Midkine Mouse

Midkine Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Mouse Models in Midkine Research

Genetic knockout models have been pivotal in elucidating midkine’s roles. Below is a comparison of key phenotypes in knockout strains:

ModelGenotypeKey DefectsReferences
MKKOMdk−/−Mild auditory deficits, reduced β-tectorin expression in cochlea .
PTNKOPtn−/−Moderate auditory impairments, partial β-tectorin expression .
DKOMdk−/− Ptn−/−Severe auditory deficits, complete loss of β-tectorin, female sterility .
Wild-typeMdk+/+ Ptn+/+Normal auditory function, full β-tectorin expression .

Notable observations:

  • 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 .

Neurogenesis and Neuroprotection

  • 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 .

Immunomodulation and Neuroinflammation

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 .

Oncogenic and Angiogenic Roles

  • 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 .

Mechanistic Insights and Signaling Pathways

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 .

Downstream pathways:

PathwayFunctionReferences
Ras-Raf-MEK-ERKProliferation and survival .
PI3K-Akt-mTORAnti-apoptotic effects, protein synthesis .
NF-κBPro-inflammatory cytokine production, survival signaling .

Cancer Therapy

  • Biomarker potential: Elevated serum midkine levels correlate with tumor progression .

  • Inhibition strategies: RNA aptamers targeting nucleolin-MK interactions or ALK/PTPζ signaling .

Neurological Disorders

  • 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 .

Challenges in Translation

  • Species-specific effects: Midkine’s role in neuroprotection diverges between avian and murine models .

  • Off-target effects: Systemic inhibition may compromise physiological repair processes .

Product Specs

Introduction
Midkine (MK) belongs to a family of heparin-binding growth factors and is encoded by a gene responsive to retinoic acid. Composed of 121 amino acids, it features 10 cysteine residues that are conserved and likely form disulfide bonds. During embryonic development, MK exhibits a distinct expression pattern suggestive of roles in neurogenesis, cell migration, organ development, and interactions between mesoderm and epithelium. While its presence is largely downregulated in adult humans, MK expression resurfaces in various cancers, where it can function as both a growth factor promoting transformation and a contributor to angiogenesis. Notably, MK exhibits chemotactic activity on human neutrophils, prompting their migration, and can trigger calcium mobilization within these cells. Moreover, it acts as a potent stimulator of histamine release from rat peritoneal mast cells in a rapid and dose-dependent fashion. Furthermore, MK demonstrates the ability to stimulate the production of collagen and glycosaminoglycans.
Description
Recombinant Mouse Midkine, produced in E. coli, is a single-chain polypeptide lacking glycosylation. It consists of 120 amino acids and has a molecular weight of 13.3 kDa. The purification process for this recombinant protein involves proprietary chromatographic techniques.
Physical Appearance
White, lyophilized (freeze-dried) powder, sterile-filtered.
Formulation
Lyophilized from a 0.2 µm filtered solution in phosphate-buffered saline (PBS) at pH 7.4.
Solubility
To reconstitute lyophilized Mouse Midkine, it is recommended to dissolve it in sterile 18M-cm H₂O at a concentration of at least 100 µg/ml. The resulting solution can be further diluted in other aqueous solutions.
Stability
Lyophilized Mouse Midkine, though stable at room temperature for up to 3 weeks, should be stored in a dry environment below -18°C for long-term preservation. Once reconstituted, it can be kept at 4°C for 2-7 days. For extended storage, freezing at -18°C is recommended. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95.0%, as determined by high-performance liquid chromatography (HPLC) and sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE).
Biological Activity
The biological activity is comparable to the standard, as assessed by its chemotactic effect on human neutrophils. Within a concentration range of 10-100 ng/ml, it exhibits a specific activity of 10,000-100,000 IU/mg.
Synonyms
NEGF-2, Neurite Growth-Promoting Factor 2, MK, Neurite outgrowth-promoting protein, Midgestation and kidney protein, Amphiregulin-associated protein, ARAP, Neurite outgrowth-promoting factor 2, FLJ27379, Midkine, MK1, NEGF2.
Source
Escherichia Coli.
Amino Acid Sequence
VAKKKEKVKK GSECSEWTWG PCTPSSKDCG MGFREGTCGA QTQRVHCKVP CNWKKEFGAD CKYKFESWGA CDGSTGTKAR QGTLKKARYN AQCQETIRVT KPCTSKTKSK TKAKKGKGKD

Q&A

What is Midkine and why is it important in mouse developmental studies?

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 .

How does the structure of mouse Midkine compare to human Midkine?

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 .

What are the primary receptors for Midkine in mouse models?

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 .

What is the temporal and spatial expression pattern of Midkine during mouse development?

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 .

How does Midkine expression differ across cell types in the mouse brain?

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 .

What are the phenotypic consequences of Midkine knockout in mice?

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

What techniques are recommended for studying Midkine expression in mouse models?

Several complementary techniques are recommended for comprehensive analysis of Midkine expression:

  • RNA-sequencing and transcriptomics:

  • 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

How should Midkine knockout or knockdown experiments be designed and validated?

When designing Midkine knockout or knockdown experiments:

  • Generation of knockout models:

    • Consider both conventional and conditional knockout approaches

    • Currently, there are no conditional KO mice for Midkine, which limits the ability to study nuanced functions

    • Double knockout with Pleiotrophin should be considered due to functional redundancy

  • 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:

    • Include appropriate littermate controls

    • Consider the impact of genetic background on phenotype

    • Due to potential embryonic lethality in double knockouts, carefully time experiments and consider using heterozygous breeding schemes

What methodological approaches are effective for studying Midkine's neuroprotective functions?

To investigate Midkine's neuroprotective functions:

  • In vitro approaches:

    • Primary neuronal cultures subjected to oxygen-glucose deprivation

    • Excitotoxicity models using glutamate exposure

    • Assessment of apoptotic markers (cleaved caspase-3, TUNEL staining)

    • Measurement of key signaling pathways (e.g., pS6, cFos expression)

  • In vivo approaches:

    • Traumatic brain injury models - assess apoptosis via cleaved caspase-3 expression

    • Retinal damage models - quantify dying neurons and reactive microglia

    • Exogenous Midkine administration with appropriate delivery methods

    • Inhibition of Midkine signaling using Na₃VO₄ to block neuroprotective effects

  • Analysis methods:

    • Flow cytometry to segregate and analyze specific cell populations

    • Assessment of microglial/macrophage polarization using markers for M1 (CD16/32+) and M2 (arginase1+) phenotypes

    • mRNA analysis of inflammatory markers (TNFα, CD11b)

    • Quantification of cell death and survival

How does Midkine influence neuroinflammation in mouse models of brain injury?

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:

    • Age-dependent differences in blood-brain barrier permeability significantly impact Midkine's effects

    • In contrast to adult models, developmental models may show different inflammatory responses to Midkine modulation

What is known about Midkine's role in retinal development and injury response?

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:

    • In both chick and mouse retinas, exogenous Midkine induces expression of cFos and pS6 in Müller glia

    • In chick retina, Midkine significantly decreases:

      • Numbers of dying neurons

      • Reactive microglia

      • Proliferating Müller glia-derived progenitor cells (MGPCs)

    • In mice, Midkine:

      • Promotes a small but significant increase in proliferating MGPCs in damaged retinas

      • Potently decreases the number of dying cells

  • Signaling pathway interactions:

    • Inhibition of Midkine signaling with Na₃VO₄ blocks neuroprotective effects and increases cell death

    • Inhibitors of PP2A and Pak1, which are associated with Midkine signaling through integrin β1, suppressed the formation of MGPCs in damaged chick retinas

How can Midkine be utilized as a therapeutic agent in mouse models of brain injury?

Midkine shows promise as a therapeutic agent for brain injury, though methodological considerations are important:

  • Delivery methods:

    • Direct intracerebroventricular or intranasal administration

    • Modified Midkine peptides with enhanced stability and brain penetration

    • Nanoparticle encapsulation for targeted delivery

    • Recombinant Midkine can be produced in various expression systems and tagged with fluorophores for tracking

  • Therapeutic applications:

    • Amelioration of cell death in hypoxic-ischemic injury models

    • Modulation of glial reactivity after injury

    • Enhancement of proliferation and migration of neural precursor cells

    • Promotion of hypoxia-induced angiogenesis

  • 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

How do Midkine expression patterns differ between mouse and human brain development?

Important species differences exist in Midkine expression patterns:

  • Quantitative differences:

    • Across cell types, Midkine expression is approximately 10-fold greater in mouse compared to humans

    • This substantial difference should be considered when translating findings from mouse models to human applications

  • 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

What methodological challenges exist in studying Midkine function across species?

Several key challenges complicate cross-species studies of Midkine:

  • Receptor redundancy and complexity:

    • Midkine interacts with several receptors, each with numerous potential ligands

    • This limits the ability to delineate activity through receptor blockade or knockout studies

    • Structural and functional similarities between Midkine and Pleiotrophin create redundancy in signaling systems

  • Technical limitations:

    • Lack of conditional knockout models for Midkine limits nuanced investigation of functions

    • Complex feedback loops between Midkine and Pleiotrophin complicate interpretation of single-protein manipulations

    • Antibody specificity issues may arise when detecting Midkine across different species

  • 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

What should researchers consider when designing translational studies using mouse Midkine models?

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:

    • Validate key findings in human cell cultures or organoids when possible

    • Consider the species differences in receptor expression when interpreting results

    • Account for the 10-fold difference in Midkine expression levels between mice and humans

  • 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

Product Science Overview

Discovery and Structure

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 .

Biological Functions

Midkine is involved in a variety of biological functions, including:

  • Cell Proliferation and Migration: MK promotes cell proliferation, migration, and differentiation through interactions with multiple cell surface receptors .
  • Neuroprotection and Neuroregeneration: MK is upregulated in the adult central nervous system (CNS) after various types of experimental injury. It has neuroprotective and neuroregenerative properties, making it a promising candidate for therapeutic applications in brain injuries .
  • Developmental Roles: During embryonic and fetal development, MK is highly expressed and plays a significant role in the structural and functional development of the brain .
Recombinant Mouse Midkine

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 .

Therapeutic Potential

The potential for MK as a therapeutic agent is being explored in various fields, including:

  • Perinatal Brain Injury: MK has shown promise as a neurotrophic therapy for perinatal brain injury, where it can ameliorate the effects of brain damage caused by hypoxia and inflammation .
  • Cancer: MK expression is induced in many forms of cancer, where it mediates hypoxic or inflammatory-driven cell response pathways . Research is ongoing to explore its role in cancer therapy.

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 2024 Thebiotek. All Rights Reserved.