Recombinant Mouse Microcephalin (Mcph1), partial

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a reference.
Shelf Life
Shelf life depends on storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. To prioritize a specific tag, please inform us during your order.
Synonyms
Mcph1; Microcephalin
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Mus musculus (Mouse)
Target Names
Mcph1
Uniprot No.

Target Background

Function
Mouse Microcephalin (Mcph1) is implicated in chromosome condensation and cellular responses to DNA damage. It may also play a role in neurogenesis and cerebral cortex size regulation.
Gene References Into Functions
  1. MCPH1 acts as a bona fide tumor suppressor gene. PMID: 25362854
  2. Mcph1-deficient mice studies indicate a role for MCPH1 in otitis media, with Mcph1 expression in middle ear epithelial cells suggesting involvement in otitis media development. PMID: 23516444
  3. MCPH1's role in the Chk1-Cdc25-Cdk1 pathway couples the centrosome cycle with mitosis, regulating precise mitotic spindle orientation and progenitor cell division mode for maintaining brain size. PMID: 21947081
  4. VIP blockade induces microcephaly via Mcph1 signaling; VIP/Mcph1/Chk1 signaling is crucial for normal cortical development. PMID: 21737879
  5. SET regulates chromosome condensation/decondensation, and disruption of the MCPH1-SET interaction may contribute to primary microcephaly pathogenesis. PMID: 21515671
  6. Studies did not reveal clear evidence of accelerated malignant disease development due to impaired Mcph1 function. PMID: 20169082
  7. Craniosynostosis-microcephaly with chromosomal breakage and other abnormalities is associated with a truncating MCPH1 mutation. PMID: 20101680
  8. MCPH1 is essential for mitotic and meiotic recombination DNA repair and maintaining genomic stability. PMID: 20107607
Database Links
Subcellular Location
Cytoplasm, cytoskeleton, microtubule organizing center, centrosome.
Tissue Specificity
High levels of expression are found in the developing forebrain and, in particular, in the walls of the lateral ventricles.

Q&A

What is Microcephalin (Mcph1) and what is its significance in research?

Microcephalin (Mcph1) is one of the causative genes responsible for autosomal recessive primary microcephaly in humans, a neurological disorder characterized by reduced head size (between -5 and -10 standard deviations below the mean) and mental retardation . The gene has gained significant research attention due to its multifaceted functions, including but not limited to brain development, DNA damage repair, chromosome condensation, cancer suppression, and germline function . Mouse models with Mcph1 mutations have been developed to study these functions, making recombinant Mcph1 an important tool for investigating the molecular mechanisms underlying microcephaly and broader aspects of brain development and evolution .

What phenotypes are observed in Mcph1-deficient mouse models?

Mcph1-deficient mouse models exhibit several distinctive phenotypes:

Importantly, these phenotypes create valuable opportunities for investigating the gene's role in development and disease mechanisms.

How do mouse Mcph1 models compare to human MCPH1 mutations?

FeatureHuman MCPH1 MutationsMouse Mcph1 Models
Brain SizeSevere microcephaly (-5 to -10 SD)~20% reduction in brain weight
Body SizeVariable short stature~20% reduction in body weight
Brain:Body RatioDisproportionately small brainProportionate reduction (no significant difference in brain:body ratio)
Chromosome CondensationPCC in G2 phasePCC present
Known MutationsS25X, 427insA, T27R (many in N-terminal BRCT domain)Various engineered knockouts and domain deletions

These comparative data raise questions about Mcph1's specificity as a brain-specific regulator in mouse models, as the proportionate reduction in both brain and body weight differs from the human presentation . This suggests potential species-specific differences in Mcph1 function or compensatory mechanisms.

What are the recommended approaches for generating Mcph1-deficient mouse models?

When designing Mcph1-deficient mouse models, researchers have successfully employed several strategies, each with specific advantages for investigating different aspects of Mcph1 function:

  • Complete gene knockout: Several mouse lines have been generated through targeted deletion of critical exons. For example, deletion of exons 4-5 (Mcph1 tm1.1Zqw) and exon 2 (Mcph1 tm1.2Kali) have produced viable models with microcephaly phenotypes .

  • Domain-specific deletions: Particularly valuable is the targeted deletion of the N-terminal BRCT domain (Mcph1-ΔBR1), which specifically investigates this domain's function. This approach involves constructing a targeting vector with homologous arms containing exon 1 and exons 4-6, effectively removing the N-terminal BRCT domain .

  • Hypomorphic mutations: Gene trap approaches have been used to create hypomorphic alleles (e.g., Mcph1 Gt(RRO608)Byg) that reduce but do not eliminate Mcph1 function .

Methodologically, these models are typically generated through electroporation of targeting vectors into embryonic stem cells, followed by screening for correctly targeted clones and injection into blastocysts . When designing your experimental approach, consider that complete knockout models may have reduced viability, as homozygous mice are born at lower-than-expected Mendelian ratios (10-15% vs. expected 25%) .

What cell-based assays are optimal for studying Mcph1 function?

To effectively investigate Mcph1 function at the cellular level, several complementary assays have proven particularly informative:

  • Chromosome condensation assays:

    • DAPI staining of nuclei to visualize abnormal chromatin condensation patterns

    • Metaphase spread analysis to quantify premature chromosome condensation (PCC)

    • These assays directly assess the characteristic PCC phenotype of Mcph1-deficient cells

  • DNA damage response (DDR) assays:

    • Homologous recombination (HR) assays using DR-GFP/I-sceI system

    • Evaluation of γ-H2AX foci formation after irradiation

    • These approaches evaluate the role of Mcph1 in DNA repair mechanisms

  • Protein interaction studies:

    • Tandem affinity purification techniques using tagged Mcph1 constructs

    • Co-immunoprecipitation assays to identify protein binding partners

    • These methods have successfully identified interactions with the Condensin II complex

  • Cell proliferation and cell cycle analysis:

    • BrdU incorporation assays

    • Flow cytometry for cell cycle analysis

    • These assays help understand Mcph1's role in cell proliferation and cell cycle regulation

When designing these experiments, it is crucial to include appropriate controls, particularly wild-type cells and cells reconstituted with various Mcph1 constructs to map domain-specific functions.

How should recombinant Mcph1 constructs be designed for functional studies?

When designing recombinant Mcph1 constructs for functional studies, consider the following guidelines based on established research practices:

  • Domain structure consideration: Mcph1 contains three BRCT domains—one N-terminal and two C-terminal—with distinct functions. Constructs should be designed to systematically investigate these domains:

    • Full-length Mcph1

    • N-terminal BRCT domain deletion (ΔN-BRCT)

    • Middle domain deletion (residues 376-485, which mediates Condensin II binding)

    • C-terminal BRCT domains deletion

  • Expression systems:

    • Retroviral vectors (e.g., pEF1A-HA/FLAG) have been successfully used for Mcph1 expression in mammalian cells

    • Gateway cloning systems facilitate efficient generation of multiple constructs

    • Consider including epitope tags (HA, FLAG, SFB) for detection and purification purposes

  • Delivery methods:

    • For stable expression, viral transduction followed by antibiotic selection (e.g., puromycin) is effective

    • For complementation assays in Mcph1-deficient cells, viral particles can be collected 48-72 hours post-transfection of packaging cells

  • Validation approaches:

    • Western blotting to confirm expression levels

    • Functional rescue assays to verify biological activity

    • Subcellular localization assessment through immunofluorescence

When designing deletion constructs, careful consideration of protein folding and stability is essential, as improper design may result in misfolded proteins that could confound functional studies.

What is the molecular basis for Mcph1's role in chromosome condensation?

The molecular mechanism underlying Mcph1's role in chromosome condensation involves a specific interaction with the Condensin II complex. Research has revealed several key components of this relationship:

  • Direct interaction with Condensin II: Mcph1 physically associates with the Condensin II complex, which is a major regulator of chromosome condensation during cell division. This interaction is specifically mediated through the CAPG2 subunit of Condensin II binding to a middle domain (residues 376-485) of Mcph1 .

  • Competitive inhibition mechanism: The N-terminal domain of human MCPH1 specifically inhibits the action of Condensin II by competing for its chromosomal binding sites. This competitive binding prevents premature chromosome condensation during G2 phase .

  • Domain-specific regulation: While the middle domain (residues 376-485) mediates binding to Condensin II, surprisingly, it is the N-terminal BRCT domain that is required for preventing premature chromosome condensation. Deletion of this N-terminal domain fails to rescue the chromosome condensation defect in Mcph1-deficient cells, despite not being directly involved in Condensin II binding .

  • Temporal regulation: This inhibitory relationship appears to be cell-cycle dependent, ensuring proper timing of chromosome condensation in preparation for mitosis.

This mechanistic understanding explains why MCPH1-deficient cells exhibit premature chromosome condensation and provides insight into one of the molecular pathways disturbed in primary microcephaly.

How does Mcph1 influence neuroprogenitor cell dynamics during brain development?

Mcph1 plays a critical role in regulating neuroprogenitor cell dynamics during brain development through several interconnected mechanisms:

  • Neuroprogenitor pool maintenance: Studies of Mcph1-ΔBR1 mice (with deleted N-terminal BRCT domain) reveal a reduction in the neuroprogenitor pool during brain development. This suggests Mcph1 is essential for maintaining the proper number of neural stem and progenitor cells .

  • Prevention of premature differentiation: Mcph1-deficient models show premature neuronal differentiation, indicating that Mcph1 normally acts to prevent the untimely transition from proliferative progenitors to post-mitotic neurons .

  • Cell cycle regulation: The observed thinner neocortex in Mcph1-deficient mice likely results from alterations in the balance between symmetric (proliferative) and asymmetric (neurogenic) divisions of neural progenitors. Mcph1 may influence this balance by regulating cell cycle dynamics and the timing of cell fate decisions .

  • Possible region-specific effects: While detailed spatial expression patterns of Mcph1 during cortical development remain incompletely characterized, it is important to determine if Mcph1 is expressed in the outer subventricular zone (OSVZ) progenitors, which are considered crucial for neocortical surface area expansion in humans .

Future research should focus on conditional knockout models with specific inactivation in the brain or neocortex to better distinguish brain-specific from systemic effects of Mcph1 deficiency, as current models show proportionate reductions in both brain and body size .

What are the challenges in analyzing Mcph1's evolutionary role in brain size determination?

Analyzing Mcph1's evolutionary role in brain size determination presents several significant challenges:

  • Distinguishing brain-specific from pleiotropic effects: Mcph1 has multifaceted functions across various systems—including DNA repair, germline function, and tumor suppression—making it difficult to isolate brain-specific evolutionary adaptations. Mouse models show proportionate reduction in both brain and body weight, questioning Mcph1's specificity as a brain size regulator .

  • Positive selection interpretation: While evidence suggests positive selection of Mcph1 in the primate lineage, this selection may not necessarily relate to brain evolution. Large-scale comparative studies have shown that genes involved in tumor suppression, apoptosis, and spermatogenesis (all functions where Mcph1 is implicated) frequently show positive selection .

  • Expression pattern uncertainties: The lack of detailed information on Mcph1 expression patterns during cortical development hampers the interpretation of phenotypic effects. It remains unclear whether Mcph1 is expressed in outer subventricular zone (OSVZ) progenitors, which are considered crucial for neocortical expansion in primates .

  • Species-specific differences: Molecular and genetic networks controlled by Mcph1 may have been tuned or co-opted differently across species, particularly in primates. Understanding these species-specific adaptations requires comparative expression and functional studies across diverse species .

  • Correlation versus causation: While Mcph1 mutations cause microcephaly, establishing its direct role in evolutionary brain size expansion requires additional evidence beyond correlation with increased brain size in certain lineages.

Addressing these challenges requires integrative approaches combining evolutionary genomics, comparative developmental biology, and functional studies across multiple species.

What are the optimal conditions for working with recombinant Mcph1 in cellular systems?

When working with recombinant Mcph1 in cellular systems, several methodological considerations can optimize experimental success:

  • Expression system selection:

    • For stable expression in mammalian cells, retroviral vectors (e.g., pEF1A-HA/FLAG) have proven effective

    • For biochemical studies, tagged versions (SFB-tagged, HA-tagged) facilitate purification and detection

    • Expression levels should be carefully monitored, as overexpression may cause artifacts

  • Cell line considerations:

    • MEFs derived from Mcph1-deficient mice (isolated from E14.5 embryos) provide an excellent system for complementation studies

    • HeLa and 293T cells are commonly used for overexpression studies

    • Culture conditions: MEFs require Dulbecco's modified Eagle's medium with 20% fetal bovine serum, while HeLa and 293T cells can be maintained in RPMI 1640 with 10% bovine serum

  • Transfection and viral transduction optimization:

    • For transient expression: standard transfection protocols are suitable

    • For stable expression: viral transduction followed by antibiotic selection (puromycin 2 μg/ml) for 2-3 weeks ensures consistent expression

    • Viral particles should be collected 48 and 72 hours after transfection of packaging cells

  • Functional validation approaches:

    • Western blotting to confirm expression levels

    • DAPI staining and metaphase spreads to assess chromosome condensation phenotypes

    • Complementation assays to verify functional rescue

  • Protein interaction studies:

    • For tandem affinity purification: lyse cells with NETN buffer on ice for 20 minutes

    • Centrifuge at 14,000 rpm at 4°C for 10 minutes to clear lysates

    • Follow established protocols for streptavidin bead binding and subsequent elution with biotin

Following these validated methodological approaches will increase reproducibility and reliability when working with recombinant Mcph1 in cellular systems.

How can Mcph1 domain-specific functions be effectively mapped?

Mapping domain-specific functions of Mcph1 requires systematic approaches that isolate the contributions of individual domains to particular cellular processes. Based on published research, the following strategies are most effective:

  • Domain deletion strategy:

    • Create a panel of constructs with specific domain deletions:

      • N-terminal BRCT domain deletion (ΔN-BRCT)

      • Middle domain deletion (residues 376-485, which mediates Condensin II binding)

      • C-terminal tandem BRCT domains deletion

    • Express these constructs in Mcph1-deficient cells and assess rescue of specific phenotypes

  • Phenotype-specific readouts:

    • For chromosome condensation: Use DAPI staining patterns and metaphase spread analysis

    • For DNA damage response: Employ HR repair assays using the DR-GFP/I-sceI system

    • For neuroprogenitor function: Analyze cortical development in domain-specific knockout mice

  • Protein interaction mapping:

    • Perform co-immunoprecipitation studies with domain deletion constructs to map binding interfaces

    • Use yeast two-hybrid or in vitro binding assays to confirm direct interactions

    • The middle domain (residues 376-485) has been identified as the binding region for Condensin II

  • In vivo domain function validation:

    • Generate domain-specific knockout mice (e.g., Mcph1-ΔBR1) to assess in vivo function

    • Compare phenotypes to complete knockout models to determine domain-specific contributions

Results from these approaches have revealed important functional specificity:

  • The N-terminal BRCT domain is essential for preventing premature chromosome condensation and microcephaly

  • The middle domain (376-485) mediates Condensin II binding and is required for HR repair

  • Domain functions may be partially independent, as constructs that cannot bind Condensin II can still rescue chromosome condensation defects

What are the best approaches for analyzing contradictory data in Mcph1 research?

When confronting contradictory data in Mcph1 research, several analytical approaches can help reconcile discrepancies and advance understanding:

  • Model system differences assessment:

    • Systematically compare experimental conditions, genetic backgrounds, and cell types

    • Consider species differences, as Mcph1 functions may vary between human and mouse models

    • For example, while Mcph1 mutant mice show proportionate reduction in brain and body size, human MCPH1 mutations primarily affect brain size

  • Domain-specific function analysis:

    • Apparent contradictions may reflect domain-specific functions

    • For example, the middle domain (376-485) binds Condensin II but cannot rescue PCC phenotypes, whereas the N-terminal domain doesn't bind Condensin II but is required for preventing PCC

    • Construct complementation experiments using domain-specific deletions can resolve such contradictions

  • Temporal and context-dependent effects consideration:

    • Analyze developmental timing differences, as Mcph1 may have stage-specific functions

    • Consider cell-cycle phase-specific effects, particularly for chromosome condensation phenotypes

    • Document environmental conditions that might influence experimental outcomes

  • Technical approach harmonization:

    • Standardize analytical methods, particularly for phenotype assessment

    • For chromosome condensation studies, use both DAPI staining and metaphase spread analysis

    • For brain development, employ consistent histological and imaging techniques

  • Integrative data analysis:

    • Combine biochemical, cellular, and in vivo data to generate comprehensive models

    • When contradictions persist, design experiments that specifically test competing hypotheses

    • Consider compensatory mechanisms that may mask phenotypes in certain contexts

One example of resolving contradictory data comes from understanding how Mcph1 regulates chromosome condensation: while Mcph1 binds Condensin II through its middle domain, the prevention of premature chromosome condensation depends on the N-terminal BRCT domain, suggesting a more complex regulatory mechanism than simple protein-protein interaction .

What are the promising therapeutic applications of Mcph1 research?

While Mcph1 research is primarily fundamental in nature, several promising therapeutic applications are emerging that merit further investigation:

  • Microcephaly treatment approaches:

    • Understanding the molecular pathways regulated by Mcph1 may reveal therapeutic targets to mitigate microcephaly

    • Particularly promising is the potential to modulate neuroprogenitor proliferation and differentiation to preserve brain development in affected individuals

  • Cancer therapy applications:

    • Given Mcph1's role in DNA repair, chromosome stability, and tumor suppression, it represents a potential target for cancer therapeutics

    • Female Mcph1-ΔBR1 mice develop ovary tumors, suggesting a role in gynecological cancer suppression

    • Two MCPH1 polymorphisms have been associated with breast cancer risk, indicating potential diagnostic or therapeutic relevance

  • Chromosome condensation modulators:

    • The elucidation of how Mcph1 regulates chromosome condensation through interaction with Condensin II provides a framework for developing compounds that could modulate this process

    • Such modulators could have applications in treating chromosome instability syndromes

  • DNA damage response enhancement:

    • Mcph1's role in homologous recombination repair suggests potential applications in enhancing DNA repair in contexts of radiation exposure or chemotherapy

    • Understanding the molecular basis of Mcph1's function in DNA repair could inform development of compounds that enhance repair mechanisms

  • Neurodevelopmental disorder insights:

    • Beyond primary microcephaly, Mcph1 research may provide insights into other neurodevelopmental disorders characterized by brain size abnormalities

    • Comparative studies could reveal shared pathways and potential therapeutic targets across related conditions

These therapeutic directions remain largely exploratory and will require further fundamental research before clinical applications can be developed.

What unresolved questions remain about Mcph1's molecular mechanisms?

Despite significant progress, several critical questions about Mcph1's molecular mechanisms remain unresolved:

  • Spatio-temporal expression pattern clarification:

    • Detailed information on the pattern of Mcph1 expression during cortical development remains incomplete

    • Particularly important is determining whether Mcph1 is expressed in outer subventricular zone (OSVZ) progenitors, which are considered crucial for neocortical expansion in primates

  • Cell-type specificity:

    • The cell types expressing Mcph1 during development and in adult tissues are incompletely characterized

    • Understanding cell-type specific expression patterns could explain the pleiotropic effects of Mcph1 mutations

  • Subcellular localization dynamics:

    • The subcellular localization of Mcph1 protein at different cell cycle stages and under different conditions (e.g., DNA damage) requires further investigation

    • This information is crucial for understanding Mcph1's diverse functions

  • Regulatory network integration:

    • How Mcph1 integrates with other microcephaly-associated genes (ASPM, CDK5RAP2, CENPJ, etc.) in regulatory networks remains unclear

    • Understanding these networks could reveal convergent mechanisms underlying microcephaly

  • Domain-function relationships:

    • While some domain-specific functions have been identified, a comprehensive understanding of how each domain contributes to Mcph1's diverse functions is lacking

    • Particularly intriguing is how the N-terminal BRCT domain prevents premature chromosome condensation without directly binding Condensin II

  • Species-specific adaptations:

    • How Mcph1's functions may have been tuned or co-opted differently across species remains largely unexplored

    • Comparative functional studies across diverse species, including primates, would address this question

Addressing these questions will require integrative approaches combining genomics, biochemistry, cell biology, and developmental neuroscience.

How might emerging technologies advance Mcph1 research?

Emerging technologies offer exciting opportunities to address unresolved questions in Mcph1 research:

  • Single-cell technologies:

    • Single-cell RNA sequencing can reveal cell-type specific expression patterns of Mcph1 during development

    • Single-cell ATAC-seq could identify chromatin accessibility changes in Mcph1-deficient cells

    • These approaches would help resolve the spatio-temporal expression patterns of Mcph1 and its impact on gene regulation

  • Advanced imaging techniques:

    • Super-resolution microscopy can provide detailed visualization of Mcph1's subcellular localization

    • Live-cell imaging of fluorescently tagged Mcph1 would reveal its dynamics during cell cycle progression

    • Three-dimensional imaging of neuroprogenitor divisions in Mcph1-mutant mice could clarify its role in division plane orientation and fate determination

  • CRISPR-Cas9 gene editing:

    • Generation of precise point mutations corresponding to human disease variants

    • Development of conditional knockout models with spatial and temporal control

    • Creation of domain-specific mutations for fine-mapping function

    • These approaches would overcome limitations of current mouse models and enable more precise dissection of Mcph1 function

  • Organoid technologies:

    • Brain organoids derived from Mcph1-mutant stem cells could model human microcephaly

    • This system would allow investigation of species-specific aspects of Mcph1 function in human neural development

    • Comparative organoid studies across species could illuminate Mcph1's evolutionary role

  • Structural biology approaches:

    • Cryo-EM and X-ray crystallography could resolve the structure of Mcph1 and its complexes

    • Understanding the structural basis of interactions with Condensin II and other partners would clarify molecular mechanisms

    • This information could guide future therapeutic development

  • Multi-omics integration:

    • Combining transcriptomics, proteomics, and epigenomics data from Mcph1-deficient models

    • Systems biology approaches to integrate these datasets

    • This integration would reveal broader networks and pathways affected by Mcph1 dysfunction

These technological advances promise to address fundamental questions about Mcph1's diverse functions and potentially lead to therapeutic applications for conditions involving Mcph1 dysfunction.

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.