TMEFF1 Human

TMEFF1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Structure and Expression of TMEFF1

TMEFF1 is a 374-amino-acid protein with three key domains:

  • N-terminal extracellular segment: Contains two Kazal-like domains and one EGF-like domain, critical for receptor interaction .

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

  • C-terminal intracellular segment: Short cytoplasmic tail with unclear functional significance .

Tissue specificity:

  • Predominantly expressed in brain cortical neurons, with minimal presence in other tissues .

  • mRNA and protein levels are highest in the human central nervous system (CNS), as confirmed by GTEx and Protein Atlas databases .

Antiviral Mechanism Against HSV-1

TMEFF1 restricts HSV-1 at the entry stage through two mechanisms:

Interaction with NECTIN-1

  • TMEFF1 binds directly to NECTIN-1, the primary HSV-1 receptor in neurons, disrupting glycoprotein D (gD)-mediated fusion of the viral envelope with the cell membrane .

  • The extracellular domain of TMEFF1 is sufficient to block viral entry, even when overexpressed in non-neuronal cells .

Disruption of Viral Capsid Transport

  • TMEFF1 interacts with non-muscle myosin heavy chains IIA/IIB (NMHC-IIA/B), which facilitate HSV-1 capsid movement toward the nucleus. This interaction delays viral translocation .

Genetic Evidence Linking TMEFF1 Deficiency to HSE

Two unrelated HSE patients were found to harbor homozygous deleterious variants in TMEFF1:

VariantImpact on ProteinFunctional Consequence
P44A (Patient 1)Missense mutation in N-terminal domainReduced interaction with NECTIN-1
c.1059-2A>G (Patient 2)Splice-site mutation causing exon skippingTruncated protein lacking transmembrane domain

Key findings:

  • Cortical neurons derived from TMEFF1 knockout human pluripotent stem cells (hPSCs) showed 10-fold higher HSV-1 replication rates compared to wild-type neurons .

  • Pretreatment with type I interferon (IFN-β) rescued viral restriction in TMEFF1-deficient neurons, but not in IFNAR1-deficient cells, indicating an IFN-independent mechanism .

Experimental Models Validating TMEFF1 Function

In Vitro Studies

  • hPSC-derived neurons: TMEFF1 knockout increased HSV-1 replication and neuronal death .

  • LUHMES neurons: CRISPR-Cas9 knockout of TMEFF1 enhanced viral gene expression and capsid nuclear localization .

In Vivo Studies

  • Tmeff1⁻/⁻ mice exhibited accelerated weight loss, higher viral loads in brain neurons, and 100% mortality post-HSV-1 infection, unlike wild-type mice .

Therapeutic Implications

  • Soluble TMEFF1 fragments: The extracellular domain of TMEFF1 could be engineered as a decoy receptor to block HSV-1 entry .

  • Gene therapy: Restoring TMEFF1 expression in susceptible neurons may prevent HSE in genetically at-risk individuals .

Unanswered Questions and Future Directions

  • Glial cell role: TMEFF1’s restriction mechanism appears neuron-specific; its function in astrocytes or microglia remains unexplored .

  • Broader antiviral activity: Whether TMEFF1 inhibits other neurotropic viruses (e.g., HSV-2, SARS-CoV-2) is unknown .

Product Specs

Introduction
TMEFF1, a type I transmembrane glycoprotein, plays a crucial role in cellular signaling. Its structure encompasses two follistatin modules, an EGF domain in the extracellular region, a transmembrane domain, and a short cytoplasmic tail. Notably, the extracellular domain can be released as a soluble protein. TMEFF1 exhibits predominant expression in the brain but experiences downregulation in brain tumors. Functionally, it selectively regulates nodal signaling, without affecting activin signaling, by directly interacting with the nodal co-receptor, Cripto. TMEFF1 exerts inhibitory effects on NODAL and BMP signaling pathways, influencing neural patterning. Furthermore, it is implicated as a potential tumor suppressor in brain cancers.
Description
Recombinant TMEFF1 Human, produced in E. coli, is a single polypeptide chain comprising 314 amino acids (40-330) with a molecular weight of 33.9 kDa. This protein is fused to a 23 amino acid His-tag at the N-terminus and purified using proprietary chromatographic methods.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
TMEFF1 is supplied as a 0.25 mg/ml solution in 20mM Tris-HCl buffer (pH 8.0), containing 0.1M NaCl and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, freeze at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity exceeds 90% as determined by SDS-PAGE analysis.
Synonyms
C9orf2, CT120.1, H7365, TR-1, Tomoregulin-1, Transmembrane protein with EGF-like and one follistatin-like domain, TMEFF1.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSSNQPPGG GGGSGGDCPG GKGKSINCSE LNVRESDVRV CDESSCKYGG VCKEDGDGLK CACQFQCHTN YIPVCGSNGD TYQNECFLRR AACKHQKEIT VIARGPCYSD NGSGSGEGEE EGSGAEVHRK HSKCGPCKYK AECDEDAENV GCVCNIDCSG YSFNPVCASD GSSYNNPCFV REASCIKQEQ IDIRHLGHCT DTDDTSLLGK KDDGLQYRPD VKDASDQRED VYIGNHMPCP ENLNGYCIHG KCEFIYSTQK ASCRCESGYT GQHCEKTDFS ILYVVPSRQK LTHV.

Q&A

What is the basic structure of human TMEFF1 protein?

Human TMEFF1 is a type I transmembrane glycoprotein with a complex domain structure. The protein contains two follistatin modules and an EGF domain in its extracellular region, followed by a transmembrane domain and a short cytoplasmic tail . The extracellular domain can be released as a soluble protein through proteolytic cleavage, suggesting potential dual functionality in both membrane-bound and soluble forms . The full protein spans from Ser40 to Thr328 in the human sequence (accession number Q8IYR6), with the full-length construct being critical for appropriate experimental studies of its function .

What is the predominant tissue expression pattern of TMEFF1 in humans?

TMEFF1 demonstrates distinct tissue specificity with predominant expression in the brain and nervous system tissues . This expression pattern is particularly notable during embryonic development, suggesting important roles in neural development and function . When investigating TMEFF1 expression in research settings, it's advisable to use positive control tissues such as epididymis, which has been validated for TMEFF1 expression studies . Expression analysis requires careful consideration of tissue specificity when designing experiments to understand TMEFF1's physiological roles.

How does TMEFF1 interact with signaling pathways in normal cellular contexts?

In normal physiological conditions, TMEFF1 selectively regulates nodal signaling pathways through direct binding to the nodal co-receptor Cripto, without affecting activin signaling . This selective regulatory function demonstrates TMEFF1's discriminatory capacity in signal transduction pathways. When investigating these interactions, co-immunoprecipitation experiments coupled with functional assays that measure pathway activation (such as reporter gene assays) provide the most reliable approach to characterize these selective binding interactions.

How does TMEFF1 expression differ across various human cancers?

TMEFF1 exhibits remarkable context-dependent expression and function across different cancer types, creating a complex research landscape:

Cancer TypeTMEFF1 Expression PatternFunctional RoleReference
Brain tumorsDownregulatedTumor suppressor
Ovarian cancerUpregulatedTumor promoter
Endometrial carcinomaUpregulatedAssociated with advanced disease

This divergent expression pattern necessitates cancer-specific investigation approaches. For brain tumors, restoration of expression may be a therapeutic strategy, while in gynecological cancers, inhibition might be more appropriate. Expression analysis should include both protein (immunohistochemistry, western blot) and mRNA (qRT-PCR) measurements to fully characterize TMEFF1 status in different tumor types .

What role does TMEFF1 play in brain defense mechanisms against viral infections?

Recent research has uncovered a previously unknown defense mechanism involving TMEFF1 that protects the brain against herpesvirus infections . Despite herpesvirus being present in 50-80% of the population, severe brain infections occur in only 1 in 250,000 cases. TMEFF1 produces a protein that specifically blocks herpesvirus from entering nerve cells, creating a critical barrier of protection . This protective role represents a novel function distinct from TMEFF1's involvement in cancer pathways. Research methodologies for studying this phenomenon should include viral challenge models in cells with modulated TMEFF1 expression, measuring viral entry, replication, and cellular response markers.

How do MAPK and PI3K/AKT pathways relate to TMEFF1 function in cancer cells?

TMEFF1 overexpression in ovarian cancer cells activates both MAPK and PI3K/AKT signaling pathways, with significant increases in phosphorylated node proteins within these pathways . This activation directly correlates with increased cellular proliferation, migration, and invasion, while reducing apoptosis in experimental models. The causative relationship was confirmed when adding specific pathway inhibitors (PD98059 for MAPK and GDC-0941 for PI3K) suppressed the malignant behavior in TMEFF1-overexpressing cells .

For investigating these pathway interactions, researchers should:

  • Use both pharmacological inhibitors and genetic knockdown approaches

  • Measure phosphorylation status of multiple nodes in each pathway

  • Confirm functional outcomes with cellular assays (proliferation, migration, invasion)

  • Employ rescue experiments to establish causal relationships

What experimental techniques are most effective for modulating TMEFF1 expression in research models?

Based on the research literature, several effective approaches for modulating TMEFF1 expression have been validated:

ApproachMethodAdvantagesApplications
OverexpressionPlasmid transfection with Lipofectamine 3000 followed by G418 selectionStable expression for long-term studiesOVCAR3, ES-2 cell lines
KnockdownVirus-mediated shRNA delivery with Pour method selectionEfficient reduction of expressionCAOV3, SKOV3 cell lines
Transient knockdownsiRNA transfection with lipofectionRapid effect assessment without selectionApoptosis studies

For optimal results, expression modulation should be confirmed at both mRNA and protein levels. When using fluorescent markers (e.g., GFP), researchers should be aware that strong fluorescence might interfere with certain downstream assays like flow cytometry with fluorescent antibodies .

What is the recommended methodology for studying TMEFF1's impact on cell migration and invasion?

For investigating TMEFF1's effects on migration and invasion, researchers have successfully employed multiple complementary techniques:

  • Scratch assay: Culture cells to 90% confluence, create a scratch with a 100-μL pipette tip, wash gently with PBS, replace with serum-free medium, and measure scratch width at 0 and 24 hours . This provides a straightforward assessment of collective cell migration.

  • Transwell assay: More appropriate for quantifying individual cell invasion through extracellular matrix components, particularly relevant for understanding TMEFF1's role in metastatic potential .

  • Epithelial-mesenchymal transition (EMT) marker analysis: Western blot assessment of E-cadherin (epithelial marker) and vimentin/N-cadherin (mesenchymal markers) provides mechanistic insight into migration/invasion changes .

For comprehensive characterization, all three approaches should be employed in parallel, with at least three biological replicates per experiment.

How should researchers interpret the apparently contradictory roles of TMEFF1 across different cancer types?

The dual nature of TMEFF1 as both tumor suppressor (in brain tumors) and oncogene (in gynecological cancers) presents a significant interpretative challenge . This contradiction likely reflects tissue-specific contexts, interacting partners, and downstream effectors. When investigating these seemingly contradictory functions:

  • Always include appropriate tissue-specific controls

  • Examine the complete signaling network, not just TMEFF1 in isolation

  • Consider potential splice variants or post-translational modifications

  • Evaluate both membrane-bound and soluble forms of TMEFF1

  • Examine the epigenetic landscape and transcriptional regulation in each tissue type

The most informative approach involves parallel studies in multiple cancer types using identical methodologies to directly compare effects.

What is the relationship between p53 and TMEFF1, and how should it be investigated?

Evidence suggests p53 may directly regulate TMEFF1 expression, with chromatin immunoprecipitation (ChIP) assays confirming p53 binding to the TMEFF1 promoter region . Further support comes from observations that inhibiting TP53 in ovarian cancer cell lines (CAOV3 and ES-2) downregulates TMEFF1 protein expression .

For investigating this regulatory relationship:

  • ChIP assays should include multiple primer sets spanning the putative p53 binding sites

  • Complement with luciferase reporter assays using wild-type and mutated TMEFF1 promoter constructs

  • Employ p53 modulation through both genetic (knockdown/overexpression) and pharmacological (nutlin-3a) approaches

  • Confirm findings across multiple cell types with different p53 status (wild-type, mutant, null)

This multifaceted approach provides the most comprehensive understanding of this potentially important regulatory mechanism.

What are the technical considerations for studying the soluble versus membrane-bound forms of TMEFF1?

TMEFF1 exists in both membrane-bound and soluble forms, with the extracellular domain capable of being released as a soluble protein . This dual localization creates technical challenges requiring specific experimental approaches:

  • For membrane-bound TMEFF1:

    • Surface biotinylation followed by precipitation

    • Membrane fractionation before western blotting

    • Immunofluorescence with non-permeabilized cells

  • For soluble TMEFF1:

    • Concentration of conditioned media before analysis

    • ELISA development with antibodies specific to the extracellular domain

    • Analysis of different proteolytic processing events

  • For functional discrimination:

    • Rescue experiments with either full-length or soluble-only constructs

    • Domain-specific mutations to determine functional regions

    • Co-culture systems separating producer and responder cells

Understanding the differential functions of these forms may help resolve some of the apparently contradictory roles observed across different cancer types.

What is the potential connection between TMEFF1's role in brain defense and neurodegenerative diseases?

Recent discoveries about TMEFF1's protection against herpesvirus in the brain suggest possible implications for neurodegenerative conditions like Alzheimer's disease . This connection warrants investigation through:

  • Examination of TMEFF1 expression in neurodegenerative disease tissues

  • Analysis of viral presence in relation to TMEFF1 levels in these tissues

  • Animal models with conditional TMEFF1 knockout in specific brain regions

  • Long-term viral exposure studies in models with modulated TMEFF1

These approaches could reveal whether TMEFF1 dysregulation contributes to neurodegenerative processes through impaired viral defense mechanisms.

How can single-cell approaches advance our understanding of TMEFF1 in heterogeneous tissues?

Given TMEFF1's context-dependent functions, single-cell technologies offer powerful approaches to resolve cell-specific expression and functions:

  • Single-cell RNA sequencing to identify specific cell populations expressing TMEFF1

  • Spatial transcriptomics to understand TMEFF1 expression in tissue architecture context

  • CyTOF or multiplexed immunofluorescence for protein-level single-cell analysis

  • Single-cell ATAC-seq to examine chromatin accessibility at the TMEFF1 locus

These technologies can help resolve contradictory findings by identifying specific cellular contexts in which TMEFF1 exhibits different functions, potentially reconciling its dual roles in cancer and normal physiology.

Product Science Overview

Structure and Function

TMEFF1 is a transmembrane protein that is primarily located in the plasma membrane. It is involved in several critical biological processes, including neural patterning and tumor suppression. The protein is predicted to inhibit NODAL and BMP signaling pathways, which are crucial during neural development . Additionally, TMEFF1 has been suggested to act as a tumor suppressor in brain cancers .

Expression and Localization

The expression of TMEFF1 is predominantly observed in the central nervous system (CNS). It plays a significant role in the development and maintenance of neural tissues. The protein’s localization to the plasma membrane allows it to participate in cell signaling processes that are essential for neuron projection development and tissue morphogenesis .

Clinical Significance

TMEFF1 has been associated with various diseases, including acute promyelocytic leukemia and leukoencephalopathy with vanishing white matter . Its role as a tumor suppressor highlights its potential importance in cancer research, particularly in understanding the mechanisms underlying brain cancers .

Recent Research

Recent studies have identified TMEFF1 as a critical factor in controlling herpes simplex virus 1 (HSV-1) replication in the CNS. Depletion of TMEFF1 in human neurons and mice led to increased viral replication and neuronal death following HSV-1 infection . This discovery underscores the importance of TMEFF1 in the innate antiviral defense mechanisms of neurons and its potential therapeutic implications .

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.