DLK1 Human

Delta-Like 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to DLK1

Delta-like 1 homolog (DLK1), also known as Fetal Antigen 1 (FA1) or Preadipocyte Factor 1 (Pref-1), is a single-pass transmembrane protein belonging to the Notch/Delta/Serrate family. Encoded by the imprinted DLK1-DIO3 gene cluster on human chromosome 14q32, DLK1 plays critical roles in embryonic development, stem cell regulation, and cancer progression . While highly expressed during fetal development, its expression in adults is largely restricted to endocrine tissues, progenitor cells, and malignancies .

Molecular Structure and Isoforms

DLK1 is synthesized as a 383-amino acid precursor with distinct domains:

  • Extracellular region: Six epidermal growth factor (EGF)-like repeats and a juxtamembrane ADAM17 cleavage site .

  • Transmembrane domain: Anchors the protein to the cell membrane.

  • Intracellular tail: Short cytoplasmic region with no known signaling motifs .

Key Isoforms:

SpeciesIsoformsFeatures
MouseA, B, C, C2, D, D2Only A/B isoforms are cleavable; others remain membrane-tethered .
Human2 isoformsOne cleavable (50 kDa soluble form) and one membrane-tethered .

Post-translational cleavage by ADAM17 releases soluble DLK1, which exerts distinct biological effects compared to its membrane-bound form .

Expression Patterns in Physiology

DLK1 exhibits tissue-specific expression:

Tissue/Cell TypeExpression Pattern
EmbryonicHigh in liver, pancreas, adrenal glands, and CNS .
AdultLow; restricted to adrenal cortex, pancreatic β-cells, and osteoblasts .
Stem/Progenitor CellsHepatoblasts, mesenchymal stem cells, and neural precursors .

In the adult hippocampus, DLK1 maintains neural stem cell (NSC) quiescence and promotes neurogenesis, with dosage sensitivity impacting cognitive functions like spatial memory .

Skeletal Muscle:

  • Ablation: Reduces myofiber numbers and impairs regeneration by enhancing NF-κB signaling .

  • Overexpression: Inhibits myoblast proliferation but enhances differentiation .

Adipogenesis:

  • Soluble DLK1 suppresses preadipocyte differentiation, while membrane-bound DLK1 inhibits proliferation .

Neurogenesis:

  • Biallelic DLK1 expression in hippocampal NSCs is essential for postnatal neurogenesis and cognitive function .

DLK1 in Cancer

DLK1 is overexpressed in multiple malignancies, correlating with poor prognosis:

Cancer TypeRole of DLK1Mechanism
Hepatocellular CarcinomaPromotes tumor invasionActivates Notch signaling; marker of cancer stem cells .
Adrenocortical CarcinomaAssociated with metastasisMaintains undifferentiated phenotype .
Breast CancerEnhances chemoresistanceInhibits differentiation via Notch cis-inhibition .

DLK1 is detectable in serum, making it a potential biomarker for malignancies and heart failure .

Notch Pathway Interaction:

DLK1 lacks the DSL domain required for canonical Notch activation but modulates signaling via:

  • Cis-inhibition: Competes with canonical ligands (e.g., DLL1) for Notch receptor binding .

  • Non-canonical signaling: Promotes β-catenin and Akt pathways in muscle regeneration .

Soluble vs. Membrane-Bound Forms:

FormFunction
Soluble DLK1Inhibits adipogenesis and myotube formation .
Membrane-Bound DLK1Promotes myotube hypertrophy and cancer stemness .

Heart Failure:

CRISPRa-mediated DLK1 activation in engineered human myocardium attenuates contractile dysfunction by inducing a regenerative transcriptome (e.g., enhanced ECM remodeling and metabolism) .

Immunomodulation:

DLK1-expressing mesenchymal stromal cells enhance T-cell development and antiviral responses in humanized mice .

Recombinant DLK1 Products:

ProductSourceApplications
Recombinant Human DLK1 (AdipoGen)HEK293 cellsUsed in studies on adipogenesis, myogenesis, and cancer .
Recombinant Human Pref-1 (R&D Systems)Mammalian expressionBioactivity assays for Notch signaling .

Key Research Findings

  1. Muscle Regeneration: DLK1 ablation increases inflammatory cytokines (e.g., TNF-α) and impairs satellite cell differentiation .

  2. Neurocognitive Effects: DLK1 haploinsufficiency in mice causes spatial memory deficits and reduced hippocampal neurogenesis .

  3. Cancer Biomarker: Serum DLK1 levels correlate with tumor burden in hepatocellular carcinoma .

Product Specs

Introduction
Delta-Like 1, also called DLK1, is a protein that belongs to the Notch/Delta/Serrate family. This family is known for its role in the Notch signaling pathway, where the proteins act as ligands (like keys) that activate the pathway. DLK1 has six special regions called epidermal-growth-factor motifs, which are important for its function. It also has specific sites where sugar molecules can attach (glycosylation), and these sugar modifications can vary.
Description
This product contains human DLK1 protein made in insect cells (Sf9 Baculovirus). It's a single chain of 288 amino acids, with a molecular weight of 30.7kDa. The protein has a small tag (His-tag) attached to aid in purification, and it's purified to a high degree. The protein is glycosylated, meaning it has sugars attached, which is important for its natural properties.
Physical Appearance
The product appears as a clear liquid that has been sterilized by filtration.
Formulation
The DLK1 protein is provided in a solution at a concentration of 0.25mg/ml. The solution contains Phosphate Buffered Saline (PBS) with a pH of 7.4 and 10% glycerol for stability.
Stability
For short-term storage (up to 4 weeks), keep the product refrigerated at 4°C. For longer storage, freeze the product at -20°C. It's best to add a carrier protein (0.1% HSA or BSA) before freezing to protect the DLK1 protein. Avoid repeatedly freezing and thawing the product as this can damage the protein.
Purity
The purity of the DLK1 protein is greater than 95%, as determined by SDS-PAGE, a common method for analyzing protein purity.
Synonyms
DLK1, Delta-Like 1 Homolog (Drosophila), PREF1, DLK-1, FA1, DLK, PG2, Delta-Like Homolog (Drosophila), Protein Delta Homolog 1, Preadipocyte Factor 1, Fetal Antigen 1, Secredeltin, DELTA1, Pref-1, ZOG.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
AECFPACNPQ NGFCEDDNVC RCQPGWQGPL CDQCVTSPGC LHGLCGEPGQ CICTDGWDGE LCDRDVRACS SAPCANNGTC VSLDDGLYEC SCAPGYSGKD CQKKDGPCVI NGSPCQHGGT CVDDEGRASH ASCLCPPGFS GNFCEIVANS CTPNPCENDG VCTDIGGDFR CRCPAGFIDK TCSRPVTNCA SSPCQNGGTC LQHTQVSYEC LCKPEFTGLT CVKKRALSPQ QVTRLPSGYG LAYRLTPGVH ELPVQQPEHR ILKVSMKELN KKTPLLTEGQ LEHHHHHH.

Q&A

What is the genomic location and structure of human DLK1?

DLK1 is located within an imprinted domain on chromosome band 14q32 in humans (12qF1 in mice). It is a paternally expressed gene within the DLK1-DIO3 imprinted domain, which contains three paternally expressed protein-coding genes (DLK1, RTL1, and DIO3) and several maternally expressed non-coding RNAs including MEG3 and MEG8 .

Structurally, DLK1 encodes a 383 amino acid, single-pass, transmembrane glycoprotein with notable similarity to canonical Notch ligand Delta-like 1 (DLL1). The protein contains six EGF-like repeats, a juxtamembrane region with a cleavage site, a transmembrane domain, and a small intracellular tail. The cleavage site is mediated by ADAM17 (also known as TNFα converting enzyme or TACE), which enables the release of a soluble form of DLK1 .

Two main isoforms of DLK1 have been identified in humans: one that is membrane-tethered and another that is cleavable. This contrasts with mice, which have six recognized isoforms (Dlk1 A, B, C, C2, D, D2), with only A and B containing the ADAM17 cleavage site .

How should researchers approach DLK1 expression analysis in different human tissues?

When studying DLK1 expression patterns, researchers should consider both developmental stage and tissue specificity. During embryonic development, DLK1 is widely expressed across multiple human tissues, whereas in adults, expression becomes restricted primarily to neuroendocrine tissues and immature stem/progenitor cell populations .

For experimental design, consider these methodological approaches:

  • Immunohistochemistry/Immunofluorescence: Essential for spatial localization in tissues. For example, in adrenal glands, DLK1+ cells remodel from a continuous subcapsular layer during development and childhood to form cell clusters in the third decade of life .

  • Single-cell RNA sequencing: Particularly valuable for heterogeneous tissues to identify specific DLK1-expressing cell populations.

  • Imprinting analysis: Use allele-specific expression analysis techniques to assess imprinting status, as DLK1 shows varied imprinting patterns. For instance, while canonically imprinted in most tissues, DLK1 exhibits biallelic expression in the subgranular zone (SGZ) of the hippocampus .

  • Temporal studies: Design longitudinal analyses as expression patterns change throughout development and aging. In adrenal tissues, for example, significant remodeling of DLK1+ cells occurs over decades .

When comparing normal versus pathological tissues, researchers should control for cell type and developmental stage, as high DLK1 expression in cancer may reflect a reversion to developmental programs rather than simply upregulation .

What experimental techniques are effective for modulating DLK1 expression in human cell models?

When experimentally manipulating DLK1 expression, several approaches have proven effective:

  • Inducible shRNA systems: Tetracycline-inducible short hairpin RNA systems allow for controlled knockdown of DLK1 expression. This approach has successfully inhibited proliferation, spheroid formation, and xenograft tumor growth in human hepatocellular carcinoma (HCC) cell models .

  • Adenoviral vector delivery: This has been effectively used for both human and mouse models. In orthotopic xenograft models, adenovirus-mediated DLK1 knockdown significantly reduced tumor size .

  • Exogenous DLK1 application: Adding DLK1 as an extrinsic factor to cell culture promotes neurogenesis in human and mouse ESC-derived neural progenitors .

  • Transgene expression: Stable expression of DLK1 transgenes provides an alternative approach for studying gain-of-function effects .

For analytical validation:

  • Verify knockdown efficiency via qRT-PCR and Western blotting

  • Assess functional outcomes through cell cycle analysis (flow cytometry)

  • Monitor differentiation markers (e.g., AFP, EpCAM for hepatic progenitors; KRT18, KRT19 for differentiated cells)

  • For in vivo models, combine interventions with imaging approaches to track tumor progression longitudinally

How does DLK1 dosage affect hippocampal neurogenesis and cognitive function?

DLK1 demonstrates remarkable dosage sensitivity in neurogenesis, with both parental alleles required for normal adult hippocampal function, unlike its canonical imprinting pattern in non-neurogenic brain regions. Researchers investigating this phenomenon should consider:

  • Allele-specific experimental design: Studies should account for DLK1's biallelic expression in the subgranular zone (SGZ) by using experimental designs that can distinguish maternal from paternal allelic contributions. Genetic deletion of DLK1 from either parental allele results in impaired neural stem cell (NSC) function .

  • Dosage quantification: Precise quantification of DLK1 levels is critical, as both under- and over-expression impair NSC function. When investigating DLK1 dosage effects, establish a dose-response relationship rather than simple presence/absence comparisons .

  • Functional readouts: Incorporate multiple assessment methods:

    • Cellular: NSC quiescence markers, proliferation rates

    • Molecular: Expression of downstream effectors

    • Behavioral: Spatial learning and memory tests, anxiety assessments

  • Controls for developmental effects: Since DLK1 plays roles in both development and adult function, use conditional knockouts or inducible systems to distinguish between developmental defects and adult-specific functions .

The experimental approach should include behavioral testing combined with cellular and molecular analyses to establish causation between DLK1 dosage, neurogenesis levels, and cognitive outcomes.

What mechanisms underlie DLK1's role in promoting neurogenesis in human neural progenitors?

DLK1 promotes neurogenesis in human neural progenitors through at least two distinct but interconnected molecular pathways. Researchers investigating these mechanisms should focus on:

  • Notch signaling antagonism: DLK1 inhibits Hes1-mediated Notch signaling, as demonstrated through luciferase reporter assays. This antagonism specifically counteracts the cell proliferation activity of canonical Notch ligands Delta 1 and Jagged .

  • BMP/Smad signaling modulation: DLK1 promotes neurogenic potential of human neural progenitors via suppression of Smad activation when cells are challenged with BMP. This represents a novel regulatory pathway distinct from typical Notch interactions .

  • Cell cycle regulation: DLK1 induces cell cycle exit of progenitors, evidenced by increased numbers of young neurons retaining BrdU labeling and enhanced expression of cycling inhibitor P57Kip2 .

Experimental approaches should include:

  • Parallel assessment of both signaling pathways within the same experimental system

  • Temporal analysis to determine the sequence of these regulatory events

  • Rescue experiments to establish causal relationships

  • Application of both exogenous DLK1 and transgene expression to distinguish cell-autonomous from non-cell-autonomous effects

For comprehensive mechanistic studies, researchers should monitor changes in the expression of downstream effectors in both pathways simultaneously and consider potential crosstalk between Notch and BMP/Smad signaling.

What are the methodological challenges in targeting DLK1 for therapeutic interventions in human cancers?

Targeting DLK1 for cancer therapy presents several methodological challenges that researchers must address:

The table below summarizes experimental approaches that have shown efficacy in preclinical models:

Therapeutic ApproachCancer TypeDelivery MethodExperimental ModelOutcome Measures
shRNA knockdownHCCTet-inducible systemHuman cell lines, xenograftsProliferation, spheroid formation, tumor growth
Adenoviral vectorHCCDirect tumor injectionOrthotopic xenograftsTumor size by imaging
Adenoviral vectorHCCSystemic deliveryDEN-induced mouse HCCTumor number, size, weight

For clinical translation, combinations with existing therapies should be explored to enhance efficacy while mitigating potential adverse effects on normal stem cell populations .

How can researchers effectively analyze DLK1's role in adrenocortical stem cells and adrenocortical carcinoma?

Investigating DLK1's role in adrenocortical stem cells and adrenocortical carcinoma (ACC) requires specialized methodological approaches:

  • Developmental timeline analysis: DLK1+ adrenocortical cells undergo significant remodeling from a continuous subcapsular layer during development to form distinctive cell clusters by the third decade of life. Longitudinal studies across different age groups are essential for capturing these developmental dynamics .

  • Genetic fate mapping: This approach has successfully identified DLK1-expressing adrenocortical stem cells that are active during development, nearly dormant postnatally, but reactivated in ACC. Researchers should implement Cre-lox systems with DLK1 promoter-driven expression to track cellular lineages .

  • Spatial transcriptomics: This technique has revealed that DLK1-expressing cell populations in human ACC have increased steroidogenic potential—a finding observed in both human and murine ACC cell lines. Integrating spatial information with expression data provides insights impossible to obtain through bulk sequencing approaches .

  • Prognostic correlation: When studying ACC samples, researchers should correlate DLK1 expression with clinical outcomes. Analysis of over 200 human ACC samples has demonstrated that DLK1 expression is an independent prognostic marker of recurrence-free survival .

  • Paradoxical function analysis: Despite its role as a progenitor marker, DLK1-expressing cells in ACC show increased steroidogenic potential—a characteristic typically associated with differentiated cells. This paradox requires careful experimental design that simultaneously assesses stemness and differentiation markers .

For comprehensive investigation, combine these approaches with standard techniques such as immunohistochemistry, flow cytometry, and genetic manipulation. Consider the unique embryological origin of the adrenal cortex when interpreting results in the context of cancer stem cell biology.

What experimental controls are essential when studying DLK1 imprinting in different human tissues?

DLK1 demonstrates unusual imprinting patterns across tissues, being canonically imprinted (paternally expressed) in most tissues but biallelically expressed in specific regions like the hippocampal subgranular zone (SGZ) . This complexity necessitates rigorous controls:

  • Allele-specific expression analysis: Use single nucleotide polymorphisms (SNPs) in the DLK1 locus to distinguish maternal and paternal allele expression. Both RNA-seq and allele-specific qPCR are suitable when properly controlled .

  • Tissue specificity controls: Always include multiple tissue types within the same experiment, with at least one tissue known to maintain canonical imprinting (e.g., liver) as a positive control .

  • Cell type resolution: Single-cell approaches are preferred in heterogeneous tissues like brain and adrenal gland, as bulk tissue analysis can mask cell type-specific imprinting patterns .

  • Developmental stage controls: Include samples from multiple developmental timepoints when possible, as imprinting status may change during development or aging .

  • Methylation analysis: Complement expression studies with analysis of differential methylation regions (DMRs) controlling imprinted expression. This provides mechanistic validation of expression findings .

  • Species comparisons: When possible, parallel analysis in human and mouse tissues provides insight into evolutionary conservation of imprinting patterns .

When interpreting results showing biallelic expression, researchers should establish whether this represents complete loss of imprinting or a partial relaxation of imprinting control by quantifying the relative contribution of each parental allele.

How can researchers differentiate between DLK1's direct effects and secondary consequences on cellular phenotypes?

Distinguishing direct effects of DLK1 from indirect consequences requires careful experimental design:

  • Temporal analysis: Implement time-course experiments following DLK1 manipulation to establish the sequence of molecular and cellular events. For example, in neural progenitors, analyze whether Notch signaling changes precede or follow alterations in cell cycle regulators .

  • Domain-specific mutations: Create DLK1 constructs with specific mutations in functional domains (EGF-like repeats, cleavage sites) to determine which protein regions mediate particular effects. This approach can separate membrane-bound versus soluble DLK1 functions .

  • Isoform-specific interventions: Target specific DLK1 isoforms using isoform-selective knockdown or overexpression to determine their differential contributions to phenotypes .

  • Pathway inhibition rescue experiments: Combine DLK1 manipulation with inhibitors of suspected downstream pathways. For example, when studying neurogenesis, combine DLK1 overexpression with Notch pathway activators or BMP/Smad inhibitors to test if these can rescue phenotypes .

  • Direct binding assays: Implement co-immunoprecipitation, proximity ligation assays, or FRET to establish direct protein-protein interactions between DLK1 and putative partners.

  • Transcriptional profiling with kinetic analysis: Use RNA-seq at multiple timepoints after DLK1 manipulation to distinguish immediate early gene responses (potential direct targets) from later transcriptional changes (likely secondary effects) .

When analyzing cell cycle effects, researchers should note that DLK1 knockdown delays the G1 to S phase transition, leading to G1 arrest without obvious apoptosis. This suggests a specific role in cell cycle regulation rather than general cytotoxicity .

What are promising approaches for investigating DLK1's role in cancer stem cell maintenance?

Several methodological approaches show promise for investigating DLK1's specific role in cancer stem cell (CSC) maintenance:

  • Lineage tracing in patient-derived xenografts: Implement DLK1 promoter-driven reporters combined with serial transplantation assays to track the fate of DLK1+ cells during tumor initiation, growth, and recurrence .

  • Single-cell multi-omics: Combine single-cell RNA-seq with ATAC-seq or methylation profiling to characterize the epigenetic landscape of DLK1+ cancer stem cells compared to DLK1- tumor cells .

  • Sphere-forming assays with serial passaging: When evaluating CSC properties, perform sphere formation assays with multiple passages to distinguish true self-renewal from proliferative capacity. DLK1 knockdown significantly inhibits spheroid formation in HCC models .

  • Resistance mechanism profiling: Since CSCs often mediate therapy resistance, compare gene expression profiles of DLK1+ cells before and after exposure to conventional therapies to identify resistance mechanisms.

  • In vivo imaging with CSC markers: Combine DLK1 manipulation with tracking of additional CSC markers to determine hierarchical relationships in the CSC compartment .

Studies should account for potential heterogeneity among DLK1+ cells, as not all may possess true CSC properties. For example, in adrenocortical carcinoma, DLK1+ cells paradoxically show increased steroidogenic potential despite their progenitor-like features .

How should researchers design experiments to study DLK1's interaction with the Notch signaling pathway?

DLK1's relationship with Notch signaling is complex and context-dependent. To effectively study this interaction, researchers should:

  • Use multiple readout systems: Employ complementary approaches such as:

    • Luciferase reporter assays for Hes1-mediated Notch signaling

    • Quantitative measurement of Notch target genes (Hes1, Hey1)

    • Analysis of Notch receptor cleavage products

  • Compare canonical and non-canonical effects: DLK1 antagonizes cell proliferation activity of canonical Notch ligands Delta 1 and Jagged, but may have distinct effects on different Notch receptors (Notch1-4) .

  • Distinguish membrane-bound from soluble effects: Separately assess the impacts of membrane-tethered versus soluble DLK1 on Notch signaling, as these forms may have different or even opposing effects .

  • Implement domain swap experiments: Create chimeric proteins between DLK1 and canonical Notch ligands to identify which domains mediate specific interactions with Notch receptors.

  • Analyze binding competition: Determine whether DLK1 competes with canonical ligands for Notch receptor binding through competitive binding assays.

  • Cell-autonomous versus non-cell-autonomous effects: Design co-culture experiments with cells expressing different levels of DLK1 and Notch pathway components to distinguish between these mechanisms .

In neural progenitor studies, DLK1 has been shown to inhibit Hes1-mediated Notch signaling, demonstrating a regulatory relationship that impacts differentiation . This suggests experiments should monitor both pathway activity and functional outcomes simultaneously.

What technological approaches can enhance the study of DLK1's tissue-specific functions?

Advanced technological approaches that can significantly enhance our understanding of DLK1's tissue-specific functions include:

  • Spatial transcriptomics and proteomics: These techniques provide crucial spatial context for DLK1 expression patterns. In adrenocortical carcinoma, spatial transcriptomics has revealed that DLK1-expressing cell populations have increased steroidogenic potential, a finding that would be missed in bulk analysis .

  • CRISPR-based epigenome editing: For studying imprinting regulation, targeted modification of methylation status at DLK1 regulatory regions can help establish causative relationships between epigenetic modifications and expression patterns .

  • Tissue-specific inducible knockdown/overexpression: Employ tissue-specific promoters with inducible systems to manipulate DLK1 expression with spatial and temporal precision, allowing distinction between developmental and maintenance roles .

  • Organoid models: Develop tissue-specific organoids to study DLK1 function in a physiologically relevant 3D context that better recapitulates tissue architecture than traditional 2D culture .

  • In vivo imaging of stem cell dynamics: Implement intravital microscopy with DLK1 reporters to track stem cell behavior in real-time within living tissues .

  • Cell type-specific isolation: Use DLK1-based cell sorting combined with comprehensive profiling to characterize the unique properties of DLK1+ cells across different tissues .

When implementing these technologies, researchers should maintain consistency in analytical approaches across tissue types to facilitate comparative studies while acknowledging tissue-specific nuances in DLK1 function and regulation.

Product Science Overview

Structure and Composition

Mature human DLL1 consists of:

  • A 528 amino acid extracellular domain (ECD) with one DSL domain and eight EGF-like repeats
  • A 23 amino acid transmembrane segment
  • A 155 amino acid cytoplasmic domain

The protein is widely expressed and localizes to adherens junctions on neuronal processes through its association with the scaffolding protein MAGI1 .

Functions

DLL1 is essential for:

  • Embryonic Development: It plays a significant role in embryonic somite formation and cochlear hair cell differentiation .
  • Immune System: It is involved in the differentiation of B and T lymphocytes .
  • Vascular System: The upregulation of DLL1 in arterial endothelial cells following injury or angiogenic stimulation is central to postnatal arteriogenesis .
Clinical Significance

DLL1 is overexpressed in certain types of cancers, such as cervical carcinoma and glioma, contributing to tumor progression . Additionally, a soluble form of DLL1 has been shown to inhibit the differentiation of hematopoietic progenitor cells .

Recombinant DLL1

Recombinant human DLL1 is typically produced in HEK293 cells and is used for research purposes. It is often provided as a lyophilized powder and can be reconstituted for use in various experimental setups .

Storage and Handling
  • Lyophilized Form: Stable for up to 12 months when stored at -20 to -80°C.
  • Reconstituted Form: Can be stored at 4-8°C for 2-7 days or at < -20°C for up to 3 months .

DLL1’s role in cell fate decisions and its involvement in critical developmental and pathological processes make it a significant protein for research in developmental biology and oncology.

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.