Open Access

Atonal bHLH transcription factor 1 is an important factor for maintaining the balance of cell proliferation and differentiation in tumorigenesis (Review)

  • Authors:
    • Ying Fu
    • Sha‑Sha Yuan
    • Li‑Jie Zhang
    • Zhi‑Li Ji
    • Xiao‑Jiang Quan
  • View Affiliations

  • Published online on: July 8, 2020     https://doi.org/10.3892/ol.2020.11833
  • Pages: 2595-2605
  • Copyright: © Fu 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

Establishing the link between cellular processes and oncogenesis may aid the elucidation of targeted and effective therapies against tumor cell proliferation and metastasis. Previous studies have investigated the mechanisms involved in maintaining the balance between cell proliferation, differentiation and migration. There is increased interest in determining the conditions that allow cancer stem cells to differentiate as well as the identification of molecules that may serve as novel drug targets. Furthermore, the study of various genes, including transcription factors, which serve a crucial role in cellular processes, may present a promising direction for future therapy. The present review described the role of the transcription factor atonal bHLH transcription factor 1 (ATOH1) in signaling pathways in tumorigenesis, particularly in cerebellar tumor medulloblastoma and colorectal cancer, where ATOH1 serves as an oncogene or tumor suppressor, respectively. Additionally, the present review summarized the associated therapeutic interventions for these two types of tumors and discussed novel clinical targets and approaches.

Introduction

A balance among cell proliferation, differentiation and migration is required for proper tissue development. Epidemiological studies investigating the association between potential risk factors and an increased risk of cancer have indicated that a number of factors, including diet, obesity, hormones, immunosuppression, cancer-causing substances, chronic inflammation, infectious agents and radiation, may increase the risk of imbalance (13). Furthermore, genetic and/or epigenetic changes or loss of function mutations in tumorigenesis-associated genes and signaling pathways may disrupt this balance, resulting in tumor progression and metastasis (4). Previous studies investigated gene mutations of the RAS, WNT, MYC, ERK and TRK genes, that may result in tumor initiation and progression, and may assist in understanding the underlying mechanisms involved (58). However, little is known about the mechanism and exact cause of >100 types of human cancer. Current knowledge has revealed that only 5–10% of cancer cases have a genetic component, which indicates that gene mutation is not the sole cause of cancer development (914). Therefore, investigating the mechanisms underlying the alteration of protein expression profiles may aid cancer study.

Atonal bHLH transcription factor 1 (ATOH1), an evolutionarily conserved human ortholog of the Drosophila proneural basic helix-loop-helix (bHLH) transcription factor atonal, is involved in a variety of developmental processes. ATOH1 was cloned and identified as a proneural transcription factor based on its sequence, structure and functional features (15). ATOH1 serves an important role in the specification and regulation of skin mechanosensory cells and in the development of the auditory system in the inner ear (16,17). Furthermore, ATOH1 is required to establish the intestinal epithelium secretory cell lineage and for the development of rhombic lip derivatives, including respiratory rhythmogenesis and the cerebellar external granule cell precursor layer (15,1820). ATOH1 positively regulates cell type specification and differentiation, controls cell cycle arrest and maintains granule neuron progenitors depending on the developmental context. Therefore, ATOH1 plays an important role in neural development and may serve as a tumor suppressor or an oncogene (2127).

Similar to other proneural genes, including achaete-scute complex like 1 and neurogenin 2, mutations that alter the function or result in loss of function of ATOH1 are generally lethal (28). Therefore, unlike the classic oncogenes or tumor suppressor genes, ATOH1 loss of function mutations are rarely found in tumor tissue and the majority of tumors tend to exhibit abnormal increased or decreased expression of ATOH1 (21,22,26,27,29,30). Previous studies assessing the expression profile of ATOH1 in various tumor tissues revealed an alteration of ATOH1 mRNA and protein levels in brain, colon, thyroid, prostate and lung cancer (21,22,26,27,29,30). Several studies demonstrated that such alterations positively or negatively regulate tumor initiation or progression via tissue-specific mechanisms.

It is essential to identify novel molecular biomarkers for the clinical diagnosis and molecular targeting of cancer for clinical treatment. Considering the complexity of the tumorigenic progress, drug resistance, the specificity of clinical treatments and side effects, further developments are required in the field of cancer therapy. ATOH1 regulates the expression of several target genes, including BarH like homeobox 1 and hes family bHLH transcription factor 6, and influences several important signaling pathways, such as the sonic hedgehog (SHH) and notch pathways (31,32). Therefore, further investigation into the effects of ATOH1 alteration on tumorigenesis is required. The present review investigated the role of ATOH1 in cancer, with a particular emphasis on medulloblastoma (MB) and gastrointestinal cancer. Furthermore, the present review aimed to develop a clearer understanding of how alterations in ATOH1 expression and activation affect tumor initiation, progression and metastasis. Additionally, potential drug treatments for cancer therapy are discussed.

General features of ATOH1

ATOH1, also referred to as Hath1 in humans, Math1 in mice and Cath1 in chickens, encodes a class II bHLH transcription factor. The functional bHLH domain consists of a basic DNA-binding region and protein-binding region with two α-helices linked by a variable loop region. The protein-binding region is required for the formation of a heterodimer with a class I member of the bHLH family protein E47/E12. ATOH1 shares ~70% homology with atonal in the bHLH domain. However, the rest of the sequence exhibits much less similarity and the positioning of the bHLH domain varies among species (33,34). In vertebrates, protein sequence comparisons have revealed >80% similarity in the serine-rich region of the C-terminal (35). Additionally, the N-terminus of the open reading frame exhibits a high similarity among mammals (35). Studies on atonal and its orthologs have revealed that the non-bHLH domain of the protein serves an important role; for example, the conserved serine residues are involved in post-translational modifications which affect protein function (15,36). Domain sweeping experiments have demonstrated that specific motifs and their combinations are important for proper protein function (36,37). Over the last few decades, research has focused on identifying the downstream targets of ATOH1/atonal. The majority of the target gene candidates identified are involved in transcriptional regulation, chromosomal organization and cell cycle control and are associated with Wnt, SHH, notch, transforming growth factor-β signaling and Janus kinase (JNK)/mitogen-activated protein kinase (MAPK) signaling pathways (26,3846).

ATOH1 in Merkel cell carcinoma and lung cancer

ATOH1 is expressed in Merkel cell (MCs) but not in lung tissue

In vertebrates, MCs are derived from neuroendocrine cells and are located in the basal layer of the epidermis (47). Clustered MCs consist of a touch sensitive zone, which is in nervated by slowly adapting type I mechanoreceptor nerves (48). These epidermis-derived cells are required for light touch responses (48). ATOH1 is expressed in MCs during development and in adults (49,50). ATOH1 may be required for MC progenitor differentiation, but its expression is also maintained throughout development and in mature MCs (51). ATOH1 null mice exhibited a loss of type I mechanoreceptor nerve response and lacked MCs (52). ATOH1 expression in MCs is regulated by the transcription factor SRY-box transcription factor 2 (SOX2). The expression of SOX2 in MCs is controlled by the polycomb repressor complex, which exhibits histone methyltransferase activity (53), suggesting the involvement of epigenetic regulation in cell lineage development. However, to the best of the authors' knowledge, the expression of ATOH1 in lung tissue during development has not been documented.

Abnormal ATOH1 expression in Merkel cell carcinoma (MCC)

MCC is a rare malignant skin cancer derived from epithelial and neuroendocrine cell differentiation that carries a very poor prognosis (54). Approximately 80% of MCC cases are polyomavirus-positive. However, the pathogenesis involved in polyomavirus-positive and negative MCC is yet to be fully elucidated (5557). Therefore, the association between polyomavirus infection and the development of MMC remains unclear.

Loss-of-function ATOH1 mutations or epigenetic silencing via promoter methylation have been detected in a small number of MCC cases (26). However, a recent study with a larger number of MCC cases did not determine a correlation between ATOH1 expression and MCC malignancy or an association between ATOH1 mutations and MCC development (58). Interestingly, the same study revealed a significant correlation between downregulated protein expression levels of ATOH1 and MCC recurrence or mortality (58). Further studies are required to determine the signaling pathways that interact with ATOH1 to control the differentiation of epidermal progenitors into MCs and to identify novel therapeutic agents for MCC.

Abnormal expression of ATOH1 in lung cancer

Previous studies investigating ATOH1 function during development and ATOH1 expression profiles in cancer have demonstrated the tissue- and context-specific functions of ATOH1 in physiological and pathological conditions (2123,26,5962). Several gene expression analyses have determined that ATOH1 is expressed in small cell lung carcinoma (SCLC), a neuroendocrine tumor (6366). Additionally, <20% adenocarcinoma samples express ATOH1 and these tumors exhibit neuroendocrine features (63). Cytoplasmic and nuclear expression of ATOH1 has been detected in certain lung squamous cell carcinoma (SCC) samples (30). However, ATOH1 is not known to be expressed in lung tissue and has not been reported to be involved in normal lung development (63). A correlation analysis revealed that the expression of ATOH1 was inversely correlated with lung cancer growth (25). Another study revealed that the ectopic activation of ATOH1 occurred in lung cancer, resulting in a poor patient prognosis (63). However, the underlying mechanisms remain unclear and ATOH1 expression in lung cancer cells is poorly understood. The pathways linking ATOH1 and lung cancer pathogenesis have not been fully elucidated. However, the association between ATOH1 expression and neuroendocrine tumors as well as the poor prognosis of these tumors may unravel the underlying mechanisms. Future studies investigating how ATOH1 expression alters protein expression profiles in lung cancer cells are warranted and may shed light on the mechanisms maintaining the balance among cell proliferation, differentiation and migration.

ATOH1 in medulloblastoma

Expression of ATOH1 in the central nervous system

The mouse ato orthologues Math1/ATOH1 mRNA is detected in the dorsal neural tube and cranial nerve ganglia at E9.5 (15). During embryonic brain development, ATOH1 is expressed in the dorsal hindbrain neuro epithelium, rhombic lip and the developing cerebellum (15,67). These ATOH1-dependent neurons are required for the generation of dorsal commissural interneurons (68) and brainstem respiratory nuclei, and the development of cerebellar granule cell lineages (18,69). Unlike MCs, in which ATOH1 is expressed in both progenitor and mature cells, ATOH1 expression persists during granule cell lineage development and in cerebellar granule cell precursors, but disappears during differentiation and migration from the external granule layer (EGL) to the internal granule layer (IGL; Fig. 1A) (18). ATOH1-null mice have a smaller cerebellum compared with wild type or heterozygous mice, and lack an EGL (18). The balance between the protein activity of ATOH1 and signaling pathway activity of SHH and Notch has been demonstrated to regulate granule cell differentiation (40). These data indicated that ATOH1 serves a crucial role in the development of cerebellar granule cells.

Abnormal ATOH1 expression in MB

Since ATOH1 is required for the regulation of cerebellar granule neuron precursors during cerebellar development, it is not surprising that ATOH1 exerts a crucial effect in the malignant cerebellar tumor subgroup SHH-MB (70), which is closely associated with the origin of granule cell progenitors (71,72). In SHH-MB, ATOH1, as well as GLI family zinc finger (GLI) 1/2, MYCN proto-oncogene, bHLH transcription factor (MYCN), cyclin D1/2 (CCND1/2) and protease nexin-1 (PN-1), are highly expressed when compared with normal tissue (73,74) and serve to control the proliferation of granule cell precursors (2123). However, the precise mechanism of ATOH1 upregulation in MB is yet to be fully elucidated. Previous studies revealed that ATOH1 was upregulated in cases of MB with loss-of-function mutations of hypermethylated in cancer 1 (HIC1) (a ZBTB transcriptional repressor) and super-activated SHH signaling (22,59,75). Furthermore, HIC1 is required for the transcriptional inhibition of ATOH1, while cerebellar granule neuron precursors (GNPs) undergo differentiation to granule neurons, migrating from the EGL to IGL (Fig. 1A) (76). Another study revealed that the phosphorylation of tyrosine 78 in ATOH1 was present only in human colorectal cancer (CRC) and not normal tissues. This phosphorylation served to both stabilize ATOH1 and increase its transcriptional activity, hence promoting MB (77).

No evidence has demonstrated the direct activation of ATOH1 by the SHH signaling pathway, although in the majority of cases, the hyper activation of the SHH signaling pathway and high levels of ATOH1 expression are observed in SHH-MB (23,59,78). Additionally, the upregulation of ATOH1 alone does not initiate MB (21), but an increased number of MB-initiating cells were observed when both ATOH1 and Gli1 were upregulated (21). Blocking ATOH1 in GNPs limited the response of pre-proliferation genes to SHH activity and accelerated differentiation (21). This indicates that the upregulation of ATOH1 and the hyper activation of SHH may synergistically interact with each in tumorigenesis. Furthermore, ATOH1 is required for the maintenance of progenitor cells in MB progression rather than MB initiation (21,59,79,80). Gene expression data analysis has revealed a strong correlation between ATOH1 gene expression and poor survival in patients with MB (81). These data suggested that ATOH1 is a tumor progression rather than a tumor initiation oncogene in the SHH subgroup of MB.

ATOH1 in the MB-associated genetic network

Hyperactive SHH induces the expression of certain downstream genes including GLI1/2, CCND1/2 and MYCN to drive the proliferation of GNPs (8284). In addition, a high expression of ATOH1 transcriptionally induces GLI2 expression (Fig. 1B). It has been demonstrated that ATOH1 regulates the SHH signaling pathway in GNPs (22). Furthermore, the overexpression of ATOH1 in GNPs under active SHH signaling conditions accelerates MB progression, however, proliferation was decreased in the absence of SHH (21). ATOH1 may enhance GNP proliferation by activating GLI2 and indirectly increasing the activity of the SHH signaling pathway via the inhibition of the transmembrane receptor patched 1 (PTCH1; Fig. 1B) (21,59,76).

It has been reported that <25% of MB cases are associated with constantly active mutations of the SHH pathway (85). Additionally, <20% of MB cases carry a loss-of-function mutation in PTCH1 (85). Since PTCH1 and the SHH pathway have been demonstrated to have an antagonistic relationship, hyperactive SHH mutants and loss-of-function mutations in PTCH1 strongly enhanced the progression of MB (Fig. 1B) (59). The markedly increased methylation of the HIC1 promoter results in the silencing of HIC1 expression in PTCH1 heterozygous mutant mice (21,76). Furthermore, the loss of PTCH1 may cause HIC1 silencing (Fig. 1B), which in turn deregulates its function to transcriptionally inhibit ATOH1 expression, potentially causing an increased expression of ATOH1. In addition, Ptch1 not only inhibits SHH to eliminate the enhancement function of SHH in proliferation, but also induces GNP differentiation (22). Hence, ATOH1 may repress GNP cell differentiation by downregulating PTCH1 (Fig. 1B). However, ATOH1 inhibits bone morphogenetic protein (BMP) 2/4 induced GNP cell differentiation by downregulating multiple BMP target genes. These genes, including distal-less homeobox 1/2 and hedgehog interacting protein, are required for GNP cell differentiation (Fig. 1B) (8688).

Previous studies have revealed that ATOH1 expression can be transcriptionally inhibited by HIC1 and post-transcriptionally downregulated by BMP2/4 (79). Through the rapid proteasome-mediated protein degradation of ATOH1, BMP2/4 is able to inhibit MB progression (Fig. 1B) (21,79). However, BMP2/4 may also inhibit ATOH1 by activating the ATOH1 inhibitors inhibitor of DNA binding 1 HLH protein (ID1) and inhibitor of DNA binding 2 (ID2) (79,89). ID1/2 are transcriptional repressors, which interact with ATOH1 to prevent ATOH1-DNA binding (79,90). BMPs, a subgroup of the transforming growth factor-β superfamily of proteins, act as MB suppressors not only via the downregulation of ATOH1 expression, but also via its regulatory effect on cell differentiation and proliferation. It has been revealed that BMPs antagonize SHH-dependent proliferation by activating Smad phosphorylation and downregulating SHH-induced genes that are required for the proliferation of GNPs in MB. In vitro and in vivo data have also revealed that BMPs promote the differentiation of GNPs in MB by increasing the expression of multiple genes for cell differentiation (79).

Several genome-wide studies on gene expression profile assessing the up and downregulation of ATOH1 in MB GNPs have revealed that the outcome candidates are primarily involved in two biological processes: Cell differentiation and proliferation. Among these ATOH1 related genes, nearly two thirds are involved in differentiation and less than one third are associated with cell proliferation (79,91). Others are associated with cell adhesion, cell migration, metabolism and chromosome modulation (21,9294). A comparative study on the categorical differences in upregulated and down regulated gene expression between the two MB groups induced by the overexpression of ATOH1 or GLI1 revealed that genes involved in neuronal differentiation, migration and adhesion, were significantly enriched in ATOH1 overexpressing MB. However, genes that regulated cell cycle progression were similarly expressed both MB groups (21). The results indicate that ATOH1 has more profound effects on controlling neuronal differentiation to promote the viability of GNPs and maintain the progenitor.

MB therapeutic intervention

As an aggressive embryonic cerebellum tumor, MB is the most common malignant pediatric brain tumor that exhibits a high mortality. The continued analysis of the mechanism that genetically and epigenetically regulates the relative gene expression of MB has permitted a deeper elucidation of therapeutic targets. The success of using Smoothened inhibitors, cyclopamine (a plant steroid alkaloid) and HhAntag (a benzimidazole derivative) as therapeutic drugs, has revealed their important regressional effect on controlling the SHH signaling pathway in MB (95,96). These agents not only decrease the proliferation of tumor cells, but also induce apoptosis of MB cells (97). However, the efficiency of these treatments are limited for MB with hyperactive SHH signaling and/or a high expression of ATOH1, since ATOH1 directly activates GLI2 expression at the transcriptional level and high GLI1/2 expressions may reduce drug efficiency. Therefore, targeting certain downstream proteins, including GLI1/2, may be more efficient (98,99). Recent reports using melanoma cell lines have demonstrated that GLI inhibitor GANT61 treatment was able to repress melanoma cells (100), indicating that additional GANT61 treatment may be effective in multiple anti-MB targeted therapy.

Although the downregulation of SHH signaling activity does not affect ATOH1 expression and a high ATOH1 expression in SHH subgroup MB represents only 25–35% of all MB cases (101), the expression of ATOH1 should still be taken into account when SHH signaling is used as a therapeutic target in MB. Despite transcription factors being difficult targets for small molecule drug development, for SHH subgroup MB, ATOH1 may still serve as a good potential therapeutic target, due to its regulatory function on GNPs in MB.

Epigenetic therapy, which reverses DNA promoter methylation in clinical treatment of myelodysplastic syndrome, has been reported (21,96). This method may be utilized to restore Hic1 function by demethylating the Hic1 promoter, thereby blocking ATOH1 expression. Another possible way to downregulate ATOH1 is via BMPs treatment. It has been demonstrated that BMPs reduce GNP proliferation and promote differentiation, as well as induce apoptosis in human MB cells (102). BMPs may therefore be used in therapeutic interventions. However, high levels of ATOH1 can override the neuronal differentiation induced by BMP by inhibiting the expression of a multiple BMP target genes (21). Therefore, similar to SHH targeting therapy, ATOH1expression levels should be considered when BMPs are used as treatment for MB.

As tumors are heterogeneous entities with different causes of disease progression, previous studies have combined two or more drugs to tackle multiple targets. For example, GNP-like MB is inhibited via effective treatment with BMPs and cyclopamine (79). Although there is no clear evidence indicating the direct effect on ATOH1 by blocking PN-1, loss of function studies on PN-1 in PTCH1+/− mice revealed the loss of ATOH1 expression and the reduction of MB formation (Fig. 1B) (74). Therefore, it may be worthwhile to combine the smoothened frizzled class receptor inhibitor with the PN-1 inhibitor to prevent relapse due to single drug resistance. For clinic treatment, theoretical and practical caution should be taken. Recent study that has performed multivariate Cox regression analysis has proposed a multi-gene model for MB risk prediction (81). They have also provided a quantitative analysis method for the identification of molecular markers and for the evaluation of these markers on influencing clinical behavior. More effort is required to optimize the outcome of therapeutic treatment.

ATOH1 in gastrointestinal cancer

Expression of ATOH1 in intestinal cells

The intestinal epithelium is a self-renewing tissue that is comprised of several cell lineages (Fig. 2A). Within the intestinal epithelium, the major cell types can be classified as absorptive (colonocytes/enterocytes) or secretory (goblet, Paneth and enteroendocrine), based on their distinct genetic programs. In the intestinal epithelium of mice, ATOH1/Math1 expression is maintained throughout embryonic and adult phases (25). The expression of ATOH1 in the intestinal epithelium is essential for the specification and regulation of proliferation of the secretory cell lineage (25). The negative regulation of intestinal epithelial cell proliferation by ATOH1 and the failure to develop all types of secretory cells in ATOH1/Math1 null mice further confirms the functions of ATOH1 in the intestinal epithelium (103,104). In addition, interaction between the ATOH1 and Notch signaling pathway regulates the lineage differentiation of different types of intestinal cells. More specifically, inhibition of ATOH1 via Hes, a Notch signaling pathway downstream gene, promoted the absorptive vs. the secretory fate (Fig. 2A) (105). Therefore, ATOH1 is required in epidermal progenitors, in their progeny to specific MCs (106) and in their commitment to neuroendocrine cells (107). These results indicate that ATOH1 is involved in the general neuroendocrine differentiation of epithelial cells.

Abnormal ATOH1 expression in CRC

It is well known that ATOH1 and Notch signaling inhibit one and another, and are involved in the development of the secretory and absorptive cell lineages. It has been demonstrated in several previous studies that intestinal ATOH1 regulates cell cycle arrest and represses proliferation, hence promoting differentiation or stimulating apoptosis (26,41). A significant decrease in ATOH1 mRNA levels has been detected in ~70% of CRC cases (24,26). In addition, the Notch signaling pathway has been determined to be associated with human colon adenocarcinomas (108). In addition to inhibiting ATOH1, Notch signaling functions to maintain stem cells in an uncommitted state (Fig. 2B) (109). Active Notch1/2 and its transcriptional target HES1 have been detected in human colon adenocarcinomas and CRC cell lines and were enriched in adenomas from adenomatous polyposis coli (APC) mutant mice (27,110113).

Previous studies have revealed that at least one copy deletion of ATOH1 is present in ~50% of tumors and ATOH1 CpG methylation has been detected in ~70% of tumors (Fig. 2B) (26). Other evidence obtained from the analysis of 48 patients with colon cancer has revealed that ATOH1 mRNA levels drop ~20-fold and goblet cell populations are markedly reduced in colon adenocarcinomas (114). These data indicate that genetic and epigenetic mechanisms may be involved in the silencing of ATOH1 expression in CRC, and that ATOH1 is required for goblet lineage development.

Not all cases of CRC exhibit a low expression of ATOH1 mRNA (24,115,116). Previous studies have reported the loss of ATOH1 protein but the presence of ATOH1 mRNA (115). In addition to the aberrant Notch signaling pathway, the proteasomal degradation of ATOH1 by glycogen synthase kinase 3 β (GSK3β) in CRC suggested a protein level regulation on ATOH1 in CRC (Fig. 2B) (117). Aberrant constitutively activated Wnt/GSK3 signaling via truncated mutations of the APC gene is observed in ~80% of CRC cases (118,119). This aberrant Wnt/GSK3β signaling induces the continuous expression of β-catenin, which promotes the proliferation of intestinal progenitors and the proteasomal degradation of the ATOH1 protein, indicating that it is the most critical trigger for colon carcinogenesis (Fig. 2B) (117). A recent structural and functional study of ATOH1 identified a highly conserved critical serine site in the second helix domain, which, when phosphorylated, results in the quick inactivation and degradation of the ATOH1 protein (120). This result further proves the existence of another layer of ATOH1 protein regulation. However, treatment with GSK inhibitors or mutating the serine residues of ATOH1to alanine on the aforementioned or multiple serine sites may stabilize the expression and activity of ATOH1 (117,120), resulting in the induction of colon cancer cell differentiation to goblet cells (117). Another previous study also demonstrated that blocking ATOH1 protein phosphorylation promoted secretory differentiation and inhibited its involvement in the self-renewal of progenitors (121). A recent study also provided evidence that microRNA-613 promoted CRC by down regulating ATOH1 (122). The results therefore indicate that ATOH1 is down regulated in CRC.

In vitro and in vivo experiments have revealed that the up regulation of ATOH1 induces progenitor cell differentiation to goblet cells, inhibits proliferation and promotes apoptosis via the JNK/MAPK pathway (26,41), as indicated by the tumor suppressive nature of ATOH1 in colon adenocarcinoma and gastrointestinal carcinoma.

ATOH1 in CRC-associated genetic networks

The majority of colorectal tumors originate from epithelial cells (123). The mucus layer produced by goblet cells is part of the innate immune system and is required for intestinal homeostasis (124,125). Mucus is composed of transmembrane and secretory proteins, such as mucin (Muc) 1 and 2, respectively, that construct a semi-permeable passage between the intestinal lumen and the underlying epithelium (126,127). Previous studies have revealed that Muc2 serves as a tumor suppressor in CRC (128,129). Although wild type ATOH1 was determined to be degraded in CRC, a stable ATOH1 form or an ATOH1 phosphorylation-null mutant form alone is sufficient to induce Muc2 expression to initiate the differentiation of colon cancer cells (114,117). Apart from the direct activation of Muc2, previous studies on ATOH1 null mutant mice revealed the absence of the ETS family transcription factor SPDEF (SAM pointed domain ETS factor) and the loss of intestinal secretory cells, indicating that ATOH1 is required for the activation of SPDEF (130132). Gain of function studies have indicated that SPDEF not only promotes the terminal differentiation of goblet/Paneth cells into goblet cells, but also inhibits the proliferation of intestinal progenitors (130132).

The cell cycle inhibitor p27 has been determined to be activated by ATOH1 and repressed by Notch signaling via Hes1 (Fig. 2B) (114,133). ATOH1 induces cell cycle exit by inhibiting the cell cycle marker gene CCND1 either directly or via p27, blocking the proliferation of intestinal progenitors (103). ATOH1 therefore serves as a tumor suppressor, which promotes differentiation and inhibits the proliferation of intestinal progenitors.

Active Notch signaling represses p27 and ATOH1 via Hes1 activation, thereby indirectly enhancing cell proliferation and maintaining cancer stem cells (130,133,134). Similarly, the aberrant activation of Wnt/GSK3 signaling promotes cell proliferation indirectly by activating the β-catenin protein (117), whilst indirectly inhibiting cell differentiation and enhancing cell proliferation by degrading ATOH1 (115).

CRC therapeutic intervention

It has been demonstrated that a high Notch activity and an accumulation of β-catenin protein as a result of aberrant Wnt/GSK3 signaling, together with the inactivation of ATOH1, induces carcinogenesis and maintains the undifferentiated state of colon cancer (135). Therefore, treatment with GSK3 inhibitors (GSK3I), such as APC, and Notch signaling inhibitors (such as Notch-targeting antibodies or γ-secretase) may represent a novel therapeutic approach (109).

GSK is considered to be a key enzyme in various biological processes. Therefore, the development of a drug targeting Wnt/GSK signaling is difficult due to its complex protein-protein interactions and various functions in different cell types (113). The risk of treatment and pathological safety should becarefully evaluated. One particular GSK3 inhibitor, lithium chloride, serves a protective effect against colon cancer and exerts no detectable pathological changes in other major organs while being used to treat bipolar disorder treatment (136). Previous studies assessing the molecular pathways and mechanisms involved in the cancer suppressive effect of GSK3I were performed in cell lines and rodent model systems (137139). In addition, inactivation of the Wnt/GSK signaling pathway via the overexpression of a full-length APC gene in colon cancer cells revealed the stabilization of ATOH1 and that the degradation of β-catenin (Fig. 2B) results in cell differentiation (115). Combination with other treatments may provide a sufficient effect when compared with single GSK3I therapies for CRC (140). For instance, the overexpression of ATOH1 or the stabilization of ATOH1 protein in combination with GSK3I treatment may serve as a potential cancer therapy (135). However, detailed analyses and evaluations are required to optimize drug dosages, multiple treatments and tissue specificity. Certain cases of CRC exhibit an undifferentiated proliferative phenotype caused by constitutively activated Notch signaling (111,112,141). Maintaining a negative regulative Notch activity is initiated via the release and entry of the active domain of Notch into the nucleus. The release of the Notch signaling receptor active domain and intracellular domain requires γ-secretase activity. Theoretically, antibody-mediated Notch inhibition and γ-secretase inhibitors (GSIs) would be good candidates to inhibit colonic cancer. However, antibody-mediated Notch inhibition exerts a weak effect on blocking CRC growth (142). A previous study on γ-secretase inhibitor treatment in colon cancer cell lines and primary human CRC cell cultures revealed that GSI treatment upregulates ATOH1 expression and increases Muc2 and p27, resulting in the reduction of anchorage independent cellular growth and increasing Muc2 positive cells in ATOH1 positive CRC, but no detectable effect in ATOH1 negative CRC (134). In addition, a significant proapoptotic effect on CRC cell lines has been observed in GSI treatment (111). These data indicate that ATOH1 is crucial to the tumorigenesis regulatory network of CRC. Undifferentiated CRC represents only a fraction of colonic cancers. There are also moderately and well-differentiated classes of CRC (143). Furthermore, the hyper activation of β-catenin signaling overrides the forced differentiation induced by GSI treatment (27). These data indicate that multiple treatments or the combination of other drugs is required to obtain improved outcomes in patients.

Currently, certain cytotoxic drugs, including taxanes or platinum compounds, have been studied in CRC cell lines (144,145). However, more detailed analyses are required for further clarification. Assessing the possibility and optimizing the proper conditions of GSK3I and GSI co-treatment in CRC should be considered. Another gene, SPDEF, has been revealed to regulate the terminal differentiation of intestinal goblet cells. In breast and prostate cancer, SPDEF acts as tumor suppressor by inhibiting invasion and metastasis (26,116) and/or tumor growth and survival (24). In CRC, SPDEF serves as a key mediator of ATOH1 for tumor suppressive activity (130). Future studies are therefore worthwhile to assess whether SPDEF upregulating treatment alone or in combination with GSK3I and/or GSI is sufficient to activate goblet cell differentiation and block proliferation in ATOH1-null CRC cells. However, since ATOH1 is a potential anti-tumor gene in CRC, whether it is sufficient to block tumor growth by elevating ATOH1 levels or by force expressing ATOH1, should be elucidated. A previous study assessing the role of adiponectin (APN) in the prevention of goblet cell apoptosis and the differentiation of epithelial cells to goblet cells revealed that APN may block goblet cell apoptosis by inhibiting TNF-α and promoting differentiation by upregulating ATOH1 and Muc2, and downregulating Hes1 (74) (Fig. 2B). Therefore, APN may serve as a potential clinical target candidate for CRC.

Conclusions

The effect of ATOH1on the tumorigenesis of different tissue organs and the possibility of targeted clinical therapy indicated that variations in the mechanisms of tumorigenesis existed in different type of tumors. ATOH1 serves as a tumor suppressor in MCC and CRC but is an oncogene in MC and SCLC/SCC. The expression profile of the ATOH1 protein also exhibits a differential change in different types of tumors. In MCC and CRC, ATOH1 expressions are downregulated. By contrast, ATOH1 expressions were upregulated in MC and SCLC/SCC. However, regardless of these specificities, ATOH1 displayed a common feature that ATOH1 influenced tumorigenesis by promoting the transcription of its target genes for cell proliferation and differentiation. In the case of MCC and CRC, these genes are required for enhancing differentiation and inhibiting proliferation. The opposite is observed in MC and SCLS/SCC. The abnormal expression of ATOH1 results in an unpaired balance between differentiation and proliferation, which promotes the progression of cancer.

Crucial proteins are more often deregulated in multiple ways at different levels, such as at the transcriptional, translational and post translational level and during mRNA and/or protein stability. The present discussion of ATOH1 in MB and CRC indicated that the temporal/spatial regulation of protein and protein functions with tissue/context specificity are observed not only during development but also in cancer progression. Clinical drug treatments should therefore address the specificity and regulatory aspects of their targets. The cross-interaction of proteins causes single drug therapeutic treatments to be ineffective. Optimal target selection or a combined treatment approach for cancer therapy should therefore consider this cross-effect. Multiple targeted treatments have revealed a more efficient effect during cancer therapy. An improved understanding of the mechanisms of tumorigenesis and cancer regulatory networks would improve clinical approaches. The core reason for the formation of cancer is the unpaired balance of biological systems to a certain extent. Instead of assessing genes or pathways as clinical targets, understanding how this balance is disturbed and subsequently elucidating an approach for adjusting this would be another direction of cancer therapy.

Acknowledgements

The authors would like to thank Professor Bassem A. Hassan (Laboratory of Brain Development, Institut du Cerveau et de la Moelle Épinière, Hôpital Pitié-Salpêtrière, Paris) for his comments on the manuscript.

Funding

The present review was supported by the National Nature Science Foundation (grant no. 81772943).

Availability of data and materials

Not applicable.

Authors' contributions

YF and SSY wrote the manuscript. LJZ wrote the manuscript and edited the figures. ZLJ and XJQ critically evaluated and revised 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.

References

1 

Kompella P and Vasquez KM: Obesity and cancer: A mechanistic overview of metabolic changes in obesity that impact genetic instability. Mol Carcinog. 58:1531–1550. 2019. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

Axelrad JE, Lichtiger S and Yajnik V: Inflammatory bowel disease and cancer: The role of inflammation, immunosuppression, and cancer treatment. World J Gastroenterol. 22:4794–4801. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Raskov H, Soby JH, Troelsen J, Bojesen RD and Gogenur I: Driver gene mutations and epigenetics in colorectal Cancer. Ann Surg. 271:75–85. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Todd R and Wong DT: Oncogenes. Anticancer Res. 19:4729–4746. 1999.PubMed/NCBI

6 

Bos JL: Ras oncogenes in human cancer: A review. Cancer Res. 49:4682–4689. 1989.PubMed/NCBI

7 

Felsher DW and Bishop JM: Reversible tumorigenesis by MYC in hematopoietic lineages. Mol Cell. 4:199–207. 1999. View Article : Google Scholar : PubMed/NCBI

8 

Cargnello M and Roux PP: Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev. 75:50–83. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Hisada M, Garber JE, Fung CY, Fraumeni JF and Li FP: Multiple primary cancers in families with Li-Fraumeni syndrome. J Natl Cancer Inst. 90:606–611. 1998. View Article : Google Scholar : PubMed/NCBI

10 

National Cancer Institute, . Genetic Testing for Hereditary Cancer Syndromes. June 21–, 2016

11 

National Cancer Institute, . Physician Data Query (PDQ). Cancer Genetics Overview. 2016. June 21–2016

12 

National Cancer Institute, . Physician Data Query (PDQ). Genetics of Breast and Ovarian Cancer. 2016. June 21–2016

13 

Chang-Claude J: Inherited genetic susceptibility to breast cancer. IARC Sci Publ. 154:177–190. 2001.PubMed/NCBI

14 

National Cancer Institute, . Physician Data Query (PDQ). Genetics of Colorectal Cancer. 2016. June 21–2016

15 

Akazawa C, Ishibashi M, Shimizu C, Nakanishi S and Kageyama R: A mammalian helix-loop-helix factor structurally related to the product of Drosophila proneural gene atonal is a positive transcriptional regulator expressed in the developing nervous system. J Biol Chem. 270:8730–8738. 1995. View Article : Google Scholar : PubMed/NCBI

16 

Cai T and Groves AK: The role of atonal factors in mechanosensory cell specification and function. Mol Neurobiol. 52:1315–1329. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Chonko KT, Jahan I, Stone J, Wright MC, Fujiyama T, Hoshino M, Fritzsch B and Maricich SM: Atoh1 directs hair cell differentiation and survival in the late embryonic mouse inner ear. Dev Biolo. 381:401–410. 2013. View Article : Google Scholar

18 

Ben-Arie N, Bellen HJ, Armstrong DL, McCall AE, Gordadze PR, Guo Q, Matzuk MM and Zoghbi HY: Math1 is essential for genesis of cerebellar granule neurons. Nature. 390:169–172. 1997. View Article : Google Scholar : PubMed/NCBI

19 

Machold R and Fishell G: Math1 is expressed in temporally discrete pools of cerebellar rhombic-lip neural progenitors. Neuron. 48:17–24. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Rose MF, Ren J, Ahmad KA, Chao HT, Klisch TJ, Flora A, Greer JJ and Zoghbi HY: Math1 is essential for the development of hindbrain neurons critical for perinatal breathing. Neuron. 64:341–354. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Ayrault O, Zhao H, Zindy F, Qu C, Sherr CJ and Roussel MF: Atoh1 inhibits neuronal differentiation and collaborates with Gli1 to generate medulloblastoma-initiating cells. Cancer Res. 70:5618–5627. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Flora A, Klisch TJ, Schuster G and Zoghbi HY: Deletion of Atoh1 disrupts Sonic Hedgehog signaling in the developing cerebellum and prevents medulloblastoma. Science. 326:1424–1427. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Yang ZJ, Ellis T, Markant SL, Read TA, Kessler JD, Bourboulas M, Schüller U, Machold R, Fishell G, Rowitch DH, et al: Medulloblastoma can be initiated by deletion of Patched in lineage-restricted progenitors or stem cells. Cancer Cell. 14:135–145. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Leow CC, Romero MS, Ross S, Polakis P and Gao WQ: Hath1, down-regulated in colon adenocarcinomas, inhibits proliferation and tumorigenesis of colon cancer cells. Cancer Res. 64:6050–6057. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Yang Q, Bermingham NA, Finegold MJ and Zoghbi HY: Requirement of Math1 for secretory cell lineage commitment in the mouse intestine. Science. 294:2155–2158. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Bossuyt W, Kazanjian A, De Geest N, Van Kelst S, De Hertogh G, Geboes K, Boivin GP, Luciani J, Fuks F, Chuah M, et al: Atonal homolog 1 is a tumor suppressor gene. PLoS Biol. 7:e392009. View Article : Google Scholar : PubMed/NCBI

27 

Peignon G, Durand A, Cacheux W, Ayrault O, Terris B, Laurent-Puig P, Shroyer NF, Van Seuningen I, Honjo T, Perret C, et al: Complex interplay between beta-catenin signalling and Notch effectors in intestinal tumorigenesis. Gut. 60:166–176. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Huang C, Chan JA and Schuurmans C: Proneural bHLH genes in development and disease. Curr Top Dev Biol. 110:75–127. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Leonard JH, Cook AL, Van Gele M, Boyle GM, Inglis KJ, Speleman F and Sturm RA: Proneural and proneuroendocrine transcription factor expression in cutaneous mechanoreceptor (Merkel) cells and Merkel cell carcinoma. Int J Cancer. 101:103–110. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Xu HT, Xie XM, Li QC, Liu SL, Dai SD, Liu Y and Wang EH: Atonal homolog 1 expression in lung cancer correlates with inhibitors of the Wnt pathway as well as the differentiation and primary tumor stage. APMIS. 121:111–119. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Hou K, Jiang H, Karim MR, Zhong C, Xu Z, Liu L, Guan M, Shao J and Huang X: A Critical E-box in Barhl1 3′ enhancer is essential for auditory hair cell differentiation. Cells. 8(pii): E4582019. View Article : Google Scholar : PubMed/NCBI

32 

Scheffer D, Sage C, Corey DP and Pingault V: Gene expression profiling identifies Hes6 as a transcriptional target of ATOH1 in cochlear hair cells. FEBS Lett. 581:4651–4666. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Jarman AP, Grau Y, Jan LY and Jan YN: Atonal is a proneural gene that directs chordotonal organ formation in the Drosophila peripheral nervous system. Cell. 73:1307–1321. 1993. View Article : Google Scholar : PubMed/NCBI

34 

Jarman AP, Grell EH, Ackerman L, Jan LY and Jan YN: Atonal is the proneural gene for Drosophila photoreceptors. Nature. 369:398–400. 1994. View Article : Google Scholar : PubMed/NCBI

35 

Mulvaney J and Dabdoub A: Atoh1, an essential transcription factor in neurogenesis and intestinal and inner ear development: Function, regulation, and context dependency. J Assoc Res Otolaryngol. 13:281–293. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Weinberger S, Topping MP, Yan J, Claeys A, Geest N, Ozbay D, Hassan T, He X, Albert JT, Hassan BA and Ramaekers A: Evolutionary changes in transcription factor coding sequence quantitatively alter sensory organ development and function. Elife. 6(pii): e264022017. View Article : Google Scholar : PubMed/NCBI

37 

Quan XJ, Denayer T, Yan J, Jafar-Nejad H, Philippi A, Lichtarge O, Vleminckx K and Hassan BA: Evolution of neural precursor selection: Functional divergence of proneural proteins. Development. 131:1679–1689. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Aerts S, Quan XJ, Claeys A, Naval Sanchez M, Tate P, Yan J and Hassan BA: Robust target gene discovery through transcriptome perturbations and genome-wide enhancer predictions in Drosophila uncovers a regulatory basis for sensory specification. PLoS Biol. 8:e10004352010. View Article : Google Scholar : PubMed/NCBI

39 

Klisch TJ, Xi Y, Flora A, Wang L, Li W and Zoghbi HY: In vivo Atoh1 targetome reveals how a proneural transcription factor regulates cerebellar development. Proc Natl Acad Sci USA. 108:3288–3293. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Gazit R, Krizhanovsky V and Ben-Arie N: Math1 controls cerebellar granule cell differentiation by regulating multiple components of the Notch signaling pathway. Development. 131:903–913. 2004. View Article : Google Scholar : PubMed/NCBI

41 

VanDussen KL and Samuelson LC: Mouse atonal homolog 1 directs intestinal progenitors to secretory cell rather than absorptive cell fate. Dev Biol. 346:215–223. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Peignon G, Durand A, Cacheux W, Ayrault O, Terris B, Laurent-Puig P, Shroyer NF, Van Seuningen I, Honjo T, Perret C and Romagnolo B: Complex interplay between β-catenin signalling and Notch effectors in intestinal tumorigenesis. Gut. 60:166–176. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Flora A, Garcia JJ, Thaller C and Zoghbi HY: The E-protein Tcf4 interacts with Math1 to regulate differentiation of a specific subset of neuronal progenitors. Proc Natl Acad Sci USA. 104:15382–15387. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Lee KJ, Dietrich P and Jessell TM: Genetic ablation reveals that the roof plate is essential for dorsal interneuron specification. Nature. 403:734–740. 2000. View Article : Google Scholar : PubMed/NCBI

45 

Zhao H, Ayrault O, Zindy F, Kim JH and Roussel MF: Post-transcriptional down-regulation of Atoh1/Math1 by bone morphogenic proteins suppresses medulloblastoma development. Genes Dev. 22:722–727. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Hu X, Huang J, Feng L, Fukudome S, Hamajima Y and Lin J: Sonic hedgehog (SHH) promotes the differentiation of mouse cochlear neural progenitors via the Math1-Brn3.1 signaling pathway in vitro. J Neurosci Res. 88:927–935. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Moll I, Roessler M, Brandner JM, Eispert AC, Houdek P and Moll R: Human Merkel cells-aspects of cell biology, distribution and functions. Eur J Cell Biol. 84:259–271. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Wellnitz SA, Lesniak DR, Gerling GJ and Lumpkin EA: The regularity of sustained firing reveals two populations of slowly adapting touch receptors in mouse hairy skin. J Neurophysiol. 103:3378–3388. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Ben-Arie N, Hassan BA, Bermingham NA, Malicki DM, Armstrong D, Matzuk M, Bellen HJ and Zoghbi HY: Functional conservation of atonal and Math1 in the CNS and PNS. Development. 127:1039–1048. 2000.PubMed/NCBI

50 

Haeberle H, Fujiwara M, Chuang J, Medina MM, Panditrao MV, Bechstedt S, Howard J and Lumpkin EA: Molecular profiling reveals synaptic release machinery in Merkel cells. Proc Natl Acad Sci USA. 101:14503–14508. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Wright MC, Reed-Geaghan EG, Bolock AM, Fujiyama T, Hoshino M and Maricich SM: Unipotent, ATOH1+ progenitors maintain the Merkel cell population in embryonic and adult mice. J Cell Biol. 208:367–379. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Maricich SM, Wellnitz SA, Nelson AM, Lesniak DR, Gerling GJ, Lumpkin EA and Zoghbi HY: Merkel cells are essential for light-touch responses. Science. 324:1580–1582. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Bardot ES, Valdes VJ, Zhang J, Perdigoto CN, Nicolis S, Hearn SA, Silva JM and Ezhkova E: Polycomb subunits Ezh1 and Ezh2 regulate the Merkel cell differentiation program in skin stem cells. EMBO J. 32:1990–2000. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Coggshall K, Tello TL, North JP and Yu SS: Merkel cell carcinoma: An update and review: Pathogenesis, diagnosis, and staging. J Am Acad Dermatol. 78:433–442. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Tello TL, Coggshall K, Yom SS and Yu SS: Merkel cell carcinoma: An update and review: Current and future therapy. J Am Acad Dermatol. 78:445–454. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Liu W, MacDonald M and You J: Merkel cell polyomavirus infection and Merkel cell carcinoma. Curr Opin Virol. 20:20–27. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Schadendorf D, Lebbé C, Zur Hausen A, Avril MF, Hariharan S, Bharmal M and Becker JC: Merkel cell carcinoma: Epidemiology, prognosis, therapy and unmet medical needs. Eur J Cancer. 71:53–69. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Gambichler T, Mohtezebsade S, Wieland U, Silling S, Hoh AK, Dreissigacker M, Schaller J, Schulze HJ, Oellig F, Kreuter A, et al: Prognostic relevance of high atonal homolog-1 expression in Merkel cell carcinoma. J Cancer Res Clin Oncol. 143:43–49. 2017. View Article : Google Scholar : PubMed/NCBI

59 

Briggs KJ, Corcoran-Schwartz IM, Zhang W, Harcke T, Devereux WL, Baylin SB, Eberhart CG and Watkins DN: Cooperation between the Hic1 and Ptch1 tumor suppressors in medulloblastoma. Genes Dev. 22:770–785. 2008. View Article : Google Scholar : PubMed/NCBI

60 

Schüller U, Heine VM, Mao J, Kho AT, Dillon AK, Han YG, Huillard E, Sun T, Ligon AH, Qian Y, et al: Acquisition of granule neuron precursor identity is a critical determinant of progenitor cell competence to form Shh-induced medulloblastoma. Cancer Cell. 14:123–134. 2008. View Article : Google Scholar : PubMed/NCBI

61 

Shroyer NF, Helmrath MA, Wang VY, Antalffy B, Henning SJ and Zoghbi HY: Intestine-specific ablation of mouse atonal homolog 1 (Math1) reveals a role in cellular homeostasis. Gastroenterology. 132:2478–2488. 2007. View Article : Google Scholar : PubMed/NCBI

62 

Peignon G, Durand A, Cacheux W, Ayrault O, Terris B, Laurent-Puig P, Shroyer NF, Van Seuningen I, Honjo T, Perret C, et al: Complex interplay between β-catenin signalling and Notch effectors in intestinal tumorigenesis. Gut. 60:166–176. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Westerman BA, Breuer RH, Poutsma A, Chhatta A, Noorduyn LA, Koolen MG, Postmus PE, Blankenstein MA and Oudejans CB: Basic helix-loop-helix transcription factor profiling of lung tumors shows aberrant expression of the proneural gene atonal homolog 1 (ATOH1, HATH1, MATH1) in neuroendocrine tumors. Int J Biol Markers. 22:114–123. 2007. View Article : Google Scholar : PubMed/NCBI

64 

Bhattacharjee A, Richards WG, Staunton J, Li C, Monti S, Vasa P, Ladd C, Beheshti J, Bueno R, Gillette M, et al: Classification of human lung carcinomas by mRNA expression profiling reveals distinct adenocarcinoma subclasses. Proc Natl Acad Sci USA. 98:13790–13795. 2001. View Article : Google Scholar : PubMed/NCBI

65 

Hiroshima K, Iyoda A, Shibuya K, Toyozaki T, Haga Y, Fujisawa T and Ohwada H: Prognostic significance of neuroendocrine differentiation in adenocarcinoma of the lung. Ann Thorac Surg. 73:1732–1735. 2002. View Article : Google Scholar : PubMed/NCBI

66 

Berendsen HH, de Leij L, Poppema S, Postmus PE, Boes A, Sluiter HJ and The H: Clinical characterization of non-small-cell lung cancer tumors showing neuroendocrine differentiation features. J Clin Oncol. 7:1614–1620. 1989. View Article : Google Scholar : PubMed/NCBI

67 

Ben-Arie N, McCall AE, Berkman S, Eichele G, Bellen HJ and Zoghbi HY: Evolutionary conservation of sequence and expression of the bHLH protein Atonal suggests a conserved role in neurogenesis. Hum Mol Genet. 5:1207–1216. 1996. View Article : Google Scholar : PubMed/NCBI

68 

Bermingham NA, Hassan BA, Wang VY, Fernandez M, Banfi S, Bellen HJ, Fritzsch B and Zoghbi HY: Proprioceptor pathway development is dependent on Math1. Neuron. 30:411–422. 2001. View Article : Google Scholar : PubMed/NCBI

69 

Iulianella A, Wingate RJ, Moens CB and Capaldo E: The generation of granule cells during the development and evolution of the cerebellum. Dev Dyn. 248:506–513. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Thompson MC, Fuller C, Hogg TL, Dalton J, Finkelstein D, Lau CC, Chintagumpala M, Adesina A, Ashley DM, Kellie SJ, et al: Genomics identifies medulloblastoma subgroups that are enriched for specific genetic alterations. J Clin Oncol. 24:1924–1931. 2006. View Article : Google Scholar : PubMed/NCBI

71 

Gibson P, Tong Y, Robinson G, Thompson MC, Currle DS, Eden C, Kranenburg TA, Hogg T, Poppleton H, Martin J, et al: Subtypes of medulloblastoma have distinct developmental origins. Nature. 468:1095–1099. 2010. View Article : Google Scholar : PubMed/NCBI

72 

Gilbertson RJ and Ellison DW: The origins of medulloblastoma subtypes. Ann Rev Pathol. 3:341–365. 2008. View Article : Google Scholar

73 

Lee Y, Miller HL, Jensen P, Hernan R, Connelly M, Wetmore C, Zindy F, Roussel MF, Curran T, Gilbertson RJ and McKinnon PJ: A molecular fingerprint for medulloblastoma. Cancer Res. 63:5428–5437. 2003.PubMed/NCBI

74 

Vaillant C, Valdivieso P, Nuciforo S, Kool M, Schwarzentruber-Schauerte A, Mereau H, Cabuy E, Lobrinus JA, Pfister S, Zuniga A, et al: Serpine2/PN-1 is required for proliferative expansion of pre-neoplastic lesions and malignant progression to medulloblastoma. PLoS One. 10:e01248702015. View Article : Google Scholar : PubMed/NCBI

75 

Blaess S, Corrales JD and Joyner AL: Sonic hedgehog regulates Gli activator and repressor functions with spatial and temporal precision in the mid/hindbrain region Development 133. 1799–1809. 2006.PubMed/NCBI

76 

Briggs KJ, Eberhart CG and Watkins DN: Just say no to ATOH: How HIC1 methylation might predispose medulloblastoma to lineage addiction. Cancer Res. 68:8654–8666. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Klisch TJ, Vainshtein A, Patel AJ and Zoghbi HY: Jak2-mediated phosphorylation of ATOH1 is critical for medulloblastoma growth. Elife. 6(pii): e311812017. View Article : Google Scholar : PubMed/NCBI

78 

Salsano E, Pollo B, Eoli M, Giordana MT and Finocchiaro G: Expression of MATH1, a marker of cerebellar granule cell progenitors, identifies different medulloblastoma sub-types. Neurosci Lett. 370:180–185. 2004. View Article : Google Scholar : PubMed/NCBI

79 

Zhao H, Ayrault O, Zindy F, Kim JH and Roussel MF: Post-transcriptional down-regulation of ATOH1/Math1 by bone morphogenic proteins suppresses medulloblastoma development. Genes Dev. 22:722–727. 2008. View Article : Google Scholar : PubMed/NCBI

80 

Garraway LA, Weir BA, Zhao X, Widlund H, Beroukhim R, Berger A, Rimm D, Rubin MA, Fisher DE, Meyerson ML and Sellers WR: ‘Lineage addiction’ in human cancer: Lessons from integrated genomics. Cold Spring Harb Symp Quant Biol. 70:25–34. 2005. View Article : Google Scholar : PubMed/NCBI

81 

Zakrzewska M, Gresner SM, Zakrzewski K, Zalewska-Szewczyk B and Liberski PP: Novel gene expression model for outcome prediction in paediatric medulloblastoma. J Mol Neurosci. 51:371–379. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Lum L and Beachy PA: The Hedgehog response network: Sensors, switches, and routers. Science. 304:1755–1759. 2004. View Article : Google Scholar : PubMed/NCBI

83 

Katoh Y and Katoh M: Integrative genomic analyses on GLI1: Positive regulation of GLI1 by Hedgehog-GLI, TGFbeta-Smads, and RTK-PI3K-AKT signals, and negative regulation of GLI1 by Notch-CSL-HES/HEY, and GPCR-Gs-PKA signals. Int J Oncol. 35:187–192. 2009. View Article : Google Scholar : PubMed/NCBI

84 

Browd SR, Kenney AM, Gottfried ON, Yoon JW, Walterhouse D, Pedone CA and Fults DW: N-myc can substitute for insulin-like growth factor signaling in a mouse model of sonic hedgehog-induced medulloblastoma. Cancer Res. 66:2666–2672. 2006. View Article : Google Scholar : PubMed/NCBI

85 

Eberhart CG: Medulloblastoma in mice lacking p53 and PARP: All roads lead to Gli. Am J Pathol. 162:7–10. 2003. View Article : Google Scholar : PubMed/NCBI

86 

Chiba S, Takeshita K, Imai Y, Kumano K, Kurokawa M, Masuda S, Shimizu K, Nakamura S, Ruddle FH and Hirai H: Homeoprotein DLX-1 interacts with Smad4 and blocks a signaling pathway from activin A in hematopoietic cells. Proc Natl Acad Sci USA. 100:15577–15582. 2003. View Article : Google Scholar : PubMed/NCBI

87 

Harris SE, Guo D, Harris MA, Krishnaswamy A and Lichtler A: Transcriptional regulation of BMP-2 activated genes in osteoblasts using gene expression microarray analysis: Role of Dlx2 and Dlx5 transcription factors. Front Biosci. 8:s1249–s1265. 2003. View Article : Google Scholar : PubMed/NCBI

88 

Katoh Y and Katoh M: Comparative genomics on HHIP family orthologs. Int J Mol Med. 17:391–395. 2006.PubMed/NCBI

89 

Kamaid A, Neves J and Giraldez F: Id gene regulation and function in the prosensory domains of the chicken inner ear: A link between Bmp signaling and ATOH1. J Neurosci. 30:11426–11434. 2010. View Article : Google Scholar : PubMed/NCBI

90 

Angley C, Kumar M, Dinsio KJ, Hall AK and Siegel RE: Signaling by bone morphogenetic proteins and Smad1 modulates the postnatal differentiation of cerebellar cells. J Neurosci. 23:260–268. 2003. View Article : Google Scholar : PubMed/NCBI

91 

Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC and Lempicki RA: DAVID: Database for annotation, visualization, and integrated discovery. Genome Biol. 4:P32003. View Article : Google Scholar : PubMed/NCBI

92 

Killeen MT and Sybingco SS: Netrin, Slit and Wnt receptors allow axons to choose the axis of migration. Dev Biol. 323:143–151. 2008. View Article : Google Scholar : PubMed/NCBI

93 

Zou YR, Kottmann AH, Kuroda M, Taniuchi I and Littman DR: Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature. 393:595–599. 1998. View Article : Google Scholar : PubMed/NCBI

94 

Adamson DC, Shi Q, Wortham M, Northcott PA, Di C, Duncan CG, Li J, McLendon RE, Bigner DD, Taylor MD and Yan H: OTX2 is critical for the maintenance and progression of Shh-independent medulloblastomas. Cancer Res. 70:181–191. 2010. View Article : Google Scholar : PubMed/NCBI

95 

Berman DM, Karhadkar SS, Hallahan AR, Pritchard JI, Eberhart CG, Watkins DN, Chen JK, Cooper MK, Taipale J, Olson JM and Beachy PA: Medulloblastoma growth inhibition by hedgehog pathway blockade. Science. 297:1559–1561. 2002. View Article : Google Scholar : PubMed/NCBI

96 

Romer JT, Kimura H, Magdaleno S, Sasai K, Fuller C, Baines H, Connelly M, Stewart CF, Gould S, Rubin LL and Curran T: Suppression of the Shh pathway using a small molecule inhibitor eliminates medulloblastoma in Ptc1(+/-)p53(−/-) mice. Cancer Cell. 6:229–240. 2004. View Article : Google Scholar : PubMed/NCBI

97 

Hallahan AR, Pritchard JI, Hansen S, Benson M, Stoeck J, Hatton BA, Russell TL, Ellenbogen RG, Bernstein ID, Beachy PA and Olson JM: The SmoA1 mouse model reveals that notch signaling is critical for the growth and survival of sonic hedgehog-induced medulloblastomas. Cancer Res. 64:7794–7800. 2004. View Article : Google Scholar : PubMed/NCBI

98 

Northcott PA, Nakahara Y, Wu X, Feuk L, Ellison DW, Croul S, Mack S, Kongkham PN, Peacock J, Dubuc A, et al: Multiple recurrent genetic events converge on control of histone lysine methylation in medulloblastoma. Nat Genet. 41:465–472. 2009. View Article : Google Scholar : PubMed/NCBI

99 

Stanton BZ and Peng LF: Small-molecule modulators of the Sonic Hedgehog signaling pathway. Mol Biosyst. 6:44–54. 2010. View Article : Google Scholar : PubMed/NCBI

100 

Vlckova K, Reda J, Ondrusova L, Krayem M, Ghanem G and Vachtenheim J: GLI inhibitor GANT61 kills melanoma cells and acts in synergy with obatoclax. Int J Oncol. 49:953–960. 2016. View Article : Google Scholar : PubMed/NCBI

101 

Northcott PA, Rutka JT and Taylor MD: Genomics of medulloblastoma: From Giemsa-banding to next-generation sequencing in 20 years. Neurosurg Focus. 28:E62010. View Article : Google Scholar : PubMed/NCBI

102 

Hallahan AR, Pritchard JI, Chandraratna RA, Ellenbogen RG, Geyer JR, Overland RP, Strand AD, Tapscott SJ and Olson JM: BMP-2 mediates retinoid-induced apoptosis in medulloblastoma cells through a paracrine effect. Nat Med. 9:1033–1038. 2003. View Article : Google Scholar : PubMed/NCBI

103 

Kazanjian A, Noah T, Brown D, Burkart J and Shroyer NF: Atonal homolog 1 is required for growth and differentiation effects of notch/gamma-secretase inhibitors on normal and cancerous intestinal epithelial cells. Gastroenterology. 139:918–928. 2010. View Article : Google Scholar : PubMed/NCBI

104 

Kim TH and Shivdasani RA: Genetic evidence that intestinal Notch functions vary regionally and operate through a common mechanism of Math1 repression. J Biol Chem. 286:11427–11433. 2011. View Article : Google Scholar : PubMed/NCBI

105 

Noah TK and Shroyer NF: Notch in the intestine: Regulation of homeostasis and pathogenesis. Annu Rev Physiol. 75:263–288. 2013. View Article : Google Scholar : PubMed/NCBI

106 

Van Keymeulen A, Mascre G, Youseff KK, Harel I, Michaux C, De Geest N, Szpalski C, Achouri Y, Bloch W, Hassan BA and Blanpain C: Epidermal progenitors give rise to Merkel cells during embryonic development and adult homeostasis. J Cell Biol. 187:91–100. 2009. View Article : Google Scholar : PubMed/NCBI

107 

Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A, Korving J, Begthel H, Peters PJ and Clevers H: Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 449:1003–1307. 2007. View Article : Google Scholar : PubMed/NCBI

108 

Reedijk M, Odorcic S, Zhang H, Chetty R, Tennert C, Dickson BC, Lockwood G, Gallinger S and Egan SE: Activation of Notch signaling in human colon adenocarcinoma. Int J Oncol. 33:1223–1229. 2008.PubMed/NCBI

109 

van Es JH, de Geest N, van de Born M, Clevers H and Hassan BA: Intestinal stem cells lacking the Math1 tumour suppressor are refractory to Notch inhibitors. Nat Commun. 1:182010. View Article : Google Scholar : PubMed/NCBI

110 

Guilmeau S, Flandez M, Mariadason JM and Augenlicht LH: Heterogeneity of Jagged1 expression in human and mouse intestinal tumors: Implications for targeting Notch signaling. Oncogene. 29:992–1002. 2010. View Article : Google Scholar : PubMed/NCBI

111 

Sikandar SS, Pate KT, Anderson S, Dizon D, Edwards RA, Waterman ML and Lipkin SM: NOTCH signaling is required for formation and self-renewal of tumor-initiating cells and for repression of secretory cell differentiation in colon cancer. Cancer Res. 70:1469–1478. 2010. View Article : Google Scholar : PubMed/NCBI

112 

Fre S, Pallavi SK, Huyghe M, Lae M, Janssen KP, Robine S, Artavanis-Tsakonas S and Louvard D: Notch and Wnt signals cooperatively control cell proliferation and tumorigenesis in the intestine. Proc Natl Acad Sci USA. 106:6309–6314. 2009. View Article : Google Scholar : PubMed/NCBI

113 

van Es JH, van Gijn ME, Riccio O, van den Born M, Vooijs M, Begthel H, Cozijnsen M, Robine S, Winton DJ, Radtke F and Clevers H: Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature. 435:959–963. 2005. View Article : Google Scholar : PubMed/NCBI

114 

Zhu DH, Niu BL, Du HM, Ren K, Sun JM and Gong JP: Hath1 inhibits proliferation of colon cancer cells probably through up-regulating expression of Muc2 and p27 and down-regulating expression of cyclin D1. Asian Pac J Cancer Prev. 13:6349–6355. 2012. View Article : Google Scholar : PubMed/NCBI

115 

Tsuchiya K, Nakamura T, Okamoto R, Kanai T and Watanabe M: Reciprocal targeting of Hath1 and beta-catenin by Wnt glycogen synthase kinase 3beta in human colon cancer. Gastroenterology. 132:208–220. 2007. View Article : Google Scholar : PubMed/NCBI

116 

Park ET, Oh HK, Gum JR Jr, Crawley SC, Kakar S, Engel J, Leow CC, Gao WQ and Kim YS: HATH1 expression in mucinous cancers of the colorectum and related lesions. Clin Cancer Res. 12:5403–5410. 2006. View Article : Google Scholar : PubMed/NCBI

117 

Aragaki M, Tsuchiya K, Okamoto R, Yoshioka S, Nakamura T, Sakamoto N, Kanai T and Watanabe M: Proteasomal degradation of ATOH1 by aberrant Wnt signaling maintains the undifferentiated state of colon cancer. Biochem Biophys Res Commun. 368:923–929. 2008. View Article : Google Scholar : PubMed/NCBI

118 

Polakis P: Wnt signaling and cancer. Genes Dev. 14:1837–1851. 2000.PubMed/NCBI

119 

Polakis P: The oncogenic activation of beta-catenin. Curr Opin Genet Dev. 9:15–21. 1999. View Article : Google Scholar : PubMed/NCBI

120 

Quan XJ, Yuan L, Tiberi L, Claeys A, De Geest N, Yan J, van der Kant R, Xie WR, Klisch TJ, Shymkowitz J, et al: Post-translational control of the temporal dynamics of transcription factor activity regulates neurogenesis. Cell. 164:460–475. 2016. View Article : Google Scholar : PubMed/NCBI

121 

Tomic G, Morrissey E, Kozar S, Ben-Moshe S, Hoyle A, Azzarelli R, Kemp R, Chilamakuri CSR, Itzkovitz S, Philpott A and Winton DJ: Phospho-regulation of ATOH1 is required for plasticity of secretory progenitors and tissue regeneration. Cell Stem Cell. 23:436–43.e7. 2018. View Article : Google Scholar : PubMed/NCBI

122 

Yang X, Zhang L, Song X, He W, Zhang D, Lu Q, Wu J, Wu C and Jiang J: MicroRNA-613 promotes colon cancer cell proliferation, invasion and migration by targeting ATOH1. Biochem Biophys Res Commun. 504:827–833. 2018. View Article : Google Scholar : PubMed/NCBI

123 

Ponz de Leon M and Di Gregorio C: Pathology of colorectal cancer. Digestive and liver disease: Official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver. 33:372–388. 2001. View Article : Google Scholar : PubMed/NCBI

124 

Hooper LV and Macpherson AJ: Immune adaptations that maintain homeostasis with the intestinal microbiota. Nat Rev Immunol. 10:159–169. 2010. View Article : Google Scholar : PubMed/NCBI

125 

Hollingsworth MA and Swanson BJ: Mucins in cancer: Protection and control of the cell surface. Nat Rev Cancer. 4:45–60. 2004. View Article : Google Scholar : PubMed/NCBI

126 

Sheng XZ, Xu GJ, Tang XQ and Zhan WB: Monoclonal antibodies recognizing mucus immunoglobulin and surface immunoglobulin-positive cells of flounder (Paralichthys olivaceus). Vet Immunol Immunopathol. 145:143–150. 2012. View Article : Google Scholar : PubMed/NCBI

127 

Corfield AP, Carroll D, Myerscough N and Probert CS: Mucins in the gastrointestinal tract in health and disease. Front Biosci. 6:D1321–D1357. 2001. View Article : Google Scholar : PubMed/NCBI

128 

Velcich A, Yang W, Heyer J, Fragale A, Nicholas C, Viani S, Kucherlapati R, Lipkin M, Yang K and Augenlicht L: Colorectal cancer in mice genetically deficient in the mucin Muc2. Science. 295:1726–1729. 2002. View Article : Google Scholar : PubMed/NCBI

129 

Byrd JC and Bresalier RS: Mucins and mucin binding proteins in colorectal cancer. Cancer Metastasis Rev. 23:77–99. 2004. View Article : Google Scholar : PubMed/NCBI

130 

Noah TK, Kazanjian A, Whitsett J and Shroyer NF: SAM pointed domain ETS factor (SPDEF) regulates terminal differentiation and maturation of intestinal goblet cells. Exp Cell Res. 316:452–265. 2010. View Article : Google Scholar : PubMed/NCBI

131 

Lo YH, Chung E, Li Z, Wan YW, Mahe MM, Chen MS, Noah TK, Bell KN, Yalamanchili HK, Klisch TJ, et al: Transcriptional regulation by ATOH1 and its Target SPDEF in the intestine. Cell Mol Gastroenterol Hepatol. 3:51–71. 2017. View Article : Google Scholar : PubMed/NCBI

132 

Noah TK, Lo YH, Price A, Chen G, King E, Washington MK, Aronow BJ and Shroyer NF: SPDEF functions as a colorectal tumor suppressor by inhibiting β-catenin activity. Gastroenterology. 144:1012–1023.e6. 2013. View Article : Google Scholar : PubMed/NCBI

133 

Riccio O, van Gijn ME, Bezdek AC, Pellegrinet L, van Es JH, Zimber-Strobl U, Strobl LJ, Honjo T, Clevers H and Radtke F: Loss of intestinal crypt progenitor cells owing to inactivation of both Notch1 and Notch2 is accompanied by derepression of CDK inhibitors p27Kip1 and p57Kip2. EMBO Rep. 9:377–383. 2008. View Article : Google Scholar : PubMed/NCBI

134 

Souaze F, Bou-Hanna C, Kandel C, Leclair F, Devalliere J, Charreau B, Bézieau S, Mosnier JF and Laboisse CL: Differential roles of Hath1, MUC2 and P27Kip1 in relation with gamma-secretase inhibition in human colonic carcinomas: A translational study. PLoS One. 8:e559042013. View Article : Google Scholar : PubMed/NCBI

135 

Aragaki M, Tsuchiya K, Okamoto R, Yoshioka S, Nakamura T, Sakamoto N, Kanai T and Watanabe M: Proteasomal degradation of Atoh1 by aberrant Wnt signaling maintains the undifferentiated state of colon cancer. Biochem Biophys Res Commun. 368:923–929. 2008. View Article : Google Scholar : PubMed/NCBI

136 

Cohen Y, Chetrit A, Cohen Y, Sirota P and Modan B: Cancer morbidity in psychiatric patients: Influence of lithium carbonate treatment. Med Oncol. 15:32–36. 1998. View Article : Google Scholar : PubMed/NCBI

137 

Gould TD, Gray NA and Manji HK: Effects of a glycogen synthase kinase-3 inhibitor, lithium, in adenomatous polyposis coli mutant mice. Pharmacol Res. 48:49–53. 2003.PubMed/NCBI

138 

Shakoori A, Mai W, Miyashita K, Yasumoto K, Takahashi Y, Ooi A, Kawakami K and Minamoto T: Inhibition of GSK-3 beta activity attenuates proliferation of human colon cancer cells in rodents. Cancer Sci. 98:1388–1393. 2007. View Article : Google Scholar : PubMed/NCBI

139 

Tan J, Zhuang L, Leong HS, Iyer NG, Liu ET and Yu Q: Pharmacologic modulation of glycogen synthase kinase-3beta promotes p53-dependent apoptosis through a direct Bax-mediated mitochondrial pathway in colorectal cancer cells. Cancer Res. 65:9012–9020. 2005. View Article : Google Scholar : PubMed/NCBI

140 

Ring DB, Johnson KW, Henriksen EJ, Nuss JM, Goff D, Kinnick TR, Ma ST, Reeder JW, Samuels I, Slabiak T, et al: Selective glycogen synthase kinase 3 inhibitors potentiate insulin activation of glucose transport and utilization in vitro and in vivo. Diabetes. 52:588–595. 2003. View Article : Google Scholar : PubMed/NCBI

141 

Veenendaal LM, Kranenburg O, Smakman N, Klomp A, Borel Rinkes IH and van Diest PJ: Differential Notch and TGFbeta signaling in primary colorectal tumors and their corresponding metastases. Cell Oncol. 30:1–11. 2008.PubMed/NCBI

142 

Wu Y, Cain-Hom C, Choy L, Hagenbeek TJ, de Leon GP, Chen Y, Finkle D, Venook R, Wu X, Ridgway J, et al: Therapeutic antibody targeting of individual Notch receptors. Nature. 464:1052–1057. 2010. View Article : Google Scholar : PubMed/NCBI

143 

Kim JW, Shin MK and Kim BC: Clinicopathologic impacts of poorly differentiated cluster-based grading system in colorectal carcinoma. J Korean Med Sci. 30:16–23. 2015. View Article : Google Scholar : PubMed/NCBI

144 

Akiyoshi T, Nakamura M, Yanai K, Nagai S, Wada J, Koga K, Nakashima H, Sato N, Tanaka M and Katano M: Gamma-secretase inhibitors enhance taxane-induced mitotic arrest and apoptosis in colon cancer cells. Gastroenterology. 134:131–144. 2008. View Article : Google Scholar : PubMed/NCBI

145 

Aleksic T and Feller SM: Gamma-secretase inhibition combined with platinum compounds enhances cell death in a large subset of colorectal cancer cells. Cell Commun Signal. 6:82008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2020
Volume 20 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fu Y, Yuan SS, Zhang LJ, Ji ZL and Quan XJ: Atonal bHLH transcription factor 1 is an important factor for maintaining the balance of cell proliferation and differentiation in tumorigenesis (Review). Oncol Lett 20: 2595-2605, 2020
APA
Fu, Y., Yuan, S., Zhang, L., Ji, Z., & Quan, X. (2020). Atonal bHLH transcription factor 1 is an important factor for maintaining the balance of cell proliferation and differentiation in tumorigenesis (Review). Oncology Letters, 20, 2595-2605. https://doi.org/10.3892/ol.2020.11833
MLA
Fu, Y., Yuan, S., Zhang, L., Ji, Z., Quan, X."Atonal bHLH transcription factor 1 is an important factor for maintaining the balance of cell proliferation and differentiation in tumorigenesis (Review)". Oncology Letters 20.3 (2020): 2595-2605.
Chicago
Fu, Y., Yuan, S., Zhang, L., Ji, Z., Quan, X."Atonal bHLH transcription factor 1 is an important factor for maintaining the balance of cell proliferation and differentiation in tumorigenesis (Review)". Oncology Letters 20, no. 3 (2020): 2595-2605. https://doi.org/10.3892/ol.2020.11833