MLF1 Human

Myeloid Leukemia Factor 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Cell Cycle Regulation

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 .

Oncogenic and Tumor-Suppressive Roles

  • Pro-Tumorigenic:

    • Promotes proliferation and metastasis in intrahepatic cholangiocarcinoma (iCCA) via EGFR/AKT and Wnt/β-catenin pathways .

    • Upregulated in lung squamous cell carcinoma and esophageal cancers .

  • Anti-Tumorigenic:

    • Silencing via promoter hypermethylation is observed in gastric and nasopharyngeal cancers .

    • Stabilizes RUNX1-ETO in leukemia, impairing leukemic cell proliferation .

Immune Regulation

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 .

Hematologic Malignancies

  • 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 .

Solid Tumors

Cancer TypeMLF1 RoleMechanismReference
Intrahepatic CholangiocarcinomaPromotes invasiveness and EMTUpregulates EGFR, AKT, WNT3, and p-GSK3β
Gastric CancerTumor suppressorHypermethylation silences MLF1, promoting tumorigenesis
NeuroblastomaOncogeneSilencing MLF1 suppresses tumor proliferation

In Vitro and In Vivo Studies

  • iCCA Models:

    • Knockdown reduces cell proliferation by 40–60% and inhibits colony formation .

    • Overexpression increases lung/liver metastasis in xenograft models .

  • Leukemia Models:

    • NPM-MLF1 fusion requires NES for oncogenic transformation .

Epigenetic Regulation

  • 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 .

Therapeutic Implications

  • Targeting MLF1 Pathways: Inhibition of EGFR/AKT or Wnt signaling may benefit iCCA patients with high MLF1 expression .

  • Epigenetic Modulators: DNMT inhibitors could reactivate MLF1 in methylation-silenced cancers .

Product Specs

Introduction
Myeloid leukemia factor 1 (MLF1), a member of the MLF family, is a widely expressed protein that acts as a negative regulator of cell cycle progression. It functions upstream of the tumor suppressor protein p53. MLF1 inhibits the differentiation of erythroid cells induced by erythropoietin by preventing cell cycle exit. This is achieved through the suppression of CDKN1B/p27Kip1 levels. Primarily active in multi-potent progenitor cells, MLF1 dysregulation is potentially implicated in the development of leukemia. Notably, translocations involving the MLF1 gene and nucleophosmin are associated with myelodysplastic syndrome and acute myeloid leukemia.
Description
This product consists of the human MLF1 protein, recombinantly produced in E. coli. A 20 amino acid His tag is fused at the N-terminus. The protein is a single, non-glycosylated polypeptide chain containing 288 amino acids (residues 1-268) with a molecular mass of 32.8 kDa. Note that the molecular weight observed on SDS-PAGE will appear higher. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The MLF1 protein is supplied in a solution at a concentration of 0.5 mg/ml. The solution contains 20mM Tris-HCl buffer (pH 8.0), 40% glycerol, 5mM DTT, and 200mM NaCl.
Stability
For short-term storage (up to 2-4 weeks), keep at 4°C. For longer periods, store frozen at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Minimize repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 85.0% by SDS-PAGE analysis.
Synonyms
Myeloid leukemia factor 1, Myelodysplasia-myeloid leukemia factor 1, MLF1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MFRMLNSSFE DDPFFSESIL AHRENMRQMI RSFSEPFGRD LLSISDGRGR AHNRRGHNDG EDSLTHTDVS SFQTMDQMVS NMRNYMQKLE RNFGQLSVDP NGHSFCSSSV MTYSKIGDEP PKVFQASTQT RRAPGGIKET RKAMRDSDSG LEKMAIGHHI HDRAHVIKKS KNKKTGDEEV NQEFINMNES DAHAFDEEWQ SEVLKYKPGR HNLGNTRMRS VGHENPGSRE LKRREKPQQS PAIEHGRRSN VLGDKLHIKG SSVKSNKK.

Q&A

What is MLF1 and what are its primary functions in human 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 .

How is MLF1 expression altered during cardiac aging?

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 .

What cellular processes does MLF1 regulate in cardiomyocytes?

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

How does MLF1 regulate chromatin structure and gene accessibility?

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 .

What is the relationship between MLF1 and the Polycomb Repressive Complex 2 (PRC2)?

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 .

How does the MLF1-EP300 axis contribute to gene regulation?

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 .

What techniques are most effective for studying MLF1 binding to chromatin?

The following methodologies have proven effective for investigating MLF1's chromatin interactions:

TechniqueApplicationKey Insights
CUT&TagMapping MLF1 binding sitesReveals preference for promoters and transcription start sites
ATAC-seqAssessing chromatin accessibilityDemonstrates MLF1's role in facilitating chromatin opening
ChIP-seq/CUT&Tag for H3K27acIdentifying acetylation patternsShows correlation between MLF1 binding and active chromatin marks
Motif analysisIdentifying recognition sequencesDiscovers repetitive 'AATGG' motif in MLF1-bound regions
Co-immunoprecipitationStudying protein interactionsConfirms MLF1 interaction with EP300 and PRC2 components

How can MLF1 expression be effectively manipulated in experimental models?

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.

What assays are most informative for measuring MLF1-related cellular phenotypes?

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

  • Chromatin fractionation to confirm MLF1 localization

How should multi-omics data be integrated to understand MLF1 function?

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 .

What bioinformatic approaches best identify MLF1 target genes?

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

  • Conduct pathway enrichment analysis of target genes

This approach has successfully identified inflammation-associated genes including IL1B, IL4R, and PDGFRA as direct MLF1 targets .

How might MLF1 serve as a therapeutic target in age-related cardiac conditions?

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

How does MLF1 function within the broader context of cardiac aging mechanisms?

MLF1 functions within a network of age-associated cardiac pathways:

PathwayRelationship to MLF1Functional Significance
Inflammatory signalingMLF1 promotes IL1B expressionDrives senescence-associated inflammation
Oxidative stress responseMLF1 sensitizes cells to H₂O₂ damageContributes to age-related oxidative damage
Chromatin remodelingMLF1 facilitates chromatin openingEnables expression of pro-senescence genes
Cell cycle regulationMLF1 affects p21 expressionInfluences senescence-associated growth arrest

These interactions position MLF1 as a central regulator in cardiac aging, connecting epigenetic mechanisms with inflammatory and senescence pathways .

What are the most promising areas for future MLF1 research?

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

What methodological advances would accelerate MLF1 research?

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

Product Science Overview

Introduction

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) .

Discovery and Structure

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.

Biological Functions

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 .

Role in Disease

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 .

Research and Therapeutic Potential

Research on MLF1 is ongoing, with studies exploring its potential as a therapeutic target. Understanding the molecular mechanisms by which MLF1 influences cell cycle regulation and apoptosis could lead to the development of novel therapies for AML and other hematologic malignancies .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.