Role of DCLK1 in oncogenic signaling (Review)

  • Authors:
    • Qin Lu
    • Hailan Feng
    • Hong Chen
    • Nathaniel Weygant
    • Jian Du
    • Zixing Yan
    • Zhiyun Cao
  • View Affiliations

  • Published online on: September 21, 2022     https://doi.org/10.3892/ijo.2022.5427
  • Article Number: 137
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Doublecortin‑like kinase 1 (DCLK1) has been identified as a novel biomarker of cancer stem cells among several different cancer types, including colon, breast, pancreas, kidney, liver, stomach and esophageal cancers. Studies have demonstrated that DCLK1 regulates tumorigenesis and epithelial‑mesenchymal transformation via several important pathways, such as Notch, Wnt/β‑catenin, RAS and multiple microRNAs. The function and biological mechanisms, including their association with the molecular structure and isoforms of DCLK1, are gradually being elucidated. However, the currently available knowledge regarding DCLK1 in terms of developing effective anti‑cancer drugs remains incomplete. In the present review, the molecular characteristics, biomarker function and biological mechanisms of DCLK1 are summarized and DCLK1 is proposed as a potential anti‑tumor target via the glucose metabolism pathway.

Introduction

In 2008, the Houchen group proposed that serine-threonine kinase, doublecortin-like kinase 1 (DCLK1, then known as DCAMKL-1), was a specific marker protein for intestinal adenoma stem cells (1), which was the first of a series of research reports providing evidence that it may be an effective target for oncology drug development. To date, DCLK1 has been reported to be a selective marker of several types of cancer stem cells (CSCs), including those in colon, breast, pancreas, kidney and esophageal cancers (2,3). The first high-level evidence for the CSC marker status of DCLK1 came in 2012, when Nakanishi et al (4) reported that DCLK1 does not mark normal stem cells, but specifically marks CSCs in the adenomatous polyposis coli (APC) loss-driven APCMin/+ model of intestinal tumorigenesis. Furthermore, in normal gastrointestinal epithelia, DCLK1 has been indicated to mark fully differentiated epithelial tuft cells among several other cell types in the gastric antrum, bile duct and pancreas (5,6). Epithelial tuft cells are characterized by microtubule bundles located at the cell apex and express DCLK1 and acetylated α-tubulin to take part in regulating the microenvironment (7). DCLK1+ tuft cells have a key role in tumorigenesis by regulating inflammation of the microenvironment via expression of proteins such as Cox1, Cox2 and hematopoietic prostaglandin-D synthase in intestinal cancer (8).

After 20 years of research, DCLK1 is accepted as a specific marker of tuft cells and several types of CSCs and evidence of its ability to regulate tumor growth and metastasis has been provided (9). DCLK1 is expressed in lung, liver, heart, spleen, thymus, prostate and intestine and strongly marks specific cell types (Fig. 1) (1012). In the present review, the molecular structure and biological mechanisms of DCLK1 in tumorigenesis and metastasis were summarized and its role in metabolism was highlighted as a potentially novel area for further exploration.

Molecular structure and function of DCLK1

The human DCLK1 gene is located on the long arm 13q12.3-q13 of the 13th chromosome, which contains two different promoter sequences to form splicing variants with different protein functional domains (13). Structural domains include two N-terminal doublecortin (DCX) domains and one C-terminal serine/threonine protein kinase domain, homologous to the protein kinase superfamily and DCX (14,15). The structural characteristics of DCX1 are similar to those of DCX, which is able to specifically bind to microtubules, and DCX2 mainly interacts with microtubules and their dimers. These two DCX domains allow DCLK1 to bind microtubules and regulate microtubule aggregation to affect neuronal migration. The C-terminal domain is similar to calmodulin dependent kinase II, but lacks a typical calmodulin binding site (16,17). At present, there are several splicing variants in the DCLK1 gene that have been identified, including a full-length type with all domains and a poly-arginine region, a DCX-like type containing only the microtubule-binding domains, and a smaller molecular-weight type containing a phosphoserine-rich region and kinase domain. These variations in protein domains resulting from alternative splicing and multiple transcriptional promoter regions are hypothesized to result in completely different molecular functions (18). Human DCLK1 includes 82- and 52-kDa isoforms, which are transcribed from an upstream (A, CpG-regulated) or downstream promoter (B, TATA-box) with differing C-terminal domains (Fig. 2). The A isoforms contain N-terminal doublecortin domains to bind to microtubules and a protein kinase domain, while the B isoforms lack N-terminal doublecortin domains. Later, DCX-like was identified and only includes N-terminal DCX domains produced from the A promoter and Camk-related peptide, a 56 amino acid B-promoter-derived peptide with unknown function, was also identified (19,20).

The biological activity of DCLK1 in cancer is different between β-promoter (alternatively termed as DCLK1-S or DCLK3/4, 45–52 kDa, isoform 3/4) and α-promoter (termed as DCLK1-L or DCLK1/2, ~82 kDa, isoform 1/2) isoforms. One study determined that hypermethylation of the α-promoter directly led to the absence of expression of DCLK1-L in 15 human colon cancer cell lines, and that the α-promoter was activated by β-catenin and T-cell factor-4/lymphoid enhancer factor (LEF), while the β-promoter was activated by NF-κB p65 in cancer cells. In this study, the majority of human CRCs were reported to express DCLK1-S, which developed an invasive phenotype and this was associated with unfavorable overall survival (19). Park et al (21) identified DCLK1-B transcription as directly activated by Wnt/β-catenin signaling and that LEF1 mediates Wnt-induced CSC properties. Sarkar et al (22) reported that DCLK1-L and DCLK1-S are in nuclear and mitochondrial fractions, as well as plasma membrane and cytosolic fractions, but DCLK1-S is in the nuclei and mitochondria in colon cancer. DCLK1 α-promoter demonstrated hypermethylation in cholangiocarcinoma, but hypomethylation in α- and β-promoter regions in renal cell carcinoma (RCC) (23). Of note, two mouse models using DCLK1 A-promoter isoforms to drive Cre-recombinase (DCLK1-CreERT and DCLK1-CreERT2) demonstrated lineage tracing of CRC tumors in the presence of APC mutation (24,25). Interestingly, Ge et al (26) reported that both DCLK1-AS (isoform 1, 82 kD) and DCLK1-BL (isoform 4) isoforms are able to efficiently activate epithelial-to-mesenchymal transition (EMT) in pancreatic ductal adenocarcinoma (PDAC) cell lines. Overall, there is a significant shortcoming in the literature regarding the function of DCLK1 isoforms in cancer. The combined evidence suggests that all major DCLK1 isoforms are oncogenic, but there may be a variation among different tumor types or even among tumor subtypes.

Biological mechanism of DCLK1 in different types of cancer

Colorectal cancer (CRC)

The biological function of DCLK1 in tumorigenesis and metastasis as a marker of tuft cells and CSCs is most thoroughly studied in CRC. In normal human intestinal tissue, stem cells are located at the base of the intestinal crypt epithelium, where they are marked by leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) without co-expressing gut endocrine markers chromogranin A and somatostatin (Fig. 3) (27). DCLK1 marks fully differentiated intestinal tuft cells located in the crypt and villus (28). Long-lived DCLK1+ tuft cells with characteristic microvilli feature self-renewal ability and potential quiescent stem-like functionality (29). Knockdown of the Wnt regulator APC does not alter this quiescence, but subsequent activation through inflammation induced by dextran sodium sulfate is sufficient to initiate colon cancer in Dclk1-Cre/Apcflox/flox (Dclk1-Cre/Apcflox/flox transgenic mice featuring knocking out APC gene in DCLK1+ cells) transgenic mice (30). These findings are supported in DCLK1-knockout mice where deficiency results in increased epithelial barrier permeability, higher levels of pro-inflammatory cytokines and chemokines, decreased levels of LGR5 and dysregulated Wnt/β-Catenin pathway genes in Villin-Cre/Dclk1flox/flox (Villin-Cre/Dclk1flox/flox mice featuring deletion of DCLK1 expression in villin-positive cells) mice (31). In 2009, Gerbe et al (32) provided conclusive evidence that DCLK1 was in fact a tuft cell rather than stem cell marker, as indicated by the position and marker co-expression (cyclooxygenase enzymes 1/2, advillin and tubulin) of DCLK1-expressing cells. Lineage tracing studies demonstrated that DCLK1-positive cells also express colorectal CSC markers, such as CD133 and CD44 (3,4,33). Of note, DCLK1-positive normal intestinal epithelial cells isolated by fluorescence-assisted cell sorting form spheroids that may assemble into glandular epithelial structures and express multiple markers of gut epithelial lineages when implanted subcutaneously in athymic nude mice (27). Self-renewal and differentiation characteristics of DCLK1-positive cells and low expression in normal tissue both led researchers to speculate that they may mark a type of stem cell (34,35), but these findings are no longer supported in the literature and previous results are likely artifacts resulting from the existence of rare DCLK1/LGR5 double-positive cells.

Certain studies reported that DCLK1 expression is significantly higher in CRC tissue and adenomas compared to normal tissue. In addition, increased levels are seen in distant metastases and it is closely associated with CRC recurrence (36,37). Key evidence demonstrated that DCLK1 specifically marked CSCs in the intestine, which continuously produce tumor progeny to prompt polyp growth, but there is no apparent effect on normal tissue after deletion of these cells (38). Furthermore, overexpression of DCLK1 was observed to enhance the percentage of stem-like human CRC cells in vitro (39,40). It has become clear that DCLK1, while not a bona fide normal stem cell marker, is instead a key marker of differentiated intestinal epithelial tuft cells (6,32), which, in the context of mutation and tumorigenesis, may identify specific CRC stem cells.

A series of reports evidence that DCLK1 regulates EMT, proliferation and CSC maintenance through the Notch, Ras and Wnt pathways via interaction with different microRNAs (miRNAs/miRs). MiR-1291 was observed to directly bind to the 3′-untranslated region sequence of DCLK1 and then inhibited the stemness and cell cycle through the cyclin-dependent kinase inhibitors p21WAF1/CIP1 and p27KIP1 (41). The levels of miR-137 and miR-15a were inversely correlated with high levels of DCLK1 detected in CRC with larger tumor size, poor differentiation and lymph node involvement (42). Knockdown of DCLK1 expression led to downregulation of miR-200a, miR-144 and miR-let7a along with downregulation of EMT-associated transcription factors [zinc finger E-box binding homeobox 1 (ZEB1), ZEB2, Snail, Slug and Twist], c-Myc, KRAS and Notch-1 in human colon cancer cells (43). In a non-tumorigenic context, a recent study indicated that miR-195 is able to directly interact with DCLK1 mRNA, resulting in suppressed function for tuft and paneth cells in the small intestinal epithelium by inhibiting DCLK1 translation (44).

In CRC cell lines, Notch pathway-regulated markers of CRC CSCs [DCLK1, LGR5, aldehyde dehydrogenase 1 family member A1 (ALDH1) and CD44] by JAK2, STAT3 and ERK1/2 phosphorylation and increased expression of Jagged 1 to promote stemness (45). On the basis of the above findings, researchers have proposed potential therapeutics to suppress proliferation, colony formation and reduce the number of DCLK1+ cells via the Notch pathway (46,47). The Wnt pathway promotes elongator acetyltransferase complex subunit 3 expression and SOX9 translation, which in turn support LGR5(+)/DCLK1(+) intestinal cancer stem cells in response to intestinal regeneration after radiation-induced injury (28). Furthermore, β-catenin nuclear translocation is increased by overexpression of RNA binding motif protein 3, which induces stemness in DCLK1(+)/LGR5(+)/CD44(+) CRC cells (48). Wnt/β-catenin pathway and pluripotency transcription factors c-Myc, KLF transcription factor 4, OCT4 and SOX2 are activated by commensal-polarized macrophages through a microbiome-induced bystander effect in E. faecalis-colonized IL10 knockout mice, leading to increases in the number of DCLK1(+)/CD44(+) cells through gene mutation, chromosomal instability and endogenous transformation to promote tumorigenicity (35). Basic research indicated that compounds, such as γ-mangostin present in the mangosteen (Garcinia mangostana) fruit, WNT5A agonist FOXY5 and niclosamide, are able to regulate chemotherapy resistance and cancer stemness by decreasing the number of DCLK1-positive cells (21,49,50). KRAS mutation was observed to upregulate DCLK1 protein levels, which was reversed by inhibiting KRAS expression (51).

Pancreatic cancer

DCLK1 marks a small subpopulation of morphologically and functionally distinct pancreatic cancer cells, which promote tumorigenesis in multiple mouse models (52,53). In normal adult pancreas, DCLK1 is expressed in ductal epithelial cells and islet cells (54) and it is upregulated in murine and human pancreatic intraepithelial neoplasia (55). It is co-expressed with neurogenin-3 and somatostatin, and pancreatic stem cell markers, but not with insulin and glucagon, which mark pancreatic α cells (24). DCLK1-positive cells isolated by flow cytometry injected into nude mice give rise to nodules with a hyperplastic appearance (56). Acetylated tubulin (AcTub), a marker of differentiation of specific pancreatic intraepithelial neoplasia, is frequently co-expressed with DCLK1 and regulates epithelial-mesenchymal transformation of pancreatic cancer cells. AcTub and DCLK1-marked cells demonstrate a typical tuft cell morphology with prominent microvilli at the apical surface of the cell and lead to increased size and number of spheroids in cancer self-renewal assays (53,57). Furthermore, these cells express high levels of ABL proto-oncogene 1, non-receptor tyrosine kinase and insulin-like growth factor 1 receptor, which are drug targets in clinical cancer therapy (58,59). DCLK1 was also reported to be a marker of a population of pancreatic cancer-initiating cells with morphological and molecular features of gastrointestinal tuft cells (53), which drive pancreatic tumor growth by immune cell-derived IL17, which in turn regulates POU class 2 homeobox 3, ALDH1A1 and IL17 receptor C (60). In the pancreas, DCLK1 marks pancreatic tuft and acinar, but rarely islet cells. DCLK1+ tuft cells expand in response to chronic injury or chronic inflammation, and DCLK1+ epithelial cells are a source of acinar-ductal metaplasia after Kras-G12D mutation. These findings indicated that DCLK1+ pancreatic cells may act as pancreatic intraepithelial neoplasia stem cells, but whether or not these arise from pancreatic DCLK1+ tuft cells or DCLK1+ acinar cells is a matter of debate (5,61).

In zebrafish and mouse models, it has been confirmed that DCLK1+ cells are enriched in pancreatic intraepithelial neoplasia and their expansion is an early event in KRAS-induced pancreatic tumorigenesis (52,62). In an established KRAS transgenic mouse model of pancreatic cancer, DCLK1-positive CSC-like cells increased, while at the same time, knockdown of DCLK1 expression in human pancreatic cells reduced c-Myc and KRAS through a let-7a miRNA-dependent mechanism (43). In clinical tissue samples of PDAC, KRAS and TP53 mutations were indicated to be associated with DCLK1 gene overexpression, which may contribute to the migration, proliferation and colony formation abilities of pancreatic cancer cells (63). Mutation of KRAS in DCLK1+ pancreatic cells does not affect cell quiescence or longevity but contributes functionally to the pathogenesis of pancreatic cancer (24). In 2019, Qu et al (64) reported that DCLK1-α-long increases invasion and drug resistance by activating the PI3K/AKT/MTOR signaling pathway through increasing KRAS activity in pancreatic and duodenal homeobox Pdx1CreKRASG12D transformation-related protein 53R172H mouse models and in the human pancreatic cancer cell lines AsPC-1 and MiaPaCa-2. The hepatocyte growth factor/c-MET axis is necessary for the expression of DCLK1 in tumor cells and the recent two papers indicated that it is strongly associated with tumor immune escape, including the promotion of M2-macrophages and decrease of CD4+ and CD8+ T cells in PDAC (26,65). It facilitates pancreatic cancer progression by mediating the interaction between PCSCs and stromal pancreatic stellate cells (66). Overall, despite these complex and emerging findings, overwhelming evidence has indicated that DCLK1 is a vital pancreatic CSC-like marker, which is upregulated and closely associated with precancerous lesions, tumorigenesis and invasion (6770).

Gastric cancer and esophageal cancer

In human normal stomach tissue, stem cells are located in the isthmus of gastric glands and DCLK1-positive cells were originally located in the gastric stem cell zone. These DCLK1-positive cells were not able to be labeled by bromodeoxyuridine, which was consistent with static stem cells lacking typical cell proliferation ability, suggesting that DCLK1 may be a marker of quiescent stem cells (71). The expression of DCLK1 in gastric cancer tissues was significantly higher than that in adjacent normal tissue and significantly correlated with lymph node metastasis and prognosis (7275). A recent study suggested that long non-coding RNA small nucleolar RNA host gene 1 promoted the effects of DCLK1/Notch1 on the EMT process through regulating miR-15b expression (76). Small extracellular vesicle (exosome) isolated from a DCLK1-overexpressing human gastric cancer cell line promoted the migration of non-transfected gastric cancer cells in a kinase-dependent manner (77). DCLK1 is also a potential biomarker to predict the survival of patients with gastric cancer (78).

DCLK1 expression progressively increases from Barrett's esophagus to dysplasia and then to esophageal adenocarcinoma (2,79). In human esophageal squamous cell carcinoma (ESCC) cells, DCLK1-S induced MMP2 expression via MAPK/ERK signaling to activate the EMT (80). Knockdown of DCLK1 inhibited the progression of ESCC by regulating proliferation, migration and invasion by suppressing the β-catenin/c-Myc pathway (81). These results indicated that DCLK1 levels are associated with the occurrence and development of esophageal cancer. In Barrett's esophageal adenocarcinoma, the expression of DCLK1 and LGR5 are significantly increased in squamous epithelial cells located at the gastric spout, which indicates that Barrett's esophageal adenocarcinoma probably comes from gastric cancer (82,83).

Breast cancer

Serum estradiol levels are an important factor of increased risk of postmenopausal breast cancer. Haakensen et al (84) detected differentially expressed genes by analysis of gene microarrays and indicated that DCLK1 was one of six influenced by serum estradiol. DCLK1 gene expression was downregulated in breast carcinoma samples compared with normal tissue samples but did not exhibit any significantly differential expression between invasive breast cancer and ductal carcinoma in situ. DCLK1 was not significant as an independent factor associated with serum estradiol in a linear regression model. A series of subsequent studies on DCLK1 expression in breast carcinomas were developed and the clinical results indicated DCLK1 was associated with clinicopathological features, estrogen receptor status and neuroendocrine markers (85). A cohort study including 1,132 cases reported that DCLK1 levels varied in several molecular subtypes. Luminal cancers had higher DCLK1 expression than HER2-overexpression and triple negative breast cancers (TNBCs). Elevated DCLK1 was associated with a lower histologic grade, absence of lymphovascular invasion, fibrotic foci, necrosis and lower pN stage. DCLK1 did not correlate with other breast CSC markers and stem cell features, but significant correlations were found with estrogen receptor and neuroendocrine markers. Zhao et al (86) used DCLK1 to devise a clinically practical method based on immunohistochemistry for the molecular subtyping of the mesenchymal subtype TNBC. Specifically, DCLK1 marked a mesenchymal subtype enriched in stem cell-related gene signatures and activated JAK/STAT3 pathway, which is highly correlated with CSC-like breast cancer cells (86). In support of these findings, Ramamoorthy et al (87) reported that DCLK1 is downregulated with ALDH and CD133 downstream of the Notch signaling pathway, which results in inhibition of TNBC stemness. In breast cancer cell lines, silencing of DCLK1 decreased the levels of Wnt/β-catenin pathway proteins such as β-catenin, c-Myc and cyclin D1 to decrease cell migration and invasion (88). Further basic studies indicated that DCLK1 is a molecular regulator of breast cancer proliferation, migration, invasion and a degradome-related metastatic stem-like profile (8890). Furthermore, miR-424-5p was indicated to act as a tumor suppressive miRNA regulating breast cancer cell proliferation, migration and invasion via binding DCLK1 in vitro (90). In combination, these findings suggest that DCLK1 is a potential therapeutic target in breast cancer, but further mechanistic studies are required.

Renal cancer

Only a small number of known markers of CSCs in kidney cancers is available. Among these are the commonly reported broad CSC markers ALDH, CD44 and CD133. Ge et al (91) reported that DCLK1 stimulated essential molecular and functional characteristics of renal CSCs, including expression of ALDH, self-renewal and resistance to approved tyrosine kinase inhibitors sunitinib, sorafenib, everolimus and temsirolimus, suggesting that DCLK1 is a potential renal CSC marker. Furthermore, they indicated that overexpression of DCLK1 was a direct regulatory factor in renal clear carcinoma progression, supporting the notion that DCLK1 is a potential CSC target to inhibit RCC metastasis in early stages (3). Of note, treatment with a DCLK1-targeted monoclonal antibody was able to inhibit tumorigenesis in ACHN renal cancer xenografts, suggesting a potential therapeutic strategy for this highly chemoresistant cancer (91). In addition, a small-molecule kinase inhibitor of DCLK1, DCLK1-IN-1, demonstrated obvious inhibition of immune checkpoint ligand programmed death ligand 1 and an apparent increase in immune-mediated cytotoxicity alone or in combination with anti-programmed death 1 therapy by suppressing DCLK1 phosphorylation and downregulating pluripotency factors and CSC- or EMT-associated markers, including c-MET, c-MYC and N-cadherin in RCC cell lines. These experimental results were consistent with the analysis of clinical populations in which DCLK1 predicted RCC survival. In addition, its expression was correlated with reduced CD8+ cytotoxic T-cell infiltration and increased in M2 immunosuppressive macrophage populations (92).

Liver cancer

To date, DCLK1 has not been identified to be a hepatocellular CSC marker. However, the expression of DCLK1 in chronic hepatitis, cirrhosis and hepatocellular carcinoma was significantly increased (93). DCLK1 is mainly expressed in epithelial and stromal cells, lymphocytes and bile duct cells of liver tissue of patients with chronic hepatitis C virus infection. Furthermore, the level of DCLK1 is related to the expression of S100A9 protein (94). S100A9 is a key protein of pro-inflammatory signaling by binding to advanced glycation end product receptor and toll-like receptor 4 to activate the NFκB pathway (95). Upregulation of DCLK1 may promote the expression of S100A9 protein, while downregulation of DCLK1 directly reduces the expression of S100A9 protein and reduces signal cell infiltration of inflammatory cells (96). Ali et al (94) reported that DCLK1 was overexpressed in liver cells infected with hepatitis C virus and further results indicated that DCLK1 was involved in the replication of hepatitis C virus. According to recent findings, tuft cells express CD300lf (a murine norovirus receptor) and are virally induced to proliferate through this receptor to improve murine norovirus infection. Although research in this area is limited, it is worth considering if tuft cells in the intestine may similarly take part in the replication of hepatitis C and other viruses (97). Liver tissues from patients with cirrhosis and HCC exhibited overexpression of DCLK1, β-catenin and cleaved E-cadherin. DCLK1-overexpressing hepatoma cells induced high levels of β-catenin, α-fetoprotein and SOX9, which led to clonogenicity and dedifferentiated phenotypes (98). In HCC tumors, DCLK1-positive cells have characteristics of CSCs and co-express marker proteins CD133, LGR5, Lin28, AFP and c-Myc (99,100). DCLK1 may be a new target for the treatment of hepatitis C virus-induced tumorigenesis. However, the stem cell characteristics of DCLK1 in hepatocellular carcinoma require confirmation by further research.

All related signaling pathways of DCLK1 in different types of cancer are illustrated in Fig. 4.

DCLK1, a promising anti-tumor target

DCLK1 is one of the most important CSC markers due to its role in promoting tumorigenesis, metastasis, invasion and drug resistance by supporting self-renewal, stemness properties and quiescence, with activating signaling pathways including Wnt, Ras and Notch (101,102). DCLK1 represents a more specific CSC marker, compared with previously studied markers for colorectal, pancreatic and possibly other cancer types, such as gastric cancer, esophageal cancer, breast cancer and renal carcinoma. Development of drugs targeting DCLK1 has been reported, including kinase inhibitors LRRK2-IN-1, XMD8-92 and DCLK1-IN-1; monoclonal antibody CBT-15 (targeting DCLK1′s extracellular C-terminus); and chimeric antigen receptor T-cell therapy (CAR-T_ CBT-511) (91,103105). These drugs exhibited anti-tumor effects via regulating EMT, angiogenesis, proliferation, migration, invasion, apoptosis, cell cycle, DNA damage and stemness in several different cancer types (106) (Table I).

Table I.

Studies reporting inhibition of DCLK1.

Table I.

Studies reporting inhibition of DCLK1.

Cancer typeCancer subtypeInterventionLevel of evidencePathways affectedFunctional effect(Refs.)
CRCNoneHonokiol with ionizing radiationIn vivo and in vitroNotch pathwayInhibition of tumor growth and enhancement of the sensitivity to radiation(115)
CRCNoneAG490 and PD98059In vivo and in vitroNotch pathwayProlactin activates the Notch pathway, then regulating the CSC population(45)
Pancreatic cancerNoneQuinomycinIn vivo and in vitroNotch pathwayTargeting of CSCs and inhibition of tumor growth(47)
CRCNoneAlcea rosea seed extractIn vivo and in vitroNotch pathway and Wnt/β-catenin pathwayInhibition of tumor growth(46)
CRCNoneXAV939, dibenzazepine, siRNA to DCLK1In vivo and in vitro miR-153-3p/Wnt/β-catenin pathwayInfection-induced signals regulate the Wnt signaling pathway(116)
CRCNoneCucurbitacin B and cucurbitacin IIn vivo and in vitroNotch pathwayInhibition of tumor growth and proliferation, induction of apoptosis(117)
Pancreatic cancerNonesiRNA-mediated knockdown of DCAMKL-1In vivo and in vitromiR-200a/ZEB1, miR-144/notch, miR-let-7a/KrasInhibition of stemness, invasion and EMT(68)
Breast cancerTriple negativeCelastrol and triptolideIn vitroNotch pathwayInhibition of proliferation and stemness(87)
CRCNone (2′Z,3′E)-6-bromoindirubin-3′-oximeIn vitroWnt/β-catenin pathwayRBM3 induces stemness(48)
CRCNoneNiclosamideIn vivo and in vitroWnt/β-catenin pathwaySuppression of tumor growth and cancer stem-like cell populations(21)
CRCNoneFoxy5In vivoWnt/β-catenin pathway and PGE2 signaling pathwayModulation of CSCs(50)
CRCNoneSilibininIn vitroWnt/β-catenin pathwayInduction of apoptosis, suppression of migration, elimination of CSCs and attenuation of EMT(118)
Pancreatic cancerNoneXMD8-92In vivo and in vitropluripotency, angiogenesis and anti-apoptotic pathwayInhibition of cell proliferation, EMT and tumor xenograft growth(119)
Pancreatic cancerNoneGemcitabine, everolimus, LY-294002 and ABT-199In vivo and in vitroPI3K/AKT/mTORInhibition of tumorigenesis, metastasis and drug resistance(64)
Pancreatic cancerNoneUNC0638In vivo and in vitroMAPK pathwayInitiation of oncogenic Kras-driven pancreatic carcinogenesis(120)

[i] AG490 is a JAK inhibitor, PD98059 is an ERK inhibitor, XAV939 is a Wnt/β-catenin inhibitor, dibenzazepine is a γ-secretase inhibitor, LRRK2-IN-1 is a DCLK1 inhibitor, (2′Z,3′E)-6-bromoindirubin-3′-oxime is a GSK3β inhibitor, XMD8-92 is a kinase inhibitor, everolimus is an mTOR inhibitor, LY-294002 is a PI3K inhibitor, ABT-199 is a BCL-2 inhibitor, UNC0638 is a G9a inhibitor and Foxy5 is a WNT5A agonist. LEF1, lymphoid enhancer-binding factor 1; RBM3, RNA binding protein RBM3; PGE, prostaglandin E; ZEB1, zinc finger E-box binding homeobox 1; CRC, colorectal cancer; CSC, cancer stem cell; EMT, epithelial-to-mesenchymal transition; siRNA, small inhibitory RNA; miR, microRNA; DCLK1, doublecortin-like kinase 1.

Notch, Wnt/β-catenin and RAS pathways are closely related to DCLK1 in regulating stemness, tumorigenesis, metastasis and drug resistance of several different cancer types. At present, increasing attention is paid to the energy metabolism of CSCs, as the common antitumor treatments aiming to decrease tumor size or reduce proliferating tumor cells may fail to target CSCs, which accounts for this therapeutic treatment resistance (107). Furthermore, the metabolic type for CSCs is primarily dominated by oxidative phosphorylation but not glycolysis, as CSCs consume more oxygen, produce higher levels of ATP and increase mitochondrial mass and membrane potential compared with the bulk of differentiated cancer cells, which rely on glycolysis. The limited but emerging data in this field suggest the importance of further investigation of the relationship between DCLK1 and DCLK1+ CSCs and metabolism (108).

Despite promising findings regarding DCLK1-targeted agents, successfully targeting DCLK1 and avoiding toxicity and other concerns will require a thorough exploration of the roles of DCLK1 in other biological aspects. In 2013, Verissimo et al (109) first reported that knockdown of DCL, a splice variant of DCLK1, is related to reduced mitochondrial activity, which significantly decreased tumor growth in neuroblastoma xenografts. In this study, DCL affected oxidative phosphorylation by interacting with the mitochondrial outer membrane protein outer membrane protein 25/synaptojanin 2 binding protein. However, DCL lacks the kinase domain and kinase catalytic and autoinhibitory activity present in other prominent DCLK1 isoforms (110). However, new evidence suggests that DCLK1 may also be important in conditions of altered metabolism. First, MCF-7 breast cancer cells deregulated the metabolism by triggering transcriptomic reprogramming closely related to DCLK1 levels (111). These findings suggest accelerated dedifferentiation towards a more stem-like state and that DCLK1 may be a key part of this process. Coincidentally, in a non-cancer context, an isoform of DCLK1, candidate plasticity gene 16 (CPG16; also known as DCLK1-BL or DCLK1-Short), was identified as a negative regulator of insulin gene expression, which was increased by long-term exposure of pancreatic β-cells to a high-glucose medium (112). In addition, CPG16 suppressed the jun dimerization protein 2-mediated upregulation of insulin promoter activity in a kinase activity-dependent manner under glucotoxic conditions (113). Of note, Zhao et al (114) reported that glycolysis promotes the expression of DCLK1 and maintains the CSC and EMT phenotypes via maintenance of low reactive oxygen species levels in gemcitabine-resistant Patu8988 pancreatic cancer cells. Together, these findings suggest that DCLK1 may be a key target of glucose metabolism inhibiting drugs such as metformin, which may be helpful in decreasing the incidence of cancer. The limited but emerging data in this field suggest the importance of further investigation of the relationship between DCLK1 function and metabolism.

Conclusion and future directions

DCLK1 as a marker of tuft cells and CSCs is closely related to tumorigenesis and metastasis in various cancer types, including gastrointestinal, breast, renal and other cancers. The DCLK1 isoforms have different functions in the development and progression of the above cancers. Furthermore, the evidence for the emergence of tumors related to various signaling pathways has been linked to DCLK1 in the literature (e.g. Notch, WNT and RAS signaling pathways). Several drugs have been developed by targeting the genetic or kinase activity of DCLK1, and in the future, metabolic regulation via glycolysis and regulation of insulin expression by targeting DCLK1 is worthy of further study.

It is well known that DCLK1 expression is obviously significant in melanoma, testicular cancer, lymphoma and endometrial cancer (9), besides the above ones, but only a small number of studies have been performed on them until now. Thus, by including these data, it is esteemed that other groups in these specific subfields of oncology may become aware of and consider researching DCLK1 in their respective projects.

Acknowledgements

Not applicable.

Funding

This research was funded by the CHEN Ke-ji Integrative Medicine Development Fund (grant nos. CKJ 2021011 and CKJ 2021010), National Natural Science Foundation of China (grant no. 81873166).

Availability of data and materials

Data sharing is not applicable.

Authors' contributions

Study conception and design: ZC, ZY and QL; Searching and selection the literature: QL, HF, HC and JD; writing the original manuscript: QL, HF, HC; revision of the manuscript: ZC, ZY and NW. All authors read and approved the final manuscript. Data authentication is not applicable.

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:

DCLK1

doublecortin-like kinase 1

CSCs

cancer stem cells

DCX

doublecortin

AcTub

acetylated tubulin

EMT

epithelial-mesenchymal transition

References

1 

May R, Riehl TE, Hunt C, Sureban SM, Anant S and Houchen CW: Identification of a novel putative gastrointestinal stem cell and adenoma stem cell marker, doublecortin and CaM kinase-like-1, following radiation injury and in adenomatous polyposis coli/multiple intestinal neoplasia mice. Stem Cells. 26:630–637. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Vega KJ, May R, Sureban SM, Lightfoot SA, Qu D, Reed A, Weygant N, Ramanujam R, Souza R, Madhoun M, et al: Identification of the putative intestinal stem cell marker doublecortin and CaM kinase-like-1 in Barrett's esophagus and esophageal adenocarcinoma. J Gastroenterol Hepatol. 27:773–780. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Weygant N, Qu D, May R, Tierney RM, Berry WL, Zhao L, Agarwal S, Chandrakesan P, Chinthalapally HR, Murphy NT, et al: DCLK1 is a broadly dysregulated target against epithelial-mesenchymal transition, focal adhesion, and stemness in clear cell renal carcinoma. Oncotarget. 6:2193–2205. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Nakanishi Y, Seno H, Fukuoka A, Ueo T, Yamaga Y, Maruno T, Nakanishi N, Kanda K, Komekado H, Kawada M, et al: Dclk1 distinguishes between tumor and normal stem cells in the intestine. Nat Genet. 45:98–103. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Delgiorno KE, Hall JC, Takeuchi KK, Pan FC, Halbrook CJ, Washington MK, Olive KP, Spence JR, Sipos B, Wright CV, et al: Identification and manipulation of biliary metaplasia in pancreatic tumors. Gastroenterology. 146:233–244.e5. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Saqui-Salces M, Keeley TM, Grosse AS, Qiao XT, El-Zaatari M, Gumucio DL, Samuelson LC and Merchant JL: Gastric tuft cells express DCLK1 and are expanded in hyperplasia. Histochem Cell Biol. 136:191–204. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Gerbe F, van Es JH, Makrini L, Brulin B, Mellitzer G, Robine S, Romagnolo B, Shroyer NF, Bourgaux JF, Pignodel C, et al: Distinct ATOH1 and Neurog3 requirements define tuft cells as a new secretory cell type in the intestinal epithelium. J Cell Biol. 192:767–780. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Howitt MR, Lavoie S, Michaud M, Blum AM, Tran SV, Weinstock JV, Gallini CA, Redding K, Margolskee RF, Osborne LC, et al: Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the gut. Science. 351:1329–1333. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Westphalen CB, Quante M and Wang TC: Functional implication of Dclk1 and Dclk1-expressing cells in cancer. Small GTPases. 8:164–171. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Yi J, Bergstrom K, Fu J, Shan X, McDaniel JM, McGee S, Qu D, Houchen CW, Liu X and Xia L: Dclk1 in tuft cells promotes inflammation-driven epithelial restitution and mitigates chronic colitis. Cell Death Differ. 26:1656–1669. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Patel O, Dai W, Mentzel M, Griffin MD, Serindoux J, Gay Y, Fischer S, Sterle S, Kropp A, Burns CJ, et al: Biochemical and structural insights into doublecortin-like kinase domain 1. Structure. 24:1550–1561. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Cheung AS, de Rooy C, Levinger I, Rana K, Clarke MV, How JM, Garnham A, McLean C, Zajac JD, Davey RA and Grossmann M: Actin alpha cardiac muscle 1 gene expression is upregulated in the skeletal muscle of men undergoing androgen deprivation therapy for prostate cancer. J Steroid Biochem Mol Biol. 174:56–64. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Matsumoto N, Pilz DT and Ledbetter DH: Genomic structure, chromosomal mapping, and expression pattern of human DCAMKL1 (KIAA0369), a homologue of DCX (XLIS). Genomics. 56:179–183. 1999. View Article : Google Scholar : PubMed/NCBI

14 

Burgess HA and Reiner O: Cleavage of doublecortin-like kinase by calpain releases an active kinase fragment from a microtubule anchorage domain. J Biol Chem. 276:36397–36403. 2001. View Article : Google Scholar : PubMed/NCBI

15 

Kim MH, Cierpicki T, Derewenda U, Krowarsch D, Feng Y, Devedjiev Y, Dauter Z, Walsh CA, Otlewski J, Bushweller JH and Derewenda ZS: The DCX-domain tandems of doublecortin and doublecortin-like kinase. Nat Struct Biol. 10:324–333. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Lin PT, Gleeson JG, Corbo JC, Flanagan L and Walsh CA: DCAMKL1 encodes a protein kinase with homology to doublecortin that regulates microtubule polymerization. J Neurosci. 20:9152–9161. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Engels BM, Schouten TG, van Dullemen J, Gosens I and Vreugdenhil E: Functional differences between two DCLK splice variants. Brain Res Mol Brain Res. 120:103–114. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Burgess HA and Reiner O: Alternative splice variants of doublecortin-like kinase are differentially expressed and have different kinase activities. J Biol Chem. 277:17696–17705. 2002. View Article : Google Scholar : PubMed/NCBI

19 

O'Connell MR, Sarkar S, Luthra GK, Okugawa Y, Toiyama Y, Gajjar AH, Qiu S, Goel A and Singh P: Epigenetic changes and alternate promoter usage by human colon cancers for expressing DCLK1-isoforms: Clinical Implications. Sci Rep. 5:149832015. View Article : Google Scholar : PubMed/NCBI

20 

Walker TL, Yasuda T, Adams DJ and Bartlett PF: The doublecortin-expressing population in the developing and adult brain contains multipotential precursors in addition to neuronal-lineage cells. J Neurosci. 27:3734–3742. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Park SY, Kim JY, Choi JH, Kim JH, Lee CJ, Singh P, Sarkar S, Baek JH and Nam JS: Inhibition of LEF1-mediated DCLK1 by niclosamide attenuates colorectal cancer stemness. Clin Cancer Res. 25:1415–1429. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Sarkar S, Popov VL, O'Connell MR, Stevenson HL, Lee BS, Obeid RA, Luthra GK and Singh P: A novel antibody against cancer stem cell biomarker, DCLK1-S, is potentially useful for assessing colon cancer risk after screening colonoscopy. Lab Invest. 97:1245–1261. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Andresen K, Boberg KM, Vedeld HM, Honne H, Hektoen M, Wadsworth CA, Clausen OP, Karlsen TH, Foss A, Mathisen O, et al: Novel target genes and a valid biomarker panel identified for cholangiocarcinoma. Epigenetics. 7:1249–1257. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Westphalen CB, Takemoto Y, Tanaka T, Macchini M, Jiang Z, Renz BW, Chen X, Ormanns S, Nagar K, Tailor Y, et al: Dclk1 defines quiescent pancreatic progenitors that promote injury-induced regeneration and tumorigenesis. Cell Stem Cell. 18:441–455. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Yamaga Y, Fukuda A, Nakanishi Y, Goto N, Matsumoto Y, Yoshioka T, Maruno T, Chiba T and Seno H: Gene expression profile of Dclk1+ cells in intestinal tumors. Dig Liver Dis. 50:1353–1361. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Ge Y, Liu H, Zhang Y, Liu J, Yan R, Xiao Z, Fan X, Huang X and An G: Inhibition of DCLK1 kinase reverses epithelial-mesenchymal transition and restores T-cell activity in pancreatic ductal adenocarcinoma. Transl Oncol. 17:1013172022.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

27 

May R, Sureban SM, Hoang N, Riehl TE, Lightfoot SA, Ramanujam R, Wyche JH, Anant S and Houchen CW: Doublecortin and CaM kinase-like-1 and leucine-rich-repeat-containing G-protein-coupled receptor mark quiescent and cycling intestinal stem cells, respectively. Stem Cells. 27:2571–2579. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Ladang A, Rapino F, Heukamp LC, Tharun L, Shostak K, Hermand D, Delaunay S, Klevernic I, Jiang Z, Jacques N, et al: Elp3 drives Wnt-dependent tumor initiation and regeneration in the intestine. J Exp Med. 212:2057–2075. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Leppänen J, Helminen O, Huhta H, Kauppila JH, Miinalainen I, Ronkainen VP, Saarnio J, Lehenkari PP and Karttunen TJ: Doublecortin-like kinase 1-positive enterocyte-a new cell type in human intestine. APMIS. 124:958–965. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Westphalen CB, Asfaha S, Hayakawa Y, Takemoto Y, Lukin DJ, Nuber AH, Brandtner A, Setlik W, Remotti H, Muley A, et al: Long-lived intestinal tuft cells serve as colon cancer-initiating cells. J Clin Invest. 124:1283–1295. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Qu D, Weygant N, May R, Chandrakesan P, Madhoun M, Ali N, Sureban SM, An G, Schlosser MJ and Houchen CW: Ablation of doublecortin-like kinase 1 in the colonic epithelium exacerbates dextran sulfate sodium-induced colitis. PLoS One. 10:e01342122015. View Article : Google Scholar : PubMed/NCBI

32 

Gerbe F, Brulin B, Makrini L, Legraverend C and Jay P: DCAMKL-1 expression identifies Tuft cells rather than stem cells in the adult mouse intestinal epithelium. Gastroenterology. 137:2179–2181. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Eini L, Naseri M, Karimi-Busheri F, Bozorgmehr M, Ghods R and Madjd Z: Primary colonospheres maintain stem cell-like key features after cryopreservation. J Cell Physiol. 235:2452–2463. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Chandrakesan P, Yao J, Qu D, May R, Weygant N, Ge Y, Ali N, Sureban SM, Gude M, Vega K, et al: Dclk1, a tumor stem cell marker, regulates pro-survival signaling and self-renewal of intestinal tumor cells. Mol Cancer. 16:302017. View Article : Google Scholar : PubMed/NCBI

35 

Wang X, Yang Y and Huycke MM: Commensal-infected macrophages induce dedifferentiation and reprogramming of epithelial cells during colorectal carcinogenesis. Oncotarget. 8:102176–102190. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Gagliardi G, Goswami M, Passera R and Bellows CF: DCLK1 immunoreactivity in colorectal neoplasia. Clin Exp Gastroenterol. 5:35–42. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Vedeld HM, Skotheim RI, Lothe RA and Lind GE: The recently suggested intestinal cancer stem cell marker DCLK1 is an epigenetic biomarker for colorectal cancer. Epigenetics. 9:346–450. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Takiyama A, Tanaka T, Kazama S, Nagata H, Kawai K, Hata K, Otani K, Nishikawa T, Sasaki K, Kaneko M, et al: DCLK1 expression in colorectal polyps increases with the severity of dysplasia. In Vivo. 32:365–371. 2018.PubMed/NCBI

39 

Ahmed I, Roy BC, Raach RT, Owens SM, Xia L, Anant S, Sampath V and Umar S: Enteric infection coupled with chronic Notch pathway inhibition alters colonic mucus composition leading to dysbiosis, barrier disruption and colitis. PLoS One. 13:e02067012018. View Article : Google Scholar : PubMed/NCBI

40 

Mirzaei A, Tavoosidana G, Modarressi MH, Rad AA, Fazeli MS, Shirkoohi R, Tavakoli-Yaraki M and Madjd Z: Upregulation of circulating cancer stem cell marker, DCLK1 but not Lgr5, in chemoradiotherapy-treated colorectal cancer patients. Tumour Biol. 36:4801–4810. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Wang J, Yokoyama Y, Hirose H, Shimomura Y, Bonkobara S, Itakura H, Kouda S, Morimoto Y, Minami K, Takahashi H, et al: Functional assessment of miR-1291 in colon cancer cells. Int J Oncol. 60:132022. View Article : Google Scholar : PubMed/NCBI

42 

Razi S, Sadeghi A, Asadi-Lari Z, Tam KJ, Kalantari E and Madjd Z: DCLK1, a promising colorectal cancer stem cell marker, regulates tumor progression and invasion through miR-137 and miR-15a dependent manner. Clin Exp Med. 21:139–147. 2021. View Article : Google Scholar : PubMed/NCBI

43 

Sureban SM, May R, Mondalek FG, Qu D, Ponnurangam S, Pantazis P, Anant S, Ramanujam RP and Houchen CW: Nanoparticle-based delivery of siDCAMKL-1 increases microRNA-144 and inhibits colorectal cancer tumor growth via a Notch-1 dependent mechanism. J Nanobiotechnology. 9:402011. View Article : Google Scholar : PubMed/NCBI

44 

Kwon MS, Chung HK, Xiao L, Yu TX, Wang SR, Piao JJ, Gorospe M and Wang JY: MicroRNA-195 regulates Tuft cell function in the intestinal epithelium by altering translation of DCLK1. Am J Physiol Cell Physiol. 320:C1042–C1054. 2021. View Article : Google Scholar : PubMed/NCBI

45 

Neradugomma NK, Subramaniam D, Tawfik OW, Goffin V, Kumar TR, Jensen RA and Anant S: Prolactin signaling enhances colon cancer stemness by modulating Notch signaling in a Jak2-STAT3/ERK manner. Carcinogenesis. 35:795–806. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Ahmed I, Roy BC, Subramaniam D, Ganie SA, Kwatra D, Dixon D, Anant S, Zargar MA and Umar S: An ornamental plant targets epigenetic signaling to block cancer stem cell-driven colon carcinogenesis. Carcinogenesis. 37:385–396. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Ponnurangam S, Dandawate PR, Dhar A, Tawfik OW, Parab RR, Mishra PD, Ranadive P, Sharma R, Mahajan G, Umar S, et al: Quinomycin A targets Notch signaling pathway in pancreatic cancer stem cells. Oncotarget. 7:3217–3232. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Venugopal A, Subramaniam D, Balmaceda J, Roy B, Dixon DA, Umar S, Weir SJ and Anant S: RNA binding protein RBM3 increases β-catenin signaling to increase stem cell characteristics in colorectal cancer cells. Mol Carcinog. 55:1503–1516. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Krishnamachary B, Subramaniam D, Dandawate P, Ponnurangam S, Srinivasan P, Ramamoorthy P, Umar S, Thomas SM, Dhar A, Septer S, et al: Targeting transcription factor TCF4 by γ-mangostin, a natural xanthone. Oncotarget. 10:5576–5591. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Osman J, Bellamkonda K, Liu Q, Andersson T and Sjölander A: The WNT5A agonist Foxy5 reduces the number of colonic cancer stem cells in a xenograft mouse model of human colonic cancer. Anticancer Res. 39:1719–1728. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Hammond DE, Mageean CJ, Rusilowicz EV, Wickenden JA, Clague MJ and Prior IA: Differential reprogramming of isogenic colorectal cancer cells by distinct activating KRAS mutations. J Proteome Res. 14:1535–1546. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Qiu W, Remotti HE, Tang SM, Wang E, Dobberteen L, Lee Youssof A, Lee JH, Cheung EC and Su GH: Pancreatic DCLK1+ cells originate distinctly from PDX1+ progenitors and contribute to the initiation of intraductal papillary mucinous neoplasm in mice. Cancer Lett. 423:71–79. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Bailey JM, Alsina J, Rasheed ZA, McAllister FM, Fu YY, Plentz R, Zhang H, Pasricha PJ, Bardeesy N, Matsui W, et al: DCLK1 marks a morphologically distinct subpopulation of cells with stem cell properties in preinvasive pancreatic cancer. Gastroenterology. 146:245–256. 2014. View Article : Google Scholar : PubMed/NCBI

54 

May R, Sureban SM, Lightfoot SA, Hoskins AB, Brackett DJ, Postier RG, Ramanujam R, Rao CV, Wyche JH, Anant S and Houchen CW: Identification of a novel putative pancreatic stem/progenitor cell marker DCAMKL-1 in normal mouse pancreas. Am J Physiol Gastrointest Liver Physiol. 299:G303–G310. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Sureban SM, May R, Qu D, Weygant N, Chandrakesan P, Ali N, Lightfoot SA, Pantazis P, Rao CV, Postier RG and Houchen CW: DCLK1 regulates pluripotency and angiogenic factors via microRNA-dependent mechanisms in pancreatic cancer. PLoS One. 8:e739402013. View Article : Google Scholar : PubMed/NCBI

56 

Yao ZX, Qin ML, Liu JJ, Chen XS and Zhou DS: In vitro cultivation of human fetal pancreatic ductal stem cells and their differentiation into insulin-producing cells. World J Gastroenterol. 10:1452–1456. 2004. View Article : Google Scholar : PubMed/NCBI

57 

Seeley ES, Carrière C, Goetze T, Longnecker DS and Korc M: Pancreatic cancer and precursor pancreatic intraepithelial neoplasia lesions are devoid of primary cilia. Cancer Res. 69:422–430. 2009. View Article : Google Scholar : PubMed/NCBI

58 

Lee H, Basso IN and Kim DDH: Target spectrum of the BCR-ABL tyrosine kinase inhibitors in chronic myeloid leukemia. Int J Hematol. 113:632–641. 2021. View Article : Google Scholar : PubMed/NCBI

59 

Zhang Y, Gao C, Cao F, Wu Y, Chen S, Han X, Mo J, Qiu Z, Fan W, Zhou P and Shen L: Pan-cancer analysis of IGF-1 and IGF-1R as potential prognostic biomarkers and immunotherapy targets. Front Oncol. 11:7553412021. View Article : Google Scholar : PubMed/NCBI

60 

Zhang Y, Zoltan M, Riquelme E, Xu H, Sahin I, Castro-Pando S, Montiel MF, Chang K, Jiang Z, Ling J, et al: Immune cell production of interleukin 17 induces stem cell features of pancreatic intraepithelial neoplasia cells. Gastroenterology. 155:210–223.e3. 2018. View Article : Google Scholar : PubMed/NCBI

61 

DelGiorno KE, Naeem RF, Fang L, Chung CY, Ramos C, Luhtala N, O'Connor C, Hunter T, Manor U and Wahl GM: Tuft cell formation reflects epithelial plasticity in pancreatic injury: Implications for modeling human pancreatitis. Front Physiol. 11:882020. View Article : Google Scholar : PubMed/NCBI

62 

Park JT and Leach SD: Zebrafish model of KRAS-initiated pancreatic cancer. Anim Cells Syst (Seoul). 22:353–359. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Zhou B, Irwanto A, Guo YM, Bei JX, Wu Q, Chen G, Zhang TP, Lei JJ, Feng QS, Chen LZ, et al: Exome sequencing and digital PCR analyses reveal novel mutated genes related to the metastasis of pancreatic ductal adenocarcinoma. Cancer Biol Ther. 13:871–879. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Qu D, Weygant N, Yao J, Chandrakesan P, Berry WL, May R, Pitts K, Husain S, Lightfoot S, Li M, et al: Overexpression of DCLK1-AL increases tumor cell invasion, drug resistance, and KRAS activation and can be targeted to inhibit tumorigenesis in pancreatic cancer. J Oncol. 2019:64029252019. View Article : Google Scholar : PubMed/NCBI

65 

Chandrakesan P, Panneerselvam J, May R, Weygant N, Qu D, Berry WR, Pitts K, Stanger BZ, Rao CV, Bronze MS and Houchen CW: DCLK1-isoform2 alternative splice variant promotes pancreatic tumor immunosuppressive M2-macrophage polarization. Mol Cancer Ther. 19:1539–1549. 2020. View Article : Google Scholar : PubMed/NCBI

66 

Xu Z, Pang TCY, Liu AC, Pothula SP, Mekapogu AR, Perera CJ, Murakami T, Goldstein D, Pirola RC, Wilson JS and Apte MV: Targeting the HGF/c-MET pathway in advanced pancreatic cancer: A key element of treatment that limits primary tumour growth and eliminates metastasis. Br J Cancer. 122:1486–1495. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Rieder S, Michalski CW, Friess H and Kleeff J: Insulin-like growth factor signaling as a therapeutic target in pancreatic cancer. Anticancer Agents Med Chem. 11:427–433. 2011. View Article : Google Scholar : PubMed/NCBI

68 

Sureban SM, May R, Lightfoot SA, Hoskins AB, Lerner M, Brackett DJ, Postier RG, Ramanujam R, Mohammed A, Rao CV, et al: DCAMKL-1 regulates epithelial-mesenchymal transition in human pancreatic cells through a miR-200a-dependent mechanism. Cancer Res. 71:2328–2338. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Bjerknes M, Khandanpour C, Moroy T, Fujiyama T, Hoshino M, Klisch TJ, Ding Q, Gan L, Wang J, Martín MG and Cheng H: Origin of the brush cell lineage in the mouse intestinal epithelium. Dev Biol. 362:194–218. 2012. View Article : Google Scholar : PubMed/NCBI

70 

Ali Y, Lin Y, Gharibo MM, Gounder MK, Stein MN, Lagattuta TF, Egorin MJ, Rubin EH and Poplin EA: Phase I and pharmacokinetic study of imatinib mesylate (Gleevec) and gemcitabine in patients with refractory solid tumors. Clin Cancer Res. 13:5876–5882. 2007. View Article : Google Scholar : PubMed/NCBI

71 

Giannakis M, Stappenbeck TS, Mills JC, Leip DG, Lovett M, Clifton SW, Ippolito JE, Glasscock JI, Arumugam M, Brent MR and Gordon JI: Molecular properties of adult mouse gastric and intestinal epithelial progenitors in their niches. J Biol Chem. 281:11292–11300. 2006. View Article : Google Scholar : PubMed/NCBI

72 

Weygant N, Ge Y, Qu D, Kaddis JS, Berry WL, May R, Chandrakesan P, Bannerman-Menson E, Vega KJ, Tomasek JJ, et al: Survival of patients with gastrointestinal cancers can be predicted by a surrogate microRNA signature for cancer stem-like cells marked by DCLK1 kinase. Cancer Res. 76:4090–4099. 2016. View Article : Google Scholar : PubMed/NCBI

73 

Zhang Y and Huang X: Investigation of doublecortin and calcium/calmodulin-dependent protein kinase-like-1-expressing cells in the mouse stomach. J Gastroenterol Hepatol. 25:576–582. 2010. View Article : Google Scholar : PubMed/NCBI

74 

Meng QB, Yu JC, Kang WM, Ma ZQ, Zhou WX, Li J, Zhou L, Cao ZJ and Tian SB: Expression of doublecortin-like kinase 1 in human gastric cancer and its correlation with prognosis. Zhongguo Yi Xue Ke Xue Yuan Xue Bao. 35:639–644. 2013.(In Chinese). PubMed/NCBI

75 

Sureban SM, Qu D and Houchen CW: Regulation of miRNAs by agents targeting the tumor stem cell markers DCLK1, MSI1, LGR5, and BMI1. Curr Pharmacol Rep. 1:217–222. 2015. View Article : Google Scholar : PubMed/NCBI

76 

Liu ZQ, He WF, Wu YJ, Zhao SL, Wang L, Ouyang YY and Tang SY: LncRNA SNHG1 promotes EMT process in gastric cancer cells through regulation of the miR-15b/DCLK1/Notch1 axis. BMC Gastroenterol. 20:1562020. View Article : Google Scholar : PubMed/NCBI

77 

Carli ALE, Afshar-Sterle S, Rai A, Fang H, O'Keefe R, Tse J, Ferguson FM, Gray NS, Ernst M, Greening DW and Buchert M: Cancer stem cell marker DCLK1 reprograms small extracellular vesicles toward migratory phenotype in gastric cancer cells. Proteomics. 21:e20000982021. View Article : Google Scholar : PubMed/NCBI

78 

Dai J, Li ZX, Zhang Y, Ma JL, Zhou T, You WC, Li WQ and Pan KF: Whole genome messenger RNA profiling identifies a novel signature to predict gastric cancer survival. Clin Transl Gastroenterol. 10:e000042019. View Article : Google Scholar : PubMed/NCBI

79 

Schellnegger R, Quante A, Rospleszcz S, Schernhammer M, Höhl B, Tobiasch M, Pastula A, Brandtner A, Abrams JA, Strauch K, et al: Goblet cell ratio in combination with differentiation and stem cell markers in barrett esophagus allow distinction of patients with and without esophageal adenocarcinoma. Cancer Prev Res (Phila). 10:55–66. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Ge Y, Fan X, Huang X, Weygant N, Xiao Z, Yan R, Liu H, Liu J, An G and Yao J: DCLK1-short splice variant promotes esophageal squamous cell carcinoma progression via the MAPK/ERK/MMP2 pathway. Mol Cancer Res. 19:1980–1991. 2021. View Article : Google Scholar : PubMed/NCBI

81 

Zhang L, Zhou S, Guo E, Chen X, Yang J and Li X: DCLK1 inhibition attenuates tumorigenesis and improves chemosensitivity in esophageal squamous cell carcinoma by inhibiting β-catenin/c-Myc signaling. Pflugers Arch. 472:1041–1049. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Whorton J, Sureban SM, May R, Qu D, Lightfoot SA, Madhoun M, Johnson M, Tierney WM, Maple JT, Vega KJ and Houchen CW: DCLK1 is detectable in plasma of patients with Barrett's esophagus and esophageal adenocarcinoma. Dig Dis Sci. 60:509–513. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Quante M, Bhagat G, Abrams JA, Marache F, Good P, Lee MD, Lee Y, Friedman R, Asfaha S, Dubeykovskaya Z, et al: Bile acid and inflammation activate gastric cardia stem cells in a mouse model of Barrett-like metaplasia. Cancer Cell. 21:36–51. 2012. View Article : Google Scholar : PubMed/NCBI

84 

Haakensen VD, Bjøro T, Lüders T, Riis M, Bukholm IK, Kristensen VN, Troester MA, Homen MM, Ursin G, Børresen-Dale AL and Helland Å: Serum estradiol levels associated with specific gene expression patterns in normal breast tissue and in breast carcinomas. BMC Cancer. 11:3322011. View Article : Google Scholar : PubMed/NCBI

85 

Liu YH, Tsang JY, Ni YB, Hlaing T, Chan SK, Chan KF, Ko CW, Mujtaba SS and Tse GM: Doublecortin-like kinase 1 expression associates with breast cancer with neuroendocrine differentiation. Oncotarget. 7:1464–1476. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Zhao S, Ma D, Xiao Y, Li XM, Ma JL, Zhang H, Xu XL, Lv H, Jiang WH, Yang WT, et al: Molecular subtyping of triple-negative breast cancers by immunohistochemistry: Molecular Basis and clinical relevance. Oncologist. 25:e1481–e1491. 2020. View Article : Google Scholar : PubMed/NCBI

87 

Ramamoorthy P, Dandawate P, Jensen RA and Anant S: Celastrol and triptolide suppress stemness in triple negative breast cancer: Notch as a therapeutic target for stem cells. Biomedicines. 9:4822021. View Article : Google Scholar : PubMed/NCBI

88 

Wang YL, Li Y, Ma YG and Wu WY: DCLK1 promotes malignant progression of breast cancer by regulating Wnt/β-catenin signaling pathway. Eur Rev Med Pharmacol Sci. 23:9489–9498. 2019.PubMed/NCBI

89 

Liu H, Wen T, Zhou Y, Fan X, Du T, Gao T, Li L, Liu J, Yang L, Yao J, et al: DCLK1 plays a metastatic-promoting role in human breast cancer cells. Biomed Res Int. 2019:10619792019.PubMed/NCBI

90 

Wang J, Wang S, Zhou J and Qian Q: miR-424-5p regulates cell proliferation, migration and invasion by targeting doublecortin-like kinase 1 in basal-like breast cancer. Biomed Pharmacother. 102:147–152. 2018. View Article : Google Scholar : PubMed/NCBI

91 

Ge Y, Weygant N, Qu D, May R, Berry WL, Yao J, Chandrakesan P, Zheng W, Zhao L, Zhao KL, et al: Alternative splice variants of DCLK1 mark cancer stem cells, promote self-renewal and drug-resistance, and can be targeted to inhibit tumorigenesis in kidney cancer. Int J Cancer. 143:1162–1175. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Ding L, Yang Y, Ge Y, Lu Q, Yan Z, Chen X, Du J, Hafizi S, Xu X, Yao J, et al: Inhibition of DCLK1 with DCLK1-IN-1 suppresses renal cell carcinoma invasion and stemness and promotes cytotoxic T-cell-mediated anti-tumor immunity. Cancers (Basel). 13:57292021. View Article : Google Scholar : PubMed/NCBI

93 

Sureban SM, Madhoun MF, May R, Qu D, Ali N, Fazili J, Weygant N, Chandrakesan P, Ding K, Lightfoot SA and Houchen CW: Plasma DCLK1 is a marker of hepatocellular carcinoma (HCC): Targeting DCLK1 prevents HCC tumor xenograft growth via a microRNA-dependent mechanism. Oncotarget. 6:37200–37215. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Ali N, Chandrakesan P, Nguyen CB, Husain S, Gillaspy AF, Huycke M, Berry WL, May R, Qu D, Weygant N, et al: Inflammatory and oncogenic roles of a tumor stem cell marker doublecortin-like kinase (DCLK1) in virus-induced chronic liver diseases. Oncotarget. 6:20327–20344. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Girotto G, Vuckovic D, Buniello A, Lorente-Cánovas B, Lewis M, Gasparini P and Steel KP: Expression and replication studies to identify new candidate genes involved in normal hearing function. PLoS One. 9:e853522014. View Article : Google Scholar : PubMed/NCBI

96 

Srikrishna G: S100A8 and S100A9: New insights into their roles in malignancy. J Innate Immun. 4:31–40. 2012. View Article : Google Scholar : PubMed/NCBI

97 

Wilen CB, Lee S, Hsieh LL, Orchard RC, Desai C, Hykes BL Jr, McAllaster MR, Balce DR, Feehley T, Brestoff JR, et al: Tropism for tuft cells determines immune promotion of norovirus pathogenesis. Science. 360:204–208. 2018. View Article : Google Scholar : PubMed/NCBI

98 

Ali N, Nguyen CB, Chandrakesan P, Wolf RF, Qu D, May R, Goretsky T, Fazili J, Barrett TA, Li M, et al: Doublecortin-like kinase 1 promotes hepatocyte clonogenicity and oncogenic programming via non-canonical β-catenin-dependent mechanism. Sci Rep. 10:105782020. View Article : Google Scholar : PubMed/NCBI

99 

Ali N, Allam H, May R, Sureban SM, Bronze MS, Bader T, Umar S, Anant S and Houchen CW: Hepatitis C virus-induced cancer stem cell-like signatures in cell culture and murine tumor xenografts. J Virol. 85:12292–12303. 2011. View Article : Google Scholar : PubMed/NCBI

100 

Ali N, Allam H, Bader T, May R, Basalingappa KM, Berry WL, Chandrakesan P, Qu D, Weygant N, Bronze MS, et al: Fluvastatin interferes with hepatitis C virus replication via microtubule bundling and a doublecortin-like kinase-mediated mechanism. PLoS One. 8:e803042013. View Article : Google Scholar : PubMed/NCBI

101 

Pattabiraman DR and Weinberg RA: Tackling the cancer stem cells-what challenges do they pose? Nat Rev Drug Discov. 13:497–512. 2014. View Article : Google Scholar : PubMed/NCBI

102 

Brooks MD, Burness ML and Wicha MS: Therapeutic implications of cellular heterogeneity and plasticity in breast cancer. Cell Stem Cell. 17:260–271. 2015. View Article : Google Scholar : PubMed/NCBI

103 

Weygant N, Qu D, Berry WL, May R, Chandrakesan P, Owen DB, Sureban SM, Ali N, Janknecht R and Houchen CW: Small molecule kinase inhibitor LRRK2-IN-1 demonstrates potent activity against colorectal and pancreatic cancer through inhibition of doublecortin-like kinase 1. Mol Cancer. 13:1032014. View Article : Google Scholar : PubMed/NCBI

104 

Ferguson FM, Nabet B, Raghavan S, Liu Y, Leggett AL, Kuljanin M, Kalekar RL, Yang A, He S, Wang J, et al: Discovery of a selective inhibitor of doublecortin like kinase 1. Nat Chem Biol. 16:635–643. 2020. View Article : Google Scholar : PubMed/NCBI

105 

Sureban SM, Berahovich R, Zhou H, Xu S, Wu L, Ding K, May R, Qu D, Bannerman-Menson E, Golubovskaya V and Houchen CW: DCLK1 monoclonal antibody-based CAR-T cells as a novel treatment strategy against human colorectal cancers. Cancers (Basel). 12:542019. View Article : Google Scholar : PubMed/NCBI

106 

Cao Z, Weygant N, Chandrakesan P, Houchen CW, Peng J and Qu D: Tuft and cancer stem cell marker DCLK1: A new target to enhance anti-tumor immunity in the tumor microenvironment. Cancers (Basel). 12:38012020. View Article : Google Scholar : PubMed/NCBI

107 

Chae YC and Kim JH: Cancer stem cell metabolism: Target for cancer therapy. BMB Rep. 51:319–326. 2018. View Article : Google Scholar : PubMed/NCBI

108 

Sancho P, Barneda D and Heeschen C: Hallmarks of cancer stem cell metabolism. Br J Cancer. 114:1305–1312. 2016. View Article : Google Scholar : PubMed/NCBI

109 

Verissimo CS, Elands R, Cheng S, Saaltink DJ, ter Horst JP, Alme MN, Pont C, van de Water B, Håvik B, Fitzsimons CP and Vreugdenhil E: Silencing of doublecortin-like (DCL) results in decreased mitochondrial activity and delayed neuroblastoma tumor growth. PLoS One. 8:e757522013. View Article : Google Scholar : PubMed/NCBI

110 

Patel O, Roy MJ, Kropp A, Hardy JM, Dai W and Lucet IS: Structural basis for small molecule targeting of doublecortin like kinase 1 with DCLK1-IN-1. Commun Biol. 4:11052021. View Article : Google Scholar : PubMed/NCBI

111 

Oliveras-Ferraros C, Vazquez-Martin A, Cuyàs E, Corominas-Faja B, Rodríguez-Gallego E, Fernández-Arroyo S, Martin-Castillo B, Joven J and Menendez JA: Acquired resistance to metformin in breast cancer cells triggers transcriptome reprogramming toward a degradome-related metastatic stem-like profile. Cell Cycle. 13:1132–1144. 2014. View Article : Google Scholar : PubMed/NCBI

112 

Nakane T, Ido A, Higuchi T, Todaka H, Morisawa K, Nagamine T, Fukunaga K, Sakamoto S, Murao K and Sugiyama Y: Candidate plasticity gene 16 mediates suppression of insulin gene expression in rat insulinoma INS-1 cells under glucotoxic conditions. Biochem Biophys Res Commun. 512:189–195. 2019. View Article : Google Scholar : PubMed/NCBI

113 

Nakane T, Matsumoto S, Iida S, Ido A, Fukunaga K, Murao K and Sugiyama Y: Candidate plasticity gene 16 and jun dimerization protein 2 are involved in the suppression of insulin gene expression in rat pancreatic INS-1 β-cells. Mol Cell Endocrinol. 527:1112402021. View Article : Google Scholar : PubMed/NCBI

114 

Zhao H, Duan Q, Zhang Z, Li H, Wu H, Shen Q, Wang C and Yin T: Up-regulation of glycolysis promotes the stemness and EMT phenotypes in gemcitabine-resistant pancreatic cancer cells. J Cell Mol Med. 21:2055–2067. 2017. View Article : Google Scholar : PubMed/NCBI

115 

Ponnurangam S, Mammen JM, Ramalingam S, He Z, Zhang Y, Umar S, Subramaniam D and Anant S: Honokiol in combination with radiation targets notch signaling to inhibit colon cancer stem cells. Mol Cancer Ther. 11:963–972. 2012. View Article : Google Scholar : PubMed/NCBI

116 

Ahmed I, Roy BC, Rao Jakkula LUM, Subramaniam D, Dandawate P, Anant S, Sampath V and Umar S: Infection-induced signals generated at the plasma membrane epigenetically regulate Wnt signaling in vitro and in vivo. J Biol Chem. 295:1021–1035. 2020. View Article : Google Scholar : PubMed/NCBI

117 

Dandawate P, Subramaniam D, Panovich P, Standing D, Krishnamachary B, Kaushik G, Thomas SM, Dhar A, Weir SJ, Jensen RA and Anant S: Cucurbitacin B and I inhibits colon cancer growth by targeting the Notch signaling pathway. Sci Rep. 10:12902020. View Article : Google Scholar : PubMed/NCBI

118 

Sameri S, Saidijam M, Bahreini F and Najafi R: Cancer chemopreventive activities of silibinin on colorectal cancer through regulation of E-cadherin/β-catenin pathway. Nutr Cancer. 73:1389–1399. 2021. View Article : Google Scholar : PubMed/NCBI

119 

Sureban SM, May R, Weygant N, Qu D, Chandrakesan P, Bannerman-Menson E, Ali N, Pantazis P, Westphalen CB, Wang TC and Houchen CW: XMD8-92 inhibits pancreatic tumor xenograft growth via a DCLK1-dependent mechanism. Cancer Lett. 351:151–161. 2014. View Article : Google Scholar : PubMed/NCBI

120 

Kato H, Tateishi K, Fujiwara H, Ijichi H, Yamamoto K, Nakatsuka T, Kakiuchi M, Sano M, Kudo Y, Hayakawa Y, et al: Deletion of histone methyltransferase G9a suppresses mutant kras-driven pancreatic carcinogenesis. Cancer Genomics Proteomics. 17:695–705. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2022
Volume 61 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lu Q, Feng H, Chen H, Weygant N, Du J, Yan Z and Cao Z: Role of DCLK1 in oncogenic signaling (Review). Int J Oncol 61: 137, 2022
APA
Lu, Q., Feng, H., Chen, H., Weygant, N., Du, J., Yan, Z., & Cao, Z. (2022). Role of DCLK1 in oncogenic signaling (Review). International Journal of Oncology, 61, 137. https://doi.org/10.3892/ijo.2022.5427
MLA
Lu, Q., Feng, H., Chen, H., Weygant, N., Du, J., Yan, Z., Cao, Z."Role of DCLK1 in oncogenic signaling (Review)". International Journal of Oncology 61.5 (2022): 137.
Chicago
Lu, Q., Feng, H., Chen, H., Weygant, N., Du, J., Yan, Z., Cao, Z."Role of DCLK1 in oncogenic signaling (Review)". International Journal of Oncology 61, no. 5 (2022): 137. https://doi.org/10.3892/ijo.2022.5427