MLF1 suppresses COP1 activity via CSN3, stabilizing p53 and inducing cell cycle arrest . It inhibits erythroid differentiation by downregulating CDKN1B/p27Kip1, favoring myeloid lineage commitment .
Pro-Tumorigenic:
Anti-Tumorigenic:
MLF1 interacts with DnaJ-1 to modulate RUNX transcription factors and Notch signaling, influencing hematopoietic stem cell development . In shrimp, MLF1 homologs exhibit antiviral and antibacterial activity .
Leukemia: The t(3;5)(q25.1;q34) translocation generates the NPM-MLF1 fusion protein, linked to myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) .
Prognostic Marker: High MLF1 expression correlates with poor survival in iCCA (median OS: 14.2 vs. 34.4 months) and AML .
iCCA Models:
Leukemia Models:
MLF1 expression in iCCA is driven by miR-29c-3p downregulation due to DNMT1/3A/3B hypermethylation .
5-AZA-dC (demethylating agent) restores MLF1 expression in gastric cancer cells .
MLF1 is a transcription regulator highly expressed in the heart, testis, lung, and skeletal muscle. Research demonstrates that MLF1 primarily functions as a transcription activator that governs chromatin accessibility, particularly at promoter regions . In human cardiomyocytes, MLF1 plays a significant role in cell senescence processes, with its downregulation serving as a protective mechanism against oxidative stress-induced senescence . Molecularly, MLF1 facilitates gene expression related to inflammatory responses, TGF-beta, Wnt, and interleukin-1 signaling pathways .
MLF1 expression significantly decreases in the aged heart compared to young heart tissue, as well as in H₂O₂-induced senescent AC16 cardiomyocytes . This downregulation appears to be a compensatory surveillance mechanism, as experimental evidence shows that reduced MLF1 expression protects cardiomyocytes against senescence . MLF1 has been identified as one of four reliable biomarkers for age-associated cardiac hypertrophy (AACH) that are also sensitive to anti-aging treatments .
In human AC16 cardiomyocytes, MLF1 primarily regulates:
Cellular senescence (silencing MLF1 suppresses H₂O₂-induced senescence)
Expression of senescence markers including P21, IL1B, and IL6
Inflammatory responses (MLF1 knockdown downregulates inflammation-related genes)
Apoptosis (MLF1 silencing alleviates late apoptosis after H₂O₂ treatment)
Chromatin accessibility, particularly at promoter regions of target genes
MLF1 functions as a critical epigenetic regulator that promotes chromatin opening. ATAC-seq analysis has demonstrated that MLF1 knockdown results in 8825 closed chromatin peaks (30.8%) versus only 140 open peaks (0.05%), with pronounced effects at promoter regions . MLF1 primarily recognizes a repetitive 'AATGG' motif and binds predominantly at transcription start sites . Functionally, MLF1 recruits the histone acetyltransferase EP300 to deposit H3K27ac at target promoters, creating an open chromatin environment that facilitates transcription of genes involved in inflammatory responses and senescence pathways .
MLF1 physically interacts with components of the PRC2 complex, including EZH2, SUZ12, EED, and RBBP4/RbAp48 . Structural docking analyses suggest MLF1 binds to PRC2 through SUZ12 with an interface area of 3388.7 Ų and a ΔiG of 20.1 kcal/mol . Co-immunoprecipitation experiments confirm this interaction, and immunofluorescence assays show remarkable co-localization between MLF1 and EZH2/SUZ12 within the nucleus . Genes regulated by MLF1 show enrichment for H3K27me3-mediated regulations, suggesting a potential interplay between MLF1 and PRC2-mediated silencing .
MLF1 recruits EP300 to facilitate transcription of its target genes, as demonstrated by:
Global bound peaks of both EP300 and H3K27ac showing strong correlations with MLF1 distribution across the genome
Substantial reduction in EP300 and H3K27ac enrichment after MLF1 knockdown, both genome-wide and at specific promoters
Significant overlap between genes affected by MLF1 knockdown and EP300 knockdown (38.6% of upregulated genes and 48.2% of downregulated genes)
This axis is particularly important for regulating inflammation-associated genes, with IL1B and p21 identified as prominent targets .
The following methodologies have proven effective for investigating MLF1's chromatin interactions:
For functional studies of MLF1, researchers have successfully employed:
RNA interference (siRNA) for transient knockdown in AC16 cardiomyocytes
Plasmid-based overexpression systems
Combined approaches to validate phenotypic effects (e.g., rescue experiments)
When manipulating MLF1, researchers should verify expression changes at both mRNA and protein levels, and design experiments to control for off-target effects.
The following assays have proven valuable for assessing MLF1's cellular effects:
β-galactosidase staining to quantify senescent cells
qRT-PCR measurement of senescence markers (P21, IL1B, IL6)
Flow cytometry for cell cycle and apoptosis analysis
Inflammatory cytokine expression profiling
Comprehensive understanding of MLF1 function requires integration of multiple data types:
Transcriptome data (RNA-seq) to identify MLF1-regulated genes
Chromatin accessibility data (ATAC-seq) to determine effects on chromatin structure
Protein-DNA interaction data (CUT&Tag) to map direct binding sites
Histone modification profiles (H3K27ac, H3K27me3) to understand epigenetic mechanisms
Successful integration has revealed that approximately 10.72% of genes with closed chromatin peaks after MLF1 knockdown show corresponding transcriptional downregulation, identifying direct MLF1 targets .
The following analytical pipeline has proven effective:
Identify genes with altered expression after MLF1 manipulation (RNA-seq)
Overlay with chromatin accessibility changes (ATAC-seq)
Filter for direct MLF1 binding at promoters (CUT&Tag)
Combine with histone modification data (H3K27ac CUT&Tag)
Perform motif analysis to identify sequence preferences
This approach has successfully identified inflammation-associated genes including IL1B, IL4R, and PDGFRA as direct MLF1 targets .
MLF1 represents a promising therapeutic target based on several observations:
MLF1 silencing protects cardiomyocytes against H₂O₂-induced senescence
MLF1 knockdown suppresses expression of inflammatory mediators
MLF1 promotes chromatin accessibility at senescence-related gene promoters
MLF1 is downregulated in aged hearts, suggesting a natural compensatory mechanism
Potential therapeutic strategies might include:
Small molecule inhibitors disrupting MLF1-EP300 interaction
Targeted approaches to reduce MLF1 expression in cardiac tissue
Blockade of downstream effectors like IL1B, which partially mediates MLF1's pro-senescence effects
MLF1 functions within a network of age-associated cardiac pathways:
These interactions position MLF1 as a central regulator in cardiac aging, connecting epigenetic mechanisms with inflammatory and senescence pathways .
Based on current findings, several research directions warrant further investigation:
Tissue-specific functions of MLF1 beyond cardiomyocytes
MLF1's role in other age-related pathologies
Detailed structural studies of MLF1-EP300 and MLF1-PRC2 interactions
Development of small molecule modulators of MLF1 activity
In vivo validation of MLF1 as a therapeutic target using animal models
Investigation of potential MLF1 genetic variants associated with cardiac aging
Advancements that would significantly enhance MLF1 research include:
Development of high-quality, highly specific antibodies for MLF1
Single-cell multi-omics approaches to understand cellular heterogeneity in MLF1 function
CRISPR-based screening to identify genetic modifiers of MLF1 activity
Advanced protein structure prediction and modeling to enable drug development
In vivo chromatin profiling techniques to confirm MLF1 functions in intact tissues
Myeloid Leukemia Factor 1 (MLF1) is a small nucleocytoplasmic shuttling protein that plays a significant role in cell cycle regulation, apoptosis, and immune functions. It is associated with both normal hematopoiesis and malignancies, particularly acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) .
MLF1 was initially identified in the leukemic fusion protein NPM-MLF1, which is generated by a rare t(3;5)(q25.1;q34) chromosomal translocation in patients with AML . The protein is characterized by its ability to shuttle between the nucleus and cytoplasm, which is crucial for its function in regulating various cellular processes.
MLF1 has diverse biological functions, acting as either a tumor suppressor or an oncogene depending on the cellular context. In hematopoietic cells, MLF1 serves as a protective factor for lineage development. It regulates cell cycle exit and differentiation, promotes apoptosis, inhibits proliferation, and enhances immune function .
MLF1’s role in disease is complex and context-dependent. In the context of AML and MDS, MLF1 is implicated in oncogenesis. The fusion protein NPM-MLF1 disrupts normal cellular functions, contributing to the development of these malignancies . However, MLF1 can also act as a tumor suppressor in certain contexts, highlighting its dual role as a “double-edged sword” in health and disease .