubMed/NCBI

151 

Ilango S, Paital B, Jayachandran P, Padma PR and Nirmaladevi R: Epigenetic alterations in cancer. Front Biosci (Landmark Ed). 25:1058–1109. 2020. View Article : Google Scholar : PubMed/NCBI

152 

Zhang K, Chen J, Song H and Chen LB: SNHG16/miR-140-5p axis promotes esophagus cancer cell proliferation, migration and EMT formation through regulating ZEB1. Oncotarget. 9:1028–1040. 2017. View Article : Google Scholar : PubMed/NCBI

153 

Zhang L, Liang H, Zhang J, Yang Y, Ling X and Jiang H: Long non-coding RNA SNHG16 facilitates esophageal cancer cell proliferation and self-renewal through the microRNA-802/PTCH1 axis. Curr Med Chem. 29:6084–6099. 2022. View Article : Google Scholar : PubMed/NCBI

154 

Ren L, Fang X, Shrestha SM, Ji Q, Ye H, Liang Y, Liu Y, Feng Y, Dong J and Shi R: LncRNA SNHG16 promotes development of oesophageal squamous cell carcinoma by interacting with EIF4A3 and modulating RhoU mRNA stability. Cell Mol Biol Lett. 27:892022. View Article : Google Scholar : PubMed/NCBI

155 

Xie X, Xu X, Sun C and Yu Z: Long intergenic noncoding RNA SNHG16 interacts with miR-195 to promote proliferation, invasion and tumorigenesis in hepatocellular carcinoma. Exp Cell Res. 383:1115012019. View Article : Google Scholar : PubMed/NCBI

156 

Li S, Qi Y, Huang Y, Guo Y, Huang T and Jia L: Exosome-derived SNHG16 sponging miR-4500 activates HUVEC angiogenesis by targeting GALNT1 via PI3K/Akt/mTOR pathway in hepatocellular carcinoma. J Physiol Biochem. 77:667–682. 2021. View Article : Google Scholar : PubMed/NCBI

157 

Li W, Xu W, Song JS, Wu T and Wang WX: LncRNA SNHG16 promotes cell proliferation through miR-302a-3p/FGF19 axis in hepatocellular carcinoma. Neoplasma. 66:397–404. 2019. View Article : Google Scholar : PubMed/NCBI

158 

Li S, Peng F, Ning Y, Jiang P, Peng J, Ding X, Zhang J, Jiang T and Xiang S: SNHG16 as the miRNA let-7b-5p sponge facilitates the G2/M and epithelial-mesenchymal transition by regulating CDC25B and HMGA2 expression in hepatocellular carcinoma. J Cell Biochem. 121:2543–2558. 2020. View Article : Google Scholar : PubMed/NCBI

159 

Jing Z, Ye X, Ma X, Hu X, Yang W, Shi J, Chen G and Gong L: SNGH16 regulates cell autophagy to promote sorafenib resistance through suppressing miR-23b-3p via sponging EGR1 in hepatocellular carcinoma. Cancer Med. 9:4324–4338. 2020. View Article : Google Scholar : PubMed/NCBI

160 

Xu Y, Luan G, Li Z, Liu Z, Qin G and Chu Y: Tumour-derived exosomal lncRNA SNHG16 induces telocytes to promote metastasis of hepatocellular carcinoma via the miR-942-3p/MMP9 axis. Cell Oncol (Dordr). 46:251–264. 2023.PubMed/NCBI

161 

Zhang J and Lou W: A key mRNA-miRNA-lncRNA competing endogenous RNA triple sub-network linked to diagnosis and prognosis of hepatocellular carcinoma. Front Oncol. 10:3402020. View Article : Google Scholar : PubMed/NCBI

162 

Chen S, Zhao Z, Wang X, Zhang Q, Lyu L and Tang B: The predictive competing endogenous RNA regulatory networks and potential prognostic and immunological roles of cyclin A2 in pan-cancer analysis. Front Mol Biosci. 9:8095092022. View Article : Google Scholar : PubMed/NCBI

163 

Shao X, Zhu J, Shi Y, Fang H, Chen J, Zhang Y, Wang J, Jian H, Lan S, Jiang F, et al: Upregulated UBE4B expression correlates with poor prognosis and tumor immune infiltration in hepatocellular carcinoma. Aging (Albany NY). 14:9632–9646. 2022.PubMed/NCBI

164 

Lou W, Wang W, Chen J, Wang S and Huang Y: ncRNAs-mediated high expression of SEMA3F correlates with poor prognosis and tumor immune infiltration of hepatocellular carcinoma. Mol Ther Nucleic Acids. 24:845–855. 2021. View Article : Google Scholar : PubMed/NCBI

165 

Xu F, Zha G, Wu Y, Cai W and Ao J: Overexpressing lncRNA SNHG16 inhibited HCC proliferation and chemoresistance by functionally sponging hsa-miR-93. Onco Targets Ther. 11:8855–8863. 2018. View Article : Google Scholar : PubMed/NCBI

166 

Xu H, Miao X, Li X, Chen H, Zhang B and Zhou W: LncRNA SNHG16 contributes to tumor progression via the miR-302b-3p/SLC2A4 axis in pancreatic adenocarcinoma. Cancer Cell Int. 21:512021. View Article : Google Scholar : PubMed/NCBI

167 

Yu Y, Zou YF, Hong RQ, Chen WJ, Chen L, Chen WQ, Wang HP and Yu Y: Long non-coding RNA SNHG16 decreased SMAD4 to induce gemcitabine resistance in pancreatic cancer via EZH2-mediated epigenetic modification. Kaohsiung J Med Sci. 38:981–991. 2022. View Article : Google Scholar : PubMed/NCBI

168 

Yu Y, Dong JT, He B, Zou YF, Li XS, Xi CH and Yu Y: LncRNA SNHG16 induces the SREBP2 to promote lipogenesis and enhance the progression of pancreatic cancer. Future Oncol. 15:3831–3844. 2019. View Article : Google Scholar : PubMed/NCBI

169 

Ding Y, Gao S, Zheng J and Chen X: Blocking lncRNA-SNHG16 sensitizes gastric cancer cells to 5-Fu through targeting the miR-506-3p-PTBP1-mediated glucose metabolism. Cancer Metab. 10:202022. View Article : Google Scholar : PubMed/NCBI

170 

Lian D, Amin B, Du D and Yan W: Enhanced expression of the long non-coding RNA SNHG16 contributes to gastric cancer progression and metastasis. Cancer Biomark. 21:151–160. 2017. View Article : Google Scholar : PubMed/NCBI

171 

Zhao JJ, Liu JJ, Zhang YY, Xia Y, Du H, Yan ZQ, Zhou CH, Xia WS, Zellmer L, Liao DJ, et al: SNHG16 lncRNAs are overexpressed and may be oncogenic in human gastric cancer by regulating cell cycle progression. Neoplasma. 69:49–58. 2022. View Article : Google Scholar : PubMed/NCBI

172 

Zhou C, Zhao J, Liu J, Wei S, Xia Y, Xia W, Bi Y, Yan Z and Huang H: LncRNA SNHG16 promotes epithelial-mesenchymal transition via down-regulation of DKK3 in gastric cancer. Cancer Biomark. 26:393–401. 2019. View Article : Google Scholar : PubMed/NCBI

173 

Yang Z, Pu M, Dong X, Yang H, Chang W, Liu T and Zhang X: CTCF-activated SNHG16 facilitates gastrointestinal stromal tumor by targeting miR-128-3p/CASC3 axis. Exp Cell Res. 417:1131312022. View Article : Google Scholar : PubMed/NCBI

174 

Wang X, Kan J, Han J, Zhang W, Bai L and Wu H: LncRNA SNHG16 functions as an oncogene by sponging MiR-135a and promotes JAK2/STAT3 signal pathway in gastric cancer. J Cancer. 10:1013–1022. 2019. View Article : Google Scholar : PubMed/NCBI

175 

Zhou W, Zhang S, Li HB, Cai Z, Tang S, Chen LX, Lang JY, Chen Z and Chen XL: Development of prognostic indicator based on autophagy-related lncRNA analysis in colon adenocarcinoma. Biomed Res Int. 2020:98079182020. View Article : Google Scholar : PubMed/NCBI

176 

Chen ZY, Wang XY, Yang YM, Wu MH, Yang L, Jiang DT, Cai H and Peng Y: LncRNA SNHG16 promotes colorectal cancer cell proliferation, migration, and epithelial-mesenchymal transition through miR-124-3p/MCP-1. Gene Ther. 29:193–205. 2022. View Article : Google Scholar : PubMed/NCBI

177 

Tan P, Xu M, Nie J, Qin J, Liu X, Sun H, Wang S and Pan Y: LncRNA SNHG16 promotes colorectal cancer proliferation by regulating ABCB1 expression through sponging miR-214-3p. J Biomed Res. 36:231–241. 2022. View Article : Google Scholar : PubMed/NCBI

178 

He X, Ma J, Zhang M, Cui J and Yang H: Long non-coding RNA SNHG16 activates USP22 expression to promote colorectal cancer progression by sponging miR-132-3p. Onco Targets Ther. 13:4283–4294. 2020. View Article : Google Scholar : PubMed/NCBI

179 

Ke D and Wang Q, Ke S, Zou L and Wang Q: Long-non coding RNA SNHG16 supports colon cancer cell growth by modulating miR-302a-3p/AKT axis. Pathol Oncol Res. 26:1605–1613. 2020. View Article : Google Scholar : PubMed/NCBI

180 

Huang E, Ma T, Zhou J, Ma N, Yang W, Liu C, Hou Z, Chen S, Zong Z, Zeng B, et al: A novel senescence-associated LncRNA signature predicts the prognosis and tumor microenvironment of patients with colorectal cancer: A bioinformatics analysis. J Gastrointest Oncol. 13:1842–1863. 2022. View Article : Google Scholar : PubMed/NCBI

181 

Duan L, Xia Y, Li C, Lan N and Hou X: Identification of autophagy-related LncRNA to predict the prognosis of colorectal cancer. Front Genet. 13:9069002022. View Article : Google Scholar : PubMed/NCBI

182 

Christensen LL, True K, Hamilton MP, Nielsen MM, Damas ND, Damgaard CK, Ongen H, Dermitzakis E, Bramsen JB, Pedersen JS, et al: SNHG16 is regulated by the Wnt pathway in colorectal cancer and affects genes involved in lipid metabolism. Mol Oncol. 10:1266–1282. 2016. View Article : Google Scholar : PubMed/NCBI

183 

Xiang Z, Huang G, Wu H, He Q, Yang C, Dou R, Liu Q, Song J, Fang Y, Wang S and Xiong B: SNHG16 upregulation-induced positive feedback loop with YAP1/TEAD1 complex in colorectal cancer cell lines facilitates liver metastasis of colorectal cancer by modulating CTCs epithelial-mesenchymal transition. Int J Biol Sci. 18:5291–5308. 2022. View Article : Google Scholar : PubMed/NCBI

184 

Wu T, Lei MS, Gao XZ, Xiong TG, Yang K, Gong Q, Tang R, Tian YP and Fu XH: lncRNA SNHG16 mediates cell proliferation and apoptosis in cholangiocarcinoma by directly targeting miR-146a-5p/GATA6 axis. Biochem Genet. 59:1311–1325. 2021. View Article : Google Scholar : PubMed/NCBI

185 

Dasari A, Shen C, Halperin D, Zhao B, Zhou S, Xu Y, Shih T and Yao JC: Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol. 3:1335–1342. 2017. View Article : Google Scholar : PubMed/NCBI

186 

He W, Li Q, Lu Y, Ju D, Gu Y, Zhao K and Dong C: Cancer treatment evolution from traditional methods to stem cells and gene therapy. Curr Gene Ther. 22:368–385. 2022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2024
Volume 52 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao L, Kan Y, Wang L, Pan J, Li Y, Zhu H, Yang Z, Xiao L, Fu X, Peng F, Peng F, et al: Roles of long non‑coding RNA SNHG16 in human digestive system cancer (Review). Oncol Rep 52: 106, 2024
APA
Zhao, L., Kan, Y., Wang, L., Pan, J., Li, Y., Zhu, H. ... Ren, H. (2024). Roles of long non‑coding RNA SNHG16 in human digestive system cancer (Review). Oncology Reports, 52, 106. https://doi.org/10.3892/or.2024.8765
MLA
Zhao, L., Kan, Y., Wang, L., Pan, J., Li, Y., Zhu, H., Yang, Z., Xiao, L., Fu, X., Peng, F., Ren, H."Roles of long non‑coding RNA SNHG16 in human digestive system cancer (Review)". Oncology Reports 52.2 (2024): 106.
Chicago
Zhao, L., Kan, Y., Wang, L., Pan, J., Li, Y., Zhu, H., Yang, Z., Xiao, L., Fu, X., Peng, F., Ren, H."Roles of long non‑coding RNA SNHG16 in human digestive system cancer (Review)". Oncology Reports 52, no. 2 (2024): 106. https://doi.org/10.3892/or.2024.8765