Fact-checked by Grok 2 weeks ago

CEBPA

CEBPA is a protein-coding gene located on chromosome 19q13.11 that encodes CCAAT/enhancer-binding protein alpha (C/EBPα), a basic leucine zipper (bZIP) transcription factor essential for regulating gene expression during cellular differentiation, particularly in myeloid progenitors, adipocytes, hepatocytes, and lung cells. The protein recognizes the CCAAT motif in promoter regions and functions by forming homodimers or heterodimers with other CEBP family members, such as CEBPB and CEBPG, to control processes like proliferation arrest, granulopoiesis, adipogenesis, and gluconeogenesis. CEBPA acts as a tumor suppressor by inhibiting uncontrolled cell growth and promoting terminal differentiation, with its expression modulated through differential translation starting from GUG or AUG codons, producing full-length and truncated isoforms. Mutations in CEBPA are strongly associated with (AML), occurring in approximately 10% of cases overall and up to 18% of cytogenetically normal AML subtypes. These mutations, often biallelic and involving both N-terminal and C-terminal regions, disrupt the protein's DNA-binding or dimerization capabilities, impairing myeloid differentiation and leading to leukemogenesis. In familial AML, heterozygous mutations in the combined with C-terminal mutations follow a two-hit , conferring a high lifetime (>90%) of early-onset AML with favorable prognosis and specific features like normal and FAB M1/M2 subtypes. Beyond hematopoiesis, CEBPA influences metabolic regulation, including body weight and insulin sensitivity in adipose and liver tissues.

Molecular Biology

Gene Structure and Expression

The CEBPA is located on 19q13.1 in humans, spanning positions 33,299,934 to 33,302,564 (GRCh38.p14) on the reverse strand. It encompasses approximately 2.6 kb and is organized as an intronless gene with a single encoding the full coding sequence. This compact structure facilitates direct transcription of the coding region without splicing interruptions. Orthologs of CEBPA are conserved across mammals, including the Cebpa gene in mice on , reflecting evolutionary preservation of its regulatory roles. The promoter region upstream of the CEBPA coding sequence lacks a canonical but contains multiple binding sites, such as those for CREB, deltaCREB, E2F-2, E2F-3a, and STAT5A, enabling context-specific regulation. C/EBPα autoregulates its own promoter, contributing to feedback control of expression levels. CEBPA exhibits tissue-specific expression patterns, with high levels in the liver, and adipose tissues, and myeloid cells, where it drives programs. It is also prominently expressed during embryonic development in organs such as the liver and adipose precursors, but remains at low basal levels in most other adult tissues like muscle and . Although the gene produces a single primary mRNA transcript, alternative translation initiation sites within this transcript generate distinct protein isoforms, such as the full-length p42 and truncated p30 forms.

Protein Structure and Isoforms

The CEBPA protein, also known as CCAAT/enhancer-binding protein alpha (C/EBPα), is a basic leucine zipper (bZIP) transcription factor consisting of 358 amino acids in its full-length form, with a calculated molecular weight of approximately 37.6 kDa, though it often migrates at around 42 kDa on SDS-PAGE due to post-translational modifications. The protein features an N-terminal transactivation domain (TAD) responsible for recruiting coactivators and initiating transcription, followed by a central bZIP domain that facilitates both DNA binding via its basic region and homodimerization or heterodimerization with other family members via the leucine zipper motif. This modular architecture allows C/EBPα to regulate gene expression by binding to specific DNA sequences, such as the CCAAT box, in a dimer-dependent manner. C/EBPα exists in two primary isoforms generated through alternative translation initiation from a single mRNA: the full-length p42 isoform, which initiates at the first AUG codon and encompasses both TAD1 (residues 1–127) and TAD2 (residues 175–206) along with the bZIP domain (residues 276–358), enabling robust transcriptional activation and differentiation programs; and the truncated p30 isoform, which starts at a downstream AUG codon (residue 152), lacking TAD1 but retaining TAD2 and the bZIP domain, thereby functioning primarily as a dominant-negative regulator by forming non-productive heterodimers with p42 that impair DNA binding and transactivation. The p42 isoform is the predominant activator in normal cellular contexts, while p30 levels can increase under stress or oncogenic conditions, shifting the balance toward proliferation. These isoforms arise without alternative splicing, relying instead on ribosomal scanning and uORF regulation for their relative expression ratios. Post-translational modifications further modulate C/EBPα activity, including phosphorylation at serine 21 (Ser21) within the TAD by mitogen-activated protein kinase (MAPK/ERK) pathways, which alters protein conformation, reduces DNA binding affinity, and promotes a shift from granulocytic to monocytic differentiation in myeloid cells. Sumoylation, primarily at lysine residues in the TAD and bZIP regions by SUMO1 or SUMO2/3, enhances transcriptional repression and stability but can inhibit activation depending on the isoform and cellular context, with p42 being more susceptible to sumoylation-mediated inhibition than p30. These modifications integrate extracellular signals to fine-tune C/EBPα function without altering isoform production. The bZIP domain of C/EBPα exhibits high evolutionary conservation across vertebrate species, sharing over 90% sequence identity with homologs in mammals, , and , which underscores its critical role in DNA recognition and dimerization preserved since early chordate divergence. This conservation extends to the leucine zipper heptad repeats and basic region residues essential for major groove interactions, highlighting the domain's ancient origin within the C/EBP family.

Biological Functions

Role in Differentiation and Development

CEBPA plays a pivotal role in promoting the terminal of myeloid progenitors into granulocytes, particularly neutrophils and monocytes, during . It acts as a key that drives the maturation of common myeloid progenitors into granulocyte-monocyte progenitors and subsequently into mature granulocytes by activating genes essential for neutrophil , such as those involved in colony-stimulating factor signaling. In the absence of CEBPA, myeloid is severely impaired, as evidenced by studies showing that conditional disruption of CEBPA in adult mice leads to a profound block in granulocyte and monocyte production by preventing formation of granulocyte-monocyte progenitors. Knockout studies in mice have underscored CEBPA's indispensable function in hematopoiesis and other developmental processes. CEBPA-null mice exhibit a complete absence of mature , highlighting its necessity for granulocyte commitment and maturation from fetal liver hematopoietic cells. These mice also display severe defects in liver development, including impaired and leading to neonatal lethality within hours of birth, as well as a lack of due to failed differentiation. Such phenotypes confirm CEBPA's broad regulatory influence beyond the hematopoietic system, extending to metabolic . In , CEBPA is crucial for the maturation of preadipocytes into adipocytes by inducing the expression of PPARγ and cooperating with C/EBPβ to activate downstream adipocyte-specific genes. This hierarchical regulation ensures the commitment and terminal of mesenchymal precursors into functional fat cells, with CEBPA acting downstream of early inducers like C/EBPβ to sustain lipid accumulation and insulin sensitivity. Embryonic roles of CEBPA further emphasize its developmental importance; it regulates fetal liver hematopoiesis by directing multipotential progenitors toward myeloid fates and restricting erythroid expansion, while in the , it promotes the of alveolar type II cells essential for production and respiratory maturation at birth. Recent findings indicate that CEBPA is also required for hematopoietic stem and generation during embryogenesis and for the development of Ly6Chigh monocytes, further emphasizing its role in early myeloid commitment. CEBPA's functions in are maintained through intricate feedback loops, including its auto-regulation via binding to its own promoter and enhancers to sustain expression during lineage commitment. Additionally, CEBPA engages in cross-talk with PU.1, where it modulates PU.1 activity to favor granulocytic over monocytic fates, and interacts with GFI-1 to reinforce myeloid priming in hematopoietic stem and progenitor cells. These regulatory circuits ensure stable lineage decisions and prevent aberrant proliferation in developing tissues.

Transcriptional Regulation Mechanisms

CEBPA functions as a transcription factor through its C-terminal basic leucine zipper (bZIP) domain, which enables DNA binding and dimerization. The bZIP domain recognizes the consensus DNA sequence 5'-T[TG]NNGNAA[TG]-3', a variant of the CAAT box, allowing CEBPA to bind target promoters either as homodimers or heterodimers with other C/EBP family members. This binding specificity facilitates precise regulation of gene expression in myeloid cells, where CEBPA predominantly operates during differentiation. The full-length p42 isoform of CEBPA serves as the primary activator, containing N-terminal domains that recruit co-activators such as CBP and p300. These co-activators promote , particularly at residues on histones and H4, leading to and enhanced transcriptional initiation at target loci. In contrast, the truncated p30 isoform, initiated from an internal , lacks these full domains and acts as a dominant-negative . By forming non-productive heterodimers with p42 or competing for the same DNA binding sites, p30 inhibits activation without supporting robust transcription, thereby fine-tuning or suppressing CEBPA-dependent gene expression in immature cells. In myeloid cells, CEBPA directly regulates key target genes essential for , including the receptor (CSF3R), (LTF), and c-fos (FOS). Binding to elements in the promoters of CSF3R and LTF drives their expression in cooperation with factors like PU.1 and RUNX1, promoting maturation. Similarly, CEBPA interacts with c-Fos via heterodimerization to modulate FOS expression, influencing lineage commitment toward granulocytes over monocytes. Beyond direct targets, CEBPA exerts indirect effects through lineage-specific networks, such as inducing secondary transcription factors that amplify myeloid differentiation programs. Upstream regulation of CEBPA expression integrates signals to control its availability during . In certain contexts, such as , CEBPε can induce CEBPA to promote granulocytic fate. Conversely, miR-124 inhibits CEBPA post-transcriptionally by binding its 3' , reducing protein levels and dampening in cells where miR-124 is often epigenetically silenced. These mechanisms ensure context-dependent activation of CEBPA's transcriptional output.

Genetic and Epigenetic Alterations

Somatic Mutations

Somatic mutations in the CEBPA gene, encoding the CCAAT/enhancer-binding protein alpha transcription factor, are acquired alterations that disrupt its function and contribute to leukemogenesis, particularly in acute myeloid leukemia (AML). These mutations are predominantly biallelic, involving both alleles of the gene, and occur in approximately 10% of all AML cases, with about 4% featuring biallelic changes. Recent classifications, such as the ELN 2022 guidelines and WHO 5th edition (as of 2022), recognize in-frame mutations in the basic leucine zipper (bZIP) domain—whether monoallelic or biallelic—as defining a favorable-risk subgroup in AML. They are most prevalent in cytogenetically normal AML (CN-AML), where frequencies range from 5% to 15%, and are rare in other leukemia subtypes such as acute lymphoblastic leukemia or chronic myeloid leukemia. The characteristic biallelic mutations consist of an N-terminal frameshift mutation, often insertions or deletions in the early coding region of exon 1, paired with a C-terminal in-frame mutation in the basic leucine zipper (bZIP) domain. N-terminal mutations, typically occurring in the transactivation domains (TAD1 and TAD2), lead to premature stop codons and production of a truncated 30-kDa isoform (p30) that acts as a dominant-negative protein by competing with the full-length 42-kDa isoform (p42) for binding partners without retaining transcriptional activity. C-terminal mutations, such as in-frame insertions or deletions, disrupt the alpha-helical structure of the bZIP domain, impairing DNA binding to target sites and homodimerization or heterodimerization with other proteins. Hotspots for these mutations are concentrated in codons 178-182 within the bZIP region for C-terminal changes and the initial 100-200 base pairs of the coding sequence for N-terminal frameshifts. Detection of these mutations relies on targeted sequencing of 1 for N-terminal alterations and the bZIP-encoding region ( 3 and 4) for C-terminal changes, traditionally using , though next-generation sequencing panels have improved sensitivity in clinical settings. Biallelic status is confirmed by assessing both alleles, as monoallelic mutations are less common and often lack the same leukemogenic potential. These mutations collectively abolish CEBPA's role in , promoting a block in .

Promoter Methylation and Epigenetics

Hypermethylation of CpG islands within the CEBPA promoter region leads to transcriptional silencing of the gene, contributing to dysregulated myeloid in cancers such as (AML) and . In AML, this epigenetic modification occurs in approximately 12-51% of cases, with prevalence estimates around 20-30% among patients lacking CEBPA mutations, and it is associated with reduced CEBPA protein levels and adverse clinical outcomes, including poorer overall survival. Similarly, in non-small cell , promoter hypermethylation correlates with downregulated CEBPA expression, promoting tumor progression by impairing signals. The underlying mechanisms involve recruitment of DNA methyltransferases (DNMTs), such as DNMT1 and DNMT3A, to the CEBPA promoter, often mediated by oncogenic fusion proteins. In acute promyelocytic leukemia (APL), a subtype of AML characterized by the PML-RARA fusion, this fusion protein directly interacts with DNMTs to enforce hypermethylation at target loci, thereby suppressing gene expression and blocking granulocytic maturation. This process is compounded by histone modifications, where histone deacetylases (HDACs) are recruited to the promoter via corepressor complexes associated with PML-RARA, leading to chromatin condensation and further gene repression. In APL, CEBPA expression is downregulated, with hypermethylation observed in the upstream promoter region. Therapeutic strategies targeting this epigenetic silencing have shown promise in restoring CEBPA function. Hypomethylating agents like inhibit DNMT activity, reducing methylation at the CEBPA promoter and reactivating , which in turn promotes myeloid and in AML cells. Clinical studies demonstrate that such treatment can elevate CEBPA mRNA levels in responsive patients, highlighting its potential as a mechanism for overcoming epigenetic resistance. Beyond DNA methylation, other epigenetic regulators influence CEBPA expression post-transcriptionally. For instance, microRNAs such as miR-182 target the 3' untranslated region (3'UTR) of CEBPA mRNA, promoting its degradation or translational repression, which exacerbates silencing in hepatocellular carcinoma and AML contexts. This miRNA-mediated control adds a layer of complexity to CEBPA dysregulation, distinct from promoter modifications but synergistic in sustaining oncogenic states.

Protein Interactions

Key Binding Partners

CEBPA, a basic leucine zipper (bZIP) transcription factor, primarily exerts its effects through dimerization, forming both homodimers and heterodimers with other family members such as C/EBPβ, C/EBPγ, and C/EBPδ. These interactions occur via the conserved bZIP domain, which facilitates DNA binding to CCAAT/enhancer motifs and modulates target gene specificity during processes like granulocyte differentiation. For instance, CEBPA:AP-1 heterodimers are favored in monocytic differentiation, where AP-1 proteins influence dimer preference over CEBPA homodimers. Among co-activators, CEBPA physically associates with and p300, histone acetyltransferases that enhance transcriptional activation by acetylating at target promoters and also modifying CEBPA itself to regulate its activity. This interaction promotes granulopoietic , as demonstrated in myeloid models where CBP/p300 recruitment correlates with increased accessibility. Additionally, CEBPA interacts with CDK9, a component of the P-TEFb complex, which phosphorylates to facilitate transcriptional elongation of differentiation genes. CEBPA also engages repressors that limit its function in specific contexts. E2F1, through its dimerization partner , directly binds CEBPA via protein-protein interactions, repressing CEBPA transcriptional activity and contributing to control in progenitors; this was confirmed by co-immunoprecipitation and yeast two-hybrid assays showing CEBPA-E2F/DP complex formation independent of pocket proteins. In early hematopoietic progenitors, CEBPA forms inactive complexes with PU.1 (encoded by SPI1), where high PU.1 levels antagonize CEBPA DNA binding and differentiation potential, as evidenced by functional studies in myeloid cells. Beyond these, CEBPA interacts with RUNX1 to coordinate hematopoiesis, with co-immunoprecipitation revealing direct binding that recruits epigenetic modifiers like TET2 to myeloid enhancers. In TGF-β signaling, CEBPA associates with SMAD3, where SMAD3 binding represses CEBPA expression during epithelial-mesenchymal transition, supported by showing SMAD3 occupancy at the CEBPA locus post-TGF-β stimulation. These interactions have been validated through experimental approaches including co-immunoprecipitation, which captures endogenous complexes in cell lysates (e.g., for - and -), and yeast two-hybrid screening, which identifies binary interactions like those in the bZIP dimerization domain. Such methods confirm the direct physical nature of these partnerships, distinguishing them from indirect regulatory effects.

Functional Interaction Pathways

CEBPA plays a pivotal role in the hematopoietic regulatory network through its mutual antagonism with PU.1, which governs the switch between myeloid and lymphoid cell fates. In early hematopoietic progenitors, low PU.1 levels favor lymphoid differentiation, while higher levels promote myeloid commitment; CEBPA reinforces myeloid granulocytic fate by counteracting PU.1 activity. Specifically, PU.1 induces the expression of and Egr2, which repress CEBPA, whereas CEBPA activates Gfi1, inhibiting PU.1-dependent genes, thereby directing progenitors toward granulocytes over s. In the inflammatory response, CEBPA cooperates with to induce in macrophages, enhancing acute-phase responses during or stress. p50 directly binds the CEBPA promoter to upregulate its expression in response to inflammatory signals, allowing CEBPA to synergize with at promoters of genes like IL-6 and G-CSF, thereby amplifying and immune activation. Additionally, CEBPA interacts with in signaling pathways, where STAT3-mediated IL-6 induction stabilizes CEBPA binding to shared enhancers, promoting sustained transcription of pro-inflammatory mediators such as in activated macrophages. CEBPA contributes to cell cycle control by repressing target genes, facilitating G0 arrest essential for terminal in myeloid lineages. Through its N-terminal and basic region motifs, CEBPA directly inhibits E2F-dependent transcription of proliferation genes like cyclin E and α, coupling growth arrest with granulocytic maturation. studies of CEBPA mutants defective in E2F repression demonstrate impaired development and persistent proliferation, underscoring this mechanism's necessity for proper hematopoietic . In disease contexts, such as (AML), the AML1-ETO disrupts the CEBPA-RUNX1 axis, blocking granulocytic . AML1-ETO, arising from t(8;21) translocation, indirectly suppresses CEBPA expression by inhibiting its positive autoregulation on the CEBPA promoter, leading to reduced CEBPA protein levels and failure to activate RUNX1-dependent myeloid genes. This dysregulation maintains leukemic blasts in a proliferative state, as evidenced by restored upon CEBPA re-expression in AML1-ETO-positive cells. Systems biology models of CEBPA interactions reveal like feed-forward loops that stabilize hematopoietic fate decisions. In , CEBPA forms coherent feed-forward loops with RUNX1, where both factors co-bind and mutually reinforce targets such as G-CSF receptor, ensuring robust activation during differentiation.

Clinical Significance

Involvement in

CEBPA mutations play a central role in the pathogenesis of (AML), particularly through biallelic alterations that disrupt normal myeloid differentiation. These mutations, often involving both N-terminal and C-terminal regions of the gene, lead to the production of truncated or dominant-negative CEBPA proteins that fail to activate target genes essential for granulocytic maturation, resulting in the accumulation of immature myeloid blasts. This differentiation block is a key driver of leukemogenesis, as evidenced by functional studies showing that mutant CEBPA impairs the transcriptional regulation of genes like those involved in the GFI-1/PU.1 pathway, promoting self-renewal and proliferation of leukemic progenitors. CEBPA-mutated AML is recognized as a distinct diagnostic entity in the (WHO) classification, with updates in 2016 and 2022 specifying that biallelic , including at least one in the basic (bZIP) domain, define this subtype among cases with ≥20% blasts and normal . These are enriched in cytogenetically normal AML, comprising approximately 5-14% of adult cases and 5-15% of pediatric cases, with higher frequencies observed in younger patients and those lacking other recurrent abnormalities like . The subtype typically presents with M1 or M2 morphology, monocytic features, and expression of and CD7 on blasts. Full leukemic transformation in CEBPA-mutated AML often requires cooperating genetic , such as in FLT3 (particularly internal tandem duplications, ITD) or , which enhance proliferative signaling and further impair differentiation. For instance, mutations occur in up to 28% of CEBPA double-mutant cases and contribute to dysregulated hematopoiesis, while FLT3-ITD co-occurrences, seen in about 16% of cases, provide additional oncogenic drive. These secondary hits collaborate with CEBPA alterations to initiate and maintain the leukemic state, as demonstrated in patient-derived samples where combined accelerate disease progression. Preclinical model systems have confirmed the oncogenic potential of CEBPA . Conditional knock-in models expressing patient-derived biallelic CEBPA develop a recapitulating human AML, including arrest at the stage, , and reduced to upon secondary hits like FLT3-ITD. These models highlight CEBPA as an initiating lesion and provide insights into the stepwise accumulation of required for full transformation.

Implications in Solid Tumors and Other Cancers

In solid tumors, CEBPA is frequently downregulated, often through promoter , which contributes to uncontrolled . In lung adenocarcinoma, a subtype of non-small cell , CEBPA expression is reduced in approximately 40-50% of cases, primarily due to epigenetic silencing via upstream promoter hypermethylation, leading to loss of its antiproliferative and -inducing effects. This downregulation promotes tumor growth by disrupting cell cycle arrest and pathways, as evidenced by studies showing that forced CEBPA re-expression in cell lines inhibits proliferation and induces type pneumocyte-like . in CEBPA are rare in solid tumors but have been reported in hepatocellular carcinoma (HCC), where they may coexist with expression changes; however, altered CEBPA activity more commonly arises from other mechanisms like miRNA-mediated suppression or proteasomal degradation, exacerbating HCC progression. Beyond AML, CEBPA alterations occur in other hematologic malignancies, influencing disease biology. Monoallelic mutations in the CEBPA gene have been identified in (T-ALL), potentially disrupting and contributing to leukemogenesis, though they are less frequent than in myeloid disorders. In , CEBPA silencing, often through indirect epigenetic or posttranscriptional mechanisms such as lncRNA interference, correlates with disease progression and resistance to therapy, highlighting its role as a potential tumor suppressor in plasma cell neoplasms. Prognostically, biallelic CEBPA in AML are associated with favorable outcomes, with 5-year overall rates around 55% in intensively treated patients, reflecting lower rates compared to wild-type cases. In contrast, CEBPA promoter methylation serves as a marker of adverse across malignancies, predicting higher risk in AML by sustaining epigenetic silencing and impairing , as observed in cohorts where methylated cases showed inferior event-free . Diagnostically, CEBPA alterations are integral to AML risk stratification per the 2022 European LeukemiaNet (ELN) guidelines, where in-frame bZIP domain (mono- or biallelic) classify cases as favorable risk, guiding decisions on intensive therapy versus allogeneic transplantation. Next-generation sequencing (NGS) panels routinely detect these , enabling precise in clinical workflows for AML and emerging applications in solid tumors. Therapeutically, strategies targeting CEBPA dysfunction emphasize restoring its function to induce . All-trans retinoic acid (ATRA)-like agents promote myeloid maturation in CEBPA-altered AML by enhancing transcriptional activity, while (HDAC) inhibitors reverse promoter silencing, reactivating CEBPA expression and synergizing with to reduce proliferation in preclinical models. Ongoing trials post-2020, such as NCT06458257 evaluating allogeneic in high-relapse-risk CEBPA-mutant AML and NCT06529250 assessing intermediate-dose regimens, aim to optimize outcomes by addressing relapse vulnerability in these patients.

References

  1. [1]
    CEBPA CCAAT enhancer binding protein alpha [ (human)] - NCBI
    Aug 25, 2025 · CEBPA mutations, and their co-occurrence with FLT3-ITD, FLT3-D835, and NPM1 mutations, are reliable genetic markers for acute myeloid leukemia.
  2. [2]
    CEBPA gene: MedlinePlus Genetics
    Jan 1, 2014 · The CEBPA gene provides instructions for making a protein called CCAAT enhancer-binding protein alpha. Learn about this gene and related ...
  3. [3]
    CEBPA-associated Familial AML (PDQ®) - National Cancer Institute
    Nov 25, 2024 · The CEBPA gene is a granulocytic transcription factor (C/EBPα) that serves an important role in myeloid cell differentiation.
  4. [4]
  5. [5]
    116897 - CCAAT/ENHANCER-BINDING PROTEIN, ALPHA; CEBPA
    A novel member of the C/EBP gene family, encodes a liver-enriched transcriptional activator protein. Genes Dev. 4: 1541-1551, 1990.<|control11|><|separator|>
  6. [6]
    Cebpa CCAAT/enhancer binding protein alpha [ (house mouse)]
    Sep 24, 2025 · Cebpa is a key regulator within the apoptotic network activated in pancreatic beta cells during insulitis, and Arl6ip5, Tnfrsf10b, Traf2, and ...
  7. [7]
    CEBPA Gene - CCAAT Enhancer Binding Protein Alpha - GeneCards
    CEBPA (CCAAT Enhancer Binding Protein Alpha) is a Protein Coding gene. Diseases associated with CEBPA include Leukemia, Acute Myeloid and Acute Myeloid Leukemia ...
  8. [8]
    C/EBPα in normal and malignant myelopoiesis - PMC
    C/EBPα auto-activates its own promoter, and RUNX1 activates the Cebpa promoter modestly, via two conserved non-consensus binding sites, and strongly activates ...Missing: TATA | Show results with:TATA
  9. [9]
    Expression and regulation of C/EBPα in normal myelopoiesis and in ...
    Apr 13, 2017 · CEBPA is an intronless gene located on chromosome 19q in humans and on chromosome 7 in mice, which encodes a 42-kD and a 30-kD DNA-binding ...
  10. [10]
    C/EBPαp30 plays transcriptional regulatory roles distinct from C ...
    Apr 16, 2007 · C/EBPα is highly expressed in a variety of tissues including liver, lung, placenta, and white and brown adipose tissues. As a transcription ...<|separator|>
  11. [11]
    Mutant CEBPA directly drives the expression of the targetable tumor ...
    Jul 10, 2019 · The intron-less CEBPA gene is transcribed into a single mRNA, which is translated into two isoforms by alternative start codon usage: the full- ...
  12. [12]
    Gain-of-Function Effects of N-Terminal CEBPA Mutations in Acute ...
    Dec 23, 2019 · This review outlines direct and indirect effects of the C/EBPα p30 variant on oncogenic transformation of hematopoietic progenitor cells.<|control11|><|separator|>
  13. [13]
    Block of C/EBPα function by phosphorylation in acute myeloid ... - NIH
    Expression of C/EBPα mutant lacking Ser21 phosphorylation relieves the differentiation block in FLT3-ITD human AML cells. We have demonstrated that C/EBPα with ...Missing: sumoylation | Show results with:sumoylation
  14. [14]
    Differential effects of sumoylation on the activities of CCAAT ...
    In this study, we have examined the role of post-translational modification of the myeloid master regulator C/EBPα by small ubiquitin-related modifier (SUMO).
  15. [15]
    CEBPA (CCAAT/enhancer binding protein (C/EBP), alpha)
    Jul 1, 2014 · Human C/EBPα is an intronless gene located on the minus strand of chromosome 19q. Transcription. The C/EBPa mRNA (RNA messenger) consists of ...Missing: structure exon intron<|control11|><|separator|>
  16. [16]
    Acetylation of C/EBPα inhibits its granulopoietic function - Nature
    Mar 23, 2016 · The role of C/EBPα in granulopoiesis is demonstrated through Mx1-Cre-driven conditional disruption of C/EBPα in adult mice, resulting in a ...
  17. [17]
    Impaired Energy Homeostasis in C/EBPα Knockout Mice - Science
    This study demonstrates that C/EBPα is critical for the establishment and maintenance of energy homeostasis in neonates.
  18. [18]
    C/EBPα induces adipogenesis through PPARγ: a unified pathway
    These results indicate that C/EBPα and PPARγ participate in a single pathway of fat cell development with PPARγ being the proximal effector of adipogenesis.
  19. [19]
    C/EBPα determines hematopoietic cell fate in multipotential ... - NIH
    We found increased numbers of erythroid progenitors and erythroid cells in C/EBPα–/– fetal liver (FL). Also, enforced expression of C/EBPα in hematopoietic ...
  20. [20]
    Respiratory Failure Due to Differentiation Arrest and Expansion of ...
    This differentiation arrest results in the lack of type I alveolar cells and differentiated surfactant-secreting type II alveolar cells. In addition to showing ...
  21. [21]
    Expression and regulation of C/EBPα in normal myelopoiesis and in ...
    Apr 13, 2017 · In this review, we emphasize the importance of C/EBPα for neutrophil maturation, its role in myeloid priming of hematopoietic stem and progenitor cells,
  22. [22]
  23. [23]
    A mini-review of CEBPA variants in patients with acute myeloid ... - NIH
    CEBPA mutations occur in ∼10% of AML with only 4% of patients having bi-allelic mutations [5]. Recent evidence from three large datasets demonstrated that only ...
  24. [24]
    CCAAT enhancer binding protein alpha (CEBPA) biallelic acute ...
    In normal cells, p42 is dominant, but stop and frameshift mutations within the 5′‐end of the one‐exon CEBPA gene, sustain p30 expression at the expense of p42.
  25. [25]
  26. [26]
  27. [27]
  28. [28]
  29. [29]
  30. [30]
    Methylation status of CEBPA gene promoter in chronic myeloid ...
    Nov 25, 2013 · Hypermethylation of C/EBPα promoter has also been identified in 12–51% of acute myeloid leukemia cases. In BCR–ABL-positive CML ...
  31. [31]
    The CEBPA gene is down-regulated in acute promyelocytic ... - NIH
    Epigenetic regulation of CEBPA expression through DNA methylation has been demonstrated in lung cancer and head and neck squamous cell carcinoma. Tada et al.
  32. [32]
    CEBPA methylation as a prognostic biomarker in patients ... - PubMed
    Hypermethylation of the distal CEBPA promoter region has been reported to result in the downregulation of CEBPA expression in several malignancies.
  33. [33]
    Epigenetic Modifications in Acute Myeloid Leukemia - Frontiers
    Feb 28, 2019 · The PML-RARa product works in tandem with DNA methyltransferases (DNMTs) to induce hypermethylation in PML-RARa targets, particularly RARβ ...
  34. [34]
    DNMT3A Mutations in Acute Myeloid Leukemia
    Nov 10, 2010 · The PML-RARA fusion protein, which is created by t(15;17), physically interacts with DNMT3A, and AML-ETO, which is created by t(8;21) ...
  35. [35]
    The CEBPA gene is down-regulated in acute promyelocytic ...
    Apr 1, 2011 · The CEBPA gene is down-regulated in acute promyelocytic leukemia and its upstream promoter, but not the core promoter, is highly methylated.Missing: TATA box
  36. [36]
    DNA methylation landscape reveals LIN7A as a decitabine ...
    Mar 3, 2023 · And the methylation-silencing genes, LIN7A, CEBPA, BASP1, and EMB were identified as critical decitabine-sensitive genes in t(8;21) AML.
  37. [37]
    [PDF] Impact of MLL5 expression on decitabine efficacy and DNA ...
    In addition, decitabine-induced demethylation was associated with an increase in Cebpa mRNA expression, suggesting that Cebpa is regulated through DNA methyla-.
  38. [38]
    MiR-182 is up-regulated and targeting Cebpa in hepatocellular ...
    However it is unknown whether CEBPA is regulated by miRNAs in HCC. In this study, we find that miR-182 is upregulated in HCC model rat, and represses CEBPA in ...
  39. [39]
    MicroRNAs and acute myeloid leukemia: therapeutic implications ...
    Sep 14, 2017 · Studies have identified miRNAs as playing a role in nearly all aspects of AML disease development, including cellular proliferation, survival, and ...
  40. [40]
    C/EBPα in normal and malignant myelopoiesis
    Mar 10, 2015 · In addition, C/EBPα cooperates with PU.1, c-Myb, and RUNX1 to activate genes such as myeloperoxidase, neutrophil elastase, lysozyme, lactoferrin ...
  41. [41]
    AP-1 protein induction during monopoiesis favors C/EBP - PubMed
    ... C/EBPα and C/EBPβ, indicating the feasibility of heterodimer formation. Induction of AP-1 proteins during monocytic differentiation favored formation of C ...<|separator|>
  42. [42]
    Repression of Transcriptional Activity of C/EBPα by E2F ... - NIH
    The data presented here show that, independently of pocket proteins, C/EBPα interacts with the dimerization partner (DP) of E2F and that C/EBPα-E2F/DP ...
  43. [43]
    PU.1 promotes cell cycle exit in the murine myeloid lineage ...
    Restoration of PU.1 expression in Sfpi1BN/BN myeloid cells blocked proliferation, induced differentiation, and reduced E2F1 expression. Taken together, these ...Missing: repressors | Show results with:repressors
  44. [44]
    RUNX1/CEBPA Mutation in Acute Myeloid Leukemia Promotes ...
    In a recent study, Suzuku et al. showed, through co-immunoprecipitation, that RUNX1 and CEBPA directly interacts with TET2 and, in this way, could recruit it to ...
  45. [45]
    C/EBPɑ is crucial determinant of epithelial maintenance by ... - Nature
    Feb 7, 2020 · CEBPA is a SMAD3-repressed target during TGF-β-mediated EMT. To identify novel transcription factors with a potential epithelial-gatekeeper ...
  46. [46]
    NF-κB p50 Regulates C/EBPα Expression and Inflammatory ...
    Thus, C/EBPα regulates genes that control differentiation, such as CEBPA and NE, independent of p50 but cooperates with p50 to activate genes that inhibit ...
  47. [47]
    C/EBPβ: The structure, regulation, and its roles in inflammation ...
    Dec 31, 2023 · The expression of C/EBPβ is mediated at the transcriptional and post-translational levels, such as phosphorylation, acetylation, methylation, ...
  48. [48]
  49. [49]
    RUNX1: A MicroRNA Hub in Normal and Malignant Hematopoiesis
    In one circuit, CEBPA (CCAAT/enhancer-binding protein alpha) is induced by granulocyte colony stimulating factor during granulopoiesis and is part of a feed- ...<|control11|><|separator|>
  50. [50]
    Acute Myeloid Leukemia With CEBPA Mutations - Frontiers
    Jan 31, 2022 · The frequency of CEBPA mutations in AML is 6.86%–20.33%, and a higher incidence rate is observed in AML patients from Asia compared to that in ...
  51. [51]
    Acute Myeloid Leukemia With CEBPA Mutations - NIH
    Feb 1, 2022 · Patients with CEBPA mutations had significantly improved EFS and OS, and lower cumulative incidence rate of relapse compared to those with wide- ...
  52. [52]
    What is new in acute myeloid leukemia classification? - PMC - NIH
    Apr 15, 2024 · In WHO2022, AML with CEBPA mutation requires > 20% blasts. TP53 mutation, a new entity is exclusive to ICC, diagnosed with > 20% blasts and ...
  53. [53]
    Sporadic and Familial Acute Myeloid Leukemia with CEBPA Mutations
    Jun 1, 2023 · Most AML-CEBPA cases are sporadic and harbor somatic CEBPA mutations, while approximately 10% of individuals inherit or develop de novo ...Cebpa Protein Structure · Sporadic Aml With Cebpa... · Familial Aml With Germline...<|control11|><|separator|>
  54. [54]
    CEBPA mutations in 4708 patients with acute myeloid leukemia
    Jan 6, 2022 · In contrast, with both CEBPAp42 and CEBPAp30, mutations in the DBD and bZIP domains result in loss of DNA binding as well as dimerization.Results · Association Of Cebpa... · Prognostic Classification...Missing: evolutionary | Show results with:evolutionary<|control11|><|separator|>
  55. [55]
    Prognostic impact of CEBPA mutational subgroups in adult AML
    Jan 16, 2024 · These results demonstrate bZIP InDel mutations to be the major independent determinant of outcome in CEBPA-mutant AML.Missing: evolutionary | Show results with:evolutionary
  56. [56]
    Modeling of C/EBPα Mutant Acute Myeloid Leukemia Reveals a ...
    Apr 8, 2008 · These results demonstrate that, in a mouse model, patient-derived CEBPA mutations are AML-initiating mutations, providing direct genetic validation of their ...