Genetic knockout models have been pivotal in elucidating midkine’s roles. Below is a comparison of key phenotypes in knockout strains:
Auditory deficits: DKO mice show profound hearing loss due to β-tectorin deficiency, a critical cochlear protein .
Neuroprotection: MK knockout mice exhibit impaired recovery after neurotoxicity (e.g., NMDA-induced retinal damage) .
Gene expression: DNA microarray analysis in DKO mice revealed suppressed expression of genes involved in neurogenesis and repair .
Retinal injury: Exogenous midkine reduces neuronal death and reactive microglia in chick retinas but has limited effects in mice .
Müller glia reactivity: Midkine upregulates cFos and pS6 in Müller glia, promoting proliferation in chick retinas but not in mice .
Neurotoxicity: MK knockout mice show exacerbated neuronal loss after drug-induced neurotoxicity .
Midkine regulates immune cell infiltration in the CNS:
Astrocyte-mediated signaling: Activated astrocytes secrete midkine, recruiting neutrophils and macrophages to injury sites .
T-regulatory cell expansion: Midkine modulates STAT3/STAT5 signaling, influencing IL-2-dependent FOXP3+ T-reg cells .
Tumor progression: Midkine overexpression correlates with metastasis and poor prognosis in cancers (e.g., neuroblastoma, glioblastoma) .
Angiogenesis: Promotes vascular endothelial cell migration and survival, critical for tumor vasculature .
Midkine signals through a multi-receptor complex involving:
Core receptors: LRP1 (low-density lipoprotein receptor-related protein 1) and integrins .
Secondary partners: ALK (anaplastic lymphoma kinase), PTPζ (protein tyrosine phosphatase ζ), and syndecans .
Pathway | Function | References |
---|---|---|
Ras-Raf-MEK-ERK | Proliferation and survival . | |
PI3K-Akt-mTOR | Anti-apoptotic effects, protein synthesis . | |
NF-κB | Pro-inflammatory cytokine production, survival signaling . |
Biomarker potential: Elevated serum midkine levels correlate with tumor progression .
Inhibition strategies: RNA aptamers targeting nucleolin-MK interactions or ALK/PTPζ signaling .
Traumatic brain injury: Midkine gene therapy reduces neuronal death in ischemic models .
Retinal regeneration: Midkine enhances Müller glia-derived progenitor cell proliferation in zebrafish, but efficacy in mammals remains limited .
Midkine is a small secreted heparin-binding protein (13-18 kD) that belongs to the neurite growth-promoting factor family, along with pleiotrophin (PTN). It was named "midkine" because it was originally identified as a cytokine highly expressed during mid-gestation in many organs of the mouse, particularly the kidneys, heart, and brain . Midkine promotes growth through effects on cell proliferation, migration, and differentiation, making it a critical factor in developmental biology . Mouse models are particularly valuable for studying Midkine because they demonstrate high expression levels (approximately 10-fold greater than in humans across cell types) and have well-characterized expression patterns throughout development .
Mouse and human Midkine share significant structural similarities. The mRNA and protein structures are similar, with an amino acid sequence predicted to have 83% homology between species . Both consist of 121 amino acids and are highly endowed with positively charged basic amino acids (arginine, lysine, and histidine) . The protein structure is composed of N-terminal and C-terminal halves linked by five disulfide bonds, with the C-terminal portion containing a strong conformation-dependent heparin binding site that is crucial for neurite extension and plasminogen activator activities, but not for promoting cell survival .
Midkine interacts with several receptors in mouse models, which complicates understanding its specific roles. Key receptors include:
Syndecans (particularly Syndecan-1 and Syndecan-3)
Protein tyrosine phosphatase ζ (PTPζ)
Low-density lipoprotein receptor-related protein (LRP)
Anaplastic lymphoma kinase (ALK)
Notch2
Syndecan-3 appears particularly important in neural tissues, with expression data showing it is more highly expressed than Syndecan-1 in both mouse and human brain tissues . Researchers should note that Midkine's interaction with multiple receptors (each having numerous potential ligands) limits the ability to delineate specific activities through simple receptor blockade or knockout studies .
Midkine expression during mouse development follows a distinct pattern:
Detected as early as embryonic day 5 (E5) in the ectoderm, allantois, and chorion of placental tissues
By E8.5, expression is found throughout the whole mouse embryo and extra-embryonic membranes
Strong mRNA expression throughout the developing cortical plate at E14.5 and E15.5
Expression in jaw, hindlimb bud, skin, placental capillary endothelial cells, brain, and spinal cord at E14.5
By E17 in rats (comparable developmental stage in mice), Midkine immunoreactivity emerges radially from the ventricular zone into the telencephalon, with most intense expression in the intermediate zone and subventricular zones
Expression decreases after birth, unlike Pleiotrophin which increases from birth and persists into adulthood
For reference, gene expression profiling data across different brain regions and developmental stages is available in Figures 2 and 5 of the cited research .
Cell-type specific analysis using RNA-sequencing has revealed considerable variation in Midkine expression across neural cell types. Data from mouse brain tissues shows that Midkine expression is approximately 10-fold greater across all cell types compared to human counterparts . In the embryonic mouse brain at E14.5-E15.5, Midkine mRNA is strongly expressed throughout the developing cortical plate . During later developmental stages, single-cell RNA-sequencing has revealed that Midkine and Pleiotrophin are upregulated by Müller glia during later stages of development in retinal tissues . Following injury, Midkine is dramatically upregulated in mature Müller glia in chick retinas but interestingly downregulated in mouse retinas .
Midkine knockout (KO) mice exhibit several distinctive phenotypic characteristics:
Delayed hippocampal development, shown by a transient abnormal increase in calretinin in the granule cell layer of the dentate gyrus
Increased anxiety and impaired working memory as assessed via elevated plus maze test and y-maze test, respectively
Reduced striatal dopamine content, suggesting increased vulnerability for behavioral disorders such as schizophrenia and autism
Auditory deficits, which are more severe in Midkine/Pleiotrophin double knockout mice
Most notably, Midkine/Pleiotrophin double knockout (DKO) mice:
Are born at one-third the expected frequency based on Mendelian segregation, suggesting embryonic lethality prior to E14.5
Present with severe postnatal growth retardation (50% reduction at 4 weeks of age) that is not corrected with high-calorie feeding
Show 40-50% reduction in spontaneous locomotor activity at 4 weeks of age compared to wild-type mice
Several complementary techniques are recommended for comprehensive analysis of Midkine expression:
RNA-sequencing and transcriptomics:
Single-cell RNA-sequencing offers high-resolution cell-type specific expression data
Resources like http://www.brainrnaseq.org/ provide valuable reference datasets
For embryonic expression patterns, resources like Genepaint (https://gp3.mpg.de/) and GENSAT (http://www.gensat.org/) provide validated expression data
Immunohistochemistry/Immunofluorescence:
Effective for spatial localization within tissues
Should include controls for antibody specificity
Can be combined with cell-type specific markers for colocalization studies
In situ hybridization:
Particularly valuable for embryonic tissues
Allows precise localization of mRNA expression patterns
Can be performed at high resolution using RNAscope technology
Western blotting:
For quantitative analysis of protein levels
Should be normalized to appropriate housekeeping proteins
Can detect different Midkine isoforms
When designing Midkine knockout or knockdown experiments:
Generation of knockout models:
Validation recommendations:
Confirm knockout at both mRNA (RT-qPCR) and protein (Western blot, immunohistochemistry) levels
Validate across multiple tissues due to tissue-specific expression
Check for compensatory upregulation of related proteins (particularly Pleiotrophin)
Assess developmental timepoints carefully, as effects may be stage-specific
Experimental considerations:
To investigate Midkine's neuroprotective functions:
In vitro approaches:
In vivo approaches:
Analysis methods:
Midkine plays a complex role in neuroinflammation after brain injury:
Microglial/macrophage response:
In traumatic brain injury (TBI) models, Midkine knockout mice show reduced microglial/macrophage Iba1-immunoreactivity at 3 days post-injury compared to wild-type mice
Midkine appears to modulate the polarization of microglia, with knockout mice showing fewer pro-inflammatory M1 (CD16/32+) cells in perilesional sites and reduced mRNA levels of M1 markers (TNFα, CD11b)
Flow cytometry analysis reveals that Midkine knockout mice have increased levels of anti-inflammatory M2 arginase1+ microglia and M2 CD163+ macrophages
Chemoattractant properties:
Midkine serves as a chemoattractant for leukocytes, potentially amplifying peripheral immune cell recruitment to injury sites
This may explain why Midkine knockout mice had reduced apoptosis and improved neurological outcomes following TBI
The effect appears to be age-dependent, with different impacts on blood-brain barrier permeability, chemokine function, and leukocyte recruitment after CNS injury
Developmental considerations:
Recent research on Midkine in retinal tissues has revealed:
Expression patterns:
Single-cell RNA-sequencing shows that Midkine and Pleiotrophin are upregulated by Müller glia during later stages of development in chick retina
After retinal damage or FGF2 and insulin treatment, Midkine is dramatically upregulated in mature Müller glia in chick retinas
Interestingly, Midkine is downregulated by Müller glia in damaged mouse retinas, showing species-specific responses
Functional effects:
Signaling pathway interactions:
Midkine shows promise as a therapeutic agent for brain injury, though methodological considerations are important:
Delivery methods:
Therapeutic applications:
Research gaps and future directions:
Most studies have been conducted in vitro with limited in vivo validation
Need for studies in models that incorporate the maternal-placental-fetal unit, such as the precocial spiny mouse or fetal and newborn sheep
Investigation of potential Midkine deficiency in preterm birth scenarios, given high levels in amniotic fluid
Development of improved drug delivery platforms to enhance therapeutic efficacy and bioavailability
Important species differences exist in Midkine expression patterns:
Quantitative differences:
Receptor expression differences:
RNA sequencing suggests that Syndecan-1 expression is approximately 100-fold less in humans than in mice
Syndecan-3 expression is about 50-fold less in humans compared to mice
These differences may indicate fundamental physiological variations that prevent simple extrapolation of findings between species
Research implications:
Results from rodent experiments must be interpreted cautiously due to these expression differences
Protein abundance or stability differences in humans may potentially counteract higher expression of mouse mRNA
Validation in human cell or tissue models is strongly recommended before clinical translation
Several key challenges complicate cross-species studies of Midkine:
Receptor redundancy and complexity:
Technical limitations:
Developmental timing differences:
Cross-species comparisons must account for differences in developmental timing and maturation
Expression patterns that appear similar may occur at different relative developmental stages
These differences necessitate careful experimental design when comparing developmental processes across species
For translational studies involving Midkine in mouse models:
Model selection considerations:
Consider models that incorporate the maternal-placental-fetal unit for developmental studies
The precocial spiny mouse or fetal and newborn sheep may provide better translational value for certain questions
Use appropriate developmental timepoints that correspond to human developmental stages of interest
Validation requirements:
Therapeutic development suggestions:
Focus on functional outcomes rather than mechanistic details that may differ across species
Consider the potential impact of species-specific inflammatory responses
Develop improved drug delivery platforms to enhance therapeutic efficacy and bioavailability in target tissues
Evaluate dose-response relationships carefully, given the substantial expression differences between species
Midkine was originally identified as a cytokine highly expressed during mid-gestation in many organs of the mouse, particularly the kidneys, heart, and brain . It is a highly basic, non-glycosylated polypeptide consisting of two domains stabilized by five intrachain disulfide bonds . The protein has a molecular weight of approximately 13 kDa .
Midkine is involved in a variety of biological functions, including:
Recombinant mouse midkine is produced using E. coli expression systems. The recombinant protein is typically purified to a high degree of purity (>95%) and is used in various research applications . It is often used to study its effects on neurite outgrowth, cell proliferation, and other biological processes .
The potential for MK as a therapeutic agent is being explored in various fields, including: