Skip to main content

Retinal determination gene networks: from biological functions to therapeutic strategies

Abstract

The retinal determinant gene network (RDGN), originally discovered as a critical determinator in Drosophila eye specification, has become an important regulatory network in tumorigenesis and progression, as well as organogenesis. This network is not only associated with malignant biological behaviors of tumors, such as proliferation, and invasion, but also regulates the development of multiple mammalian organs. Three members of this conservative network have been extensively investigated, including DACH, SIX, and EYA. Dysregulated RDGN signaling is associated with the initiation and progression of tumors. In recent years, it has been found that the members of this network can be used as prognostic markers for cancer patients. Moreover, they are considered to be potential therapeutic targets for cancer. Here, we summarize the research progress of RDGN members from biological functions to signaling transduction, especially emphasizing their effects on tumors. Additionally, we discuss the roles of RDGN members in the development of organs and tissue as well as their correlations with the pathogenesis of chronic kidney disease and coronary heart disease. By summarizing the roles of RDGN members in human diseases, we hope to promote future investigations into RDGN and provide potential therapeutic strategies for patients.

Introduction

The normal developmental programs, including cell proliferation, differentiation, morphogenesis, and tissue homeostasis, are regulated by many intra-cellular molecular regulatory networks [1, 2]. Generally, the coordination of these programs maintains the balance of physiological functions in the organism. Dysfunction of genes and signaling pathways in the developmental program will break the homeostatic balance and lead to the occurrence of disease, including tumors [3]. The retinal determinant gene network (RDGN) was initially discovered in the study of Drosophila eye specification and primarily consisted of dachshund (dac/DACH), so/SIX, eya/EYA, and the Pax6 homologs. Dac/DACH is a structural relative of the Ski/Sno gene family, a dominant suppressor of elipse; eya/EYA, a tyrosine phosphatase eyes absent; so/SIX, the SIX family transcription factor (TF) sine oculis; and the Pax6 homologs, eyeless (ey) and twin of eyeless (toy) [4, 5]. Many previous studies have found that this regulatory network is essential for the development of the mammalian organ system and tumor progression by encoding nuclear TFs and cofactors [6].

The DACH family is composed of DACH1 and DACH2 [7]. DACH1, a putative tumor suppressor, is involved in the oncogenesis of various cancers, such as breast cancer (BC), prostate cancer, and so on [8, 9]. Furthermore, DACH1 has also been implicated in a variety of developmental diseases, mainly including renal diseases and cardiac coronary arteries development diseases, such as chronic kidney disease (CKD), familial young-onset diabetes, pre-diabetes, coronary heart disease (CHD), and coronary arteriosclerosis [10, 11]. SIX, is a supergene family which includes SIX1-6 [12, 13]. SIX1 was found to synergize with the transcriptional cofactor EYA, which had tyrosine/threonine-phosphatase activity, to promote tumor initiation and progression [14, 15].

It has been well demonstrated that the RDGN has been involved in tumorigenesis and the progression of BC. The balance of DACH1 and SIX1/EYA function keeps the homeostasis of luminal cell proliferation and apoptosis, which maintains the stability of the luminal structure. The abnormal expression of DACH1 and SIX/EYA caused a disorder in the balance of physiological functions [16]. Subsequently, the luminal may undergo proliferation, epithelial-mesenchymal transition (EMT), and progression to ductal carcinoma. It is generally accepted that a few malignant tumor cells, acquiring the features of cancer stem cells (CSCs) via EMT, trigger tumor metastasis by invading the basement membrane and entering blood vessels [16] (Fig. 1).

Fig. 1
figure 1

The Role of RDGN in the progression of breast cancer. The functional balance of DACH1 and SIX1/EYA keeps the homeostasis of luminal cell proliferation and apoptosis, which maintains the stability of the luminal structure. The abnormal expression of DACH1 and SIX/EYA caused a disorder of physiological functions. Subsequently, the luminal cells may undergo hyperproliferation, and transform to form ductal cancer in situ (DCIS). Then some cells acquire epithelial-mesenchymal transition (EMT) and trigger tumor metastasis by invading the basement membrane and entering blood vessels. “↓” means downregulated function, while “↑” means up-regulated functions. (The role of RDGN in the progression of RDGN was adapted from Fig. 1 in [16].)

Here, we summarize the research progress on RDGN members from biological behaviors to therapeutic applications, especially emphasizing their functions in tumors. In addition, we discuss the roles of RDGN members in the development of organs and tissue, which are correlated with the pathogenesis of chronic kidney disease and coronary heart disease. By summarizing the functions of RDGN members in human diseases, we hope to promote future investigations into RDGN and provide new insights into therapeutic strategies for those diseases.

The structures of RDGN members

RDGN has been discovered in the process of Drosophila eye determination. It attracted scholars' attention because it is a crucial signaling pathway in tissue specification and organogenesis of multiple organs. Furthermore, the abnormal expression and regulation of this network is linked to a range of different diseases, including cancers and congenital abnormalities or developmental defects [4, 5] (Table 1). This network mainly includes DACH, EYA, and SIX families.

Table 1 Summarization of targets regulated by RDGN

DACH

DACH is divided into two paralogs, including DACH1 and DACH2, encoding nuclear proteins with two deeply conserved domains: dachshund domain 1 (DD1) and dachshund domain 2 (DD2). DD1, also known as box-N, is able to bind to DNA. Another conserved domain is DD2, referred to as box-C, which is responsible for protein–protein interaction. Moreover, the X-ray crystal structure analysis has demonstrated that DD1 concludes a conserved domain, which is homologized with the Sno/Ski oncogene [7] (Fig. 2A). The DACH1 is capable of combining DNA sequences and chromatin or binding to other TFs (such as c-Jun, Smads, SIX, and estrogen receptor-α (ER-α)), which lead to the inhibition of the specific target genes transcription [14, 70,71,72]. Besides, the DACH1 DNA-binding site, which resembled the binding site of FOX (forkhead box-containing protein), competitively bound to FOX and inhibited forkhead signaling. Therefore, DACH1 replaced the function of FOXM1 in the promoter of the target gene and competed with FOXC2 to inhibit the expression of genes involved in the invasion and metastasis of tumor cells [71, 73, 74]. Moreover, it's reported that DACH1 remodels chromatin by the recruitment of NcoR, Sin3, and HDACs or CBP, resulting in the suppression or activation of transcription [25, 73, 75].

Fig. 2
figure 2

Structure of DACH, SIX, and EYA. A Key domain of DACH, SIX, and EYA. The working model of DACH, SIX, and EYA cooperate with transcriptional activator/repressor and cofactor. Abbreviations: DACH, dachshund; EYA, eyes absent; ED, Eya domain; SD, Six domain; HD, Homeobox domain; ER, estrogen receptor; NcoR, nuclear corepressor; Gro, Groucho; HDAC, histone deacetylase; P/S/T, proline, serine and threonine rich region. (The structure of RDGN was adapted from Fig. 1 in [76].)

DACH1 is not only involved in tumor progression but also participates in CKD, cardiovascular diseases, and bilateral cystic renal dysplasia [10, 11, 45]. Although the significance of DACH1 in human diseases has been widely investigated in the past decades, the roles of DACH1 in disease pathogenesis and the underlying mechanisms remain largely unknown.

SIX

SIX gene family, a homeobox gene superfamily, plays a role in tumor initiation and progression and organ development by encoding TFs to activate or inhibit the transcription of downstream target genes [77]. SIX family members consist of the SIX1/2, SIX3/6, and SIX4/5 superfamilies, which correspond to the homologs of sine oculis, optix, and D-SIX4, respectively [12, 13]. Similar to DACH1, SIX family also has two highly conserved sequences, including the homeoprotein domain (HD) and the SIX domain (SD) [67, 78]. The HD is related to the DNA binding process, and SD is associated with the interaction of protein–protein [67] (Fig. 2A). Moreover, A series of studies have shown that SIX2, SIX4, and SIX5 had endogenous transcriptional activation domains in addition to SD and HD. Therefore, co-activator proteins (such as EYAs) were not necessary for the transcription of these proteins [79, 80]. However, SIX1, SIX3, and SIX6 required co-activator (such as EYA1-4) to stimulate their transcriptional activity [81]. It’s well accepted that SIX proteins could promote tumor growth and other tumor biological behaviors [82, 83]. Additionally, it was reported that EYA is a co-activator of SIX while DACH is a co-suppressor of SIX [14, 76] (Fig. 2B). Hence, RDGN members play a coordinate role in the process of tumorigenesis.

EYA

The EYA family is composed of EYA1-4, most of which are transcriptional cofactors [84] (Fig. 2A). It has been indicated that the EYA family plays a role in the regulation of transcription and signal transduction processes owning to it containing three separate biochemical activities: tyrosine phosphatase, threonine phosphatase, and transactivation [85]. EYA has two tyrosine phosphatase activity targets, including histone variant H2AX [86, 87] and ERβ Y36 residue [88]. By dephosphorylating H2AX, EYAs such as EYA1, EYA2, and EYA3 induce DNA repair in cells after DNA damage [86, 87]. EYA1 stimulated the activity of Cyclin D1, and EYA2 inhibited the transcriptional activity of the ER-β by dephosphorylating the Y36 residue, thus promoting cancer cellular proliferation and tumor growth [88, 89]. EYA4 has threonine phosphatase activity and has a significant role in the innate immune response [90]. As a member of the RDGN family, EYA functions by regulating transcription and tyrosine phosphatase activity. The abnormal expression or activity of EYA is related to tumorigenesis, such as Ewing sarcoma and lung adenocarcinoma [91,92,93]. Moreover, mutations of SIX1 or EYA contribute to branchio-oto-renal (BOR) syndrome, muscle defects, asthma, etc. [59, 94, 95].

The role of RDGN in cancer

It is well recognized that DACH1, SIX, and EYA, the key members of RDGN, have abnormal expression in various cancers [76, 96]. RDGN played a key role in tumor initiation and progression by interacting with multiple signaling pathways in a coordinated manner, regulating the proliferation, apoptosis, movement, and stemlike property [63, 76, 96,97,98]. In general, DACH1 is down-regulated as a tumor suppressor, while SIX and EYA are up-regulated in tumors, which promotes tumorigenesis and tumor progression [99]. Watanabe et al. proposed that DACH1 could be a unique tumor suppressor [39]. A comprehensive and deeper understanding of how RDGN works in tumors will contribute to new therapeutic approaches by targeting RDGN members.

Proliferation

The dysregulation of mitosis signals is one of the prominent features of cancer cells, which leads to a disordered cell cycle and abnormal proliferation and eventually results in tumor initiation and progression [100, 101]. The RDGN maintained the dynamic balance of cells in the progression of the cell cycle (Fig. 3). Overexpression of DACH1 promoted renal cell arrest in the G1/S or G2/M phase by downregulating cyclin D1 and cyclin A and upregulating P21 and P53 [102]. Further analysis proved that DACH1 inhibited the transcription of cyclin D1 and cyclin A by directly binding to its corresponding promoter region, thereby further repressing the proliferation of cells and tumor growth in various cancer types, such as BC and renal cancer [34, 70, 103,104,105]. However. Lee et al. found that DACH1 induced cyclin D1 expression while inhibiting P21 expression in myeloid progenitor cells [106], which is fairly contradicted by previous findings in solid tumors. We speculate that there may exist significant functional differences between solid and hematologic tumors, as well as between progenitor and differentiated cells. The clinical database showed that the expression of Kruppel-like factor 5 (KLF5) was positively correlated with Y-box binding protein 1 (YB-1) while negatively correlated with DACH1 in patients with BC. Furthermore, YB-1 regulated KLF5 expression by inhibiting DACH1 transcriptional activation or stabilizing KLF5 mRNA construction, resulting in the progression of triple-negative breast cancer (TNBC). Ribosomal S6 kinase 2 (RSK2) mediated YB-1 phosphorylation, promoted the formation of the YB-1/KLF5 transcription complex, and further enhanced the proliferation of tumor cells. Consequently, the RSK inhibitor LJH685 primarily inhibited BLBC tumorigenesis by interfering with the YB-1-KLF5 axis ​[107].

Fig. 3
figure 3

The Role of RDGN in the Cell Cycle. DACH1 inhibits cyclin D1 transcription, resulting in G1-phase arrest. SIX1/EYA facilitates G1 progression via promoting the transcription of cyclin D1. Moreover, DACH1 also inhibits cyclin A, but SIX1 enhances cyclin A in S-phase. DACH1 increases the protein abundance of p21 and p27, further suppressing cdk2/cyclin E activity and finally preventing cells from entering S-phase. DACH1 also binds with p53 and synergizes p21 induction, and inhibits Cdc25, reducing the activity of Cdc2 to arrest cells in G2/M. (The role of RDGN in the cell cycle was adapted from Fig. 2 in [76].)

Xu et al. confirmed that SIX1 was highly expressed in rhabdomyosarcoma cells, inhibited rhabdomyosarcoma cell differentiation, and promoted tumor growth in vivo through MYOD1 and MYOG-mediated transcriptional genomic changes [108]. SIX1 promoted cyclin D1 transcription in rhabdomyosarcoma cells, leading to tumor initiation [57], and SIX1 also facilitated BC cell proliferation via inducing cyclin A expression [109]. Furthermore, EYA regulates the cell cycle and consequently promotes BC cell proliferation by increasing cyclin D1 transcription [89].

In addition to changing cyclin D1 expression, RDGN also regulates cell cycle progression via affecting P53 transcription. DACH1 binds to P53, enhancing P53-mediated cell cycle arrest to inhibit tumor growth [24, 29]. In contrast, SIX1 negatively regulates P53 to promote BC cell proliferation through upregulating microRNA-27a-3p and downregulating ribosomal protein L26 (RPL26) [110]. A recent study showed that SIX1 knockdown promoted DACH1 expression in vitro and in vivo in hepatocellular carcinoma (HCC) cells, which synergistically induced P53 expression and inhibited HCC progression [35]. The imbalance between the tumor-suppressor function of DACH1 and the oncogenic effect of SIX/EYA not only accelerates cell cycle disorder but also decreases apoptosis, providing the basis for tumorigenesis.

CSCs promote tumor progression and induce immune evasion through facilitating angiogenesis, proliferation, and colonization in other sites, which are strongly related to therapeutic resistance [111,112,113]. It has been demonstrated that RDGN is involved in the regulation of CSCs, especially in BC. Counteracting transcriptional activation of NANOG, KLF4, and SOX2, DACH1 reduced the proportion of breast CSCs [97]. The overexpression of SIX1 increases phenotypic CSCs in some cancers, such as breast, esophageal, colorectal, and pancreatic cancers [49, 54, 114, 115]. SIX1 mediate the CSCs phenotype by activating MAPK and TGF signaling pathway [49, 52]. Similarly, EYA also increases the proportion of breast CSCs via phosphatase activity [89]. Thus, RDGN may be a potential target to address therapeutic resistance by reducing the proportion of stem cells.

Invasion and metastasis

EMT is a dynamic process in which cells lose their epithelial phenotype and acquire the characteristics of a mesenchymal phenotype [116]. EMT renders cells high mobility, thus contributing to tumor cell migration and invasion [117]. Tumor cells detach themselves from the primary tumor site to colonize the secondary sites (distant organs), forming metastasis [118]. EMT is induced by activating Wnt/TGF-β signaling, which enhances the ability of tumor cells to migrate and invade [119,120,121,122]. Many studies demonstrated that RDGN is involved in the process of EMT through mediating Wnt/TGF-β signaling pathway [6] (Fig. 4).

Fig. 4
figure 4

The interaction of RDGN with key signaling in cancer. TGFβ and Wnt signaling pathways are vital for tumorigenesis and progression. RDGN is involved in the regulation of TGF-β and Wnt signaling pathways. DACH1 inhibits smad complex, while SIX/EYA activates TGF-ßR. DACH1 represses ß -catenin, while SIX/EYA activates Wnt signaling. EYA could switch DACH1 repressional function and enhance activation of SIX. Silencing the expression of SIX or interfering with the SIX/EYA complex using small molecule compounds can block the function of SIX. Moreover, EYA phosphatase activity can be inhibited by biochemical inhibitors. The use of demethylating agents can reactivate DACH1 expression. Therefore, targeting RDGN members holds promise as a cancer treatment strategy

DACH1 indirectly activates E-cadherin via inhibiting the transcription of SNAI1 in BC, which suppresses tumor cell migration and invasion [21]. Similarly, Bu et al. demonstrated that DACH1 inhibited pancreatic cancer cell invasion via activating E-cadherin and repressing N-cadherin and vimentin [123]. Furthermore, DACH1 suppressed tumor growth and EMT of mouse BC models via blocking YB-1 [23] and inhibited BC cell initiation by decreasing CD44 levels [22]. In addition, DACH1 suppressed MMP-9 transcription to reduce BC invasion and metastasis [20]. Overexpression of DACH1 inhibited the TGF-β-mediated EMT, thus suppressing colorectal cancer (CRC) cell growth, invasion, and metastasis. These results suggest that DACH1 is a prospective target for cancer therapies [19].

SIX1 also plays an important role in tumor invasion and metastasis. The upregulation of SIX1 was related to poor prognosis in many malignancies, such as cervical cancer, CRC, prostate cancer, and so on. Kong et al. found that SIX1 facilitated epithelial cancer cell proliferation, migration, and invasion via mediating the VEGF-C/NRP2/GLI signaling axis [124]. The knockdown of SIX1 suppressed the process of EMT in papillary thyroid carcinoma through inhibiting the TGF-β/SMAD2/3 signaling pathway [125]. Meanwhile, Huang et al. demonstrated that SIX1 facilitated migration and invasion in non-small cell lung cancer via activating the Notch pathway. The γ-secretase, a Notch pathway inhibitor, reversed the SIX1-mediated phenomenon [126]. Intriguingly, SIX1 was found to facilitate the EMT process in BC by forming a negative feedback loop with miR-204-5p [127]. Micalizzi et al. found that SIX1 contributed to BC cell invasion and metastasis via regulating TGF-β-induced EMT [128]. SIX1 promotes EMT in cervical cancer cells via the TGF-β/SMAD signaling pathway, in contrast to DACH1 [50, 129]. By enhancing NF‐κB/p65 pathway in macrophages, SIX1 enhanced the expression of matrix metalloproteinase 9 (MMP-9) and further promoted the invasion of cancer cells. Moreover, SIX1 triggered the activation of the IL-6/STAT3/MMP-9 signaling pathway, which further facilitated HCC invasion [56]. In CRC, it has been found that overexpression of SIX1 promotes cell invasion by mediating the expression of TEAD4 and ZEB1, or inhibiting miR-200 expression [130,131,132]. Furthermore, SIX1 contributed to the proliferation, invasion, and EMT of gastric cancer cells via multiple pathways, including cyclin D1, ERK, MMP-2, and E-cadherin [133]. Similarly, the up-regulation of SIX1 expression in cervical cancer induces EMT to enhance the proliferation, invasion, and migration of cells [134]. A study has shown that SIX1 promoted EMT and stem cell phenotype, leading to the activation of the Wnt pathway, such as cyclin D1 [128]. Smith et al. indicated that SIX1 activated the pro-tumor TGF-β signaling through regulating the expression of the miR-106b-25 cluster. At the same time, the miR-106p-25 cluster also could induce tumor cell EMT [135]. CircNHSL1 was shown to be highly expressed in gastric cancer tissues and cell lines and was associated with poor prognosis in patients. Mechanistically, circNHSL1 promoted gastric cancer invasion and metastasis by acting as a miR-1306-3p sponge to derepress its target SIX1. Meanwhile, SIX1 directly binds to vimentin promoter to further enhance vimentin expression This result suggested that CircNHSL1 promoted gastric cancer progression through the miR-1306-3p/SIX1/Vimentin axis [136].

As mentioned above, EYA function as a co-factor for SIX1. EYA enhanced the stemness of cancer stem cells and induced EMT by synergizing with SIX1 [137]. SIX1 in cooperation with EYA2 promoted tumor cell metastasis by inducing TGF-β signaling and EMT [63]. Zhang et al. found that EYA3 enhanced the stability of c-Myc via the dephosphorylation of protein phosphatase 2A-B55α, which increased the metastasis capability of BC cells [64]. Moreover, other studies also showed that EYA3 stabilized Myc through threonine phosphatase activity to dephosphorylate Myc at pT58, then MYC upregulated PD-L1 and decreased CD8+ T cell activity. Given the immunosuppressive role of EYA3, w inhibition of EYA3 may enhance the effect of immune checkpoint therapy [138].

However, there are some contrary reports on RDGN’s function in different cancers. DACH1 was up-regulated in ovarian cancer, which might facilitate the resistance to the TGF-β signaling pathway. Moreover, DACH1 protein levels were increased with the invasiveness of ovarian cancer and the subcellular distribution of DACH1 changed from nucleus in normal tissue to cytoplasm in cancer [38, 139]. Zheng et al. demonstrated that SIX3 was downregulated in numerous cancers, such as breast, glioma, lung adenocarcinoma, and gastric cancers [140,141,142,143]. As a negative regulator of the Wnt pathway, SIX3 inhibited breast cancer carcinogenesis and metastasis by recruiting the LSD1/NuRD (MTA3) complex. Moreover, expression profile analysis indicated that high SIX3 mRNA level was a protective factor for OS and RFS of basal-like breast cancer patients [141]. EYA4 functions as a tumor suppressor in pancreatic ductal adenocarcinoma (PDAC) by inhibiting the activation of β-catenin and ID2, representing a favorable prognostic factor in PDAC [144]. Moreover, EYA4 was also associated with a favorite prognosis in HCC and intrahepatic cholangiocarcinoma [145, 146].

Therefore, RDGN plays a different role in various cancers, and further exploration will clarify the organ/tissue-specific function of RDGN in cancer.

The reciprocal regulation of RDGN components

Recent studies have shown that there is a crosstalk among the components of RDGN and RDGN work coordinated to regulate tumor progression. The work by Cheng et al. found that the overexpression of DACH1 inhibited tumor initiation via up-regulating p53 expression, while SIX1 enhanced tumorigenesis via downregulation of p53 expression. Consistently, it was observed that the knockdown of SIX1 enhanced the expression of DACH1, which further activated the expression of p53 and ultimately inhibited hepatocellular carcinoma progression [35]. In addition, Farabaugh et al. showed that in BC, SIX1 cooperated with its co-activator EYA2 to induce the activation of the TGF-β signaling pathway, thereby promoting the malignant biological behavior of cancer cells, such as EMT, and the proliferation of cancer stem cells [63]. Similarly, SIX1/EYA signaling also transformed normal hematopoietic progenitors into leukemia [147]. SIX1 up-regulated VEGF-C expression and promoted lymph angiogenesis and lymphatic metastasis. Moreover, SIX1 induced the transcription of VEGF-C by enhancing the activation of the TGF-β/SMAD signaling pathway [51, 148]. In papillary thyroid cancer tissues, protein expressions of SIX1 and EYA1 are positively correlated, SIX1 stabilizes EYA1 at the post-transcriptional level and activates STAT3 signaling requiring EYA1 [15].

Crystal structure study showed that SIX1 helix interacted with HAD catalytic domain of EYA, and SIX1-EYA interaction was required for the activation of TGF signaling and EMT-like phenotypes. Moreover, disruption of SIX1-EYA interaction abolished SIX1-induced metastasis [67]. SIX1 was highly expressed in meningiomas. SIX1 and EYA2 functioned together to support the expression of leptin receptors, which promoted the proliferation of meningioma patient-derived cell lines [149]. Exploring the interaction of RDGN will help us better understand its role in tumors and the facility to develop effective therapeutic strategies.

RDGN in other diseases

In addition to the function of RDGN in tumors, recent studies have found that RDGN plays an important regulatory role in the occurrence and development of multiple human diseases (Fig. 5).

Fig. 5
figure 5

The role of RDGN in human diseases. DACH1 is required for the elongation of the coronary artery. DACH1 stimulates coronary artery endothelial cells (CECs) proliferation and promotes the migration of CECs by up-regulating CXCL12. Moreover, the overexpressed DACH1 in capillary endothelial cells promotes arterial normalization and prolongs arterial branches. The overexpression of EYA2 directly activates mTOR, leading to cardiac hypertrophy. In the hormonal system, DACH1 is linked to islet development, and insulin secretion, proving that DACH1 is involved in the pathogenesis of type-2 diabetes. In the urinary system, knocked down the expression of DACH1 caused damage to the kidney podocytes and tubular cells, eventually leading to CKD. Moreover, DACH1 knockout mice are more likely to aggravate the progress of diabetes nephropathy. DACH1 inhibits the expression of ATF6 and reduces tissue-type plasminogen activator (tPA) in mouse liver cells, reducing the risk of thrombosis. In the nervous system, DACH1 is a specific marker for neuroepithelial and ventricular radial glial cells. DACH1 is identified as one of the driver genes for postaxial polydactyly in the locomotor system. After denervation, lower DACH2 is observed in adult muscle fibers, allowing myogenin and nAchR expression to be up-regulated. While overexpression of DACH2 in denervated muscle reduced Mgn, nAChR, and MuSK gene induction depending on HDAC inhibitor. In the respiratory system, overexpression of SIX1 increases macrophage migration inhibitory factor (MIF) levels and promotes pulmonary fibrosis. Six1 is up-regulated in the chronic asthma mouse model. Silencing of Six1 reduced the expression and secretion of the airway remodeling-related mediators MMP-2 and MMP-9 through inhibition of the NF-KB pathway

Kidney

Diabetes and hypertension are the most common CKD risk factors [150, 151]. Moreover, it’s well-accepted that diabetic kidney disease (DKD) is one of the crucial causes of end-stage renal disease (ESRD) [151]. With the transcriptomic studies of human DKD and transcriptome-wide association studies of human CKD [38, 139], DACH1 in glomerular and tubular cells was demonstrated to be a protective factor for their function [152, 153].

The CKDGen consortium identified DACH1 as one of 13 novel loci influencing renal function and CKD through genome-wide association studies [10]. And DACH1 has also been identified as a gene with a therapeutic intervention function in CKD. In murine folate and diabetic kidney injury models, mice with tubule-specific DACH1 deletion were more susceptible to severe renal fibrosis. In contrast, fibrosis was rarely observed in mice with renal tubular-specific overexpression of DACH1. These results indicated that DACH1 could be used as a risk gene for kidney disease [152]. DACH1 regulated podocyte cell cycle and podocyte structure and function. Reduced DACH1 expression is associated with albuminuria and CKD [154]. DACH1-deficient mice were susceptible to hyperglycemia, streptozotocin, and adriamycin-induced kidney injury. Podocyte-specific overexpression of DACH1 protects mice from diabetic kidney disease. However, the mechanisms that reduce the expression of DACH1 are unknown [153]. Davis also found that mice with loss of function mutation in DACH1 exhibit postnatal lethality associated with respiratory dysfunction [155]. Meanwhile, one case report reported that the patient with concomitant mutations in DACH1 and BMP4 developed renal dysplasia [45]. Now it is well accepted that DACH1 protects podocyte and tubular cells from injury expression and its expression correlates with kidney function [156]. Considering that DACH1 is a protective factor for CKD, we speculate restoring DACH1 to normal levels may be a therapeutic target for kidney injury.

SIX1 and SIX2 are expressed in human fetal nephron progenitor cells. Although 80% of the target sites of SIX1 are shared with SIX2, and the expression of SIX1 in the posterior renal mass spectrometry system overlaps with SIX2, SIX1 couldn’t compensate for renal phenotype for the SIX2 deficiency. These results indicated that there were differences between SIX1 and SIX2 in the physiological role of kidney development stems from the transcriptional regulation of genes and the different biochemical characteristics of proteins [157].

Cardiovascular system

RDGN also plays a vital role in the cardiovascular system. Chang et al. found that the mice lacking endothelial DACH1 could lead to the reduction of coronary artery diameter and overexpression of DACH1 stimulated endothelial cell polarization and migration predominantly by up-regulating CXCL12/CXCR4 signaling [41]. Further study indicated that overexpression of DACH1 in capillary endothelial cells promoted coronary artery differentiation and prolonged arterial branches. In addition, the overexpression of DACH1 enhanced the ejection fraction and cardiac function, and improved survival in myocardial infarction mice [42]. Ischemic heart disease is majorly caused by atherosclerotic plaque or thrombotic vessel occlusion to block the blood supply. The observation that DACH1 stimulates arterial regeneration and improves cardiac function support a potential therapeutic approach by modifying the DACH1 signaling in heart endothelial cells.

Nervous system

DACH1 is involved in the development of the nervous system. The Castiglioni team found that DACH1 was overexpressed in proliferating neural progenitor cells in developing ventricles, subventricular regions and striatum from 5–21 weeks post-conception human brain samples. Single-cell transcriptome analysis identified DACH1 as a specific marker for neuroepithelial and ventricular radial glial cells [158, 159]. Genome-wide association study suggested that there was an age-dependent association between DACH1 and cerebral white matter volume [158, 159]. Antineuronal nuclear antibodies, a kind of novel neural autoantibody, are used as a biomarker of paraneoplastic neural autoimmunity [160]. Zekeridou et al. reported that DACH1 could be used as the autoantigen of ANNA3 determined by immunohistochemical colocalization and antigen specificity assay, and DACH1-IgG was identified as a new biomarker of neural autoimmunity [161].

Hepatocyte

The expression of DACH1 was reported to be increased in the liver of obese mice and humans. Liver-specific silencing of DACH1 improves hepatocyte insulin signaling, and protects against hyperinsulinemia, which suggested that DACH1 was also involved in the metabolism of hepatocytes. Molecular analysis demonstrated that silencing of DACH1 in the hepatocytes increased the expression of ATF6 by inhibiting the nuclear rejection of calcium/calmodulin-dependent protein kinase II (CaMKII) or histone deacetylase 4 (HDAC4) [162]. Tissue-type plasminogen activator (tPA) prevents excessive coagulation without compromising hemostasis. ATF6 is an inducer of tPA and inverse correlation between DACH1 and ATF6 in the human liver. Increased circulating tPA, fibrinolytic activity, and bleeding time were observed in DACH1-knock-out mice, which can be reversed by silencing ATF6 [163]. The balance between plasma PAI-1 and tPA proteins controls the fibrinolysis. DACH1 expression in hepatocytes between obese and lean affects the balance though regulating ATF6 ​[164]. Those studies provide a prospective therapeutic method to reduce the risk of thrombosis and improve fibrinolysis.

Development of limb, ear, and muscles

Polydactyly is a common congenital limb anomaly disorder caused by patterning defects in the developing anterior–posterior axis [165].​ DACH1 was identified as one of the causative genes for postaxial polydactyly by whole-exome sequencing and Sanger sequencing of DNA from individuals with postaxial polydactyly [166].​

Branchio-oto-renal syndrome (BOR) is characterized by hearing loss, and craniofacial and/or renal defects. So-binding protein (Sobp) binds as a SIX1 cofactor and inhibits the transcriptional activation of SIX1 and EYA1, which changes the developmental process of ear follicles and leads to craniofacial cartilage defects. Therefore, Sobp was determined as a candidate gene for BOR syndrome and other deafness syndromes [94]. Sox2+ proneurosensory progenitors within otic ectoderm form sensory cells and neurons for hearing. EYA1-SIX1 signaling facilitated the binding of Brg1 to enhancers of Sox2, which was correlated with the differentiation of neurosensory cells within the otic ectoderm [167]. Depleting Brg1 or EYA1-SIX1 signaling inhibited the expression of Sox2, and the subsequent deficiency of neurosensory identity resulted in the aberrant apoptosis of otic neurosensory cells [167]. The SIX1 mutant fetal mice display primary defects in myogenesis and muscle [95]. Pairing box 3 (Pax3) plays an important role in the development of lower axons and limb muscles [168]. The overexpression of SIX1 not only increased the expression of Pax3, but also elevated the proliferation of Pax7 ( +) cells by up-regulating Smad1/5/8 [169, 170]. EYA1 activates multiple target genes, including Pax3, MyoD, and myogenin. Consequently, EYA1/EYA2 mutant mice show delayed myogenesis [14, 171]. Lee et al. confirmed that the four transcription factors, SIX1, EYA1, Esrrb, and Pax3, transform fibroblasts into induced myogenic stem cells (iMSCs). Moreover, those iMSCs were effectively different into multinucleated myotubes and had a stronger proliferative capacity than muscle-derived stem cells. Thus, iMSCs provide a new therapy strategy for muscle atrophy and muscle regeneration [172].

Lung

Asthma is a chronic disease caused by inflammation in the respiratory tract [173]. Yang et al. identified that SIX1 was up-regulated in a chronic asthma murine model. In addition, with SIX1 knockdown, the expression and secretion of MMP-2 and MMP-9, mediators related to airway remodeling, were reduced, and the activation of the NF-KB pathway in the lung was simultaneously suppressed, which effectively suppressed airway inflammation and remodeling in asthmatic mice [59]. In addition, studies have also found that miR-448-5p overexpression inhibited TGF-β1-induced EMT and pulmonary fibrosis in asthmatic mice by targeting SIX1 expression. The results suggested that the miR-448-5p/TGF-β1/SIX1 axis may be a novel target for strategies to prevent airway remodeling in asthma [174]. Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and fibrotic lung disease. Increased expressions of SIX1 and EYA were observed in IPF lung tissue samples and IPF-derived alveolar type II cells (AT2). SIX1-driven expression of macrophage migration inhibitory factor (MIF) was attributed to the process of lung fibrosis. ISO-1, a MIF inhibitor, reduces SIX1-mediated cell proliferation and the expression levels of α-SMA and Col1a1, providing a potential pathway for IPF therapy [58].

Hematologic cells

It is known that the GATA and PAX-SIX-EYA-DACH transcriptional networks (PSEDNs) are essential for proper development. Creed et al. indicated that SIX1 and SIX2 strengthened GATA1 function and accelerated erythropoiesis in human erythroleukemia cell lines and primary hematopoietic stem-progenitor cells (HPCs). Conversely, the erythropoietin (EPO)-stimulated erythropoiesis process was impaired when SIX1 was knocked out [60]. MLL encodes a histone methyltransferase that maintains key gene expressions during embryonic development and hematopoiesis. MLL-AF9 is known to be a driver of acute myeloid leukemia (AML). Zhang et al. confirmed that SIX1 was the target gene of leukemic initiating cells (LICs) confusion protein (MLL-AF9), and blocking WNT/SIX1 signaling by Wnt signaling inhibitors suppressed the progression of AML [61]. EYA1 and SIX1 are induced by MLL-ENL, EYA1 could immortalize hematopoietic progenitor cells, and SIX1 enhanced the transforming capacity of EYA1 [175].

Others

Immune disorders are associated with non-classical NF-KB pathway dysfunction, and expression of NF-κB-inducing kinase (NIK) is the rate-limiting step in non-canonical NF-κB activation [176, 177]. NIK activates SIX protein in macrophages. SIX1 and SIX2 are identified as integral components of the non-canonical NF-κB signaling cascade. SIX1 and SIX2 inhibit the trans-activating function of RELA and RELB, the inflammatory gene promoters, leading to sustained non-classical NF-KB pathway activation. In addition, this research illustrated that SIX1 suppressed inflammation and promoted recovery ability from endotoxic shock in mice. Moreover, SIX1 and SIX2 preserved RAS/P53-driven non-small cell lung cancer from SMAC mimetic chemotherapeutic drug-induced inflammation [178]. Thus, NIK-SIX signaling axis programs inflammatory gene expressions in both physiological and pathological conditions.

The potential therapeutic targets of RDGN

It is generally accepted that cancer drugs can target particular molecular proteins to inhibit the malignant behavior of cancer cells, such as BCL2 [179,180,181,182]. Currently, molecular target therapeutics are mainly inhibitors of oncogenes, and it's challenging to design or discover an effective cancer drug targeting tumor suppressor [183,184,185]. The changes in RDGN members' expression and physiological functions have been observed in various tumors. Moreover, a series of studies found that the aberrant expression of RDGN contributed to the malignant behaviors of tumors, as discussed in the previous sections. Several studies have proved that targeting RDGN members maybe a potential therapeutic approach for cancer therapy [86]. Here, we summarize potential therapeutic strategies targeting RDGN (Fig. 4).

DACH1

Studies have extensively demonstrated that the down-expression of DACH1 is related to poor prognoses in many tumors, such as BC, prostate cancer, lung cancer, and hepatocellular carcinoma [8, 103, 105, 186,187,188]. Moreover, Nuclear DACH1 expression could be a luminal biomarker and independent prognosis factor [189]. Importantly, circulating cell-free DNA-based four methylation markers (RASGRF1, CPXM1, HOXA10, and DACH1) and two parameters (cfDNA concentration and the mean of 12 methylation markers) could detect early breast cancer. The sensitivity and specificity are comparable to mammography screening [190]. Therefore, DACH1 can be used as a potential molecule in cancer diagnosis and therapy.

Chu et al. also found that the reactivation of DACH1 might be a novel renal clear cell cancer therapy because it suppressed the clear renal cell cancer cell growth and proliferation by down-expressing the cyclin D1 [34]. The overexpression of miR-217 enhanced cell cycle progression to promote the proliferation of BC cells, and DACH1 is one of the targets of miR-217. Therefore, targeting miR-217/DACH1 axis is a plausible strategy for BC therapy [191]. Liu et al. found that DACH1 was negatively correlated with CXCL8 via evaluating the lung adenocarcinoma cell lines and tissues. Meanwhile, it had been demonstrated that AP-1 and NF-kB activation sites of CXCL8 promoter were repressed by DACH1. This result demonstrated that the DACH1-CXCL8 axis regulated lung cancer progression and affected the prognosis of lung adenocarcinoma patients [28]. However, there are studies showing that DACH1 plays an oncogenic role in endometrial cancer and leukemia [76, 104]. Similarly, Hu et al. showed that DACH1 could be identified as a tumor promoter based on the organoid model. Overexpression of DACH1 increased stem cell proliferation by suppressing bone morphogenetic protein (BMP) via SMAD4. Therefore, they proposed that DACH1 could be a potential therapeutic target for CRC patients [17]. This discrepancy may attribute to the activation of specific signaling pathways by DACH1 and the heterogeneity in different organs.

Restoration of DACH1 expression may be an innovative approach to cancer treatment. For example, the restoration of DACH1 expression could sensitize HCCs to 5-fluorouracil [36], and it also enhanced the sensitivity to docetaxel in CRC cells and gastric cancer cells [18, 33]. 5-Aza-2'-deoxyazacytidine, a demethylating agent, reactivated DACH1 expression, which would manipulate for tumor therapy [33]. Furthermore, a recent study indicated that DACH1 methylation might be an early marker in esophageal cancer and correlated with progression. DACH1 hyper-methylation was associated with poor differentiation and advanced stage. Ectopic expression of DACH1 activated TGF-β signaling and inhibited cancer cell proliferation and tumor growth. Therefore, restoration of DACH1 by demethylase may be a promising therapeutic target for patients with esophageal cancer [31]. A similar phenomenon is also observed in lung adenocarcinoma cells [192].

Metformin, a common medicine for diabetes, possesses an anti-tumor role in several kinds of cancer cells, such as BC and pancreatic cancer [193, 194]. The study showed that metformin increased the expression of DACH1 primarily via activating AMPK phosphorylation, then DACH1 inhibited the secretion of CXCL1 though the inhibition of NF-kB in esophageal squamous carcinoma cells. Thus, the antitumor effect of metformin was attributed to the reduced PMN-MDSC accumulation in the tumor microenvironment via AMPK-DACH1-CXCL1 signaling [32].

SIX1

Members of the SIX family, especially SIX1, play oncogenic roles in various cancers, which suggests that SIX1 may be a potential therapeutic target in cancer therapy.

SIX1 promoted stem or progenitor cell phenotype and induced EMT in tumor cells, indicating that SIX1 knockdown may be a method of inhibiting tumor growth and delaying tumor progression [98]. The study showed that SIX1 strengthened the special subgroup of cancer stem/progenitor cells via binding to transcriptional factors [76, 98]. Several studies in recent years have demonstrated that targeting the SIX/EYA transcriptional complex delayed tumor progression, indicating that the inhibitors’ development of this complex could be effective in cancer therapy [96]. Meanwhile, Zhou et al. found a small molecule SIX1/EYA inhibitor compound named NCGC00378430, which effectively suppressed the metastasis of BC in the murine model [195]. It’s been widely demonstrated that SIX1 was over-expressed in prostate cancer. The study has found that the GRP75-USP1-SIX1 complex inhibited the growth of prostate cancer. Moreover, this compound could also alleviate the drug resistance effect of prostate cancer cells in mouse models, which may provide novel ideas for the future treatment of prostate cancer [196]. Many studies have illustrated that aerobic glycolysis accelerated tumor growth. SIX1 was found to boost the ability of aerobic glycolysis. Meanwhile, miRNA-548a-3p was shown to be negatively correlated with SIX1 and could inhibit the glycolytic function of SIX1. This research showed that targeting the miRNA-548a-3p/SIX1 axis would presumably serve as a therapeutic target in BC [197]. The short-form recepteur d'origine nantais (sf-RON) was found to enhance glucose metabolism in gastric cancer by activating the β-catenin/SIX1 signaling axis, which promotes tumor cell proliferation, which indicated that sf-RON might serve as a promising therapeutic target for gastric cancer [198]. SNS-032 was able to induce the degradation of SIX1 through the inactivation of the EGFR-AKT-USP1 axis, and SNS-032 in combination with sorafenib was found to inhibit tumor growth in a mouse model of HCC [199]. Zhang et al. revealed that SIX1 was one of the WNT-controlled target genes in MLL-AF9-transformed LICs. MLL-AF9 was found to induce both the expression of SIX1 and EYA1 [175, 200]. MLL-AF9 promotes WNT/-linked protein-dependent growth of LICs by binding TCF7L2 to the TCF/LEF, a regulating element of SIX1. Furthermore, Zhang et al. illustrated that SIX1 directly affected the expression activity of SIX1 and EYA1 in MLL-AF9. With the administration of WNT signaling inhibitors, WNT/SIX1 signaling was disrupted, which delayed the progression of AML [61]. Monteiro et al. proposed that the methylation of the SIX1 promoter could be an independent prognostic factor for overall survival in melanoma patients [201].

Liu et al. fund that SIX1 expressions were high in human tumor samples and inversely correlated with immune cell infiltration in the TME. Tumor growth was decreased in an immune-dependent way, and the anti-tumor immunity was strengthened in the tumor microenvironment with the silence of SIX1. These results suggested that targeting SIX1 provided a novel idea for cancer immunotherapy [202].

EYA

EYA is a protein tyrosine phosphatase that facilitates the repair of DNA damage and tumorigenesis. Therefore, it is speculated that EYA may lead to relapse from chemotherapy and radiotherapy [86, 89]. And we propose that using EYA inhibitors may reduce chemo-resistance. Recently, it has been found that EYA2 Tyr phosphatase inhibitors could reduce the mesenchymal phenotype, enhance the immunogenicity of tumor cells, weaken tumor growth, and increase the antitumor effects of anti-PD-1 in the mouse BC model by targeting FBOX7. This experiment may provide a favorable basis for immune checkpoint block therapy via combination with FBOX7/EYA2 inhibitors [203]. Moreover, Vartuli et al. discovered that EYA3 was highly expressed in BC and reduced the number of infiltrating CD8+T cells along with T-cell depletion. Meanwhile, EYA3 was able to enhance PD-L1 expression via c-Myc to accelerate tumor growth. Hence, it is believed that the small molecule inhibitors of EYA3 could improve the curative effect of immune checkpoint blockades through their combination with an immune checkpoint inhibitor [138]. The overexpression of EGFR is associated with poor prognosis in BC. MiR-338-3p inhibited EGFR and subsequently activated EYA2, which accelerates BC tumor cell growth and lung metastasis. Moreover, the application of EYA2 inhibitors or miR-338-3p activators may be a promising therapeutic approach for BC lung metastasis [204]. The binding of EYA2 and DACH1 transcriptionally regulates the expression of SOCS3 and inhibits the progression of hepatocellular carcinoma by blocking the activation of the SOCS3-mediated JAK/STAT signaling pathway. Meanwhile, the delivery of EYA2 can be used to treat orthotopic liver cancer in nude mice, which showed that EYA2 may be a target for liver cancer treatment [205]. EYA4, a member of the EYA family, has been identified as a promising tumor suppressor in HCC, potentially inhibiting malignant behaviors in HCC cell lines by interfering with NF-κB/RAP1 signaling activation [145]. EYA4 was also identified as a tumor suppressor and abnormally hypermethylated in ESCC [206]. A class of reversible EYA inhibitors has been elucidated that selectively inhibit the activity of EYA2 phosphatase, and such compounds inhibit tumor cell migration, such as MLS00054460, MLS000585814, NCGC00249327, and NCGC00241224. And they could serve as prototypes for the development of EYA2 phosphate-specific anticancer drugs [207]. ​Moreover, another small-molecular complex was NCGC00249987, which prevented Mg2+ to bind with EYA2, and further inhibited the activity of EYA Tyr phosphatase. Anantharajan et al. indicated that NCGC00249987 specifically suppressed the migration and invasion in lung adenocarcinoma cells, but it didn't inhibit the survival and proliferation or of cancer cells [93].

Perspective

The RDGN members determine the biological behavior and therapeutic response of tumor cells and also affect the development of organs and tissues. In this work, we review the structures and functions of the RDGN gene family in tumorigenesis. We also summarize their roles in the physiological processes of tissue specification and organ development. The RDGN family has a specific expression pattern in tumors, which is associated with the clinicopathologic features and prognosis of cancer patients. There are interactions among RDGN family and a variety of tumor-related signaling pathways. These complex regulatory networks suggest that the functions of the RDGN members could be regulated by existing small-molecule targeted therapies, such as EGFR and Wnt inhibitors. In addition, the RDGN family is involved in the regulation of the tumor microenvironment and immune cell functions, shedding new light on tumor immunotherapy. However, the mechanism by which the RDGN family regulates tumorigenesis and its regulatory relationship with other molecules remain unclear.

The RDGN family regulates the expansion and differentiation of organ-specific stem cells or progenitor cells. Thus balanced expressions of RDGN determine the normal development of multiple organs. Correspondingly, their dysfunctions attribute to a series of human diseases, such as CKD, coronary disease, and lung fibrosis. With the rapid advancement of gene editing techniques, targeting RDGN for gene therapy is plausible.

From organ development to tumorigenesis, the RDGN is a conserved network. DACH1 hyper-methylation results in epigenetic silencing in various cancers, including esophageal and breast cancers. Restoring DACH1 expression may inhibit tumorigenesis via demethylase. SIX1 is overexpressed in many cancers compared to normal tissue. Inhibition of SIX1 by small interfering RNA may suppress the progression of cancer. As a co-transcriptor for SIX1, the tyrosine phosphatase activity of EYA promotes the progression of tumors. It has been confirmed that this activity could be specifically inhibited by benzbromarone and benzarone. At present, the major studies on DACH1, SIX1, and EYA have been focused on their individual biological function and underlying molecular mechanisms. The multi-omics study is needed to explore the complex interaction of RDGN and organ-specific function in different cancers.

With the development of precision medicine and genomics, molecularly targeted therapies have revolutionized the treatment strategy for several kinds of cancers. At present, researches on RDGN are still in the preclinical stage. Translational potential needs to be explored to address the clinical value of targeting the RDGN network for cancer therapeutic strategy.

Availability of data and materials

Not applicable.

Abbreviations

RDGN:

Retinal determinant gene network

TF:

Transcription factor

CKD:

Chronic kidney disease

CHD:

Coronary heart disease

EMT:

Epithelial-mesenchymal transition

CSCs:

Cancer stem cells

DD1:

Dachshund domain 1

DD2:

Dachshund domain 2

HD:

Homeoprotein domain

SD:

SIX domain

ER-α:

Estrogen receptor-α

BOR syndrome:

Branchio-oto-renal syndrome

KLF5:

Kruppel-like factor 5

YB-1:

Y-box binding protein 1

BLBC:

Basal-like breast cancer

RSK2:

Ribosomal S6 kinase 2

RPL26:

Ribosomal protein L26

HCC:

Hepatocellular carcinoma cell

CRC:

Colorectal cancer

MMP-9:

Matrix metalloproteinase 9

DKD:

Diabetic kidney disease

ESRD:

End-stage renal disease

ANNA3:

Antineuronal nuclear antibody 3

CaMKII:

Calcium/calmodulin-dependent protein kinase II

HDAC4:

Histone deacetylase 4

tPA:

Tissue-type plasminogen activator

Sobp:

So-binding protein

Pax3:

Pairing box 3

MIF:

Migration inhibitory factor

EPO:

Erythropoietin

AML:

Acute myeloid leukemia

BMP:

Bone morphogenetic protein

sf-RON:

Short-form recepteur d'origine nantais

References

  1. Abate-Shen C. Deregulated homeobox gene expression in cancer: cause or consequence? Nat Rev Cancer. 2002;2(10):777–85.

    Article  CAS  Google Scholar 

  2. Samuel S, Naora H. Homeobox gene expression in cancer: insights from developmental regulation and deregulation. Eur J Cancer. 2005;41(16):2428–37.

    Article  CAS  Google Scholar 

  3. Pearson JC, Lemons D, McGinnis W. Modulating Hox gene functions during animal body patterning. Nat Rev Genet. 2005;6(12):893–904.

    Article  CAS  Google Scholar 

  4. Kumar JP. The molecular circuitry governing retinal determination. Biochim Biophys Acta. 2009;1789(4):306–14.

    Article  CAS  Google Scholar 

  5. Jemc J, Rebay I. Targeting Drosophila eye development. Genome Biol. 2006;7(7):226.

    Article  Google Scholar 

  6. Silver SJ, Rebay I. Signaling circuitries in development: insights from the retinal determination gene network. Development. 2005;132(1):3–13.

    Article  CAS  Google Scholar 

  7. Mardon G, Solomon NM, Rubin GM. dachshund encodes a nuclear protein required for normal eye and leg development in Drosophila. Development. 1994;120(12):3473–86.

    Article  CAS  Google Scholar 

  8. Wu K, Katiyar S, Li A, et al. Dachshund inhibits oncogene-induced breast cancer cellular migration and invasion through suppression of interleukin-8. Proc Natl Acad Sci U S A. 2008;105(19):6924–9.

    Article  CAS  Google Scholar 

  9. Chen K, Wu K, Jiao X, et al. The endogenous cell-fate factor dachshund restrains prostate epithelial cell migration via repression of cytokine secretion via a cxcl signaling module. Cancer Res. 2015;75(10):1992–2004.

    Article  CAS  Google Scholar 

  10. Köttgen A, Pattaro C, Böger CA, et al. New loci associated with kidney function and chronic kidney disease. Nat Genet. 2010;42(5):376–84.

    Article  Google Scholar 

  11. Ma RC, Lee HM, Lam VK, et al. Familial young-onset diabetes, pre-diabetes and cardiovascular disease are associated with genetic variants of DACH1 in Chinese. PLoS ONE. 2014;9(1):e84770.

    Article  Google Scholar 

  12. Seimiya M, Gehring WJ. The Drosophila homeobox gene optix is capable of inducing ectopic eyes by an eyeless-independent mechanism. Development. 2000;127(9):1879–86.

    Article  CAS  Google Scholar 

  13. Seo HC, Curtiss J, Mlodzik M, et al. Six class homeobox genes in drosophila belong to three distinct families and are involved in head development. Mech Dev. 1999;83(1–2):127–39.

    Article  CAS  Google Scholar 

  14. Li X, Oghi KA, Zhang J, et al. Eya protein phosphatase activity regulates Six1-Dach-Eya transcriptional effects in mammalian organogenesis. Nature. 2003;426(6964):247–54.

    Article  CAS  Google Scholar 

  15. Kong D, Li A, Liu Y, et al. SIX1 Activates STAT3 Signaling to Promote the Proliferation of Thyroid Carcinoma via EYA1. Front Oncol. 2019;9:1450.

    Article  Google Scholar 

  16. Kong D, Liu Y, Liu Q, et al. The retinal determination gene network: from developmental regulator to cancer therapeutic target. Oncotarget. 2016;7(31):50755–65.

    Article  Google Scholar 

  17. Hu X, Zhang L, Li Y, et al. Organoid modelling identifies that DACH1 functions as a tumour promoter in colorectal cancer by modulating BMP signalling. EBioMedicine. 2020;56:102800.

    Article  Google Scholar 

  18. Yan W, Wu K, Herman JG, et al. Epigenetic regulation of DACH1, a novel Wnt signaling component in colorectal cancer. Epigenetics. 2013;8(12):1373–83.

    Article  CAS  Google Scholar 

  19. Wang P. Suppression of DACH1 promotes migration and invasion of colorectal cancer via activating TGF-β-mediated epithelial-mesenchymal transition. Biochem Biophys Res Commun. 2015;460(2):314–9.

    Article  CAS  Google Scholar 

  20. Aman S, Li Y, Cheng Y, et al. DACH1 inhibits breast cancer cell invasion and metastasis by down-regulating the transcription of matrix metalloproteinase 9. Cell Death Discov. 2021;7(1):351.

    Article  CAS  Google Scholar 

  21. Zhao F, Wang M, Li S, et al. DACH1 inhibits SNAI1-mediated epithelial-mesenchymal transition and represses breast carcinoma metastasis. Oncogenesis. 2015;4(3): e143.

    Article  CAS  Google Scholar 

  22. Xu H, Yu S, Yuan X, et al. DACH1 suppresses breast cancer as a negative regulator of CD44. Sci Rep. 2017;7(1):4361.

    Article  Google Scholar 

  23. Wu K, Chen K, Wang C, et al. Cell fate factor DACH1 represses YB-1-mediated oncogenic transcription and translation. Cancer Res. 2014;74(3):829–39.

    Article  CAS  Google Scholar 

  24. Chen K, Wu K, Gormley M, et al. Acetylation of the cell-fate factor dachshund determines p53 binding and signaling modules in breast cancer. Oncotarget. 2013;4(6):923–35.

    Article  Google Scholar 

  25. Wu K, Yang Y, Wang C, et al. DACH1 inhibits transforming growth factor-beta signaling through binding Smad4. J Biol Chem. 2003;278(51):51673–84.

    Article  CAS  Google Scholar 

  26. Yu S, Yi M, Xu L, et al. CXCL1 as an Unfavorable Prognosis Factor Negatively Regulated by DACH1 in Non-small Cell Lung Cancer. Front Oncol. 2019;9:1515.

    Article  Google Scholar 

  27. Han N, Yuan X, Wu H, et al. DACH1 inhibits lung adenocarcinoma invasion and tumor growth by repressing CXCL5 signaling. Oncotarget. 2015;6(8):5877–88.

    Article  Google Scholar 

  28. Liu Q, Li A, Yu S, et al. DACH1 antagonizes CXCL8 to repress tumorigenesis of lung adenocarcinoma and improve prognosis. J Hematol Oncol. 2018;11(1):53.

    Article  Google Scholar 

  29. Chen K, Wu K, Cai S, et al. Dachshund binds p53 to block the growth of lung adenocarcinoma cells. Cancer Res. 2013;73(11):3262–74.

    Article  CAS  Google Scholar 

  30. Zhu J, Wu C, Li H, et al. DACH1 inhibits the proliferation and invasion of lung adenocarcinoma through the downregulation of peroxiredoxin 3. Tumour Biol. 2016;37(7):9781–8.

    Article  CAS  Google Scholar 

  31. Wu L, Herman JG, Brock MV, et al. Silencing DACH1 promotes esophageal cancer growth by inhibiting TGF-β signaling. PLoS ONE. 2014;9(4): e95509.

    Article  Google Scholar 

  32. Qin G, Lian J, Huang L, et al. Metformin blocks myeloid-derived suppressor cell accumulation through AMPK-DACH1-CXCL1 axis. Oncoimmunology. 2018;7(7): e1442167.

    Article  Google Scholar 

  33. Yan W, Wu K, Herman JG, et al. Epigenetic silencing of DACH1 induces the invasion and metastasis of gastric cancer by activating TGF-β signalling. J Cell Mol Med. 2014;18(12):2499–511.

    Article  CAS  Google Scholar 

  34. Chu Q, Han N, Yuan X, et al. DACH1 inhibits cyclin D1 expression, cellular proliferation and tumor growth of renal cancer cells. J Hematol Oncol. 2014;7:73.

    Article  Google Scholar 

  35. Cheng Q, Ning D, Chen J, et al. SIX1 and DACH1 influence the proliferation and apoptosis of hepatocellular carcinoma through regulating p53. Cancer Biol Ther. 2018;19(5):381–90.

    Article  CAS  Google Scholar 

  36. Zhu H, Wu K, Yan W, et al. Epigenetic silencing of DACH1 induces loss of transforming growth factor-β1 antiproliferative response in human hepatocellular carcinoma. Hepatology. 2013;58(6):2012–22.

    Article  CAS  Google Scholar 

  37. Zhou Q, Li W, Kong D, et al. DACH1 suppresses epithelial to mesenchymal transition (EMT) through Notch1 pathway and reverses progestin resistance in endometrial carcinoma. Cancer Med. 2019;8(9):4380–8.

    Article  CAS  Google Scholar 

  38. Sunde JS, Donninger H, Wu K, et al. Expression profiling identifies altered expression of genes that contribute to the inhibition of transforming growth factor-beta signaling in ovarian cancer. Cancer Res. 2006;66(17):8404–12.

    Article  CAS  Google Scholar 

  39. Watanabe A, Ogiwara H, Ehata S, et al. Homozygously deleted gene DACH1 regulates tumor-initiating activity of glioma cells. Proc Natl Acad Sci U S A. 2011;108(30):12384–9.

    Article  CAS  Google Scholar 

  40. Zheng X, Liu Q, Yi M, et al. The regulation of cytokine signaling by retinal determination gene network pathway in cancer. Onco Targets Ther. 2018;11:6479–87.

    Article  CAS  Google Scholar 

  41. Chang AH, Raftrey BC, D’Amato G, et al. DACH1 stimulates shear stress-guided endothelial cell migration and coronary artery growth through the CXCL12-CXCR4 signaling axis. Genes Dev. 2017;31(13):1308–24.

    Article  CAS  Google Scholar 

  42. Raftrey B, Williams M, Rios Coronado PE, et al. Dach1 Extends Artery Networks and Protects Against Cardiac Injury. Circ Res. 2021;129(7):702–16.

    Article  CAS  Google Scholar 

  43. Iso T, Maeda K, Hanaoka H, et al. Capillary endothelial fatty acid binding proteins 4 and 5 play a critical role in fatty acid uptake in heart and skeletal muscle. Arterioscler Thromb Vasc Biol. 2013;33(11):2549–57.

    Article  CAS  Google Scholar 

  44. Jabs M, Rose AJ, Lehmann LH, et al. Inhibition of Endothelial Notch Signaling Impairs Fatty Acid Transport and Leads to Metabolic and Vascular Remodeling of the Adult Heart. Circulation. 2018;137(24):2592–608.

    Article  CAS  Google Scholar 

  45. Schild R, Knüppel T, Konrad M, et al. Double homozygous missense mutations in DACH1 and BMP4 in a patient with bilateral cystic renal dysplasia. Nephrol Dial Transplant. 2013;28(1):227–32.

    Article  CAS  Google Scholar 

  46. Macpherson PC, Farshi P, Goldman D. Dach2-Hdac9 signaling regulates reinnervation of muscle endplates. Development. 2015;142(23):4038–48.

    CAS  Google Scholar 

  47. Cohen TJ, Waddell DS, Barrientos T, et al. The histone deacetylase HDAC4 connects neural activity to muscle transcriptional reprogramming. J Biol Chem. 2007;282(46):33752–9.

    Article  CAS  Google Scholar 

  48. Moresi V, Williams AH, Meadows E, et al. Myogenin and class II HDACs control neurogenic muscle atrophy by inducing E3 ubiquitin ligases. Cell. 2010;143(1):35–45.

    Article  CAS  Google Scholar 

  49. Iwanaga R, Wang CA, Micalizzi DS, et al. Expression of Six1 in luminal breast cancers predicts poor prognosis and promotes increases in tumor initiating cells by activation of extracellular signal-regulated kinase and transforming growth factor-beta signaling pathways. Breast Cancer Res. 2012;14(4):R100.

    Article  CAS  Google Scholar 

  50. Micalizzi DS, Wang CA, Farabaugh SM, et al. Homeoprotein Six1 increases TGF-beta type I receptor and converts TGF-beta signaling from suppressive to supportive for tumor growth. Cancer Res. 2010;70(24):10371–80.

    Article  CAS  Google Scholar 

  51. Wang CA, Jedlicka P, Patrick AN, et al. SIX1 induces lymphangiogenesis and metastasis via upregulation of VEGF-C in mouse models of breast cancer. J Clin Invest. 2012;122(5):1895–906.

    Article  CAS  Google Scholar 

  52. Xu H, Zhang Y, Altomare D, et al. Six1 promotes epithelial-mesenchymal transition and malignant conversion in human papillomavirus type 16-immortalized human keratinocytes. Carcinogenesis. 2014;35(6):1379–88.

    Article  CAS  Google Scholar 

  53. Liu D, Li L, Zhang XX, et al. Correction: SIX1 Promotes Tumor Lymphangiogenesis by Coordinating TGFβ Signals That Increase Expression of VEGF-C. Cancer Res. 2019;79(7):1715.

    Article  Google Scholar 

  54. Nishimura T, Tamaoki M, Komatsuzaki R, et al. SIX1 maintains tumor basal cells via transforming growth factor-β pathway and associates with poor prognosis in esophageal cancer. Cancer Sci. 2017;108(2):216–25.

    Article  CAS  Google Scholar 

  55. Behbakht K, Qamar L, Aldridge CS, et al. Six1 overexpression in ovarian carcinoma causes resistance to TRAIL-mediated apoptosis and is associated with poor survival. Cancer Res. 2007;67(7):3036–42.

    Article  CAS  Google Scholar 

  56. Zhang Y, Wang S, Liu Z, et al. Increased Six1 expression in macrophages promotes hepatocellular carcinoma growth and invasion by regulating MMP-9. J Cell Mol Med. 2019;23(7):4523–33.

    Article  CAS  Google Scholar 

  57. Yu Y, Davicioni E, Triche TJ, et al. The homeoprotein six1 transcriptionally activates multiple protumorigenic genes but requires ezrin to promote metastasis. Cancer Res. 2006;66(4):1982–9.

    Article  CAS  Google Scholar 

  58. Wilson C, Mertens TC, Shivshankar P, et al. Sine oculis homeobox homolog 1 plays a critical role in pulmonary fibrosis. JCI Insight. 2022;7(10):e142984.

    Article  Google Scholar 

  59. Yang ZC, Yi MJ, Shan YC, et al. Targeted inhibition of Six1 attenuates allergic airway inflammation and remodeling in asthmatic mice. Biomed Pharmacother. 2016;84:1820–5.

    Article  CAS  Google Scholar 

  60. Creed Creed, Baldeosingh R, Eberly CL, et al. The PAX-SIX-EYA-DACH network modulates GATA-FOG function in fly hematopoiesis and human erythropoiesis. Development. 2020;147(1):dev177022.

    CAS  Google Scholar 

  61. Zhang LS, Kang X, Lu J, et al. Installation of a cancer promoting WNT/SIX1 signaling axis by the oncofusion protein MLL-AF9. EBioMedicine. 2019;39:145–58.

    Article  Google Scholar 

  62. Maire P, Dos Santos M, Madani R, et al. Myogenesis control by SIX transcriptional complexes. Semin Cell Dev Biol. 2020;104:51–64.

    Article  CAS  Google Scholar 

  63. Farabaugh SM, Micalizzi DS, Jedlicka P, et al. Eya2 is required to mediate the pro-metastatic functions of Six1 via the induction of TGF-β signaling, epithelial-mesenchymal transition, and cancer stem cell properties. Oncogene. 2012;31(5):552–62.

    Article  CAS  Google Scholar 

  64. Zhang L, Zhou H, Li X, et al. Eya3 partners with PP2A to induce c-Myc stabilization and tumor progression. Nat Commun. 2018;9(1):1047.

    Article  Google Scholar 

  65. Lee SH, Kim J, Ryu JY, et al. Transcription coactivator Eya2 is a critical regulator of physiological hypertrophy. J Mol Cell Cardiol. 2012;52(3):718–26.

    Article  CAS  Google Scholar 

  66. Schönberger J, Wang L, Shin JT, et al. Mutation in the transcriptional coactivator EYA4 causes dilated cardiomyopathy and sensorineural hearing loss. Nat Genet. 2005;37(4):418–22.

    Article  Google Scholar 

  67. Patrick AN, Cabrera JH, Smith AL, et al. Structure-function analyses of the human SIX1-EYA2 complex reveal insights into metastasis and BOR syndrome. Nat Struct Mol Biol. 2013;20(4):447–53.

    Article  CAS  Google Scholar 

  68. Abdelhak S, Kalatzis V, Heilig R, et al. A human homologue of the Drosophila eyes absent gene underlies branchio-oto-renal (BOR) syndrome and identifies a novel gene family. Nat Genet. 1997;15(2):157–64.

    Article  CAS  Google Scholar 

  69. Ruf RG, Xu PX, Silvius D, et al. SIX1 mutations cause branchio-oto-renal syndrome by disruption of EYA1-SIX1-DNA complexes. Proc Natl Acad Sci U S A. 2004;101(21):8090–5.

    Article  CAS  Google Scholar 

  70. Wu K, Liu M, Li A, et al. Cell fate determination factor DACH1 inhibits c-Jun-induced contact-independent growth. Mol Biol Cell. 2007;18(3):755–67.

    Article  CAS  Google Scholar 

  71. Ikeda K, Watanabe Y, Ohto H, et al. Molecular interaction and synergistic activation of a promoter by Six, Eya, and Dach proteins mediated through CREB binding protein. Mol Cell Biol. 2002;22(19):6759–66.

    Article  CAS  Google Scholar 

  72. Popov VM, Zhou J, Shirley LA, et al. The cell fate determination factor DACH1 is expressed in estrogen receptor-alpha-positive breast cancer and represses estrogen receptor-alpha signaling. Cancer Res. 2009;69(14):5752–60.

    Article  CAS  Google Scholar 

  73. Kim SS, Zhang RG, Braunstein SE, et al. Structure of the retinal determination protein Dachshund reveals a DNA binding motif. Structure. 2002;10(6):787–95.

    Article  CAS  Google Scholar 

  74. Zhou J, Liu Y, Zhang W, et al. Transcription elongation regulator 1 is a co-integrator of the cell fate determination factor Dachshund homolog 1. J Biol Chem. 2010;285(51):40342–50.

    Article  CAS  Google Scholar 

  75. Li X, Perissi V, Liu F, et al. Tissue-specific regulation of retinal and pituitary precursor cell proliferation. Science. 2002;297(5584):1180–3.

    Article  CAS  Google Scholar 

  76. Liu Y, Han N, Zhou S, et al. The DACH/EYA/SIX gene network and its role in tumor initiation and progression. Int J Cancer. 2016;138(5):1067–75.

    Article  CAS  Google Scholar 

  77. Wu W, Ren Z, Li P, et al. Six1: a critical transcription factor in tumorigenesis. Int J Cancer. 2015;136(6):1245–53.

    Article  CAS  Google Scholar 

  78. Anderson AM, Weasner BM, Weasner BP, et al. Dual transcriptional activities of SIX proteins define their roles in normal and ectopic eye development. Development. 2012;139(5):991–1000.

    Article  CAS  Google Scholar 

  79. Kawakami K, Ohto H, Ikeda K, et al. Structure, function and expression of a murine homeobox protein AREC3, a homologue of Drosophila sine oculis gene product, and implication in development. Nucleic Acids Res. 1996;24(2):303–10.

    Article  CAS  Google Scholar 

  80. Brodbeck S, Besenbeck B, Englert C. The transcription factor Six2 activates expression of the Gdnf gene as well as its own promoter. Mech Dev. 2004;121(10):1211–22.

    Article  CAS  Google Scholar 

  81. Rebay I, Silver SJ, Tootle TL. New vision from Eyes absent: transcription factors as enzymes. Trends Genet. 2005;21(3):163–71.

    Article  CAS  Google Scholar 

  82. Zou D, Silvius D, Fritzsch B, et al. Eya1 and Six1 are essential for early steps of sensory neurogenesis in mammalian cranial placodes. Development. 2004;131(22):5561–72.

    Article  CAS  Google Scholar 

  83. Liu Q, Li A, Tian Y, et al. The expression profile and clinic significance of the SIX family in non-small cell lung cancer. J Hematol Oncol. 2016;9(1):119.

    Article  Google Scholar 

  84. Zhou H, Zhang L, Vartuli RL, et al. The Eya phosphatase: Its unique role in cancer. Int J Biochem Cell Biol. 2018;96:165–70.

    Article  CAS  Google Scholar 

  85. Hegde RS, Roychoudhury K, Pandey RN. The multi-functional eyes absent proteins. Crit Rev Biochem Mol Biol. 2020;55(4):372–85.

    Article  CAS  Google Scholar 

  86. Cook PJ, Ju BG, Telese F, et al. Tyrosine dephosphorylation of H2AX modulates apoptosis and survival decisions. Nature. 2009;458(7238):591–6.

    Article  CAS  Google Scholar 

  87. Krishnan N, Jeong DG, Jung SK, et al. Dephosphorylation of the C-terminal tyrosyl residue of the DNA damage-related histone H2A.X is mediated by the protein phosphatase eyes absent. J Biol Chem. 2009;284(24):16066–70.

    Article  CAS  Google Scholar 

  88. Yuan B, Cheng L, Chiang HC, et al. A phosphotyrosine switch determines the antitumor activity of ERβ. J Clin Invest. 2014;124(8):3378–90.

    Article  CAS  Google Scholar 

  89. Wu K, Li Z, Cai S, et al. EYA1 phosphatase function is essential to drive breast cancer cell proliferation through cyclin D1. Cancer Res. 2013;73(14):4488–99.

    Article  CAS  Google Scholar 

  90. Okabe Y, Sano T, Nagata S. Regulation of the innate immune response by threonine-phosphatase of Eyes absent. Nature. 2009;460(7254):520–4.

    Article  CAS  Google Scholar 

  91. Soni UK, Roychoudhury K, Hegde RS. The Eyes Absent proteins in development and in developmental disorders. Biochem Soc Trans. 2021;49(3):1397–408.

    Article  CAS  Google Scholar 

  92. Wang Y, Pandey RN, Roychoudhury K, et al. Targeting EYA3 in Ewing Sarcoma Retards Tumor Growth and Angiogenesis. Mol Cancer Ther. 2021;20(5):803–15.

    Article  CAS  Google Scholar 

  93. Anantharajan J, Zhou H, Zhang L, et al. Structural and Functional Analyses of an Allosteric EYA2 Phosphatase Inhibitor That Has On-Target Effects in Human Lung Cancer Cells. Mol Cancer Ther. 2019;18(9):1484–96.

    Article  CAS  Google Scholar 

  94. Tavares ALP, Jourdeuil K, Neilson KM, et al. Sobp modulates the transcriptional activation of Six1 target genes and is required during craniofacial development. Development. 2021;148(17):dev199684.

    Article  CAS  Google Scholar 

  95. Laclef C, Hamard G, Demignon J, et al. Altered myogenesis in Six1-deficient mice. Development. 2003;130(10):2239–52.

    Article  CAS  Google Scholar 

  96. Blevins MA, Towers CG, Patrick AN, et al. The SIX1-EYA transcriptional complex as a therapeutic target in cancer. Expert Opin Ther Targets. 2015;19(2):213–25.

    Article  CAS  Google Scholar 

  97. Wu K, Jiao X, Li Z, et al. Cell fate determination factor Dachshund reprograms breast cancer stem cell function. J Biol Chem. 2011;286(3):2132–42.

    Article  CAS  Google Scholar 

  98. McCoy EL, Iwanaga R, Jedlicka P, et al. Six1 expands the mouse mammary epithelial stem/progenitor cell pool and induces mammary tumors that undergo epithelial-mesenchymal transition. J Clin Invest. 2009;119(9):2663–77.

    Article  CAS  Google Scholar 

  99. Miller SJ, Lan ZD, Hardiman A, et al. Inhibition of Eyes Absent Homolog 4 expression induces malignant peripheral nerve sheath tumor necrosis. Oncogene. 2010;29(3):368–79.

    Article  CAS  Google Scholar 

  100. Williams GH, Stoeber K. The cell cycle and cancer. J Pathol. 2012;226(2):352–64.

    Article  CAS  Google Scholar 

  101. Schulenburg A, Blatt K, Cerny-Reiterer S, et al. Cancer stem cells in basic science and in translational oncology: can we translate into clinical application? J Hematol Oncol. 2015;8:16.

    Article  Google Scholar 

  102. Liu QQ, Zhou YQ, Liu HQ, et al. Decreased DACH1 expression in glomerulopathy is associated with disease progression and severity. Oncotarget. 2016;7(52):86547–60.

    Article  Google Scholar 

  103. Wu K, Li A, Rao M, et al. DACH1 is a cell fate determination factor that inhibits cyclin D1 and breast tumor growth. Mol Cell Biol. 2006;26(19):7116–29.

    Article  CAS  Google Scholar 

  104. Nan F, Lü Q, Zhou J, et al. Altered expression of DACH1 and cyclin D1 in endometrial cancer. Cancer Biol Ther. 2009;8(16):1534–9.

    Article  CAS  Google Scholar 

  105. Liu Y, Zhou R, Yuan X, et al. DACH1 is a novel predictive and prognostic biomarker in hepatocellular carcinoma as a negative regulator of Wnt/β-catenin signaling. Oncotarget. 2015;6(11):8621–34.

    Article  Google Scholar 

  106. Lee JW, Kim HS, Kim S, et al. DACH1 regulates cell cycle progression of myeloid cells through the control of cyclin D, Cdk 4/6 and p21Cip1. Biochem Biophys Res Commun. 2012;420(1):91–5.

    Article  CAS  Google Scholar 

  107. Jiang D, Qiu T, Peng J, et al. YB-1 is a positive regulator of KLF5 transcription factor in basal-like breast cancer. Cell Death Differ. 2022;29(6):1283–95.

    Article  CAS  Google Scholar 

  108. Hsu JY, Danis EP, Nance S, et al. SIX1 reprograms myogenic transcription factors to maintain the rhabdomyosarcoma undifferentiated state. Cell Rep. 2022;38(5):110323.

    Article  CAS  Google Scholar 

  109. Coletta RD, Christensen K, Reichenberger KJ, et al. The Six1 homeoprotein stimulates tumorigenesis by reactivation of cyclin A1. Proc Natl Acad Sci U S A. 2004;101(17):6478–83.

    Article  CAS  Google Scholar 

  110. Towers CG, Guarnieri AL, Micalizzi DS, et al. The Six1 oncoprotein downregulates p53 via concomitant regulation of RPL26 and microRNA-27a-3p. Nat Commun. 2015;6:10077.

    Article  CAS  Google Scholar 

  111. Huang T, Song X, Xu D, et al. Stem cell programs in cancer initiation, progression, and therapy resistance. Theranostics. 2020;10(19):8721–43.

    Article  CAS  Google Scholar 

  112. Dong B, Li S, Zhu S, et al. MiRNA-mediated EMT and CSCs in cancer chemoresistance. Exp Hematol Oncol. 2021;10(1):12.

    Article  CAS  Google Scholar 

  113. Li MM, Yuan J, Guan XY, et al. Molecular subclassification of gastrointestinal cancers based on cancer stem cell traits. Exp Hematol Oncol. 2021;10(1):53.

    Article  CAS  Google Scholar 

  114. Xu H, Zhang Y, Peña MM, et al. Six1 promotes colorectal cancer growth and metastasis by stimulating angiogenesis and recruiting tumor-associated macrophages. Carcinogenesis. 2017;38(3):281–92.

    Article  CAS  Google Scholar 

  115. Lerbs T, Bisht S, Schölch S, et al. Inhibition of Six1 affects tumour invasion and the expression of cancer stem cell markers in pancreatic cancer. BMC Cancer. 2017;17(1):249.

    Article  Google Scholar 

  116. Pastushenko I, Blanpain C. EMT Transition States during Tumor Progression and Metastasis. Trends Cell Biol. 2019;29(3):212–26.

    Article  CAS  Google Scholar 

  117. Nieto MA, Huang RY, Jackson RA, et al. EMT: 2016. Cell. 2016;166(1):21–45.

    Article  CAS  Google Scholar 

  118. Fidler IJ. The pathogenesis of cancer metastasis: the “seed and soil” hypothesis revisited. Nat Rev Cancer. 2003;3(6):453–8.

    Article  CAS  Google Scholar 

  119. Liu Q, Zhang H, Jiang X, et al. Factors involved in cancer metastasis: a better understanding to “seed and soil” hypothesis. Mol Cancer. 2017;16(1):176.

    Article  Google Scholar 

  120. Xu J, Lamouille S, Derynck R. TGF-beta-induced epithelial to mesenchymal transition. Cell Res. 2009;19(2):156–72.

    Article  CAS  Google Scholar 

  121. Massagué J. TGFβ signalling in context. Nat Rev Mol Cell Biol. 2012;13(10):616–30.

    Article  Google Scholar 

  122. Lin YT, Wu KJ. Epigenetic regulation of epithelial-mesenchymal transition: focusing on hypoxia and TGF-β signaling. J Biomed Sci. 2020;27(1):39.

    Article  CAS  Google Scholar 

  123. Bu XN, Qiu C, Wang C, et al. Inhibition of DACH1 activity by short hairpin RNA represses cell proliferation and tumor invasion in pancreatic cancer. Oncol Rep. 2016;36(2):745–54.

    Article  CAS  Google Scholar 

  124. Kong D, Zhou H, Neelakantan D, et al. VEGF-C mediates tumor growth and metastasis through promoting EMT-epithelial breast cancer cell crosstalk. Oncogene. 2021;40(5):964–79.

    Article  CAS  Google Scholar 

  125. Min WP, Wei XF. Silencing SIX1 inhibits epithelial mesenchymal transition through regulating TGF-β/Smad2/3 signaling pathway in papillary thyroid carcinoma. Auris Nasus Larynx. 2021;48(3):487–95.

    Article  Google Scholar 

  126. Huang S, Lin W, Wang L, et al. SIX1 Predicts Poor Prognosis and Facilitates the Progression of Non-small Lung Cancer via Activating the Notch Signaling Pathway. J Cancer. 2022;13(2):527–40.

    Article  CAS  Google Scholar 

  127. Zeng J, Wei M, Shi R, et al. MiR-204-5p/Six1 feedback loop promotes epithelial-mesenchymal transition in breast cancer. Tumour Biol. 2016;37(2):2729–35.

    Article  CAS  Google Scholar 

  128. Micalizzi DS, Christensen KL, Jedlicka P, et al. The Six1 homeoprotein induces human mammary carcinoma cells to undergo epithelial-mesenchymal transition and metastasis in mice through increasing TGF-beta signaling. J Clin Invest. 2009;119(9):2678–90.

    Article  CAS  Google Scholar 

  129. Sun SH, Liu D, Deng YT, et al. SIX1 coordinates with TGFβ signals to induce epithelial-mesenchymal transition in cervical cancer. Oncol Lett. 2016;12(2):1271–8.

    Article  CAS  Google Scholar 

  130. Yu T, Song J, Zhou H, et al. Nuclear TEAD4 with SIX1 Overexpression is an Independent Prognostic Marker in the Stage I-III Colorectal Cancer. Cancer Manag Res. 2021;13:1581–9.

    Article  CAS  Google Scholar 

  131. Ono H, Imoto I, Kozaki K, et al. SIX1 promotes epithelial-mesenchymal transition in colorectal cancer through ZEB1 activation. Oncogene. 2012;31(47):4923–34.

    Article  CAS  Google Scholar 

  132. Kahlert C, Lerbs T, Pecqueux M, et al. Overexpression of SIX1 is an independent prognostic marker in stage I-III colorectal cancer. Int J Cancer. 2015;137(9):2104–13.

    Article  CAS  Google Scholar 

  133. Xie Y, Jin P, Sun X, et al. SIX1 is upregulated in gastric cancer and regulates proliferation and invasion by targeting the ERK pathway and promoting epithelial-mesenchymal transition. Cell Biochem Funct. 2018;36(8):413–9.

    Article  CAS  Google Scholar 

  134. Xu H, Pirisi L, Creek KE. Six1 overexpression at early stages of HPV16-mediated transformation of human keratinocytes promotes differentiation resistance and EMT. Virology. 2015;474:144–53.

    Article  CAS  Google Scholar 

  135. Smith AL, Iwanaga R, Drasin DJ, et al. The miR-106b-25 cluster targets Smad7, activates TGF-β signaling, and induces EMT and tumor initiating cell characteristics downstream of Six1 in human breast cancer. Oncogene. 2012;31(50):5162–71.

    Article  CAS  Google Scholar 

  136. Zhu Z, Rong Z, Luo Z, et al. Circular RNA circNHSL1 promotes gastric cancer progression through the miR-1306-3p/SIX1/vimentin axis. Mol Cancer. 2019;18(1):126.

    Article  Google Scholar 

  137. Kingsbury TJ, Kim M, Civin CI. Regulation of cancer stem cell properties by SIX1, a member of the PAX-SIX-EYA-DACH network. Adv Cancer Res. 2019;141:1–42.

    Article  CAS  Google Scholar 

  138. Vartuli RL, Zhou H, Zhang L, et al. Eya3 promotes breast tumor-associated immune suppression via threonine phosphatase-mediated PD-L1 upregulation. J Clin Invest. 2018;128(6):2535–50.

    Article  Google Scholar 

  139. Popov VM, Wu K, Zhou J, et al. The Dachshund gene in development and hormone-responsive tumorigenesis. Trends Endocrinol Metab. 2010;21(1):41–9.

    Article  CAS  Google Scholar 

  140. Zhang Z, Tang H, Wang Z, et al. MiR-185 targets the DNA methyltransferases 1 and regulates global DNA methylation in human glioma. Mol Cancer. 2011;10:124.

    Article  CAS  Google Scholar 

  141. Zheng Y, Zeng Y, Qiu R, et al. The Homeotic Protein SIX3 Suppresses Carcinogenesis and Metastasis through Recruiting the LSD1/NuRD(MTA3) Complex. Theranostics. 2018;8(4):972–89.

    Article  CAS  Google Scholar 

  142. Mo ML, Okamoto J, Chen Z, et al. Down-regulation of SIX3 is associated with clinical outcome in lung adenocarcinoma. PLoS ONE. 2013;8(8):e71816.

    Article  CAS  Google Scholar 

  143. Rajkumar T, Vijayalakshmi N, Gopal G, et al. Identification and validation of genes involved in gastric tumorigenesis. Cancer Cell Int. 2010;10:45.

    Article  Google Scholar 

  144. Mo SJ, Liu X, Hao XY, et al. EYA4 functions as tumor suppressor gene and prognostic marker in pancreatic ductal adenocarcinoma through β-catenin/ID2 pathway. Cancer Lett. 2016;380(2):403–12.

    Article  CAS  Google Scholar 

  145. Mo SJ, Hou X, Hao XY, et al. EYA4 inhibits hepatocellular carcinoma growth and invasion by suppressing NF-κB-dependent RAP1 transactivation. Cancer Commun (Lond). 2018;38(1):9.

    Article  Google Scholar 

  146. Hao XY, Cai JP, Liu X, et al. EYA4 gene functions as a prognostic marker and inhibits the growth of intrahepatic cholangiocarcinoma. Chin J Cancer. 2016;35(1):70.

    Article  Google Scholar 

  147. Chu Y, Chen Y, Li M, et al. Six1 regulates leukemia stem cell maintenance in acute myeloid leukemia. Cancer Sci. 2019;110(7):2200–10.

    Article  CAS  Google Scholar 

  148. Liu D, Li L, Zhang XX, et al. SIX1 promotes tumor lymphangiogenesis by coordinating TGFβ signals that increase expression of VEGF-C. Cancer Res. 2014;74(19):5597–607.

    Article  CAS  Google Scholar 

  149. Du Z, Brewster R, Merrill PH, et al. Meningioma transcription factors link cell lineage with systemic metabolic cues. Neuro Oncol. 2018;20(10):1331–43.

    Article  CAS  Google Scholar 

  150. Drawz P, Rahman M. Chronic kidney disease. Ann Intern Med. 2015;162(11):Itc1-16.

    Article  Google Scholar 

  151. Kovesdy CP. Epidemiology of chronic kidney disease: an update 2022. Kidney Int Suppl (2011). 2022;12(1):7–11.

    Article  Google Scholar 

  152. Doke T, Huang S, Qiu C, et al. Transcriptome-wide association analysis identifies DACH1 as a kidney disease risk gene that contributes to fibrosis. J Clin Invest. 2021;131(10):e141801.

    Article  CAS  Google Scholar 

  153. Cao A, Li J, Asadi M, et al. DACH1 protects podocytes from experimental diabetic injury and modulates PTIP-H3K4Me3 activity. J Clin Invest. 2021;131(10):e141279.

    Article  CAS  Google Scholar 

  154. Merscher S, Faul C. DACH1 as a multifaceted and potentially druggable susceptibility factor for kidney disease. J Clin Invest. 2021;131(10):e149043.

    Article  CAS  Google Scholar 

  155. Davis RJ, Shen W, Sandler YI, et al. Dach1 mutant mice bear no gross abnormalities in eye, limb, and brain development and exhibit postnatal lethality. Mol Cell Biol. 2001;21(5):1484–90.

    Article  CAS  Google Scholar 

  156. Wang M. DACH1 protects podocytes and tubules from injury. Nat Rev Nephrol. 2021;17(8):511.

    Article  CAS  Google Scholar 

  157. Xu J, Li J, Ramakrishnan A, et al. Six1 and Six2 of the Sine Oculis Homeobox Subfamily are Not Functionally Interchangeable in Mouse Nephron Formation. Front Cell Dev Biol. 2022;10:815249.

    Article  Google Scholar 

  158. Castiglioni V, Faedo A, Onorati M, et al. Dynamic and Cell-Specific DACH1 Expression in Human Neocortical and Striatal Development. Cereb Cortex. 2019;29(5):2115–24.

    Article  Google Scholar 

  159. Brouwer RM, Klein M, Grasby KL, et al. Genetic variants associated with longitudinal changes in brain structure across the lifespan. Nat Neurosci. 2022;25(4):421–32.

    Article  CAS  Google Scholar 

  160. Graus F, Vogrig A, Muñiz-Castrillo S, et al. Updated Diagnostic Criteria for Paraneoplastic Neurologic Syndromes. Neurol Neuroimmunol Neuroinflamm. 2021;8(4):e1014.

    Article  Google Scholar 

  161. Zekeridou A, Yang B, Lennon VA, et al. Anti-Neuronal Nuclear Antibody 3 Autoimmunity Targets Dachshund Homolog 1. Ann Neurol. 2022;91(5):670–5.

    Article  CAS  Google Scholar 

  162. Ozcan L, Ghorpade DS, Zheng Z, et al. Hepatocyte DACH1 Is Increased in Obesity via Nuclear Exclusion of HDAC4 and Promotes Hepatic Insulin Resistance. Cell Rep. 2016;15(10):2214–25.

    Article  CAS  Google Scholar 

  163. Zheng Z, Nayak L, Wang W, et al. An ATF6-tPA pathway in hepatocytes contributes to systemic fibrinolysis and is repressed by DACH1. Blood. 2019;133(7):743–53.

    Article  CAS  Google Scholar 

  164. Zheng Z, Nakamura K, Gershbaum S, et al. Interacting hepatic PAI-1/tPA gene regulatory pathways influence impaired fibrinolysis severity in obesity. J Clin Invest. 2020;130(8):4348–59.

    CAS  Google Scholar 

  165. Biesecker LG. Polydactyly: how many disorders and how many genes? 2010 update. Dev Dyn. 2011;240(5):931–42.

    Article  Google Scholar 

  166. Umair M, Palander O, Bilal M, et al. Biallelic variant in DACH1, encoding Dachshund Homolog 1, defines a novel candidate locus for recessive postaxial polydactyly type A. Genomics. 2021;113(4):2495–502.

    Article  CAS  Google Scholar 

  167. Xu J, Li J, Zhang T, et al. Chromatin remodelers and lineage-specific factors interact to target enhancers to establish proneurosensory fate within otic ectoderm. Proc Natl Acad Sci U S A. 2021;118(12):e2025196118.

    Article  CAS  Google Scholar 

  168. Bajard L, Relaix F, Lagha M, et al. A novel genetic hierarchy functions during hypaxial myogenesis: Pax3 directly activates Myf5 in muscle progenitor cells in the limb. Genes Dev. 2006;20(17):2450–64.

    Article  CAS  Google Scholar 

  169. Nord H, Nygård Skalman L, von Hofsten J. Six1 regulates proliferation of Pax7-positive muscle progenitors in zebrafish. J Cell Sci. 2013;126(Pt 8):1868–80.

    CAS  Google Scholar 

  170. Grifone R, Demignon J, Houbron C, et al. Six1 and Six4 homeoproteins are required for Pax3 and Mrf expression during myogenesis in the mouse embryo. Development. 2005;132(9):2235–49.

    Article  CAS  Google Scholar 

  171. Grifone R, Demignon J, Giordani J, et al. Eya1 and Eya2 proteins are required for hypaxial somitic myogenesis in the mouse embryo. Dev Biol. 2007;302(2):602–16.

    Article  CAS  Google Scholar 

  172. Lee EJ, Kim M, Kim YD, et al. Establishment of stably expandable induced myogenic stem cells by four transcription factors. Cell Death Dis. 2018;9(11):1092.

    Article  Google Scholar 

  173. Papi A, Brightling C, Pedersen SE, et al. Asthma. Lancet. 2018;391(10122):783–800.

    Article  Google Scholar 

  174. Yang ZC, Qu ZH, Yi MJ, et al. MiR-448-5p inhibits TGF-β1-induced epithelial-mesenchymal transition and pulmonary fibrosis by targeting Six1 in asthma. J Cell Physiol. 2019;234(6):8804–14.

    Article  CAS  Google Scholar 

  175. Wang QF, Wu G, Mi S, et al. MLL fusion proteins preferentially regulate a subset of wild-type MLL target genes in the leukemic genome. Blood. 2011;117(25):6895–905.

    Article  CAS  Google Scholar 

  176. Cildir G, Low KC, Tergaonkar V. Noncanonical NF-κB Signaling in Health and Disease. Trends Mol Med. 2016;22(5):414–29.

    Article  CAS  Google Scholar 

  177. Sun SC. The non-canonical NF-κB pathway in immunity and inflammation. Nat Rev Immunol. 2017;17(9):545–58.

    Article  CAS  Google Scholar 

  178. Liu Z, Mar KB, Hanners NW, et al. A NIK-SIX signalling axis controls inflammation by targeted silencing of non-canonical NF-κB. Nature. 2019;568(7751):249–53.

    Article  CAS  Google Scholar 

  179. Radha G, Raghavan SC. BCL2: A promising cancer therapeutic target. Biochim Biophys Acta Rev Cancer. 2017;1868(1):309–14.

    Article  CAS  Google Scholar 

  180. Xu Y, Ye H. Progress in understanding the mechanisms of resistance to BCL-2 inhibitors. Exp Hematol Oncol. 2022;11(1):31.

    Article  CAS  Google Scholar 

  181. Wang H, Guo H, Yang J, et al. Bruton tyrosine kinase inhibitors in B-cell lymphoma: beyond the antitumour effect. Exp Hematol Oncol. 2022;11(1):60.

    Article  CAS  Google Scholar 

  182. Cho H, Jang JE, Eom JI, et al. Arsenic trioxide synergistically promotes the antileukaemic activity of venetoclax by downregulating Mcl-1 in acute myeloid leukaemia cells. Exp Hematol Oncol. 2021;10(1):28.

    Article  CAS  Google Scholar 

  183. Morris LG, Chan TA. Therapeutic targeting of tumor suppressor genes. Cancer. 2015;121(9):1357–68.

    Article  CAS  Google Scholar 

  184. Okabe S, Tanaka Y, Gotoh A. Targeting phosphoinositide 3-kinases and histone deacetylases in multiple myeloma. Exp Hematol Oncol. 2021;10(1):19.

    Article  CAS  Google Scholar 

  185. Li J, Guo M, Chen L, et al. p53 amyloid aggregation in cancer: function, mechanism, and therapy. Exp Hematol Oncol. 2022;11(1):66.

    Article  Google Scholar 

  186. Wu K, Katiyar S, Witkiewicz A, et al. The cell fate determination factor dachshund inhibits androgen receptor signaling and prostate cancer cellular growth. Cancer Res. 2009;69(8):3347–55.

    Article  CAS  Google Scholar 

  187. Dong B, Yi M, Luo S, et al. RDGN-based predictive model for the prognosis of breast cancer. Exp Hematol Oncol. 2020;9:13.

    Article  CAS  Google Scholar 

  188. Hassan M, Zahid S, Shahzadi S, et al. Mechanistic insight of DACH1 receptor in the development of carcinoma insurgence through MD simulation studies. J Biomol Struct Dyn. 2022;40(2):742–51.

    Article  CAS  Google Scholar 

  189. Powe DG, Dhondalay GK, Lemetre C, et al. DACH1: its role as a classifier of long term good prognosis in luminal breast cancer. PLoS ONE. 2014;9(1):e84428.

    Article  Google Scholar 

  190. Uehiro N, Sato F, Pu F, et al. Circulating cell-free DNA-based epigenetic assay can detect early breast cancer. Breast Cancer Res. 2016;18(1):129.

    Article  Google Scholar 

  191. Zhang Q, Yuan Y, Cui J, et al. MiR-217 Promotes Tumor Proliferation in Breast Cancer via Targeting DACH1. J Cancer. 2015;6(2):184–91.

    Article  Google Scholar 

  192. Yu J, Jiang P, Zhao K, et al. Role of DACH1 on Proliferation, Invasion, and Apoptosis in Human Lung Adenocarcinoma Cells. Curr Mol Med. 2021;21(9):806–11.

    Article  CAS  Google Scholar 

  193. Cifarelli V, Lashinger LM, Devlin KL, et al. Metformin and Rapamycin Reduce Pancreatic Cancer Growth in Obese Prediabetic Mice by Distinct MicroRNA-Regulated Mechanisms. Diabetes. 2015;64(5):1632–42.

    Article  CAS  Google Scholar 

  194. Wu L, Oshiro-Rapley N, et al. An Ancient, Unified Mechanism for Metformin Growth Inhibition in C. elegans and Cancer. Cell. 2016;167(7):1705-18.e13.

    Article  CAS  Google Scholar 

  195. Zhou H, Blevins MA, Hsu JY, et al. Identification of a Small-Molecule Inhibitor That Disrupts the SIX1/EYA2 Complex, EMT, and Metastasis. Cancer Res. 2020;80(12):2689–702.

    Article  CAS  Google Scholar 

  196. Liao Y, Liu Y, Shao Z, et al. A new role of GRP75-USP1-SIX1 protein complex in driving prostate cancer progression and castration resistance. Oncogene. 2021;40(25):4291–306.

    Article  CAS  Google Scholar 

  197. Li L, Liang Y, Kang L, et al. Transcriptional Regulation of the Warburg Effect in Cancer by SIX1. Cancer Cell. 2018;33(3):368-85.e7.

    Article  CAS  Google Scholar 

  198. Wang Z, Yang Y, Hu S, et al. Short-form RON (sf-RON) enhances glucose metabolism to promote cell proliferation via activating β-catenin/SIX1 signaling pathway in gastric cancer. Cell Biol Toxicol. 2021;37(1):35–49.

    Article  Google Scholar 

  199. Liu Y, Kong WY, Yu CF, et al. SNS-023 sensitizes hepatocellular carcinoma to sorafenib by inducing degradation of cancer drivers SIX1 and RPS16. Acta Pharmacol Sin. 2022. https://doi.org/10.1038/s41401-022-01003-4.

    Article  Google Scholar 

  200. Stavropoulou V, Kaspar S, Brault L, et al. MLL-AF9 Expression in Hematopoietic Stem Cells Drives a Highly Invasive AML Expressing EMT-Related Genes Linked to Poor Outcome. Cancer Cell. 2016;30(1):43–58.

    Article  CAS  Google Scholar 

  201. Monteiro AC, Muenzner JK, Andrade F, et al. Gene expression and promoter methylation of angiogenic and lymphangiogenic factors as prognostic markers in melanoma. Mol Oncol. 2019;13(6):1433–49.

    Article  CAS  Google Scholar 

  202. Liu W, Gao M, Li L, et al. Homeoprotein SIX1 compromises antitumor immunity through TGF-β-mediated regulation of collagens. Cell Mol Immunol. 2021;18(12):2660–72.

    Article  CAS  Google Scholar 

  203. Shen JZ, Qiu Z, Wu Q, et al. A FBXO7/EYA2-SCF(FBXW7) axis promotes AXL-mediated maintenance of mesenchymal and immune evasion phenotypes of cancer cells. Mol Cell. 2022;82(6):1123-39.e8.

    Article  CAS  Google Scholar 

  204. Liang Y, Xu X, Wang T, et al. The EGFR/miR-338-3p/EYA2 axis controls breast tumor growth and lung metastasis. Cell Death Dis. 2017;8(7): e2928.

    Article  CAS  Google Scholar 

  205. Liu ZK, Li C, Zhang RY, et al. EYA2 suppresses the progression of hepatocellular carcinoma via SOCS3-mediated blockade of JAK/STAT signaling. Mol Cancer. 2021;20(1):79.

    Article  CAS  Google Scholar 

  206. Luo M, Li Y, Shi X, et al. Aberrant methylation of EYA4 promotes epithelial-mesenchymal transition in esophageal squamous cell carcinoma. Cancer Sci. 2018;109(6):1811–24.

    Article  CAS  Google Scholar 

  207. Krueger AB, Drasin DJ, Lea WA, et al. Allosteric inhibitors of the Eya2 phosphatase are selective and inhibit Eya2-mediated cell migration. J Biol Chem. 2014;289(23):16349–61.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (No.81874120, 82073370).

Author information

Authors and Affiliations

Authors

Contributions

SZ drafted the manuscript and prepared the figures. WL, HZ and YY collected the related references and participated in the discussion. QM and KW designed this review and revised the manuscript. All authors contributed to this manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Qi Mei or Kongming Wu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhu, S., Li, W., Zhang, H. et al. Retinal determination gene networks: from biological functions to therapeutic strategies. Biomark Res 11, 18 (2023). https://doi.org/10.1186/s40364-023-00459-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40364-023-00459-8

Keywords