Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 4152 2023-12-12 16:10:42 |
2 format change Meta information modification 4152 2023-12-13 02:56:44 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Kimura, K.; Jackson, T.L.B.; Huang, R.C.C. Interaction and Collaboration of SP1, HIF-1, and MYC. Encyclopedia. Available online: https://encyclopedia.pub/entry/52626 (accessed on 09 July 2024).
Kimura K, Jackson TLB, Huang RCC. Interaction and Collaboration of SP1, HIF-1, and MYC. Encyclopedia. Available at: https://encyclopedia.pub/entry/52626. Accessed July 09, 2024.
Kimura, Kotohiko, Tiffany L. B. Jackson, Ru Chih C. Huang. "Interaction and Collaboration of SP1, HIF-1, and MYC" Encyclopedia, https://encyclopedia.pub/entry/52626 (accessed July 09, 2024).
Kimura, K., Jackson, T.L.B., & Huang, R.C.C. (2023, December 12). Interaction and Collaboration of SP1, HIF-1, and MYC. In Encyclopedia. https://encyclopedia.pub/entry/52626
Kimura, Kotohiko, et al. "Interaction and Collaboration of SP1, HIF-1, and MYC." Encyclopedia. Web. 12 December, 2023.
Interaction and Collaboration of SP1, HIF-1, and MYC
Edit

Specificity protein 1 (SP1), hypoxia-inducible factor 1 (HIF-1), and MYC are important transcription factors (TFs). SP1, a constitutively expressed housekeeping gene, regulates diverse yet distinct biological activities; MYC is a master regulator of all key cellular activities including cell metabolism and proliferation; and HIF-1, whose protein level is rapidly increased when the local tissue oxygen concentration decreases, functions as a mediator of hypoxic signals. Systems analyses of the regulatory networks in cancer have shown that SP1, HIF-1, and MYC belong to a group of TFs that function as master regulators of cancer. Therefore, the contributions of these TFs are crucial to the development of cancer. SP1, HIF-1, and MYC are often overexpressed in tumors, which indicates the importance of their roles in the development of cancer. Thus, proper manipulation of SP1, HIF-1, and MYC by appropriate agents could have a strong negative impact on cancer development. Under these circumstances, these TFs have naturally become major targets for anticancer drug development. Accordingly, there are many SP1 or HIF-1 inhibitors available; however, designing efficient MYC inhibitors has been extremely difficult. Studies have shown that SP1, HIF-1, and MYC modulate the expression of each other and collaborate to regulate the expression of numerous genes.

hypoxia-inducible factor 1 SP1 MYC cancer

1. Introduction: Specificity Protein 1, Hypoxia-Inducible Factor-1, and MYC as Master Regulators of Cancer

Recent progress in systems biology has shown that several specific factors are participants of a network that function as master regulators of cancer [1][2][3]. Wilson and Volker Filipp investigated complementary omics in human cancer, and discovered a close teamwork of transcriptional and epigenomic machinery, which is tightly connected and comprises histone lysine demethylase 3A, basic helix-loop-helix factors, MYC, hypoxia-inducible factor 1 alpha (HIF1A), and sterol regulatory element-binding transcription factor 1, as well as differentiation factors such as activator protein 1, myogenic differentiation 1, specificity protein 1 (SP1), Meis homeobox 1, zinc finger E-box-binding homeobox 1, and ETS like-1 protein (ETS1) [1]. Cao et al. [2] showed that 10 long non-coding RNA (lncRNA)-transcription factor (TF) pairs including four glycolysis-related lncRNAs (FTX, long intergenic non-protein coding RNA 472, proteasome 20S subunit alpha 3 antisense RNA 1, and small nucleolar RNA host gene 14) and six TFs (forkhead box protein P1, SP1, MYC, FOX-M1, hypoxia-inducible factor 1 alpha [HIF1A], and FOS) are involved in the progression of human lung adenocarcinoma. Malik et al. [3] discovered, using a statistical method called CoMEx (Combined score of DNA Methylation and Expression) to assess differentially expressed and methylated genes/microRNAs (miRNAs) between human seminoma and normal tissues, two hub miRNAs (miR-182-5p and miR-338-3p), five hub TFs (ETS1, HIF1A, hepatocyte nuclear factor-1 alpha, MYC, and SP1), and three hub genes (cadherin 1, C-X-C chemokine receptor type 4, and Snail family transcriptional repressor 1) in the seminoma-specific regulatory network. Interestingly, in all of these studies, three TFs, namely SP1, HIF1A, and MYC, were among the factors that participated in the cancer regulatory network. In addition, many studies have shown that SP1, HIF1A, and MYC are often upregulated in cancer [4][5][6][7][8][9]. Together, these data suggest that SP1, HIF1A, and MYC have crucial roles in cancer development, and that interfering with their activity could negatively impact cancer development and progressions. For this reason, enormous efforts have been undertaken to develop inhibitors for SP1, HIF1A, and MYC. Accordingly, numerous inhibitors of SP1 or HIF1A have been developed [10][11][12][13][14]; however, designing MYC inhibitors has been extremely difficult [15].

2. What Are SP1, HIF-1, and MYC, and How Do These TFs Benefit Cancer

2.1. SP1: Housekeeping Gene That Regulates Biological Activities

SP1 is a ubiquitous TF from the Sp/Krüppel-like family (KLF) of TFs, which are the major forms of zinc finger DNA-binding proteins [16]. The defining feature of SP1-like/KLF proteins is a highly conserved DNA-binding domain (>65% sequence identity among family members) at the C-terminus that has three tandem Cys2His2 (C2H2) zinc finger motifs [17]. Likewise, SP1 contains three highly homologous C2H2 regions [18][19], which exhibit direct binding to DNA at the C-terminal regions of the protein, thus enhancing gene transcription [20]. By contrast, the N-terminal regions of the proteins are more divergent [21]. SP1 has four unstructured domains A, B, C, and D, starting from the C-terminal region of the protein. The two main transactivating domains of SP1 are A and B, which are capable of direct interaction with the components of transcription machinery such as TATA-binding protein (TBP) and TBP-associated factor 4 [22]. The C domain is not indispensable but is highly charged and supports DNA binding and transactivation. The D domain, also known as the C-terminal region of SP1, has multimeric domains and is responsible for the binding of consensus sequences such as 5′-(G/T) GGGCGG(G/A)(G/A)(G/T)-3′ (the sequences are referred to as the GC box) [23]. The N-terminal region of SP1 is a small inhibitory domain, which mainly regulates the functions of domains A and B and is linked to the A domain with a serine/threonine-rich region [22]. The transcriptional activity and stability of SP1 are influenced by its post-translational modifications. SP1 undergoes acetylation, SUMOylation, ubiquitination, and glycosylation after its translation [24][25]. Acetylation of SP1 takes place in the DNA-binding domain [26]. Glycosylation occurs at the O-GlcNAc linkages at the serine and threonine residues in SP1, which can either induce or suppress DNA binding and transcription [27]. SUMOylation, occurring in the Lys16 region, controls the transcription of SP1 by instigating alterations in the chromatin structure, making the DNA inaccessible for transcription [28]. The proteasomal degradation of SP1 is carried out by ubiquitination, where the β-transducin repeat-containing protein (TCRP) ubiquitin ligase complex interacts with SP1 through the DSG (Asp-Ser-Gly) destruction box (β-TCRP binding motif) within the C-terminus of SP1 [29]. SP1 is critical for early embryonic development [30][31], but its expression decreases with age and there is evidence that the transformation of normal cells to cancer cells is associated with the upregulation of SP1, SP3, and SP4 [10][32]. Functional studies have demonstrated that the SP-like family of TFs regulates various genes responsible for cancer-related cellular mechanisms; SP1, SP3, and SP4 are also non-oncogene addiction (NOA) genes and thus are important drug targets [33]. NOA genes are essential for supporting the stress-burdened phenotype of tumors and thus are vital for their survival. The most important functional role of SP1 in normal cells is the regulation of cell cycle and cellular reprogramming [5]. Since cell proliferation and differentiation are the most active during the developmental stage of organisms, SP1 plays critical roles during early developmental stages perhaps for this reason [30][31]. This also indicates that SP1 is still an essential component of cellular mechanisms during adulthood although less so compared with during developmental stages.

2.2. HIF-1: Functions as a Mediator of Hypoxic Signals

HIF-1 is the most important factor involved in the cellular response to hypoxia [34][35]. The broad impact of HIF-1 on cell biology is reflected in the total number of hypoxic target genes, which is estimated to be approximately 1–2% of all human genes [36]. HIF-1 plays important roles in energy metabolism and angiogenesis, especially in cancer progression [34]. It is composed of two subunits, HIF1A and HIF1B (aryl hydrocarbon receptor nuclear translocator). Among these two subunits, only HIF1A is activated under hypoxia and HIF1B is not regulated by oxygen [35]. The dual functional protein apurinic/apyrimidinic endonuclease 1 is an enzyme in DNA base excision repair but also works as a redox factor to maintain HIF1A in the reduced state that is necessary for its transcriptional function [35]. In the presence of oxygen, prolyl hydroxylase hydroxylates HIF1A and hydroxylated HIF1A binds to the tumor suppressor von Hippel–Lindau protein (pVHL), a component of the E3 ubiquitin ligase complex. This interaction causes HIF1A to become ubiquitinated and targeted to the proteasome, where it is degraded. However, under hypoxia, HIF1A is not degraded by the proteasome since prolyl hydroxylase is not functional, so HIF1A dimerizes with HIF1B and binds to the hypoxia response element (HRE) in the promoters of target genes, initiating the expression of genes that promote adaptation to hypoxia [35]. HIF1A as well as the more cell-specific HIF2A are important regulators of the hypoxic response. Although both HIF1A and HIF2A are highly conserved at the protein level, share a similar domain structure, heterodimerize with HIF1B (HIF-2 is formed by the assembly of HIF2A and HIF1B), and bind to the same DNA sequence (the HRE), their effects on the expression of various genes differ [37].

2.3. SP1 and HIF-1

The importance of HIF-1 and SP1 in cancer development is beyond dispute. In fact, it has been shown that both HIF-1 and SP1 are involved in every aspect of cancer-related cellular mechanisms. For instance, both SP1 and HIF-1 play important roles in the regulation of cancer metabolism in carbohydrates [34][38][39][40][41] and lipids [42][43][44]. Both are involved in anticancer immunity via regulation of immune-related cells [45][46][47][48][49][50][51]; the tumor microenvironment (TME)/oncometabolites [52][53][54][55][56][57][58]; and transforming growth factor beta, which regulates the immune system [59][60][61][62][63][64]. SP1 promotes tumor angiogenesis via activation of vascular endothelial growth factor (VEGF), epidermal growth factor receptor (EGFR), and VEGF receptor 3 (VEGFR3) [65][66][67], whereas HIF-1 is a master regulator of angiogenesis, participating in vasculature formation by synergistic correlations with other proangiogenic factors such as VEGF, placental growth factor, and angiopoietins [68]. In addition, SP1 plays an important role in each of the crucial events of metastasis, namely, adhesion, invasion, migration, and angiogenesis [65][66][67][69][70][71]. Both SP1 and HIF-1 are also involved in the regulation of cellular stress mechanisms as mediators of the protection of cancer cells against various stresses [72][73][74].

2.4. MYC: A Master Regulator of Cellular Activity

MYC is a transcription factor that belongs to the basic helix-loop-helix-leucine zipper (bHLHZip) family and regulates cell growth, differentiation, metabolism, and cell death. Thus, MYC functions as a master regulator of major cellular functions [75][76][77][78]. Studies using knockout mice have shown that MYC is particularly important for cell growth (accumulation of the body mass) and is indispensable during the period of both embryogenesis and adulthood [79]. c-MYC is the prototype member of the MYC family, which also includes N-MYC and L-MYC proteins in mammalian cells. All three members of the MYC family are highly homologous but distributed differently. c-MYC is ubiquitous and highly abundant in proliferating cells, whereas N-MYC and L-MYC display more restricted expression at distinct stages of cell and tissue development. MYC proteins exist within the MYC/MAX/MXD network. To fold and become transcriptionally active, c-MYC must first heterodimerize with MAX, a process governed by the coiling of their bHLHZip domains. Once dimerized, the c-MYC/MAX complex acts as a master transcriptional regulator by binding via its basic region to the specific DNA consensus sequence 5′-CANNTG-3′. Due to the multifunctional activities of MYC in cellular functions, cancers with MYC activation elicit many of the important hallmarks essential for autonomous neoplastic growth. In fact, MYC aberrations or upregulation of MYC-related pathways occur in many cancers. In preclinical animal models, MYC inactivation can result in sustained tumor regression, a phenomenon that has been attributed to oncogene addiction [80].

3. Interactions among SP1, HIF-1, and MYC with One Another and Other TFs

3.1. Modulation of SP1, HIF-1, and MYC Activities

SP1, HIF-1, and MYC modulate the expression of numerous genes as major TFs. However, these TFs do not work independently and are in fact under the regulation of many other cellular components. For example, SP1, HIF-1, and MYC can interact and modulate the activities of each other. Figure 1 shows the promoters of human SP1, HIF1A, and MYC genes [81][82][83][84][85][86][87][88]. ‘SP1′ and ‘HRE’ in the figure indicate the locations of SP1 and the HRE consensus sequences, respectively. The SP1 consensus sequences are usually the GC boxes, whereas the HIF-1 consensus sequences (of the HRE) usually contain the nucleotide residues ‘5′-RCGTG-3′. The SP1 promoter contains numerous SP1 consensus sequences as well as NF-Y and E2F consensus sequences. SP1 binds to NF-Y and E2F consensus sequences as well as SP1 consensus sequences in the SP1 promoter [82][83]. These data suggest that SP1 can autoregulate its transcriptional activity. In addition to these consensus sequences, there is an HRE in the SP1 promoter (Figure 1) [81] to which HIF-1 binds and stimulates the transcriptional activity of the SP1 promoter [81]. It has been shown that the mRNA and protein levels of SP1 are decreased by silencing HIF1A in human cultured esophageal squamous cell carcinoma cells, whereas overexpression of HIF1A significantly increases these levels [81]. These data indicate that HIF-1 upregulates SP1 through its binding to the HRE.
Figure 1. The promoter structures of human SP1, HIF1A, and MYC genes. The consensus sequences and their potential binding proteins are shown in each promoter. SP1 binds to SP1 consensus sequences (GC box) as well as NF-Y and E2F consensus sequences. HIF-1 binds to the HRE. MYC, similarly to SP1, binds to E2F. Myc-associated zinc finger protein (MAZ) is an important regulatory protein associated with MYC gene expression and binds to MAZ consensus sequences [86][88]. The nucleotide numbers are numbered from the transcription start site (TSS). The TSS for MYC gene promoter is for the P1 promoter [86][88].

3.2. Effect of HIF-1 on SP1 Gene Expression and Vice Versa

Expression of the SP1 gene can be upregulated by HIF-1 transcriptionally by the binding of HIF-1 to its consensus sequences in the SP1 gene promoter [81]. This is shown schematically in Figure 2A. Meanwhile, Figure 2B–D schematically shows how HIF1A expression is regulated by SP1, using several examples. Insulin increases HIF1A promoter activity by reactive oxygen species (ROS) via SP1 in murine 3T3-L1 preadipocytes [89]. HIF1A transcription is downregulated by protein arginine methyltransferase 1 (PRMT1), a protein whose transcription is regulated by SP1 in human Hela cervical carcinoma and human HEK293T embryonic kidney cells [90]. In the former example, SP1 is activated by phosphoinositide 3-kinase/protein kinase C via ROS, and then induces HIF1A transcription (Figure 2B). In the latter example, the suppression of PRMT1, which prevents the recruitment of SP1/SP3 to the HIF1A gene promoter, allows SP1/SP3 to activate the transcription of HIF1A (Figure 2C). In both cases, SP1 directly induces transcriptional activity of the HIF1A gene via its binding to SP1 consensus sequences in the HIF1A gene promoter (Figure 1) and upregulates expression of the HIF1A gene [77][78][79][80][81][82][83][84][85][91][92][93][94]. Meanwhile, SP1 can indirectly regulate HIF1A expression by modulating the gene expression of histone deacetylase 4 (HDAC4) in rat cardiomyocytes (Figure 2D) [95]. SP1 upregulates the activity of the HDAC4 gene promoter, thereby promoting deacetylation and impairing the secretion of high mobility group box 1 in mouse intestinal epithelial cells [96]. Likewise, HDAC4 can prevent the acetylation of HIF1A, thereby stabilizing the protein in human pVHL-null kidney cancer cell lines [97]. In this way, SP1 upregulates HIF1A expression either directly by activating HIF1A gene expression via binding to the HIF1A gene promoter (Figure 2B,C) or indirectly by stabilizing HIF1A protein via modulation of HDAC4 gene expression (Figure 2D). Either way, SP1 increases the activity of HIF1A. Unlike the HIF1A gene, the HIF1B gene is constitutively expressed [98]. Therefore, the activity of HIF-1, which is composed of HIF1A and HIF1B, is regulated by adjusting the mRNA and protein levels of HIF1A in cells as well as by modulating the levels of co-activators for HIF-1 [37].
Figure 2. Schematic of the mechanism of activation of SP1 by HIF1A and that of HIF1A by SP1. (A) The mechanism of SP1 activation by HIF1A. (BD) The mechanism of HIF1A activation by SP1. While HIF1A regulates SP1 expression via binding to the promoter of a SP1 gene, SP1 can regulate HIF1A expression at both the mRNA level (B,C) and protein level (D). PKC: protein kinase C.

3.3. Effects of HIF-1 Compared to the Effects of SP1 on MYC Gene Activities

While HIF-1 induces SP1 gene expression, it inhibits the activity of MYC (without affecting MYC gene expression) [81][99][100][101]. Since activation of MYC is usually associated with cell growth, MYC activities must be suppressed under hypoxia, which is a condition unsuitable for rapid cell growth due to a lack of oxygen, which is required for efficient biological energy production. Thus, under hypoxia, MYC activity is inhibited by HIF1A as an adaptive response that promotes cell survival under low oxygen conditions. Since there is no HRE in the MYC gene promoter (Figure 1), HIF1A is unlikely to inhibit transcription of the MYC gene by directly binding to the MYC promoter. However, there are several mechanisms by which HIF can inhibit MYC activity. First, HIF1A can antagonize MYC transcriptional activity at MYC target genes by interfering with MYC binding to protein partners. For instance, HIF1A binds to MAX and disrupts MYC/MAX complexes, leading to reduced cyclin D2 expression, induction of p21 (CDKN1A), and G1 phase cell cycle arrest in human pVHL-null kidney cancer cell lines [102]. Meanwhile, under hypoxia, HIF-1 can induce MAX interactor 1, dimerization protein, which inhibits the transcriptional activity of MYC by competing for MAX and represses MYC target genes [103] such as peroxisome proliferator-activated receptor gamma coactivator 1-beta in human pVHL-null kidney cancer cell lines [104] or ornithine decarboxylase in multiple human cancer cell lines [105]. Second, HIF1A directly inhibits MYC transcriptional activity by DNA-binding site competition. For instance, HIF1A displaces MYC binding from the promoter of cyclin-dependent kinase inhibitor 1A (CDKN1A, p21cip1) and upregulates the expression of p21 (CDKN1A) in human HCT116 colorectal carcinoma cell line [106]. HIF1A also competes against MYC for binding to SP1, a coactivator of MYC, at the promoters of MYC target genes such as MutS homolog 2 (MSH2), MSH6, and nibirin, which encode DNA repair proteins, in human HCT116 colorectal carcinoma cell line [107][108] and the E-type prostanoid receptor in human HCA-7 colon cancer cell line [109]. Third, several studies have shown that HIF-1A promotes proteasomal degradation of MYC under chronic hypoxia conditions [104][110][111][112].
Recently, it was shown that MYC induces HIF2A expression as well. MYC has been shown to preferentially bind to the HIF2A gene promoter in mouse Sca1C+ cancer stem cells (CSCs) in a MYC-driven mouse T-cell leukemia model and the equivalent ATP-binding cassette superfamily G member 2+ CSC population in human acute lymphoblastic lymphoma, and activate HIF2A expression [113]. HIF2A regulates stem cell function by inducing the expression of octamer-binding transcription factor 4 [114] and AlkB homolog 5, an m6A demethylase that demethylates Nanog mRNA and increases Nanog expression [115]. In fact, the stem cell factors Nanog and SRY-box 2 facilitate MYC-mediated induction of HIF2A, playing a critical role in stem cell renewal and tumor stemness [116].
To date, there is limited literature on the effect of SP1 on MYC gene expression or the effect of MYC on SP1 gene expression. However, Parisi et al. [117] identified a functionally distinct signature for strong dual MYC/SP1 sites in various gene promoters. This finding indicates that although SP1 and MYC do not greatly influence each other’s expression transcriptionally or post-transcriptionally, there is a distinct mechanism by which they collaborate to regulate the transcription of specifically selected sets of target genes regulated by both SP1 and MYC.
Overall, these data suggest that there is a positive activation loop of HIF-1 (HIF1A) and SP1, which mostly occurs through induction of the transcriptional activity of the HIF1A gene via SP1 and that of the SP1 gene via HIF-1 (Figure 2). HIF-1 negatively regulates MYC through post-transcriptional mechanisms, and MYC activates HIF-1 through post-transcriptional mechanism. Interestingly, unlike HIF-1 and MYC, there is a positive activation loop of HIF-2 and MYC, which occurs via the combination of both transcriptional and post-transcriptional mechanisms. By contrast, there does not seem to be a direct effect of SP1 on MYC transcription or of MYC on SP1 transcription, although SP1 and MYC collaborate to transcriptionally regulate their target genes.

4. Collaboration of SP1, HIF-1, and MYC in Transcriptional Regulation of Their Target Genes

SP1, HIF-1, and MYC interact with each other either transcriptionally or post-transcriptionally and modulate the activity of each other, which demonstrates that there is some collaboration of these TFs in the execution of their activities. However, since SP1, HIF-1, and MYC are first and foremost TFs, their more important collaborations take place when these TFs modulate transcription of their target genes.
Many studies have investigated the mechanisms underlying how SP1 and HIF-1 collaborate in transcriptional regulation of their target genes. One example is the detailed study of the effect of SP1 and HIF-1 on the promoter activity of the human erythropoietin receptor gene [118]. That study showed that the binding of SP1 and HIF-1 to their binding sites in the promoter additively increases the transcriptional activity of the promoter. Another example is the detailed study on regulation of the human retinoic acid receptor-related orphan receptor alpha 4 (RORalpha) gene by the interaction between HIF-1 and SP1 [119]. In that case, it was shown that the binding sites for HIF-1 and SP1 in the promoter of this gene are situated closely to each other, and that HIF-1 functionally interacts with SP1 [119]. It was also shown that the HIF2A/SP1/HDAC4 network is involved in transcriptional activation of the human coagulation factor VII gene promoter [120]. Although HIF2A instead of HIF1A is involved in this case, these data suggest that the complex network of HIF1A/HIF2A/SP1/HDAC4 exists, as there is a link between SP1 and HIF1A via HDAC4 (Figure 2D) [95].
The collaboration of HIF-1 and MYC in transcriptional regulation of their target genes has already been described in the previous section. As aforementioned, since HIF-1 and MYC do not modify the expression of each other transcriptionally, the interaction between HIF-1 and MYC occurs either post-transcriptionally (HIF-1 usually suppresses MYC while MYC usually activates HIF-1) or through their collaboration to regulate the expression of their target genes. As an example of HIF-1 modulating the MYC-regulated transcription of genes, for instance, HIF-1 inhibits MYC-dependent induction of the transcriptional activity of the human CDKN1A gene promoter via a HIF1A–MYC mechanism [106]. This involves functional antagonism of the transcription repressor MYC via protein–protein interactions. This mechanism is independent of HIF1A DNA binding and transcriptional activity; instead, HIF1A displaces MYC from binding to the CDKN1A promoter. A similar mechanism also works for regulation of the human MSH2 gene promoter [107]. In this case, neither HIF1A nor MYC binds directly to the MSH2 promoter. Rather, both HIF1A and MYC discretely interact with the constitutively bound TF SP1 on the MSH2 promoter, whereas HIF1A dominates SP1 binding in hypoxia by competing with MYC. As a result, SP1 acts as a molecular switch by recruiting HIF1A for the hypoxic repression of MSH2. This mechanism is a good example of how HIF-1 can suppress rather than induce gene expression under hypoxia. In addition, this mechanism also shows the diversity of how HIF-1, SP1, and MYC collaborate to control the transcriptional activity.
There is no evidence to suggest that SP1 and MYC directly affect the transcription of each other. However, the collaboration of SP1 and MYC in the regulation of their target genes has been well described in the literature [121][122][123][124][125][126]. Among the genes whose transcription is regulated by the collaboration of MYC and SP1, there are various genes involved in the regulation of CSCs such as telomerase reverse transcriptase (TERT), BMI1, cluster of differentiation 133 (CD133), and CD147 [123][124][125][126]. These genes are often upregulated in cancer. In fact, most of the genes involved in the regulation of CSCs are regulated by HIF-1 as well [127][128][129][130]. Hence, these data indicate that the genes involved in the regulation of CSCs are in most cases regulated by SP1, HIF-1, and MYC. Since CSCs possess ‘stemness’ properties, which are reflected in their capacity to self-renew and generate differentiated cells that contribute to tumor heterogeneity [131][132], the contribution of CSCs has fundamental importance in the development of cancer; therefore, the eradication of CSCs is crucial for the success of anticancer therapy. As aforementioned, SP1, HIF-1, and MYC are all participants of cancer regulatory networks. The fact that the genes involved in the regulation of CSCs are all controlled by SP1, HIF-1, and MYC indicates that the very reason why these TFs are important participants of cancer regulatory networks might be because they regulate CSCs.
Based on the current knowledge about the transcription factors SP1, HIF-1, and MYC, the following conclusions can be drawn. First, all HIF-1, SP1, and MYC are deeply involved in cancer-related cellular mechanisms including metabolism, angiogenesis, anticancer immunity, and regulation of TME. Importantly, HIF-1 and SP1 usually induce the expression of each other while HIF-1 suppresses the expression of MYC and MYC induces that of HIF-1. This indicates that HIF-1 and SP1 can cooperatively activate cancer-related cellular mechanisms while the relationship between HIF-1 and MYC regarding the regulation of cancer-related cellular mechanisms can be variable depending on the context. Second, the CSC-related genes, which have fundamental importance in oncogenesis, are all positively regulated by HIF-1, SP1, and MYC at the transcriptional level. Overall, these results suggest that inhibitors for HIF-1 and SP1 likely induce anticancer effects in cooperation by suppressing the activity of cancer-related cellular mechanisms (including the mechanisms underlying CSC regulation) while using MYC inhibitors as anticancer drugs requires some cautions.

References

  1. Wilson, S.; Filipp, F.V. A network of epigenomic and transcriptional cooperation encompassing an epigenomic master regulator in cancer. NPJ Syst. Biol. Appl. 2018, 1, 24.
  2. Cao, P.; Zhao, B.; Xiao, Y.; Hu, S.; Kong, K.; Han, P.; Yue, J.; Deng, Y.; Zhao, Z.; Wu, D.; et al. Understanding the Critical Role of Glycolysis-Related lncRNAs in Lung Adenocarcinoma Based on Three Molecular Subtypes. Biomed Res. Int. 2022, 2022, 7587398.
  3. Mallik, S.; Qin, G.; Jia, P.; Zhao, Z. Molecular signatures identified by integrating gene expression and methylation in non-seminoma and seminoma of testicular germ cell tumours. Epigenetics 2021, 16, 162–176.
  4. Sankpal, U.T.; Maliakal, P.; Bose, D.; Kayaleh, O.; Buchholz, D.; Basha, R. Expression of specificity protein transcription factors in pancreatic cancer and their association in prognosis and therapy. Curr. Med. Chem. 2012, 19, 3779–3786.
  5. Vellingiri, B.; Iyer, M.; Devi Subramaniam, M.; Jayaramayya, K.; Siama, Z.; Giridharan, B.; Narayanasamy, A.; Abdal Dayem, A.; Cho, S.G. Understanding the Role of the Transcription Factor Sp1 in Ovarian Cancer: From Theory to Practice. Int. J. Mol. Sci. 2020, 21, 1153.
  6. Gao, Y.; Gan, K.; Liu, K.; Xu, B.; Chen, M. SP1 Expression and the Clinicopathological Features of Tumors: A Meta-Analysis and Bioinformatics Analysis. Pathol. Oncol. Res. 2021, 27, 581998.
  7. Abu, E.L.; Maaty, M.A.; Terzic, J.; Keime, C.; Rovito, D.; Lutzing, R.; Yanushko, D.; Parisotto, M.; Grelet, E.; Namer, I.J.; et al. Hypoxia-mediated stabilization of HIF1A in prostatic intraepithelial neoplasia promotes cell plasticity and malignant progression. Sci. Adv. 2022, 8, eabo2295.
  8. Wu, Q.; You, L.; Nepovimova, E.; Heger, Z.; Wu, W.; Kuca, K.; Adam, V. Hypoxia-inducible factors: Master regulators of hypoxic tumor immune escape. J. Hematol. Oncol. 2022, 15, 77.
  9. Mahauad-Fernandez, W.D.; Felsher, D.W. The Myc and Ras Partnership in Cancer: Indistinguishable Alliance or Contextual Relationship? Cancer Res. 2020, 80, 3799–3802.
  10. Vizcaíno, C.; Mansilla, S.; Portugal, J. Sp1 transcription factor: A long-standing target in cancer chemotherapy. Pharmacol. Ther. 2015, 152, 111–124.
  11. Safe, S.; Abbruzzese, J.; Abdelrahim, M.; Hedrick, E. Specificity Protein Transcription Factors and Cancer: Opportunities for Drug Development. Cancer Prev. Res. 2018, 11, 371–382.
  12. Semenza, G.L. Evaluation of HIF-1 inhibitors as anticancer agents. Drug Discov. Today 2007, 12, 853–859.
  13. Joshi, E.; Pandya, M.; Desai, U. Current Drugs and Their Therapeutic Targets for Hypoxia-inducible Factors in Cancer. Curr. Protein Pept. Sci. 2023, 24, 447–464.
  14. Shirai, Y.; Chow, C.C.T.; Kambe, G.; Suwa, T.; Kobayashi, M.; Takahashi, I.; Harada, H.; Nam, J.M. An Overview of the Recent Development of Anticancer Agents Targeting the HIF-1 Transcription Factor. Cancers 2021, 13, 2813.
  15. Llombart, V.; Mansour, M.R. Therapeutic targeting of “undruggable: MYC. EBioMedicine 2022, 75, 103756.
  16. Szpirer, J.; Szpirer, C.; Riviere, M.; Levan, G.; Marynen, P.; Cassiman, J.J.; Wiese, R.; DeLuca, H.F. The Sp1transcription factor gene (SP1) and the 1,25-dihydroxyvitamin D3 receptor gene (VDR) are colocalized on human chromosome arm 12q and rat chromosome 7. Genomics 1991, 11, 168–173.
  17. Kaczynski, J.; Cook, T.; Urrutia, R. Sp1- and Krüppel-like transcription factors. Genome Biol. 2003, 4, 206.
  18. Narayan, V.A.; Kriwacki, R.W.; Caradonna, J.P. Structures of zinc finger domains from transcription factor Sp1. Insights into sequence–specific protein–DNA recognition. J. Biol. Chem. 1997, 272, 7801–7809.
  19. Oka, S.; Shiraishi, Y.; Yoshida, T.; Ohkubo, T.; Sugiura, Y.; Kobayashi, Y. NMR structure of transcription factor Sp1 DNA binding domain. Biochemistry 2004, 43, 16027–16035.
  20. Nagaoka, M.; Shiraishi, Y.; Sugiura, Y. Selected base sequence outside the target binding site of zinc finger protein Sp1. Nucleic Acids Res. 2001, 29, 4920–4929.
  21. Cook, T.; Gebelein, B.; Belal, M.; Mesa, K.; Urrutia, R. Three conserved transcriptional repressor domains are a defining feature of the TIEG subfamily of Sp1-like zinc finger proteins. J. Biol. Chem. 1999, 274, 29500–29504.
  22. Kadonag, J.T.; Courey, A.J.; Ladika, J.; Tjian, R. Distinct regions of Sp1 modulate DNA binding and transcriptional activation. Science 1988, 242, 1566–1570.
  23. Kadonaga, J.T.; Jones, K.A.; Tjian, R. Promoter specific activation of RNA polymerase II transcription by Sp1. Trends Biochem. Sci. 1986, 11, 20–23.
  24. Suske, G. The Sp-family of transcription factors. Gene 1999, 238, 291–300.
  25. Tan, N.; Khachigian, L. Sp1 phosphorylation and its regulation of gene transcription. Mol. Cell. Biol. 2009, 29, 2483–2488.
  26. Suzuki, T.; Kimura, A.; Nagai, R.; Horikoshi, M. Regulation of interaction of the acetyltransferase region of p300 and the DNA-binding domain of Sp1 on and through DNA binding. Genes Cells 2000, 5, 29–41.
  27. Jackson, S.P.; Tjian, R. O-glycosylation of eukaryotic transcription factors: Implications for mechanisms of transcriptional regulation. Cell 1988, 55, 125–133.
  28. Stielow, B.; Sapetschnig, A.; Wink, C.; Kruger, I.; Suske, G. SUMO-modified Sp3 represses transcription by provoking local heterochromatic gene silencing. EMBO Rep. 2008, 9, 899–906.
  29. Wei, S.; Chuang, H.C.; Tsai, W.C.; Yang, H.C.; Ho, S.R.; Paterson, A.J.; Kulp, S.K.; Chen, C.S. Thiazolidinediones mimic glucose starvation in facilitating Sp1 degradation through the up-regulation of beta-transducin repeat-containing protein. Mol. Pharmacol. 2009, 76, 47–57.
  30. Marin, M.; Karis, A.; Visser, P.; Grosveld, F.; Philipsen, S. Transcription factor Sp1 is essential for early embryonic development but dispensable for cell growth and differentiation. Cell 1997, 89, 619–628.
  31. Gilmour, J.; Assi, S.A.; Jaegle, U.; Kulu, D.; van de Werken, H.; Clarke, D.; Westhead, D.R.; Philipsen, S.; Bonifer, C. A crucial role for the ubiquitously expressed transcription factor Sp1 at early stages of hematopoietic specification. Development 2014, 141, 2391–2401.
  32. Li, L.; Davie, J.R. The role of Sp1 and Sp3 in normal and cancer cell biology. Ann. Anat.-Anat. Anz. 2010, 192, 275–283.
  33. Hjaltelin, J.X.; Izarzugaza, J.M.G.; Jensen, L.J.; Russo, F.; Westergaard, D.; Brunak, S. Identification of hyper-rewired genomic stress non-oncogene addiction genes across 15 cancer types. NPJ Syst. Biol. Appl. 2019, 5, 27.
  34. Wicks, E.E.; Semenza, G.L. Hypoxia-inducible factors: Cancer progression and clinical translation. J. Clin. Investig. 2022, 132, e159839.
  35. Harris, A.L. Hypoxia—A key regulatory factor in tumour growth. Nat. Rev. Cancer 2002, 2, 38–47.
  36. Mazure, N.M.; Brahimi-Horn, M.C.; Berta, M.A.; Benizri, E.; Bilton, R.L.; Dayan, F.; Ginouvès, A.; Berra, E.; Pouysségur, J. HIF-1: Master and commander of the hypoxic world. A pharmacological approach to its regulation by siRNAs. Biochem. Pharmacol. 2004, 68, 971–980.
  37. Loboda, A.; Jozkowicz, A.; Dulak, J. HIF-1 and HIF-2 transcription factors--similar but not identical. Mol. Cells 2010, 29, 435–442.
  38. Archer, M.C. Role of sp transcription factors in the regulation of cancer cell metabolism. Genes Cancer 2011, 2, 712–719.
  39. Beishline, K.; Azizkhan-Clifford, J. Sp1 and the ‘hallmarks of cancer’. FEBS J. 2015, 282, 224–258.
  40. Semenza, G.L. HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J. Clin. Investig. 2013, 123, 3664–3671.
  41. Paredes, F.; Williams, H.C.; San Martin, A. Metabolic adaptation in hypoxia and cancer. Cancer Lett. 2021, 502, 133–142.
  42. Zhu, J.; Sun, Y.; Luo, J.; Wu, M.; Li, J.; Cao, Y. Specificity protein 1 regulates gene expression related to fatty acid metabolism in goat mammary epithelial cells. Int. J. Mol. Sci. 2015, 16, 1806–1820.
  43. Shimano, H.; Sato, R. SREBP-regulated lipid metabolism: Convergent physiology—Divergent pathophysiology. Nat. Rev. Endocrinol. 2017, 13, 710–730.
  44. Mylonis, I.; Simos, G.; Paraskeva, E. Hypoxia-Inducible Factors and the Regulation of Lipid Metabolism. Cells 2019, 8, 214.
  45. Leone, R.D.; Powell, J.D. Metabolism of immune cells in cancer. Nat. Rev. Cancer 2020, 20, 516–531.
  46. Ho, P.-C.; Bihuniak, J.D.; Macintyre, A.N.; Staron, M.; Liu, X.; Amezquita, R.; Tsui, Y.-C.; Cui, G.; Micevic, G.; Perales, J.C.; et al. Phosphoenolpyruvate is a metabolic checkpoint of anti- tumor T cell responses. Cell 2015, 162, 1217–1228.
  47. Chang, C.-H.; Qiu, J.; O’Sullivan, D.; Buck, M.D.; Noguchi, T.; Curtis, J.D.; Chen, Q.; Gindin, M.; Gubin, M.M.; van der Windt, G.J.W.; et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell 2015, 162, 1229–1241.
  48. Doedens, A.L.; Stockmann, C.; Rubinstein, M.P.; Liao, D.; Zhang, N.; DeNardo, D.G.; Coussens, L.M.; Karin, M.; Goldrath, A.W.; Johnson, R.S. Macrophage expression of hypoxia-inducible factor-1 alpha suppresses T-cell function and promotes tumor progression. Cancer Res. 2010, 70, 7465–7475.
  49. Takeda, N.; O’Dea, E.L.; Doedens, A.; Kim, J.W.; Weidemann, A.; Stockmann, C.; Asagiri, M.; Simon, M.C.; Hoffmann, A.; Johnson, R.S. Differential activation and antagonistic function of HIF- isoforms in macrophages are essential for NO homeostasis. Genes Dev. 2010, 24, 491–501.
  50. Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014, 211, 781–790.
  51. Lee, J.H.; Elly, C.; Park, Y.; Liu, Y.-C. E3 ubiquitin ligase VHL regulates hypoxia-inducible factor-1α to maintain regulatory T cell stability and suppressive capacity. Immunity 2015, 42, 1062–1074.
  52. Yang, L.; Yang, C. Oncometabolites in Cancer: Current Understanding and Challenges. Cancer Res. 2021, 81, 2820–2823.
  53. Böttcher, M.; Renner, K.; Berger, R.; Mentz, K.; Thomas, S.; Cardenas-Conejo, Z.E.; Dettmer, K.; Oefner, P.J.; Mackensen, A.; Kreutz, M.; et al. D-2-hydroxyglutarate interferes with HIF-1α stability skewing T-cell metabolism towards oxidative phosphorylation and impairing Th17 polarization. Oncoimmunology 2018, 7, e1445454.
  54. Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell–derived lactic acid on human T cells. Blood 2007, 109, 3812–3819.
  55. Brand, A.; Singer, K.; Koehl, G.E.; Kolitzus, M.; Schoenhammer, G.; Thiel, A.; Matos, C.; Bruss, C.; Klobuch, S.; Peter, K.; et al. LDHA- associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 2016, 24, 657–671.
  56. Labadie, B.W.; Bao, R.; Luke, J.J. Reimagining IDO Pathway inhibition in cancer immunotherapy via downstream focus on the tryptophan–kynurenine–aryl hydrocarbon axis. Clin. Cancer Res. 2019, 25, 1462–1471.
  57. Liu, Y.; Liang, X.; Dong, W.; Fang, Y.; Lv, J.; Zhang, T.; Fiskesund, R.; Xie, J.; Liu, J.; Yin, X.; et al. Tumor- repopulating cells induce PD-1 expression in CD8+ T cells by transferring kynurenine and AhR activation. Cancer Cell 2018, 33, 480–494.e7.
  58. Schuettengruber, B.; Doetzlhofer, A.; Kroboth, K.; Wintersberger, E.; Seiser, C. Alternate activation of two divergently transcribed mouse genes from a bidirectional promoter is linked to changes in histone modification. J. Biol. Chem. 2003, 278, 1784–1793.
  59. Sanjabi, S.; Oh, S.A.; Li, M.O. Regulation of the Immune Response by TGF-β: From Conception to Autoimmunity and Infection. Cold Spring Harb. Perspect. Biol. 2017, 9, a022236.
  60. Kim, S.J.; Glick, A.; Sporn, M.B.; Roberts, A.B. Characterization of the promoter region of the human transforming growth factor-beta 1 gene. J. Biol. Chem. 1989, 264, 402–408.
  61. Qureshi, H.Y.; Sylvester, J.; El Mabrouk, M.; Zafarullah, M. TGF-beta-induced expression of tissue inhibitor of metalloproteinases-3 gene in chondrocytes is mediated by extracellular signal-regulated kinase pathway and Sp1 transcription factor. J. Cell. Physiol. 2005, 203, 345–352.
  62. Wang, J.P.; Wen, M.H.; Chen, Y.T.; Lee, H.H.; Chiang, E.R.; Lee, Y.T.; Liu, C.L.; Chen, T.H.; Hung, S.C. Trichostatin A inhibits TGF-β1 induced in vitro chondrogenesis of hMSCs through Sp1 suppression. Differentiation 2011, 81, 119–126.
  63. Datta, P.K.; Blake, M.C.; Moses, H.L. Regulation of plasminogen activator inhibitor-1 expression by transforming growth factor-beta-induced physical and functional interactions between smads and Sp1. J. Biol. Chem. 2000, 275, 40014–40019.
  64. Fajardo, O.A.; Thompson, K.; Parapuram, S.K.; Liu, S.; Leask, A. Mithramycin reduces expression of fibro-proliferative mRNAs in human gingival fibroblasts. Cell Prolif. 2011, 44, 166–173.
  65. Su, F.; Geng, J.; Li, X.; Qiao, C.; Luo, L.; Feng, J.; Dong, X.; Lv, M. SP1 promotes tumor angiogenesis and invasion by activating VEGF expression in an acquired trastuzumab-resistant ovarian cancer model. Oncol. Rep. 2017, 38, 2677–2684.
  66. Xu, K.; Shu, H.K. EGFR activation results in enhanced cyclooxygenase-2 expression through p38 mitogen-activated protein kinase-dependent activation of the Sp1/Sp3 transcription factors in human gliomas. Cancer Res. 2007, 67, 6121–6129.
  67. Hertel, J.; Hirche, C.; Wissmann, C.; Ebert, M.P.; Höcker, M. Transcription of the vascular endothelial growth factor receptor-3 (VEGFR3) gene is regulated by the zinc finger proteins Sp1 and Sp3 and is under epigenetic control: Transcription of vascular endothelial growth factor receptor 3. Cell Oncol. 2014, 37, 131–145.
  68. Zimna, A.; Kurpisz, M. Hypoxia-Inducible Factor-1 in Physiological and Pathophysiological Angiogenesis: Applications and Therapies. Biomed Res. Int. 2015, 2015, 549412.
  69. Nam, E.H.; Lee, Y.; Zhao, X.F.; Park, Y.K.; Lee, J.W.; Kim, S. ZEB2-Sp1 cooperation induces invasion by upregulating cadherin-11 and integrin a5 expression. Carcinogenesis 2014, 35, 302–314.
  70. Kolesnikoff, N.; Attema, J.L.; Roslan, S.; Bert, A.G.; Schwarz, Q.P.; Gregory, P.A.; Goodall, G.J. Specificity protein 1 (Sp1) maintains basal epithelial expression of the miR-200 family: Implications for epithelial-mesenchymal transition. J. Biol. Chem. 2014, 289, 11194–11205.
  71. Qian, Y.; Yao, W.; Yang, T.; Yang, Y.; Liu, Y.; Shen, Q.; Zhang, J.; Qi, W.; Wang, J. aPKC-i/P-Sp1/Snail signaling induces epithelial-mesenchymal transition and immunosuppression in cholangiocarcinoma. Hepatology 2017, 66, 1165–1182.
  72. Chhunchha, B.; Fatma, N.; Bhargavan, B.; Kubo, E.; Kumar, A.; Singh, D.P. Specificity protein, Sp1-mediated increased expression of Prdx6 as a curcumin-induced antioxidant defense in lens epithelial cells against oxidative stress. Cell Death Dis. 2011, 2, e234.
  73. Dauer, P.; Gupta, V.K.; McGinn, O.; Nomura, A.; Sharma, N.S.; Arora, N.; Giri, B.; Dudeja, V.; Saluja, A.K.; Banerjee, S. Inhibition of Sp1 prevents ER homeostasis and causes cell death by lysosomal membrane permeabilization in pancreatic cancer. Sci. Rep. 2017, 7, 1564.
  74. Akman, M.; Belisario, D.C.; Salaroglio, I.C.; Kopecka, J.; Donadelli, M.; De Smaele, E.; Riganti, C. Hypoxia, endoplasmic reticulum stress and chemoresistance: Dangerous liaisons. J. Exp. Clin. Cancer Res. 2021, 40, 28.
  75. Duffy, M.J.; O’Grady, S.; Tang, M.; Crown, J. MYC as a target for cancer treatment. Cancer Treat. Rev. 2021, 94, 102154.
  76. Das, S.K.; Lewis, B.A.; Levens, D. MYC: A complex problem. Trends Cell Biol. 2023, 33, 235–246.
  77. Madden, S.K.; de Araujo, A.D.; Gerhardt, M.; Fairlie, D.P.; Mason, J.M. Taking the Myc out of cancer: Toward therapeutic strategies to directly inhibit c-Myc. Mol. Cancer 2021, 20, 3.
  78. Cencioni, C.; Scagnoli, F.; Spallotta, F.; Nasi, S.; Illi, B. The “Superoncogene” Myc at the Crossroad between Metabolism and Gene Expression in Glioblastoma Multiforme. Int. J. Mol. Sci. 2023, 24, 4217.
  79. Pirity, M.; Blanck, J.K.; Schreiber-Agus, N. Lessons learned from Myc/Max/Mad knockout mice. Curr. Top. Microbiol. Immunol. 2006, 302, 205–234.
  80. Felsher, D.W. MYC Inactivation Elicits Oncogene Addiction through Both Tumor Cell-Intrinsic and Host-Dependent Mechanisms. Genes Cancer 2010, 1, 597–604.
  81. Hu, X.; Lin, J.; Jiang, M.; He, X.; Wang, K.; Wang, W.; Hu, C.; Shen, Z.; He, Z.; Lin, H.; et al. HIF-1α Promotes the Metastasis of Esophageal Squamous Cell Carcinoma by Targeting SP1. J. Cancer 2020, 11, 229–240.
  82. Nicolás, M.; Noé, V.; Ciudad, C.J. Transcriptional regulation of the human Sp1 gene promoter by the specificity protein (Sp) family members nuclear factor Y (NF-Y) and E2F. Biochem. J. 2003, 371 Pt 2, 265–275.
  83. Nicolás, M.; Noé, V.; Jensen, K.B.; Ciudad, C.J. Cloning and characterization of the 5′-flanking region of the human transcription factor Sp1 gene. J. Biol. Chem. 2001, 276, 22126–22132.
  84. Iyer, N.V.; Leung, S.W.; Semenza, G.L. The human hypoxia-inducible factor 1alpha gene: HIF1A structure and evolutionary conservation. Genomics 1998, 52, 159–165.
  85. Kato, A.; Ng, S.; Thangasamy, A.; Han, H.; Zhou, W.; Raeppel, S.; Fallon, M.; Guha, S.; Ammanamanchi, S. A potential signaling axis between RON kinase receptor and hypoxia-inducible factor-1 alpha in pancreatic cancer. Mol. Carcinog. 2021, 60, 734–745.
  86. Liu, J.; Levens, D. Making myc. Curr. Top. Microbiol. Immunol. 2006, 302, 1–32.
  87. Geltinger, C.; Hörtnagel, K.; Polack, A. TATA box and Sp1 sites mediate the activation of c-myc promoter P1 by immunoglobulin kappa enhancers. Gene Expr. 1996, 6, 113–127.
  88. Wierstra, I.; Alves, J. The c-myc promoter: Still MysterY and challenge. Adv. Cancer Res. 2008, 99, 113–333.
  89. Biswas, S.; Mukherjee, R.; Tapryal, N.; Singh, A.K.; Mukhopadhyay, C.K. Insulin regulates hypoxia-inducible factor-1α transcription by reactive oxygen species sensitive activation of Sp1 in 3T3-L1 preadipocyte. PLoS ONE 2013, 8, e62128.
  90. Lafleur, V.N.; Richard, S.; Richard, D.E. Transcriptional repression of hypoxia-inducible factor-1 (HIF-1) by the protein arginine methyltransferase PRMT1. Mol. Biol. Cell 2014, 25, 925–935.
  91. See, Y.X.; Chen, K.; Fullwood, M.J. MYC overexpression leads to increased chromatin interactions at super-enhancers and MYC binding sites. Genome Res. 2022, 32, 629–642.
  92. Dhanasekaran, R.; Deutzmann, A.; Mahauad-Fernandez, W.D.; Hansen, A.S.; Gouw, A.M.; Felsher, D.W. The MYC oncogene—The grand orchestrator of cancer growth and immune evasion. Nat. Rev. Clin. Oncol. 2022, 19, 23–36.
  93. Karadkhelkar, N.M.; Lin, M.; Eubanks, L.M.; Janda, K.D. Demystifying the Druggability of the MYC Family of Oncogenes. J. Am. Chem. Soc. 2023, 145, 3259–3269.
  94. Savino, M.; Annibali, D.; Carucci, N.; Favuzzi, E.; Cole, M.D.; Evan, G.I.; Soucek, L.; Nasi, S. The action mechanism of the Myc inhibitor termed Omomyc may give clues on how to target Myc for cancer therapy. PLoS ONE 2011, 6, e22284.
  95. Wu, M.; Huang, Z.; Huang, W.; Lin, M.; Liu, W.; Liu, K.; Li, C. microRNA-124-3p attenuates myocardial injury in sepsis via modulating SP1/HDAC4/HIF-1α axis. Cell Death Discov. 2022, 8, 40.
  96. Liu, Z.M.; Wang, X.; Li, C.X.; Liu, X.Y.; Guo, X.J.; Li, Y.; Chen, Y.L.; Ye, H.X.; Chen, H.S. SP1 Promotes HDAC4 Expression and Inhibits HMGB1 Expression to Reduce Intestinal Barrier Dysfunction, Oxidative Stress, and Inflammatory Response after Sepsis. J. Innate Immun. 2022, 14, 366–379.
  97. Geng, H.; Harvey, C.T.; Pittsenbarger, J.; Liu, Q.; Beer, T.M.; Xue, C.; Qian, D.Z. HDAC4 protein regulates HIF1α protein lysine acetylation and cancer cell response to hypoxia. J. Biol. Chem. 2011, 286, 38095–38102.
  98. Baddela, V.S.; Sharma, A.; Michaelis, M.; Vanselow, J. HIF1 driven transcriptional activity regulates steroidogenesis and proliferation of bovine granulosa cells. Sci. Rep. 2020, 10, 3906.
  99. Jeong, J.K.; Park, S.Y. Transcriptional regulation of specific protein 1 (SP1) by hypoxia-inducible factor 1 alpha (HIF-1α) leads to PRNP expression and neuroprotection from toxic prion peptide. Biochem. Biophys. Res. Commun. 2012, 429, 93–98.
  100. Culver, C.; Melvin, A.; Mudie, S.; Rocha, S. HIF-1α depletion results in SP1-mediated cell cycle disruption and alters the cellular response to chemotherapeutic drugs. Cell Cycle 2011, 10, 1249–1260.
  101. Li, Y.; Sun, X.X.; Qian, D.Z.; Dai, M.S. Molecular Crosstalk Between MYC and HIF in Cancer. Front. Cell Dev. Biol. 2020, 8, 590576.
  102. Gordan, J.D.; Bertout, J.A.; Hu, C.J.; Diehl, J.A.; and Simon, M.C. HIF-2alpha promotes hypoxic cell proliferation by enhancing c-myc transcriptional activity. Cancer Cell 2007, 11, 335–347.
  103. Conacci-Sorrell, M.; McFerrin, L.; Eisenman, R.N. An overview of MYC and its interactome. Cold Spring Harb. Perspect. Med. 2014, 4, a014357.
  104. Zhang, H.; Gao, P.; Fukuda, R.; Kumar, G.; Krishnamachary, B.; Zeller, K.I.; Dang, C.V.; Semenza, G.L. HIF-1 inhibits mitochondrial biogenesis and cellular respiration in VHL-deficient renal cell carcinoma by repression of C-MYC activity. Cancer Cell 2007, 11, 407–420.
  105. Corn, P.G.; Ricci, M.S.; Scata, K.A.; Arsham, A.M.; Simon, M.C.; Dicker, D.T.; El-Deiry, W.S. Mxi1 is induced by hypoxia in a HIF-1-dependent manner and protects cells from c-Myc-induced apoptosis. Cancer Biol. Ther. 2005, 4, 1285–1294.
  106. Koshiji, M.; Kageyama, Y.; Pete, E.A.; Horikawa, I.; Barrett, J.C.; Huang, L.E. HIF-1alpha induces cell cycle arrest by functionally counteracting Myc. EMBO J. 2004, 23, 1949–1956.
  107. Koshiji, M.; To, K.K.; Hammer, S.; Kumamoto, K.; Harris, A.L.; Modrich, P.; Huang, L.E. HIF-1alpha induces genetic instability by transcriptionally downregulating MutSalpha expression. Mol. Cell. 2005, 17, 793–803.
  108. To, K.K.; Sedelnikova, O.A.; Samons, M.; Bonner, W.M.; Huang, L.E. The phosphorylation status of PAS-B distinguishes HIF-1alpha from HIF-2alpha in NBS1 repression. EMBO J. 2006, 25, 4784–4794.
  109. Seira, N.; Yamagata, K.; Fukushima, K.; Araki, Y.; Kurata, N.; Yanagisawa, N.; Mashimo, M.; Nakamura, H.; Regan, J.W.; Murayama, T.; et al. Cellular density-dependent increases in HIF-1alpha compete with c-Myc to down-regulate human EP4 receptor promoter activity through Sp-1-binding region. Pharmacol. Res. Perspect. 2018, 6, e00441.
  110. Wong, W.J.; Qiu, B.; Nakazawa, M.S.; Qing, G.; Simon, M.C. MYC degradation under low O2 tension promotes survival by evading hypoxia-induced cell death. Mol. Cell. Biol. 2013, 33, 3494–3504.
  111. Li, Q.; Kluz, T.; Sun, H.; Costa, M. Mechanisms of c-myc degradation by nickel compounds and hypoxia. PLoS ONE 2009, 4, e8531.
  112. Zarrabi, A.J.; Kao, D.; Nguyen, D.T.; Loscalzo, J.; Handy, D.E. Hypoxia-induced suppression of c-Myc by HIF-2alpha in human pulmonary endothelial cells attenuates TFAM expression. Cell. Signal. 2017, 38, 230–237.
  113. Das, B.; Pal, B.; Bhuyan, R.; Li, H.; Sarma, A.; Gayan, S.; Talukdar, J.; Sandhya, S.; Bhuyan, S.; Gogoi, G.; et al. MYC Regulates the HIF2α Stemness Pathway via Nanog and Sox2 to Maintain Self-Renewal in Cancer Stem Cells versus Non-Stem Cancer Cells. Cancer Res. 2019, 79, 4015–4025.
  114. Covello, K.L.; Kehler, J.; Yu, H.; Gordan, J.D.; Arsham, A.M.; Hu, C.-J.; Labosky, P.A.; Simon, M.C.; Keith, B. HIF-2alpha regulates Oct-4: Effects of hypoxia on stem cell function, embryonic development, and tumor growth. Genes Dev. 2006, 20, 557–570.
  115. Zhang, C.; Samanta, D.; Lu, H.; Bullen, J.W.; Zhang, H.; Chen, I.; He, X.; Semenza, G.L. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m⁶A-demethylation of NANOG mRNA. Proc. Natl. Acad. Sci. USA 2016, 113, E2047–E2056.
  116. Soleymani Abyaneh, H.; Gupta, N.; Alshareef, A.; Gopal, K.; Lavasanifar, A.; Lai, R. Hypoxia Induces the Acquisition of Cancer Stem-like Phenotype Via Upregulation and Activation of Signal Transducer and Activator of Transcription-3 (STAT3) in MDA-MB-231, a Triple Negative Breast Cancer Cell Line. Cancer Microenviron. 2018, 11, 141–152.
  117. Parisi, F.; Wirapati, P.; Naef, F. Identifying synergistic regulation involving c-Myc and sp1 in human tissues. Nucleic Acids Res. 2007, 35, 1098–1107.
  118. Su, T.; Liu, P.; Ti, X.; Wu, S.; Xue, X.; Wang, Z.; Dioum, E.; Zhang, Q. HΙF1α, EGR1 and SP1 co-regulate the erythropoietin receptor expression under hypoxia: An essential role in the growth of non-small cell lung cancer cells. Cell Commun. Signal. 2019, 17, 152.
  119. Miki, N.; Ikuta, M.; Matsui, T. Hypoxia-induced activation of the retinoic acid receptor-related orphan receptor alpha4 gene by an interaction between hypoxia-inducible factor-1 and Sp1. J. Biol. Chem. 2004, 279, 15025–15031.
  120. Koizume, S.; Ito, S.; Miyagi, E.; Hirahara, F.; Nakamura, Y.; Sakuma, Y.; Osaka, H.; Takano, Y.; Ruf, W.; Miyagi, Y. HIF2α-Sp1 interaction mediates a deacetylation-dependent FVII-gene activation under hypoxic conditions in ovarian cancer cells. Nucleic Acids Res. 2012, 40, 5389–5401.
  121. Zhang, Y.; Chen, H.-X.; Zhou, S.-Y.; Wang, S.-X.; Zheng, K.; Xu, D.-D.; Liu, Y.-T.; Wang, X.-Y.; Yan, H.-Z.; Zhang, L.; et al. Sp1 and c-Myc modulate drug resistance of leukemia stem cells by regulating survivin expression through the ERK-MSK MAPK signaling pathway. Mol. Cancer 2015, 14, 56.
  122. Liu, H.; Zhou, M.; Luo, X.; Zhang, L.; Niu, Z.; Peng, C.; Ma, J.; Peng, S.; Zhou, H.; Xiang, B.; et al. Transcriptional regulation of BRD7 expression by Sp1 and c-Myc. BMC Mol. Biol. 2008, 9, 111.
  123. Kyo, S.; Takakura, M.; Taira, T.; Kanaya, T.; Itoh, H.; Yutsudo, M.; Ariga, H.; Inoue, M. Sp1 cooperates with c-Myc to activate transcription of the human telomerase reverse transcriptase gene (hTERT). Nucleic Acids Res. 2000, 28, 669–677.
  124. Wang, H.B.; Liu, G.H.; Zhang, H.; Xing, S.; Hu, L.J.; Zhao, W.F.; Xie, B.; Li, M.Z.; Zeng, B.H.; Li, Y.; et al. Sp1 and c-Myc regulate transcription of BMI1 in nasopharyngeal carcinoma. FEBS J. 2013, 280, 2929–2944.
  125. Gopisetty, G.; Xu, J.; Sampath, D.; Colman, H.; Puduvalli, V.K. Epigenetic regulation of CD133/PROM1 expression in glioma stem cells by Sp1/myc and promoter methylation. Oncogene 2013, 32, 3119–3129.
  126. Kong, L.M.; Liao, C.G.; Zhang, Y.; Xu, J.; Li, Y.; Huang, W.; Zhang, Y.; Bian, H.; Chen, Z.N. A regulatory loop involving miR-22, Sp1, and c-Myc modulates CD147 expression in breast cancer invasion and metastasis. Cancer Res. 2014, 74, 3764–3778.
  127. Nishi, H.; Nakada, T.; Kyo, S.; Inoue, M.; Shay, J.W.; Isaka, K. Hypoxia-inducible factor 1 mediates upregulation of telomerase (hTERT). Mol. Cell. Biol. 2004, 24, 6076–6083.
  128. Du, R.; Xia, L.; Ning, X.; Liu, L.; Sun, W.; Huang, C.; Wang, H.; Sun, S. Hypoxia-induced Bmi1 promotes renal tubular epithelial cell-mesenchymal transition and renal fibrosis via PI3K/Akt signal. Mol. Biol. Cell 2014, 25, 2650–2659.
  129. Ke, X.; Fei, F.; Chen, Y.; Xu, L.; Zhang, Z.; Huang, Q.; Zhang, H.; Yang, H.; Chen, Z.; Xing, J. Hypoxia upregulates CD147 through a combined effect of HIF-1α and Sp1 to promote glycolysis and tumor progression in epithelial solid tumors. Carcinogenesis 2012, 33, 1598–1607.
  130. Ohnishi, S.; Maehara, O.; Nakagawa, K.; Kameya, A.; Otaki, K.; Fujita, H.; Higashi, R.; Takagi, K.; Asaka, M.; Sakamoto, N.; et al. Hypoxia-inducible factors activate CD133 promoter through ETS family transcription factors. PLoS ONE 2013, 8, e66255.
  131. Barbato, L.; Bocchetti, M.; Di Biase, A.; Regad, T. Cancer Stem Cells and Targeting Strategies. Cells 2019, 8, 926.
  132. Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 128
Revisions: 2 times (View History)
Update Date: 13 Dec 2023
1000/1000
Video Production Service