C/EBPs: Comparison
Please note this is a comparison between Version 1 by Manlio Tolomeo and Version 3 by Rita Xu.

CCAAT-/enhancer-binding proteins (C/EBPs) isconstitute a family of six structurally homologous tratranscription factors that promote the expression of genes involved in differentcomposed of six members that are critical for normal cellular responses, such as proliferation, growth, and differentiation. These transcription factors control the differentiation of several cell types, and havedifferentiation in a variety of tissues. They promote the expression of genes through interaction with their promoters. Moreover, they have a key roles in regulating cellular proliferation, through interaction with cell cycle proteins. The molecular structure of C/EBPs and their ability to interact with a multitude ofre considered to be tumor suppressor factors determineue to their complex functions in differenability to arrest cells. In fact, C/EBPs can be activated or inhibited by a variety of intracellular or extracellular signals. In addition, post-translational modifications and interaction with other proteins can regulate their expression and activity in a complex manner. C/EBPs can activate or repress several classes of genes implicated growth (contributing to the terminal differentiation of several cell types) and for their role in cell differentiation, metabolism, inflammation, and immune ular response. Moreover, C/EBPs play an important role in cancer progression and metastasis, showing both pro-oncogenic and onco-suppressor functions. Interestingly, the same isotype of C/EBP to DNA damage, nutrient deprivation, hypoxia, and genotoxic agents. However, C/EBPs can exhibit both of theselicit completely opposite functions. This “Janus” role of C/EBPs in cancer could depend on their particular position at the crossroads between proeffects on cell proliferation and differentiation. Specific conditions such as cell type, microenvironment, type of heterodimerization, or interaction with different regulatory proteins can tip the balance towards pro- or anti-oncogenic actioncancer development and they have been described as both tumor promoters and tumor suppressors. 

  • C/EBP
  • cancer
  • tumor promoter
  • tumor suppressor

1. Introduction

1. Definition

11. C/EBPs Structure and Isoforms

CC/EBPs arAAT-e constituted by different functional and structural components, which include a C-terminal leucine-zipper (a basic DNAnhancer-binding region) and in the N-termini of most C/EBP proteins (regions that act as transactivating and regulatory domains).

The proteins (C/EBPs) is a family of six structuraleucine zipper is a highly conserved protein segment with a periodic repetition of leucine residues at every seventh position over a distance covering eight helical turns. The polypeptide segments containing these periodic arrays of leucine residues form a continuous α-helix that can dimerize through formation of a coiled-coil structure involving paired contacts between hydrophobic leucine zipper domains. Dimerization through the leucine-zipper leads to formation of homo- and heterodimers, which then bind with their two basic regions to DNA-sequences in the promoter/enhancer regions of a variety of genes. The basic residues interact in the major groove of the DNA, forming sequence-specific interactions [3].

Thhomologous transcription factors that promote the expression of genes involved in different cellular responses, such as proliferation, growth, and differentiation. These transcription factors control the differe dimerization and the localiztiation of leucines are critical for the DNA binding to the basic region. The basic regions of C/EBPs show a high binding affinity for (G/A)TTGCG(T/C)AA(T/C) or, broadly, the promoter CCAAT box sequence [4]. The N-terminseveral cell types, and have key roles in regulating cellular proli of the C/EBP proteins are quite divergent, except for three short sub-regions that are conserved in most members and that represent the activation domains. The N-termini of some C/EBP eration, through interaction with cell cycle proteins also contain regulatory domains that are conserved in most members [5]. Onc. The molecular structure bound to DNA, f C/EBPs can recruit co-activators in their activation domains that, in turn, can open up a chromatin structure or recruit basal transcription factors that stimulate transcription. 

The C/EBPand their ability to interact with a multitude of factors determine their complex functions family consists of six structurally and functionally homologous transcription factors—C/EBPαn different cells. In fact, C/EBPβ, C/EBPδ, C/EBPγ, C/EBPε, and CHOP. Apart from C/EBPγ and CHOP that are expressed ubiquitously, the other isoforms of C/EBP are distributed in specific tissues. For example, C/EBPα is prevalently expressed in the adipose tissue, blood mononuclear cells, liver, intestine, lung, adrenal gland, blood, nervous system, and placenta; C/EBPβ is expressed in the liver, adipose tissue, myelomonocytic cells, intestine, lung, spleen, nervous system, and kidney; C/EBPδ in the adipose tissue, myeloid cells, lung, nervous system, and intestine; and C/EBPε is expressed in myeloid and lymphoid cells.

1.2. C/EBPs Functions

C/EBPα ps can be activated or inhibited by a variety of intracellular or extracellular signals. In addition, post-translays a matin role during the late phases of differentiation of pre-adipocytes. Both C/EBPβ and C/EBPδ mRNA are induced during the mitotic expansion of pre-adipocite cells. When pre-adipocytes exit, the cell cycle begins to onal modifications and interaction with other proteins can regulate their express C/EBPα, which is then followed by the induction of adipocyte-specific markersion and activity in a complex manner [6][1]. It seems that C/EBPα expression is s can activated by the binding of C/EBPβ and C/EBPδ, with the C/EBPα promoter. C/EBPs are also or repress several classes of genes implicated in myeloidcell differentiation, [1]. Binding simetes fabor the C/EBPs (α, β, δ, and ε) are present in the promoter regions of numerous genes that are expressed in myeloid cells. The expression of C/EBPα is relatively high in early myeloid progenitors and decreases during granulocytic differentiation. On the other hand, C/EBPε, is preferentially expressed during granulocytic differentiation, whereas C/EBPβ is up-regulated during macrophage differentiation. C/EBP proteins also have a role in the differentiation of hepatocytes, mammary, epithelial cells, ovarian luteal cells, keratinocytes, neuronal cells, and intestinal epithelial cellslism, inflammation, and immune response. Moreover, C/EBPs play an important role in cancer progression and metastasis, showing both pro-oncogenic and onco-suppressor functions. Interestingly, the same isotype of [1]. C/EBPs are implicated in the control of metabolism and in inflammation, as shown by the identification of binding sites for the can exhibit both of these opposite functions. This “Janus” role of C/EBPs, in the regulatory regions of a battery of genes involved in the inflammatory response, including those coding for cytokines and their receptors, acute-phase plasma proteins, and components of signal transduction pathways. Moreover, C/EBPα, C/EBPβ, and C/EBPδ were shown to be widely expressed in the mammalian nervous system and these seemed to be involved in the memory process [7]. F in cancer could depend on their particular position at the crossroads between prolinally, most members of the C/EBPs family are implicated in the control of cell cycle and are involved during cancer progression, showing both tumor promoter and tumor suppressor activities.

2. C/EBPs and Cancer

C/EBPseration and differentiation. Specific are considered tumor suppressor factors for their ability to block cell growth, and for their role in cellular response to DNA damage. However, C/EBPs can elicit completely opposite effects on cell proliferation and cancer development, depending on the cell-type and the isoform present. They have also been described as both tumor promoters and tumor suppressors. This “Janus” role has been observed for all members of the C/EBP family (Table 1)ditions such as cell type, microenvironment, type of heterodimerization, or interaction with different regulatory proteins can tip the balance towards pro- or anti-oncogenic action [2].

2. Introduction

2.1. C/EBPs Structure and Isoforms

C/EBPs are constituted by different functional and structural components, which include a C-terminal leucine-zipper (a basic DNA-binding region) and in the N-termini of most C/EBP proteins (regions that act as transactivating and regulatory domains).
 
The leucine zipper is a highly conserved protein segment with a periodic repetition of leucine residues at every seventh position over a distance covering eight helical turns. The polypeptide segments containing these periodic arrays of leucine residues form a continuous α-helix that can dimerize through formation of a coiled-coil structure involving paired contacts between hydrophobic leucine zipper domains. Dimerization through the leucine-zipper leads to formation of homo- and heterodimers, which then bind with their two basic regions to DNA-sequences in the promoter/enhancer regions of a variety of genes. The basic residues interact in the major groove of the DNA, forming sequence-specific interactions [3].
 
The dimerization and the localization of leucines are critical for the DNA binding to the basic region. The basic regions of C/EBPs show a high binding affinity for (G/A)TTGCG(T/C)AA(T/C) or, broadly, the promoter CCAAT box sequence [4]. The N-termini of the C/EBP proteins are quite divergent, except for three short sub-regions that are conserved in most members and that represent the activation domains. The N-termini of some C/EBP proteins also contain regulatory domains that are conserved in most members [5]. Once bound to DNA, C/EBPs can recruit co-activators in their activation domains that, in turn, can open up a chromatin structure or recruit basal transcription factors that stimulate transcription. 
 
The C/EBP family consists of six structurally and functionally homologous transcription factors—C/EBPα, C/EBPβ, C/EBPδ, C/EBPγ, C/EBPε, and CHOP. Apart from C/EBPγ and CHOP that are expressed ubiquitously, the other isoforms of C/EBP are distributed in specific tissues. For example, C/EBPα is prevalently expressed in the adipose tissue, blood mononuclear cells, liver, intestine, lung, adrenal gland, blood, nervous system, and placenta; C/EBPβ is expressed in the liver, adipose tissue, myelomonocytic cells, intestine, lung, spleen, nervous system, and kidney; C/EBPδ in the adipose tissue, myeloid cells, lung, nervous system, and intestine; and C/EBPε is expressed in myeloid and lymphoid cells.
 

2.2. C/EBPs Functions

C/EBPα plays a main role during the late phases of differentiation of pre-adipocytes. Both C/EBPβ and C/EBPδ mRNA are induced during the mitotic expansion of pre-adipocite cells. When pre-adipocytes exit, the cell cycle begins to express C/EBPα, which is then followed by the induction of adipocyte-specific markers [21]. It seems that C/EBPα expression is activated by the binding of C/EBPβ and C/EBPδ, with the C/EBPα promoter. C/EBPs are also implicated in myeloid differentiation [1]. Binding sites for the C/EBPs (α, β, δ, and ε) are present in the promoter regions of numerous genes that are expressed in myeloid cells. The expression of C/EBPα is relatively high in early myeloid progenitors and decreases during granulocytic differentiation. On the other hand, C/EBPε, is preferentially expressed during granulocytic differentiation, whereas C/EBPβ is up-regulated during macrophage differentiation. C/EBP proteins also have a role in the differentiation of hepatocytes, mammary, epithelial cells, ovarian luteal cells, keratinocytes, neuronal cells, and intestinal epithelial cells [1]. C/EBPs are implicated in the control of metabolism and in inflammation, as shown by the identification of binding sites for the C/EBPs, in the regulatory regions of a battery of genes involved in the inflammatory response, including those coding for cytokines and their receptors, acute-phase plasma proteins, and components of signal transduction pathways. Moreover, C/EBPα, C/EBPβ, and C/EBPδ were shown to be widely expressed in the mammalian nervous system and these seemed to be involved in the memory process [22]. Finally, most members of the C/EBPs family are implicated in the control of cell cycle and are involved during cancer progression, showing both tumor promoter and tumor suppressor activities.
 

3. C/EBPs and Cancer

C/EBPs are considered tumor suppressor factors for their ability to block cell growth, and for their role in cellular response to DNA damage. However, C/EBPs can elicit completely opposite effects on cell proliferation and cancer development, depending on the cell-type and the isoform present. They have also been described as both tumor promoters and tumor suppressors. This “Janus” role has been observed for all members of the C/EBP family (Table 2).  

Table 12. Molecular mechanisms of tumor suppressor and the tumor promoting activity of each C/EBP family member.

Molecular mechanisms of tumor suppressor and the tumor promoting activity of each C/EBP family member.

2.1. C/EBPα 

3.1. C/EBPα 

C/EBPα was the first member of C/EBP family cloned. It is expressed prevalently in post-mitotic cells and it seems implicated in regulation of cell-cycle exit and differentiation in adipocytes, hepatocyte, myeloid cells, and other tissues [11,13,16,20,23]. C/EBPα induces cell growth arrest by blocking the association of CDKs with cyclins [24] and stabilizing the CDK2-p21 inhibitory complex [25]; moreover, C/EBPα can directly inhibit the activity of free CDK2/CDK4 [24]. In addition, C/EBPα might also associate with E2F complexes and convert them into repressors capable of inhibiting the S-phase gene transcription [26]. In this context, C/EBPα plays a role in the cellular response to DNA damage induced by extrinsic DNA-damaging agents. In fact, the C/EBPα gene is a p53-regulated DNA damage-inducible gene in keratinocytes and it is an important link between UVB-induced DNA damage and cell cycle arrest in epidermal keratinocytes [27].
Two isoforms of the C/EBPα are generated from its mRNA by a ribosomal scanning mechanism—the full-length 42 kDa C/EBPα (p42), which is implicated in the transcriptional activation of adipocyte genes and the 30-kDa isoform (p30). p30 is an alternative translation product initiated at the third in-frame methionine codon of the C/EBPα mRNA. Unlike p42, which inhibits cell proliferation, p30 seems not to exert an antiproliferative function [28].
 
For its ability to induce growth arrest contributing to the terminal differentiation of several cell types, and for its role in the cellular response to DNA damage, C/EBPα is considered a potent tumor suppressor factor. Deregulation of its expression can predispose to different malignancies, especially hematological neoplasms.
 
In fact, C/EBPα plays a main role in hematopoiesis. It is essential for myeloid differentiation and it has been implicated in regulating self-renewal of fetal liver and adult hematopoietic stem cells. Disruption of C/EBPα blocks the transition from myeloid committed stem cells to granulocyte/monocyte progenitors, leading to the loss of mature granulocytes [29]. In adult hematopoietic stem cells, the loss of C/EBPα causes increased proliferation, an increased number of functional long-term hematopoietic stem cells, and advanced repopulating ability [30]. N-Myc seems to be the downstream target of C/EBPα in hematopoietic stem cells. In fact, transcriptional repression of N-Myc by C/EBPα maintains the hematopoietic stem cells in a quiescence status [31]. Downregulation of C/EBPα plays a role in leukemogenesis. C/EBPα function is indeed frequently abrogated in acute myelogenous leukemias (AML) and oncogenes, such as AML1-ETO, BCR-ABL, or FLT3-ITD. This can downregulate or suppress C/EBPα, causing a block in myeloid differentiation and thereby inducing leukemogenesis [32,33,34,35]. Deregulation of the C/EBPα expression was also reported in a variety of additional human tumors, including breast and lung cancer [36
 
Discordant results were observed in hepatocellular carcinoma (HCC) and in hepatoblastoma (HBL). Tomizawa et al. examined the expression level of the C/EBPα and C/EBPβ genes between tumor and non-tumorous tissues of the same hepatocellular carcinoma patients, with quantitative real-time polymerase chain reactions showing that the expression of both the C/EBPα and C C/EBPβ genes was downregulated in the majority of the tumor specimens compared to the corresponding non-tumorous tissues. Patients whose expression of either C/EBPα or C/EBPβ was higher in tumors than non-tumorous tissues, survived longer than those whose expression was lower in tumors [37]. Similar results were obtained by Tseng et al. in a retrospective cohort study on 50 HCC patients. They observed that a reduced expression of the C/EBPα protein in HCC was associated with an advanced tumor stage and shortened patient survival [38]. In contrast, Lu et al. demonstrated that C/EBPα overexpression was correlated with poorer HCC overall survival [39]. Moreover, upregulation of C/EBPα was described in hepatoblastoma (HBL) [40]. These data suggest that C/EBPα might act as both a tumor suppressor and tumor promoter factor in liver cancers. These opposite functions might be correlated to a different posttranslational phosphorylation switch of C/EBPα. Phosphorylation of C/EBPα at ser190 (ser193 in mice homologue) is essential to maintain quiescence of hepatocytes through two pathways—inhibition of cdks and repression of E2F [41]. However, in liver tumor cells, the activation of the PI3K/Akt pathway blocks the growth inhibitory activity of C/EBPα, through the PI3K/AKT-protein phosphatase 2 (PP2A)-mediated dephosphorylation of C/EBPα on Ser 193 [41]. Dephosphorylated C/EBPα is unable to interact with and inhibit cdks and E2F with the consequent promotion of cell growth. Mutation of Ser 193 to Ala also abolishes the ability of C/EBPα to cause growth arrest, as it prevents the interaction of C/EBPα with cdk2 and E2F-Rb complexes [41]. Cast et al. investigated liver cancer in the mouse model C/EBPα-S193A, in a large cohort of human HBL samples, and in Pten/p53 double knockout mice, and found that these cancers were characterized by an elevation of C/EBPα, which was dephosphorylated at Ser190/193. They found that dephosphorylated C/EBPα creates preneoplastic foci with cancer stem cells that give rise to HCC and aggressive HBL [42]. Therefore, conversion of the tumor suppressor C/EBPα into an oncogenic isoform can create preneoplastic foci where hepatocytes dedifferentiate into cancer cells, giving rise to liver cancer expressing high levels of mutated C/EBPα.
 
The development of liver cancer can also be determined by degradation of C/EBPα. Carcinogens such as diethylnitrosamine/phenobarbital (DEN/PB) can induce specific degradation of the phosphorylated isoform of C/EBPα, through activation of the ubiquitinproteasome system (UPS). The mechanism of the UPS-mediated elimination of C/EBPα during carcinogenesis involves elevated levels of gankyrin (an oncogenic protein that was found to interact with the phosphorylated form of C/EBPα) and targets it for UPS-mediated degradation [43,44,45].
 

3.2. C/EBPβ

C/EBPβ, which initially denominated the nuclear factor for IL-6 (NF-IL6), was first described in 1990 as a factor binding to the interleukin 1 (IL-1)-responsive element in the IL-6 promoter and showed high C-terminal homology to C/EBPα [46]. Subsequent knockout experiments revealed that the C/EBPβ knockout mice were viable but exhibited female sterility, defective mammary epithelial differentiation, and impaired immune function [1].
 
In fact, C/EBPβ is implicated in cell differentiation and in the regulation of genes involved in immune and inflammatory responses, such as IL-6, IL-4, IL-5, and TNF-alpha genes [15,47,48,49]. Moreover, it is critical for macrophage and B-cell differentiation [50,51]. C/EBPβ can activate genes that have specific roles in the nervous system—it can interact with an element of the preprotachykinin-A promoter, facilitating the substance P precursor gene transcription and with the promoter P2 of the choline acetyltransferase gene, inducing the biosynthesis of acetylcholine [52]. C/EBPβ also seems to be implicated in activation of genes coding for proteins that confer multidrug resistance to the cells regulating the liver expression of the MRP2 gene and activating the MDR1 gene in the MCF-7 cells [53,54].
 
The implication of C/EBPβ in cancer and tumorigenesis is more complex than that of C/EBPα. Many biological properties of C/EBPβ are similar to those of C/EBPα (since it blocks proliferation, promotes differentiation, and suppresses tumorigenesis), and similarly to C/EBPα. C/EBPβ is able to suppress cell proliferation through repression of the E2F target genes, thereby causing cellular senescence [55].
 
Expression of oncoproteins in primary cells often provokes cellular senescence, which is a permanent state of cell growth arrest that acts as a tumor suppressor mechanism. This cytostatic response, termed oncogene-induced senescence (OIS) is implemented through induction of the p19Arf-p53 tumor suppressor pathway and CDK inhibitors, such as p16Ink4a and p21CIP1, which activate Rb-dependent checkpoints [56,57,58]. C/EBPβ exerts anti-oncogenic effects because it is required for OIS.
 
Nevertheless, C/EBPβ can also exert pro-oncogenic effects [59]. These opposite activities depend on different causes, such as homo- or -heterodimerization, presence of inhibitors or presence of different isoforms of C/EBPβ.
 
C/EBPβ is maintained in a latent state by several auto-inhibitory elements that suppress its DNA-binding and transactivation functions [5,60]. Oncogenic stimuli, such as oncogenic RAS, can activate the RAF–MEK–ERK pathway that causes the phosphorylation and activation of the C/EBPβ [60,61]. Oncogenic Ras also increases the ratio of C/EBPβ homodimers with respect to C/EBPβ:C/EBPγ heterodimers, through a mechanism involving phosphorylation on leucine zipper residue Ser273 by p90Rsk kinases [62]. The homodimeric form of C/EBPβ contributes to the Ras-induced cell-cycle arrest and senescence in primary cells, whereas β:γ heterodimers actively promote cell growth [62]. Moreover, in immortalized and transformed cells, Ras-induced post-translational activation of C/EBPβ is inhibited by the 3′ untranslated region (3′UTR) of its mRNA, suppressing the cytostatic and pro-senescence functions of C/EBPβ [63]. The 3′UTR inhibitory effect was mapped to a region bearing the G/U rich elements (GREs). Moreover, an AU-rich element (ARE) and the ARE/GRE-binding protein HuR are required for 3′UTR inhibition. These components act by directing C/EBPβ transcripts to the peripheral cytoplasm, excluding them from a perinuclear region where the C/EBPβ kinases ERK1/2 and CK2 reside in RAS-transformed cells. In this location, newly translated C/EBPβ is uncoupled from RAS signaling and fails to undergo phosphorylation and activation by the RAF–MEK–ERK pathway. Thus, the intracellular site of the C/EBPβ translation is critical for RAS-induced activation via effector kinases such as ERK. Interestingly, 3′UTR inhibition and C/EBPβ mRNA compartmentalization are not observed in primary mouse and human fibroblasts [64].
 
The pro- and anti-tumorigenic activities of C/EBPβ can be, in part, determined by the presence of different isoforms of C/EBPβ. In fact, C/EBPβ is expressed as three isoforms with distinct activities—full-length LAP1 (or C/EBPβ p38 or Liver Activating Protein* (LAP*)), LAP2 (or C/EBP-β p32) that are lacking 21 amino acids (23 in human proteins) from the N-terminus, and Liver Inhibitory Protein (LIP or C/EBPβ p20) that lacks the whole activation domain (TAD) and acts as a transcriptional repressor. LAP1 is generally involved in terminal differentiation of cells, whereas LAP2 and LIP promote cell proliferation and tumor progression [65]. The proper ratios of the three isoforms are critical for normal cell growth and development. For example, an excess of the LIP isoform was observed in tumor cells that evade the growth inhibitory action of TGFβ. In normal cells, TGF-β block the cell cycle at the G1 phase, induce differentiation, or promote apoptosis. C/EBPβ is essential for TGFβ induction of the cell cycle inhibitor p15 INK4b and repression of C-Myc in human epithelial cells. These cytostatic responses can be missing in metastatic breast cancer cells. This loss seems to depend on the excess of the C/EBPβ inhibitory isoform LIP [66]. An excess of LIP was also observed in FLT3-ITD signal transduction. FLT3 is a cytokine receptor involved in cell growth and apoptosis regulation. Mutations that cause constitutive activation of the FLT3 receptor are frequent in acute myelogenous leukemia (AML) patients. The most frequent FLT3-mutations in AML are internal tandem duplications (ITDs) that lead to a constitutive activation of this receptor. In FLT3-ITD positive cells or when ITD sequences were inserted into the FLT3-wild type receptor, the LIP and LIP/LAP ratios were significantly increased, showing enhanced proliferation rates of AML cells. In addition, incubation of the FLT3-wild type cells with the FLT3 receptor ligand also elevated the LIP, LIP/LAP ratios, and proliferation [67].
 
 

3.3. C/EBPδ 

C/EBPαδ is was the first member of C/EBP family cloned. It is expressed prevalently in post-mitotic cells and it seems implicated in regulation of cell-cycle exit and da transcription factor that modulates many biological processes including cell growth, differentiation in adipocytes, hepatocyte, myeloi, motility, and cells, and other tissues death. The [40][41][42][43][44]. C/EBPαδ gene is induces celld during growth arrest by blocking the assocand cell differentiation of CDKs with cyclins. For example, C/EBPδ regulates growth arrest [45](G0–G1) and apoptostabilizing the CDK2-p21 inhibitory complexis in mammary epithelial cells, [46];and promoreover, C/EBPα can directlytes contact inhibit the activity of free CDK2/CDK4ion in these cells [68,69] [45]. Iand addition, C/EBPα might also associate with E2F complexes and convert them into repressors capable of inhibiting the S-phase gene transcription [47]. In thin fibroblasts [70]. Due to its roles in cell-growth arrest, cell differentiation, and apoptosis context, C/EBPα plays a roleδ acts as a tumor suppressor especially in the cellular response to DNA damage induced by extrinsic DNA-damaging agents. In fact, the C/EBPα gene is a p53-regulated DNA damage-inducible gene in keratinocytes and it is an important link between UVB-induced DNA damage and cell cycle arrest in early stages of tumor development. Moreover, C/EBPδ is also implicated in the regulation of gene transcription of several inflammatory cytokines and acute-phase proteins [71].

The antipidermal keratinocytesroliferative [48].

Two isoefformsect of the C/EBPα are generated from its mRNA by a ribosomal scanning mechanism—the full-length 42 kDa C/EBPα (p42), which is implicated in the transcriptional activation of adipocyte genesC/EBPδ seems to depend on its ability to downregulate the expression of cyclin D/E and the 30-kDa isoform (p30). p30 is an alternative translation product initiated at the third in-frame methionine codon of the C/EBPα mRNA. Unlike p42, which io upregulate the expression of the cyclin-dependent kinase inhibits cell proliferation, p30 seems not to exert an antiproliferative functior p27CIP2. These effects prevent the cyclin-dependent phosphorylation [49].

Forf its ability to induce growth arrest contributing to the terminal differentiation of several cell types, and for its role in the cellular response to DNA damagethe retinoblastoma protein (Rb) and the G1-S transition [68,72]. For these reasons, C/EBPα is consideredδ acts as a potent tumor -suppressor factor. Deregulation of its expression can predispose to and is frequently downexpressed in different malignancies, .

Howespvecially hematological neoplasms.

In factr, C/EBPαδ plays a main role in hematopoiesis. It is essential for myeloid differentidoes not necessarily inhibit cell proliferation and it has been implicated in regulating self-renewal of fetal liver and adult hematopoietic stem cells. Disruption ofcan be compatible with cell proliferation. In fact, C/EBPα blockδ promotes the transition from myeloid committed stem cells to granulocyte/monocyte progenitors, leading to the loss of mature granulocytesproliferation of cultured osteoblasts by directly activating the expression of insulin-like growth factor-1 [50].(IGF-1) In[73] adult hematopoietic stem cells, the loss of C/EBPα causes increased nd promotes the proliferation, an increased number of functional long-term hematopoietic stem of vascular smooth muscle cells, and advanced repopulating ability [51]. N-Myc in responseems to be the downstream target of C/EBPα in hematopoietic stem cells. In fact, transcriptional repression of N-Myc byIL-1β, by inducing expression of the PDGFA-receptor [74]. Moreover, C/EBPα maintains the hematopoietic stem cells in a quiescence status [52]. Dδ is a tumor promoter in  gliownregublationstoma.

 

3.4. of C/EBPγ

C/EBPαγ plays a role in leukemogenesis. C/EBPα function is indeed frequently abrogated in acute myelogenous leukemias (AML) and oncogenes, such as AML1-ETO, BCR-ABL, or FLT3-ITDwas first identified by its affinity for cis-regulatory sites in the Ig heavy-chain promoter and enhancer. This can downregulate or suppress C/EBPα, causing a block in myeloid differentiation and thereby inducing leukemogenesis [53][54][55][56]. Dmember of C/EBPs family contains a C-terminal leucine-zippereg bulation of the C/EBPα expression was also reported in a variety of additional human tumors, including breast and lung cancer [3

Dt lacks an amino-terminal transactivation domain present iscordant results were observed in hepatocellular carcinoma (HCC) and in hepatoblastoma (HBL). Tomizawa et al. examined the expression level of the C/EBPα and the other C/EBPs members. Transfection assays have shown that C/EBPβ genes between tumor and non-tumorous tissues of the same hepatocellular carcinoma patients, with quantitative real-time polymerase chain reactions showing that the expression of both theγ is neither an activator nor a repressor of transcription but acts as a transdominant negative regulator of other C/EBPα and C C/EBPβ genes was downregulated in the majority of the tumor specimens compared to the corresponding non-tumorous tissues. Patients whose expression of either C/EBPα or C/EBPβ was higher in tumors than non-tumorous tissues, survived longer than those whose expression was lower in tumors [75]. C/EBPγ can repress the transcriptional activity of C/EBPβ, C/EBPα, and C/EBPδ, through heterodimerization with its [57]. Similpar results were obtained by Tseng et al. in a retrospective cohort study on 50 HCC patients. They observed that a reduced expression of thener. However, heterodimerization with C/EBPγ did not alter the DNA-binding specificity of its C/EBPα protein in HCC was associated with an advanced tumor stage and shortened patient survival partner, since both homodimers and heterodimers can efficiently bind [58].to Ina contrast, Lu et al. demonstratednsensus C/EBP element.

 

3.5. that C/EBPε

C/EBPαε is overeexpression was correlated with poorer HCC overall survival [8]ed exclusively in myeloid cells. Moreover,In upregulation of C/EBPα was described in hepatoblastoma (HBL)humans, C/EBPε is expressed as 4 [9]. These data suggeist that C/EBPα might act as both a tumor suppressor and tumor promoter factor in liver cancers. These opposite functions might be correlated to a inct isoforms expressed as proteins of 32, 30, 27, and 14 kDa, through alternative splicing, different posttial promoter usage, and translational phosphorylation switch of C/EBPα. Phosphorylation of C/EBPα at ser190 (ser193 in mice homologue) is essential to maintain quiescence of hepatocytes through two pathways—inhibition of cdks and restart sites [76,77]. They are required for the terminal differentiation of neutrophils and eosinophils and the highest levels of expression of E2F [59]. Howeveccur dur, in liver tumor cells, the activation of the PI3K/Akt pathway blocks the growth inhibitory activity of C/EBPα, through the PI3K/AKT-protein phosphatase 2 (PP2A)-mediated dephosphorylation ofg the transition from promyelocyte to myelocyte cells. However, these isoforms can have opposite functions. The C/EBPαϵ32 oan Ser 193 [59]. Dephosphorylated C/EBPαε30 isoforms unable to interactare transcriptional activators, whereas C/EBPε27 with ands an inhibit cdks and E2F with the consequent promotion of cell growth. Mutation of Ser 193 to Ala also abolishes the ability of C/EBPαor of GATA-1, a factor essential for the development of hematopoietic cells. C/EBPε14 to cause growth arrest, as it prevents the interaction of C/EBPα with cdk2 and E2F-Rb complexeontains only DNA-binding and DNA-dimerization domains and seems [59].to Cast et al. investigated liver cancer in the mouse model ct as a dominant-negative regulator.

C/EBPα-S193A,ε32 ian ad C/EBPε30 ltrarge cohort of human HBL samples, and in Pten/p53 double knockout mice, and found that these cancers were characterized by an elevation of C/EBPα, which was dephosphorylated at Ser190/193. They found that dephosphorylated C/EBPα creates preneoplastic foci with cancer stem cells that give rise to HCC and aggressive HBL [60]. Therefore, conversion of the tumor sunsduction of hematopoietic progenitor cells exclusively cause eosinophil differentiation. In contrast, the putative reppressor C/EBPαε27 into an oncogenic isoform can create preneoplastic foci where hepatocytes dedstrongly inhibits eosinophil differentiate into cancer cells, giving rise to liver cancerion and gene expressing high levels of mutated C/EBPα.

on, including GATheA-1, development of liver cancer can also be determined by degradation of C/EBPα. Carcinogens such as diethylnitrosamine/phenobarbital (DEN/PB) can induce specific degradation of the phosphorylated isoform of C/EBPα,promoting granulocyte (neutrophil)-macrophage differentiation. The C/EBPε27 through activation of the ubiquitinproteasome system (UPS). The mechanism of the UPS-mediated elimination of C/EBPα during carcinogenesis involves elevated levels of gankyrin (an oncogenic protein that was found to interact with the phosphorylated form of C/EBPα) and targets it for UPS-mediated degradaepressor isoform strongly inhibits eosinophil development but promotes erythroid differentiation [61][62][63][78]. 

 

3.6.

CHOP

2.2. C/EBPβ

C/EBPβ, which initially denominated the nuclear factor for IL-6 (NF-IL6), was first described in 1990 as a factor binding to the interleukin 1 (IL-1)-responsive element in the IL-6 promoter and showed high C-terminal homology to C/EBPα [64]. Subshomologous protein (CHOP), also known as growth arrest and DNA damagequent knockout exper-iments revealed that the C/EBPβ knockout mice were viable but exhibited female sterility, defective mammary epithelial differentiation, and impaired immune functionnducible protein 153 (GADD153), is another member of [1].

In fact,he C/EBPβ is implicated in cell differentiation and in the regulation of genes involved in immune and inflammatory responses, such as family that displays both tumor-suppressive or tumor-supporting roles. It plays IL-6, IL-4, IL-5, and TNF-alpha gcenes [65][66][67][68]. Moreover, it is critical for macrophage and B-cell differentiational role in endoplasmic reticulum [69][70]. C/(EBPβR) can activate genes that have specific roles in the nervous system—it can interact with an element of the preprotachykinin-A promoter, facilitating the substance P precursor genstress, especially in cancer ER stress [79]. It is a stress-responsive transcription and with the promoter P2 of the choline acetyltransferase gene, inducing the biosynthesis of acetylcholinefactor during growth arrest, DNA damage, nutrient deprivation, hypoxia, [71]. C/EBPβ alsond seems to be implicated in activation ofgenotoxic agents. CHOP controls numerous genes coding for proteins that confer multidrug resistance to the cells regulating the liver expression of the MRP2 gene and activating the MDR1 gene in the MCF-7 cells [72][18].

The iinvolved in different cellular processes including inflammplication of C/EBPβ in cancer and tumorigenesis is more complex than that of C/EBPα. Many biological properties of , differentiation, autophagy, and apoptosis.

C/EBHOPβ are similar to those of C/EBPα (since it blocks proliferation, promotes differentiation, and suppresses tumorigenesis), and similarly to C/EBPα. C/EBPβ is able to suppress cell proliferation through re exerts an anti-oncogenic function by activating apoptosis in damaged, mutated, or precancerous cells by modulating the expression of the E2F targetpro-apoptotic or anti-apoptotic genes, thereby causing cellular senescence. Moreover, CHOP-induced [73].

Exapression of oncoproteins in primary cells often provokes cellular senescence, which is a permanent state of cell growth arrest that acts as a tumor suppressor mechanism. This cytostatic response, termed oncogene-induced senescence (OIS) is implemented through induction of the p19Arf-p53 tumor suppressor pathway and CDK inhibitors, such as p16Ink4a and p21CIP1, which activate Rb-ptosis in ER stress has significant implications for cancers. Most cancer cells present a higher avidity for glucose than normal cells. This dependent checkpoints [74][75][10]. C/EBPβ exerts anti-oncogenic effects because it is required for OIS.

Nevera reduced inthakeless, C/EBPβ can also exert pro-oncogenic effects [11]. These oppos of oxygen and nutrite activities depend on different causes, such as homo- or -heterodimerints due to inadequate vascularization, presence of inhibitors or presence of different isoforms of C/EBPβ.

C/EBPβ is main and diffusion during tumor growtainedh, in a latent state by several auto-inhibitory elements that suppress its DNA-binding and transactivawhich, through stabilization of the hypoxia-inducible transcription functionactor HIF-1α, shifts [5][12].the Oncogenic stimuli, such as oncogenic RAS, can activate the RAF–MEK–ERK pathway thatell metabolism toward glycolysis. Glucose shortage associated with cancer cell growth causes the phosphorylation and activation of the C/EBPβ [12][13]. Onc ER stress. In fact, ER is the majogenicr Ras also increases the ratio of C/EBPβ homodimers with respect to C/EBPβ:C/EBPγ heterodimers, through a mechanism involving phosphorylation on leucine zipper residue Ser273 by p90Rsk kinases [14]. The homosite for folding and maturation of secretory and transmembrane proteins and requires glucose for protein translation, andimeric form of C/EBPβ contributes to the Ras-induced cell-cycle arrest and senescence in primary cells, whereas β:γ heterodimer protein glycosylation and folding. ER stress actively promote cell growth [14]. Moreover, iates a sign immortalized and transformed cells, Ras-induced post-translational activation of C/EBPβ is inhibited by the 3′ untranslated region (3′UTR) of its mRNA, suppressing the cytostatic and pro-senescence functions of C/EBPβ [15]. Th transduction pathway termed unfolded protein response (UPR) that induces changes in ge 3′UTR inhibitory effect was mapped to a region bearing the G/U rich elements (GREs). Moreover, an AU-rich element (ARE) and the ARE/GRE-binding protein HuR are required for 3′UTR inhibition. These components act by directing C/EBPβ transcripts to the peripheral cyte expression to restore ER homeostasis. However, if ER stress cannot be alleviated, UPR triggers apoptosis through PKR-like endoplasm, excluding them from a perinuclear region where the C/EBPβ kinases ERK1/2 and CK2 reside in RAS-transformed cells. In this location, newly translated C/EBPβ is uncoupled from RAS signaling and fails to undergo phosphorylation and activation by the RAF–MEK–ERK pathway. Thus, the intracellular site of the C/EBPβ transic reticulum kinase (PERK) induction of CHOP. This pathway seems to have an important role in preventing malignant progression [80]. In fact, hepatocyte-specific CHOP ablation is critical for RAS-induced activation via effector kinases such as ERK. Interestingly, 3′UTR inhibition and C/EBPβ mRNA compartmentalization are not observed in primaryncreases tumorigenesis in high-fat diet-induced steatohepatitis and HCC [81]. Moreover, deletion of CHOP in a mouse amodel of K-rasG12V-induced human fibroblastlung cancer increases [16].

The pro- and anti-tumorigenic activities of C/EBPβ can be, in part, determined by the presence of different isoforms of C/EBPβ incidence, supporting the tumor suppressor role of CHOP [80]. In fact,

C/EBHOPβ is expressed as three isoforms with distinct prevents leukemogenesis by activities—full-length LAP1 (or C/EBPβ p38 or Liver Activating Protein* (LAP*)), LAP2 (or C/EBP-β p32) that are lacking 21 amino acids (23 in ating the PERK–eIF2α–ATF4–CHOP–GADD34 signaling-induced apoptosis in a human proteins) from the N-terminus, and Liver Inhibitory Protein (LIP or C/EBPβ p20) that lacks the whole activation domain (TAD) and acts as a transcriptional represshematopoietic stem cell (HSC) pool, with oncogenic mutations [82]. 

Hor. LAP1 is gwenerally involved in terminal differentiation of cells, whereas LAP2 and LIP promotver, depending on the cell proliferation andtype, CHOP can show tumor progression [17]. The -supproper ratios of the three isoforms are critical for normal cell growth and development. For example, an excess of the LIP isoform was observed in tumor cells that evade the growth inhibitory action of TGFβ. In normal cells, TGF-β block the cell cycle at the G1 phase, induce differentiation, or promote apoptosis. C/EBPβ is essential for TGFβ rting functions and tumor-induced tolerance. In fact, CHOP acts as an oncoprotein inducing metastasis via transcriptional induction of the cell cycle inhibitor p15 INK4b and repression of C-Myc in human epithelial cells. These cytostatic responses can be missing in metastatic breast cancer cells. This loss seems to depend on the excess of the C/EBPβ inhibitory isoform LIP [19]umor-associated proteases, both in liposarcoma and fibrosarcoma cell lines and in in vivo models [83]. An Morexcess of LIP was also observed in FLT3-ITD signal transduction. FLT3 is a cytokine receptor involved in cell growth and apoptosis regulation. Mutations that cause constitutive activation ofover, CHOP is upregulated in different mouse models of HCC, as well as human hepatocellular carcinoma.

CHOP thsee FLT3 receptor are frequent in acute myelogenous leukemia (AML) patients. The most frequent FLT3-mutations in AML are internal tandem duplications (ITDs) that lead to a constitutive activation of this receptor. In FLT3-ITD positive cells or when ITD sequences were inserted into the FLT3-wild type receptor, the LIP and LIP/LAP ratios were significantly incms to have a critical role in the immune inhibitory activity of tumor-infiltrating myeloid-derived suppressor cells (MDSCs). MDSCs lacking CHOP have decreased, showing enhanced proliferation rates of AML cells. In addition, incubation of the FLT3-wild type cells with the FLT3 receptor ligand also elevated the LIP, LIP/LAP ratios, and proliferation [76] immune-regulatory functions and show the ability to prime T cell function and induce antitumor responses.

 

 

 

 

References

  1. Ramji, D.P.; Foka, P. CCAAT/enhancer-binding proteins: Structure, function and regulation. Biochem. J. 2002, 365, 561–575. [Google Scholar] [CrossRef] [PubMed]
  2. Nerlov, C. The C/EBP family of transcription factors: A paradigm for interaction between gene expression and proliferation control. Trends Cell Biol. 2007, 17, 318–324. [Google Scholar] [CrossRef] [PubMed]
  3. Vinson, C.R.; Sigler, P.B.; McKnight, S.L. Scissors-grip model for DNA recognition by a family of leucine zipper proteins. Science 1989, 246, 911–916. [Google Scholar] [CrossRef]
  4. Osada, S.; Yamamoto, H.; Nishihara, T.; Imagawa, M. DNA binding specificity of the CCAAT/enhancer-binding protein transcription factor family. J. Biologic. Chem. 1996, 271, 3891–3896. [Google Scholar] [CrossRef] [PubMed]
  5. Williams, S.C.; Baer, M.; Dillner, A.J.; Johnson, P.F. CRP2 (C/EBPβ) contains a bipartite regulatory domain that controls transcriptional activation, DNA binding and cell specificity. EMBO J. 1995, 14, 3170–3183. [Google Scholar] [CrossRef] [PubMed]
  6. Descombes, P.; Schibler, U. A liver-enriched transcriptional activator protein, LAP, and a transcriptional inhibitory protein, LIP, are translated from the same mRNA. Cell 1991, 67, 569–579. [Google Scholar] [CrossRef]
  7. Welm, A.L.; Timochenko, N.A.; Darlington, G.J. C/EBPα regulates generation of C/EBPβ isoforms through activation of specific proteolytic cleavage. Mol. Cell. Biol. 1999, 19, 1695–1704. [Google Scholar] [CrossRef]
  8. Yamanaka, R.; Kim, G.D.; Radomska, H.S.; Lekstrom-Himes, J.; Smith, L.T.; Antonson, P.; Tenen, D.G.; Xanthopoulos, K.G. CCAAT/enhancer binding protein ε is preferentially up-regulated during granulocyte differentiation and its functional versatility is determined by alternative use of promoters and differential splicing. Proc. Natl. Acad. Sci. USA 1997, 94, 6462–6467. [Google Scholar] [CrossRef]
  9. Wang, N.D.; Finegold, M.J.; Bradley, A.; Ou, C.N.; Abdelsayed, S.V.; Wilde, M.D.; Taylor, L.R.; Wilson, D.R.; Darlington, G.J. Impaired energy homeostasis in C/EBPα knockout mice. Science 1995, 269, 1108–1112. [Google Scholar] [CrossRef] [PubMed]
  10. Lin, F.T.; Lane, M.D. Antisense CCAAT/enhancer-binding protein RNA suppresses coordinate gene expression and triglyceride accumulation during differentiation of 3T3-L1 preadipocytes. Genes Dev. 1992, 6, 533–544. [Google Scholar] [CrossRef] [PubMed]
  11. Freytag, S.O.; Paielli, D.L.; Gilbert, J.D. Ectopic expression of the CCAAT/enhancer-binding protein α promotes the adipogenic program in a variety of mouse fibroblastic cells. Genes Dev. 1994, 15, 1654–1663. [Google Scholar] [CrossRef] [PubMed]
  12. Tanaka, T.; Yoshida, N.; Kishimoto, T.; Akira, S. Defective adipocyte differentiation in mice lacking the C/EBPβ and/or C/EBPδ gene. EMBO J. 1997, 16, 7432–7443. [Google Scholar] [CrossRef] [PubMed]
  13. Scott, L.M.; Civin, C.I.; Rorth, P.; Friedman, A.D. A novel temporal expression pattern of three C/EBP family members in differentiating myelomonocytic cells. Blood 1992, 80, 1725–1735. [Google Scholar] [CrossRef] [PubMed]
  14. Morosetti, R.; Park, D.J.; Chumakov, A.M.; Grillier, I.; Shiohara, M.; Gombart, A.F.; Nakamaki, T.; Weinberg, K.; Koeffler, H.P. A novel, myeloid transcription factor, C/EBPε, is upregulated during granulocytic, but not monocytic, differentiation. Blood 1997, 90, 2591–2600. [Google Scholar] [CrossRef]
  15. Natsuka, S.; Akira, S.; Nishio, Y.; Hashimoto, S.; Sugita, T.; Isshiki, H.; Kishimoto, T. Macrophage differentiation-specific expression of NF-IL6, transcription factor for IL-6. Blood 1992, 79, 460–466. [Google Scholar] [CrossRef]
  16. Flodby, P.; Barlow, C.; Kylefjord, H.; Ahrlund-Richter, L.; Xanthopoulos, K.G. Increased hepatic cell proliferation and lung abnormalities in mice deficient in CCAAT/enhancer binding protein α. J. Biol. Chem. 1996, 271, 24753–24760. [Google Scholar] [CrossRef]
  17. Robinson, G.W.; Johnson, P.F.; Hennighausen, L.; Sterneck, E. The C/EBPβ transcription factor regulates epithelial cell proliferation and differentiation in the mammary gland. Genes Dev. 1998, 12, 1907–1916. [Google Scholar] [CrossRef]
  18. Zhu, S.; Oh, H.S.; Shim, M.; Sterneck, E.; Johnson, P.F.; Smart, R.C. C/EBPβ modulates the early events of keratinocyte differentiation involving growth arrest and keratin 1 and keratin 10 expression. Mol. Cell. Biol. 1999, 19, 7181–7190. [Google Scholar] [CrossRef]
  19. Cortes-Canteli, M.; Pignatelli, M.; Santos, A.; Perez-Castillo, A. CCAAT/enhancer binding protein β plays a regulatory role in differentiation and apoptosis of neuroblastoma cells. J. Biol. Chem. 2002, 277, 5460–5467. [Google Scholar] [CrossRef]
  20. Chandrasekaran, C.; Gordon, J.I. Cell lineage-specific and differentiation dependent patterns of CCAAT/enhancer binding protein α in the gut epithelium of normal and transgenic mice. Proc. Natl. Acad. Sci. USA 1993, 90, 8871–8875. [Google Scholar] [CrossRef]
  21. Darlington, G.J.; Ross, S.E.; MacDougald, O.A. The role of C/EBP genes in adipocyte differentiation. J. Biol. Chem. 1988, 273, 30057–30060. [Google Scholar] [CrossRef] [PubMed]
  22. Taubenfeld, S.M.; Wiig, K.A.; Monti, B.; Dolan, B.; Pollonini, G.; Alberini, C.M. Fornix-dependent induction of hippocampal CCAAT enhancer-binding protein β and δ co-localizes with phosphorylated cAMP response element-binding protein and accompanies long-term memory consolidation. J. Neurosci. 2001, 21, 84–91. [Google Scholar] [CrossRef] [PubMed]
  23. Umek, R.M.; Friedman, A.D.; McKnight, S.L. CCAAT-enhancer binding protein: A component of a differentiation switch. Science 1991, 251, 288–292. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, H.; Iakova, P.; Wilde, M.; Welm, A.; Goode, T.; Roesler, W.J.; Timchenko, N.A. C/EBPalpha arrests cell proliferation through direct inhibition of Cdk2 and Cdk4. Mol. Cell 2001, 8, 817–828. [Google Scholar] [CrossRef]
  25. Harris, T.E.; Albrecht, J.H.; Nakanishi, M.; Darlington, G.J. CCAAT/enhancer-binding protein-alpha cooperates with p21 to inhibit cyclin-dependent kinase-2 activity and induces growth arrest independent of DNA binding. J. Biol. Chem. 2001, 276, 29200–29209. [Google Scholar] [CrossRef] [PubMed]
  26. Lomiany, B.A.; D’Arigo, K.L.; Kelly, M.M.; Kurtz, D.T. C/EBPalpha inhibits cell growth via direct repression of E2F-DP-mediated transcription. Mol. Cell. Biol. 2000, 20, 5986–5997. [Google Scholar] [CrossRef] [PubMed]
  27. Yoon, K.; Smart, R.C. C/EBPalpha is a DNA damage-inducible p53-regulated mediator of the G1 checkpoint in keratinocytes. Mol. Cell Biol. 2004, 24, 10650–10660. [Google Scholar] [CrossRef]
  28. Calkhoven, C.F.; Bouwman, P.R.; Snippe, L.; Ab, G. Translation start site multiplicity of the CCAAT/enhancer binding protein alpha mRNA is dictated by a small 5′ open reading frame. Nucleic Acids Res. 1994, 22, 5540–5547. [Google Scholar] [CrossRef]
  29. Zhang, D.E.; Zhang, P.; Wang, N.D.; Hetherington, C.J.; Darlington, G.J.; Tenen, D.G. Absence of granulocyte colony-stimulating factor signaling and neutrophil development in CCAAT enhancer binding protein alpha-deficient mice. Proc. Natl. Acad. Sci. USA 1997, 94, 569–574. [Google Scholar] [CrossRef]
  30. Zhang, P.; Iwasaki-Arai, J.; Iwasaki, H.; Fenyus, M.L.; Dayaram, T.; Owens, B.M.; Shigematsu, H.; Levantini, E.; Huettner, C.S.; Lekstrom-Himes, J.A.; et al. Enhancement of hematopoietic stem cell repopulating capacity and self-renewal in the absence of the transcription factor C/EBP alpha. Immunity 2004, 21, 853–863. [Google Scholar] [CrossRef]
  31. Ye, M.; Zhang, H.; Amabile, G.; Yang, H.; Staber, P.B.; Zhang, P.; Levantini, E.; Alberich-Jordà, M.; Zhang, J.; Kawasaki, A.; et al. C/EBPa controls acquisition and maintenance of adult haematopoietic stem cell quiescence. Nat. Cell Biol. 2013, 15, 385–394. [Google Scholar] [CrossRef] [PubMed]
  32. Pabst, T.; Mueller, B.U.; Harakawa, N.; Schoch, C.; Haferlach, T.; Behre, G.; Hiddemann, W.; Zhang, D.E.; Tenen, D.G. AML1-ETO downregulates the granulocytic differentiation factor C/EBPalpha in t(8;21) myeloid leukemia. Nat. Med. 2001, 7, 444–451. [Google Scholar] [CrossRef] [PubMed]
  33. Perrotti, D.; Cesi, V.; Trotta, R.; Guerzoni, C.; Santilli, G.; Campbell, K.; Iervolino, A.; Condorelli, F.; Gambacorti-Passerini, C.; Caligiuri, M.A.; et al. BCR-ABL suppresses C/EBPalpha expression through inhibitory action of hnRNP E2. Nat. Genet. 2002, 30, 48–58. [Google Scholar] [CrossRef] [PubMed]
  34. Rosenbauer, F.; Tenen, D.G. Transcription factors in myeloid development: Balancing differentiation with transformation. Nat. Rev. Immunol. 2007, 7, 105–117. [Google Scholar] [CrossRef] [PubMed]
  35. Zheng, R.; Friedman, A.D.; Levis, M.; Li, L.; Weir, E.G.; Small, D. Internal tandem duplication mutation of FLT3 blocks myeloid differentiation through suppression of C/EBPalpha expression. Blood 2004, 103, 1883–1890. [Google Scholar] [CrossRef] [PubMed]
  36. Lourenço, A.-R.; Coffer, P.J. A tumor suppressor role for C/EBPα in solid tumors: More than fat and blood. Oncogene 2017, 36, 5221–5230. [Google Scholar] [CrossRef]
  37. Tomizawa, M.; Watanabe, K.; Saisho, H.; Nakagawara, A.; Tagawa, M. Down-regulated expression of the CCAAT/enhancer binding protein alpha and beta genes in human hepatocellular carcinoma: A possible prognostic marker. Anticancer Res. 2003, 23, 351–354. [Google Scholar]
  38. Tseng, H.H.; Hwang, Y.H.; Yeh, K.T.; Chang, J.G.; Chen, Y.L.; Yu, H.S. Reduced expression of C/EBP alpha protein in HCC is associated with advanced tumor stage and shortened patient survival. J. Cancer Res. Clin. Oncol. 2009, 135, 241–247. [Google Scholar] [CrossRef]
  39. Lu, G.D.; Ang, Y.H.; Zhou, J.; Tamilarasi, J.; Yan, B.; Lim, Y.C.; Srivastava, S.; Salto-Tellez, M.; Hui, K.M.; Shen, H.M. CCAAT/enhancer binding protein α predicts poorer prognosis and prevents energy starvation-induced cell death in hepatocellular carcinoma. Hepatology 2015, 61, 965–978. [Google Scholar] [CrossRef]
  40. Gray, S.G.; Kytola, S.; Matsunaga, T.; Larsson, C.; Ekstrom, T.J. Comparative genomic hybridization reveals population-based genetic alterations in hepatoblastomas. Br. J. Cancer 2000, 83, 1020–1025. [Google Scholar] [CrossRef]
  41. Wang, G.L.; Iakova, P.; Wilde, M.; Awad, S.; Timchenko, N.A. Liver tumors escape negative control of proliferation via PI3K/Akt-mediated block of C/EBP alpha growth inhibitory activity. Genes Dev. 2004, 18, 912–925. [Google Scholar] [CrossRef]
  42. Cast, A.; Valanejad, L.; Wright, M.; Nguyen, P.; Gupta, A.; Zhu, L.; Shin, S.; Timchenko, N. C/EBPα-dependent preneoplastic tumor foci are the origin of hepatocellular carcinoma and aggressive pediatric liver cancer. Hepatology 2018, 67, 1857–1871. [Google Scholar] [CrossRef] [PubMed]
  43. Wang, G.L.; Shi, X.; Haefliger, S.; Jin, J.; Major, A.; Iakova, P.; Finegold, M.; Timchenko, N.A. Elimination of C/EBPα through the ubiquitin–proteasome system promotes the development of liver cancer in mice. J. Clin. Investig. 2010, 120, 2549–2562. [Google Scholar] [CrossRef] [PubMed]
  44. Jiang, Y.; Iakova, P.; Jin, J.; Sullivan, S.; Sharin, V.; Hong, I.-H.; Anakk, S.; Mayor, A.; Darlington, G.; Finegold, M.; et al. FXR inhibits gankyrin in mouse livers and prevents development of liver cancer. Hepatology 2013, 57, 1098–1106. [Google Scholar] [CrossRef] [PubMed]
  45. Valanejad, L.; Lewis, K.; Wright, M.; Jiang, Y.; D’Souza, A.; Karns, R.; Sheridan, R.; Gupta, A.; Bove, K.; Witte, D.; et al. FXR–gankyrin axis is involved in development of pediatric liver cancer. Carcinogenesis 2017, 38, 738–747. [Google Scholar] [CrossRef]
  46. Akira, S.; Isshiki, H.; Sugita, T.; Tanabe, O.; Kinoshita, S.; Nishio, Y.; Nakajima, T.; Hirano, T.; Kishimoto, T. A nuclear factor for IL-6 expression (NF-IL6) is a member of a C/EBP family. EMBO J. 1990, 9, 1897–1906. [Google Scholar] [CrossRef]
  47. Davydov, I.V.; Krammer, P.H.; Li-Weber, M. Nuclear factor-IL6 activates the human IL-4 promoter in T cells. J. Immunol. 1995, 155, 5273–5279. [Google Scholar]
  48. Van Dijk, T.B.; Baltus, B.; Raaijmakers, J.A.; Lammers, J.W.; Koenderman, L.; de Groot, R.P. A composite C/EBP binding site is essential for the activity of the promoter of the IL-3/IL-5/granulocyte-macrophage colony-stimulating factor receptor beta c gene. J. Immunol. 1999, 163, 2674–2680. [Google Scholar]
  49. Greenwel, P.; Tanaka, S.; Penkov, D.; Zhang, W.; Olive, M.; Moll, J.; Vinson, C.; Di Liberto, M.; Ramirez, F. Tumor Necrosis Factor Alpha Inhibits Type I Collagen Synthesis through Repressive CCAAT/Enhancer-Binding Proteins. Mol. Cell. Biol. 2000, 20, 912–918. [Google Scholar] [CrossRef]
  50. Ruffell, D.; Mourkioti, F.; Gambardella, A.; Kirstetter, P.; Lopez, R.G.; Rosenthal, N.; Nerlov, C. A CREB-C/EBPbeta cascade induces M2 macrophage-specific gene expression and promotes muscle injury repair. Proc. Natl. Acad. Sci. USA 2009, 106, 17475–17480. [Google Scholar] [CrossRef]
  51. Chen, X.; Liu, W.; Ambrosino, C.; Ruocco, M.R.; Poli, V.; Romani, L.; Quinto, I.; Barbieri, S.; Holmes, K.L.; Venuta, S.; et al. Impaired generation of bone marrow B lymphocytes in mice deficient in C/EBPbeta. Blood 1997, 90, 156–164. [Google Scholar] [PubMed]
  52. Kovács, K.A.; Steinmann, M.; Magistretti, P.J.; Halfon, O.; Cardinaux, J.R. C/EBPbeta couples dopamine signalling to substance P precursor gene expression in striatal neurones. J. Neurochem. 2006, 98, 1390–1399. [Google Scholar] [CrossRef] [PubMed]
  53. Tanaka, T.; Uchiumi, T.; Hinoshita, E.; Inokuchi, A.; Toh, S.; Wada, M.; Takano, H.; Kohno, K.; Kuwano, M. The human multidrug resistance protein 2 gene: Functional characterization of the 5′-flanking region and expression in hepatic cells. Hepatology 1999, 30, 1507–1512. [Google Scholar] [CrossRef] [PubMed]
  54. Chen, G.K.; Sale, S.; Tan, T.; Ermoian, R.P.; Sikic, B.I. CCAAT/enhancer-binding protein beta (nuclear factor for interleukin 6) transactivates the human MDR1 gene by interaction with an inverted CCAAT box in human cancer cells. Mol. Pharmacol. 2004, 65, 906–916. [Google Scholar] [CrossRef]
  55. Sebastian, T.; Malik, R.; Thomas, S.; Sage, J.; Johnson, P.F. C/EBPbeta cooperates with RB:E2F to implement Ras(V12)-induced cellular senescence. EMBO J. 2005, 24, 3301–3312. [Google Scholar] [CrossRef] [PubMed]
  56. Campisi, J. Senescent cells, tumor suppression, and organismal aging: Good citizens, bad neighbors. Cell 2005, 120, 513–522. [Google Scholar] [CrossRef]
  57. Collado, M.; Blasco, M.A.; Serrano, M. Cellular senescence in cancer and aging. Cell 2007, 130, 223–233. [Google Scholar] [CrossRef]
  58. Adams, P.D. Healing and hurting: Molecular mechanisms, functions, and pathologies of cellular senescence. Mol. Cell 2009, 36, 2–14. [Google Scholar] [CrossRef]
  59. Sterneck, E.; Zhu, S.; Ramirez, A.; Jorcano, J.L.; Smart, R.C. Conditional ablation of C/EBP beta demonstrates its keratinocyte-specific requirement for cell survival and mouse skin tumorigenesis. Oncogene 2006, 25, 1272–1276. [Google Scholar] [CrossRef]
  60. Kowenz-Leutz, E.; Twamley, G.; Ansieau, S.; Leutz, A. Novel mechanism of C/EBP β (NF-M) transcriptional control: Activation through derepression. Genes Dev. 1994, 8, 2781–2791. [Google Scholar] [CrossRef]
  61. Mo, X.; Kowenz-Leutz, E.; Xu, H.; Leutz, A. Ras induces mediator complex exchange on C/EBP beta. Mol. Cell 2004, 13, 241–250. [Google Scholar] [CrossRef]
  62. Lee, S.; Shuman, J.D.; Guszczynski, T.; Sakchaisri, K.; Sebastian, T.; Copeland, T.D.; Miller, M.; Cohen, M.S.; Taunton, J.; Smart, R.C.; et al. RSK-mediated phosphorylation in the C/EBP β leucine zipper regulates DNA binding, dimerization, and growth arrest activity. Mol. Cell Biol. 2010, 30, 2621–2635. [Google Scholar] [CrossRef] [PubMed]
  63. Basu, S.K.; Malik, R.; Huggins, C.J.; Lee, S.; Sebastian, T.; Sakchaisri, K.; Quiñones, O.A.; Alvord, W.G.; Johnson, P.F. 3′UTR elements inhibit Ras-induced C/EBPβ post-translational activation and senescence in tumour cells. EMBO J. 2011, 30, 3714–3728. [Google Scholar] [CrossRef]
  64. Basu, S.K.; Gonit, M.; Salotti, J.; Chen, J.; Bhat, A.; Gorospe, M.; Viollet, B.; Claffey, K.P.; Johnson, P.F. A RAS-CaMKKβ-AMPKα2 pathway promotes senescence by licensing post-translational activation of C/EBPβ through a novel 3′UTR mechanism. Oncogene 2018, 37, 3528–3548. [Google Scholar] [CrossRef] [PubMed]
  65. Zahnow, C.A. CCAAT/enhancer-binding protein beta: Its role in breast cancer and associations with receptor tyrosine kinases. Expert Rev. Mol. Med. 2009, 11, e12. [Google Scholar] [CrossRef] [PubMed]
  66. Gomis, R.R.; Alarcón, C.; Nadal, C.; Van Poznak, C.; Massagué, J. C/EBPbeta at the core of the TGFbeta cytostatic response and its evasion in metastatic breast cancer cells. Cancer Cell 2006, 10, 203–214. [Google Scholar] [CrossRef]
  67. Haas, S.C.; Huber, R.; Gutsch, R.; Kandemir, J.D.; Cappello, C.; Krauter, J.; Duyster, J.; Ganser, A.; Brand, K. ITD- and FL-induced FLT3 signal transduction leads to increased C/EBPβ-LIP expression and LIP/LAP ratio by different signalling modules. Br. J. Haematol. 2010, 148, 777–790. [Google Scholar] [CrossRef]
  68. O’Rourke, J.P.; Newbound, G.C.; Hutt, J.A.; DeWille, J. CCAAT/enhancer-binding protein delta regulates mammary epithelial cell G0 growth arrest and apoptosis. Biol. Chem. 1999, 274, 16582–16589.
  69. Dearth, L.R.; DeWille, J. Posttranscriptional and posttranslational regulation of C/EBP delta in G0 growth-arrested mammary epithelial cells. Biol. Chem. 2003, 278, 11246–11255.
  70. Huang, A.M.; Montagna, C.; Sharan, S.; Ni, Y.; Ried, T.; Sterneck, E. Loss of CCAAT/enhancer binding protein delta promotes chromosomal instability. Oncogene 2004, 23, 1549–1557.
  71. Balamurugan, K.; Sterneck, E. The many faces of C/EBPdelta and their relevance for inflammation and cancer. J. Biol. Sci. 2013, 9, 917–933.
  72. Pawar, S.A.; Sarkar, T.; Balamurugan, K.; Sharan, S.; Wang, J.; Zhang, Y.; Dowdy, S.F.; Huang, A.M.; Sterneck, E. C/EBP{delta} targets cyclin D1 for proteasome-mediated degradation via induction of CDC27/APC3 expression. Natl. Acad. Sci. USA 2010, 107, 9210–9215.
  73. Umayahara, Y.; Ji, C.; Centrella, M.; Rotwein, P.; McCarthy, T.L. CCAAT/enhancer-binding protein delta activates insulin-like growth factor-I gene transcription in osteoblasts. Identification of a novel cyclic AMP signaling pathway in bone. Biol. Chem. 1997, 272, 1793–1800.
  74. Fukuoka, T.; Kitami, Y.; Okura, T.; Hiwada, K. Transcriptional regulation of the platelet-derived growth factor alpha receptor gene via CCAAT/enhancer-binding protein-delta in vascular smooth muscle cells. Biol. Chem. 1999, 274, 25576–25582.
  75. Cooper, C.; Henderson, A.; Artandi, S.; Avitahl, N.; Calame, K. Ig/EBP (C/EBPγ) is a transdominant negative inhibitor of C/EBP family transcriptional activators. Nucleic Acids Res. 1995, 23, 4371–4377.
  76. Williamson, E.A.; Xu, H.N.; Gombart, A.F.; Verbeek, W.; Chumakov, A.M.; Friedman, A.D.; Koeffler, HP. Identification of transcriptional activation and repression domains in human CCAAT/enhancer-binding protein epsilon. Biol. Chem. 1998, 273, 14796–14804.
  77. Chumakov, A.M.; Grillier, I.; Chumakova, E.; Chih, D.; Slater, J.; Koeffler, H.P. Cloning of the novel human myeloid-cell-specific C/EBP-epsilon transcription factor. Cell. Biol. 1997, 17, 1375–1386.
  78. Bedi, R.; Du, J.; Sharma, A.K.; Gomes, I.; Ackerman, S.J. Human. C/EBP-epsilon activator and repressor isoforms differentially reprogram myeloid lineage commitment and differentiation. Blood 2009, 113, 317–327.
  79. Oyadomari, S.; Mori, M. Roles of CHOP/GADD153 in endoplasmic reticulum stress. Cell Death Differ. 2004, 11, 381–389.
  80. Huber, A.L.; Lebeau, J.; Guillaumot, P.; Petrilli, V.; Malek, M.; Chilloux, J.; Fauvet, F.; Payen, L.; Kfoury, A.; Renno, T.; et al. p58(IPK)-mediated attenuation of the proapoptotic PERK-CHOP pathway allows malignant progression upon low glucose. Cell 2013, 49, 1049–1059.
  81. Nakagawa, H.; Umemura, A.; Taniguchi, K.,; Font-Burgada, J.; Dhar, D.; Ogata, H.; Zhong, Z.; Valasek, M.A.; Seki, E.; Hidalgo, J. ER stress cooperates with hypernutrition to trigger TNF-dependent spontaneous HCC development. Cancer Cell 2014, 26, 331–343.
  82. van Galen, P.; Kreso, A.; Mbong, N.; Kent, D.G.; Fitzmaurice, T.; Chambers, J.E.; Xie, S.; Laurenti, E.; Hermans, K.; Eppert, K.; et al. The unfolded protein response governs integrity of the haematopoietic stem-cell pool during stress. Nature 2014, 510, 268–272.
  83. Patil, N.; Ahmed Kabeer Rasheed, S.; Abba, M.; Hendrik Leupold, J.; Schwarzbach, M.; Allgayer, H. A mechanistic study on the metastasis inducing function of FUS-CHOP fusion protein in liposarcoma. J. Cancer 2014, 134, 2808–2819.