Recombinant Pan troglodytes Mediator of RNA polymerase II transcription subunit 12 (MED12), 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 fulfillment.
Lead Time
Delivery times vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates. Note: All protein shipments default to 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 collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, provided as a guideline for your reference.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms maintain stability for 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during manufacturing. Tag type is determined during production. Specify your required tag type for prioritized development.
Synonyms
MED12; TNRC11; Mediator of RNA polymerase II transcription subunit 12; Mediator complex subunit 12; Trinucleotide repeat-containing gene 11 protein
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Pan troglodytes (Chimpanzee)
Target Names
Uniprot No.

Target Background

Function

Mediator complex component; a coactivator involved in the regulated transcription of nearly all RNA polymerase II-dependent genes. Mediator acts as a bridge, conveying information from gene-specific regulatory proteins to the basal RNA polymerase II transcription machinery. It is recruited to promoters through direct interactions with regulatory proteins and serves as a scaffold for assembling a functional preinitiation complex with RNA polymerase II and general transcription factors. This subunit may specifically regulate transcription of Wnt and SHH signaling pathway targets.

Database Links
Protein Families
Mediator complex subunit 12 family
Subcellular Location
Nucleus.

Q&A

What is MED12 and what are its primary functions in transcriptional regulation?

MED12 is a critical subunit of the 1.2 MDa Mediator complex, which plays an essential role in transcription initiation. As part of the CDK8 subcomplex (containing MED13, CDK8 kinase, and cyclin C), MED12 modulates Mediator-polymerase II interactions and thereby regulates transcription initiation and reinitiation rates . MED12 is essential for activating CDK8 kinase activity within this complex .

The primary functions of MED12 include:

  • Regulation of transcription initiation as part of the Mediator complex

  • Modulation of signaling pathways including Wnt/β-catenin and hedgehog signaling

  • Regulation of TGF-β receptor signaling through direct interaction with TGF-βR2

  • Control of response to multiple cancer therapeutics

How structurally and functionally conserved is MED12 between humans and Pan troglodytes?

While the search results don't specifically address the conservation between human and chimpanzee MED12, evolutionary conservation of critical transcriptional regulators like MED12 is typically high between closely related species. The functional domains of MED12 that mediate interactions with transcription factors and signaling molecules would be expected to show strong conservation.

For experimental purposes, researchers should note that:

  • Key functional domains are likely preserved between human and chimpanzee MED12

  • Critical interaction sites, particularly those mediating TGF-β signaling regulation, would be expected to be highly conserved

  • Any species-specific differences should be carefully validated when extrapolating findings between models

What experimental systems are commonly used to study recombinant MED12 function?

Several experimental systems have been employed to study MED12 function:

  • Cell line models:

    • Cancer cell lines (H3122, PC9, A375, SK-CO-1, and Huh-7)

    • Prostate cancer cell lines for studying MED12's role in cancer progression

  • Genetic manipulation:

    • shRNA-mediated knockdown of MED12 in multiple cancer cell types

    • Reconstitution with RNAi-resistant MED12 cDNA to validate specificity

  • Functional assays:

    • Drug sensitivity/resistance assays

    • Cell proliferation and cell cycle analysis

    • Protein interaction studies

    • Transcriptome analysis using RNA-Seq

What are the known binding partners of MED12 and their functional significance?

MED12 interacts with multiple proteins as part of its diverse functional roles:

  • Core Mediator complex components:

    • MED13, CDK8, and cyclin C as part of the CDK8 subcomplex

  • Signaling pathway components:

    • TGF-βR2: MED12 negatively regulates TGF-βR2 through direct physical interaction in the cytoplasm

    • Components of the Wnt/β-catenin pathway where MED12 acts as a signaling transducer

  • Functional significance:

    • The interaction with TGF-βR2 represents a non-canonical function of MED12 outside its role in the nuclear Mediator complex

    • This interaction helps explain how MED12 loss leads to TGF-β pathway activation and subsequent drug resistance

How does MED12 regulate TGF-β receptor signaling, and what methodologies can detect this interaction?

MED12 regulates TGF-β receptor signaling through direct interaction with TGF-βR2. This represents a cytoplasmic function of MED12 distinct from its nuclear role in transcriptional regulation .

Regulation mechanism:

  • MED12 primarily suppresses TGF-βR2 at the post-transcriptional level

  • MED12 knockdown results in strong induction of TGF-βR2 protein levels with only modest changes in mRNA levels

  • This leads to increased SMAD2 phosphorylation and activation of TGF-β signaling

Detection methodologies:

  • Subcellular fractionation to identify cytoplasmic MED12 (as shown in Figure 4K from search result 2)

  • Co-immunoprecipitation to detect MED12-TGF-βR2 interaction

  • 125I-TGF-β1 affinity-labeling assays to quantify cell-surface TGF-βR2 levels

  • Western blotting for downstream signaling components (p-SMAD2, p-MEK, p-ERK)

  • qRT-PCR to measure expression of TGF-β target genes

What is the experimental evidence linking MED12 to drug resistance mechanisms in cancer?

MED12 has been extensively characterized as a determinant of response to multiple cancer therapeutics. Loss of MED12 confers resistance through TGF-β pathway activation .

Evidence from functional genetic screens:

  • Large-scale RNAi screen identified MED12 as a gene whose suppression confers resistance to crizotinib in EML4-ALK translocation-positive NSCLC

  • Multiple independent shRNAs targeting MED12 consistently produced resistance phenotypes

  • Reconstitution with RNAi-resistant MED12 restored drug sensitivity, confirming specificity

Drugs affected by MED12 loss:

Drug ClassSpecific AgentsCancer TypesReference
ALK inhibitorsCrizotinib, NVP-TAE684NSCLC
EGFR inhibitorsGefitinib, ErlotinibNSCLC
BRAF inhibitorsPLX4032 (Vemurafenib)Melanoma
MEK inhibitorsAZD6244 (Selumetinib)Melanoma, CRC
Multikinase inhibitorsSorafenibHCC
ChemotherapyCisplatin, 5-FluorouracilVarious

Molecular mechanism:

  • MED12 knockdown leads to TGF-βR2 upregulation

  • Activated TGF-β signaling causes MEK/ERK activation

  • This bypasses the inhibitory effects of various targeted therapies

  • Suppression of TGF-βR2 restores drug sensitivity in MED12 knockdown cells

What experimental approaches can be used to study differential roles of nuclear versus cytoplasmic MED12?

MED12 has distinct functions in the nucleus (as part of the Mediator complex) and cytoplasm (regulating TGF-βR2) . Studying these compartment-specific functions requires specialized approaches:

  • Subcellular fractionation:

    • Nuclear and cytoplasmic fractionation followed by western blotting

    • Controls: Lamin A/C and SP1 for nuclear fractions; α-TUBULIN and HSP90 for cytoplasmic fractions

  • Localization-specific variants:

    • Generation of MED12 constructs with nuclear localization signal (NLS) mutations

    • Creation of MED12 fused to nuclear export signals (NES)

    • Expression of domain-specific MED12 variants

  • Proximity labeling approaches:

    • BioID or APEX2 fusion proteins to identify compartment-specific interaction partners

    • Comparison of nuclear versus cytoplasmic MED12 interactomes

  • Functional rescue experiments:

    • Expressing NLS-MED12 versus NES-MED12 in MED12 knockdown cells

    • Assessing rescue of specific phenotypes (drug sensitivity, TGF-β signaling, transcriptional changes)

How does MED12 contribute to EMT and what are the implications for cancer progression?

MED12 suppression induces an epithelial-mesenchymal transition (EMT)-like phenotype through activation of TGF-β signaling .

Evidence for MED12-regulated EMT:

  • MED12 knockdown induces expression of mesenchymal markers Vimentin (VIM) and N-cadherin (CDH2)

  • Protein products of these mesenchymal-specific genes are detected in MED12 knockdown cells at levels similar to those induced by TGF-β treatment

  • E-cadherin (CDH1) expression is not lost in MED12 knockdown cells, suggesting induction of a partial EMT

Experimental approaches to study MED12-regulated EMT:

  • RNA-Seq analysis of MED12 knockdown cells to identify EMT signature genes

  • qRT-PCR validation of mesenchymal markers (VIM, CDH2) and epithelial markers (CDH1)

  • Western blotting and immunofluorescence to assess protein-level changes

  • Migration and invasion assays to characterize functional consequences

Implications for cancer progression:

  • EMT is associated with chemotherapy resistance in colon cancer patients and to gefitinib in lung cancer

  • MED12 loss-induced EMT may contribute to therapeutic resistance and metastatic potential

  • The partial EMT phenotype may represent an intermediate state with distinct properties

What methods can be used to study MED12 in the context of transcriptional regulation at specific enhancers?

MED12 regulates HSC-specific enhancers , and studying its role in enhancer regulation requires specialized approaches:

  • Chromatin immunoprecipitation approaches:

    • ChIP-seq to map genome-wide MED12 binding sites

    • CUT&RUN or CUT&Tag for improved resolution of binding sites

    • ChIP-qPCR for targeted analysis of specific enhancers

  • Transcriptional analysis:

    • RNA-seq to assess global transcriptional changes upon MED12 manipulation

    • GRO-seq or PRO-seq to measure nascent transcription rates

    • 4C, Hi-C, or HiChIP to study enhancer-promoter interactions

  • CRISPR-based approaches:

    • CRISPR activation (CRISPRa) or inhibition (CRISPRi) at MED12-bound enhancers

    • CRISPR-mediated deletion of specific enhancers

    • CRISPR screening targeting enhancer regions

  • Reporter assays:

    • Luciferase reporter assays with wild-type and mutant enhancer sequences

    • STARR-seq for massively parallel enhancer activity testing

What expression systems are optimal for producing functional recombinant Pan troglodytes MED12?

While the search results don't specifically address expression systems for Pan troglodytes MED12, general considerations for recombinant MED12 expression include:

  • Expression system selection:

    • Mammalian expression systems (HEK293, CHO) may provide appropriate post-translational modifications

    • Baculovirus-insect cell systems offer high yields of complex mammalian proteins

    • Bacterial systems may be suitable for expression of specific domains

  • Construct design considerations:

    • Full-length MED12 is large (approximately 240 kDa) and may present expression challenges

    • Domain-specific constructs may be more amenable to recombinant expression

    • Fusion tags (His, GST, MBP) can aid in purification and solubility

  • Validation approaches:

    • Functional assays to confirm activity (e.g., ability to activate CDK8)

    • Interaction studies with known binding partners (TGF-βR2, CDK8 subcomplex components)

    • Structural analyses to confirm proper folding

What methods are most effective for analyzing MED12-dependent transcriptional changes?

MED12 knockdown significantly alters gene expression profiles across multiple cell types . Effective methods for analyzing these changes include:

  • Transcriptome analysis approaches:

    • RNA-Seq provides comprehensive analysis of transcriptional changes

    • MED12 knockdown signature genes have been identified across multiple cell lines

    • qRT-PCR validation of key target genes

  • Pathway analysis:

    • Gene set enrichment analysis (GSEA) to identify affected pathways

    • Comparison with known TGF-β response signatures

    • Analysis of EMT-related transcriptional programs

  • Integration with epigenomic data:

    • Correlation of transcriptional changes with chromatin accessibility (ATAC-seq)

    • Integration with histone modification profiles (ChIP-seq for H3K27ac, H3K4me1)

    • Analysis of enhancer activity at differentially expressed genes

  • Single-cell approaches:

    • scRNA-seq to identify cell populations differentially affected by MED12 manipulation

    • Trajectory analysis to map transcriptional state transitions

What are the key considerations for designing loss-of-function experiments targeting MED12?

Based on the search results, several important considerations emerge for MED12 loss-of-function studies:

  • RNAi approaches:

    • Multiple independent shRNAs should be used to confirm specificity

    • Both retroviral and lentiviral delivery systems have been successfully employed

    • Different cell types may require optimization of knockdown conditions

  • Rescue experiments:

    • Expression of RNAi-resistant MED12 constructs is essential to confirm specificity

    • Species-specific differences (e.g., using murine Med12 to rescue human MED12 knockdown)

    • Domain-specific variants can help dissect functional requirements

  • Phenotypic assessment:

    • Drug sensitivity assays require appropriate dose-response analysis

    • Cell cycle analysis to assess proliferation effects

    • Signaling pathway activation (Western blots for p-MEK, p-ERK, p-SMAD2)

    • Expression of TGF-β target genes and EMT markers

  • CRISPR-based approaches:

    • Complete knockout may be lethal in some contexts

    • Inducible or cell type-specific knockout systems

    • CRISPR interference for tunable repression

How can knowledge of MED12's role in drug resistance inform cancer treatment strategies?

Understanding MED12's role in drug resistance has significant implications for cancer therapy:

  • Biomarker development:

    • MED12 expression levels could serve as a biomarker for drug response

    • Nuclear overexpression of MED12 occurs in 40% of distant metastatic castration-resistant prostate cancer and 21% of local-recurrent CRPC

    • Loss of MED12 could predict resistance to multiple targeted therapies

  • Combination therapy strategies:

    • Co-targeting TGF-β signaling could overcome resistance in MED12-deficient tumors

    • TGF-β receptor inhibitors may resensitize MED12-low tumors to various cancer drugs

    • MEK inhibitors might address the downstream activation caused by MED12 loss

  • Patient stratification:

    • Screening for MED12 alterations could help guide treatment selection

    • MED12 status could inform decisions about TGF-β pathway inhibitors

  • Resistance monitoring:

    • Changes in MED12 expression during treatment might indicate emerging resistance

    • Analysis of circulating tumor DNA for MED12 alterations

What are the optimal methods for detecting MED12 alterations in patient samples?

Several approaches can be used to assess MED12 status in clinical samples:

  • Protein expression analysis:

    • Immunohistochemistry (IHC) on tissue microarrays to assess MED12 expression levels and subcellular localization

    • Western blotting of tumor lysates when sufficient material is available

    • Multiplex immunofluorescence to simultaneously evaluate MED12 and pathway components

  • Genetic analysis:

    • Targeted sequencing for MED12 mutations (present in 5.4% of primary prostate cancer)

    • RNA-seq to detect altered MED12 expression and splice variants

    • Whole-exome sequencing for comprehensive mutation profiling

  • Functional readouts:

    • Assessment of TGF-β pathway activation (SMAD3 phosphorylation)

    • Analysis of EMT markers as potential surrogates for MED12 dysfunction

    • Proliferation markers to correlate with MED12 status

  • Emerging approaches:

    • Digital spatial profiling to assess MED12 and pathway components with spatial context

    • Single-cell analysis of tumor samples to detect subpopulations with MED12 alterations

How does MED12 function compare between primates and other model organisms?

While the search results don't specifically address cross-species comparisons of MED12, understanding evolutionary conservation can provide insights into critical functional domains:

  • Functional conservation:

    • Core functions in the Mediator complex are likely conserved across vertebrates

    • Non-canonical roles (e.g., TGF-β regulation) may show more species-specific variation

    • Understanding conservation can help identify essential functional domains

  • Model organism considerations:

    • Mouse models have been valuable for studying Med12 function in development and disease

    • Non-mammalian models may reveal evolutionarily conserved core functions

    • Primate-specific functions may not be fully recapitulated in lower organisms

  • Experimental approaches:

    • Sequence alignment and structural modeling to identify conserved domains

    • Cross-species complementation studies

    • Comparative transcriptomics following MED12 manipulation in different species

What is the relationship between MED12 mutations in developmental disorders and its role in cancer?

MED12 mutations are associated with both developmental disorders and cancer:

  • Developmental disorders:

    • Mutations in MED12 cause X-linked Opitz-Kaveggia syndrome (FG syndrome)

    • Lujan-Fryns syndrome and X-linked Ohdo syndrome are also linked to MED12 mutations

    • These conditions involve intellectual disability and multiple congenital anomalies

  • Cancer relevance:

    • MED12 is recurrently mutated in 5.4% of primary prostate cancer

    • MED12 overexpression occurs in castration-resistant prostate cancer

    • Loss of MED12 function contributes to drug resistance in multiple cancer types

  • Molecular connections:

    • Both contexts may involve dysregulation of similar developmental signaling pathways

    • TGF-β, Wnt/β-catenin, and hedgehog signaling are implicated in both development and cancer

    • Investigating the specific mutations may reveal domain-specific functions

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.