ErbB4 Human Recombinant produced in HEK293 Cells is a single, glycosylated polypeptide chain containing 863 amino acids (26-649 a.a) and having a molecular mass of 96.6 kDa.
ErbB4 is fused to a 239 amino acid hIgG-His-Tag at C-terminus & purified by proprietary chromatographic techniques.
The epidermal growth factor receptor (EGFR) family, comprising receptor tyrosine kinases, plays a crucial role in mediating the effects of EGF family growth factors. ErbB4, a member of this family, functions as a receptor tyrosine kinase. Structurally, it exists as a single-pass type I transmembrane protein characterized by multiple furin-like cysteine-rich domains, a PDZ domain binding motif, a tyrosine kinase domain, and a phosphotidylinositol-3 kinase binding site. ErbB4 exhibits low affinity for its ligands and is found in various tissues including normal skeletal muscle, heart, pituitary, brain, and certain breast carcinomas. In its monomeric form, ErbB4 contributes to neuronal development, heart development, and cancer progression.
Recombinant Human ErbB4, expressed in HEK293 cells, is a single, glycosylated polypeptide chain. It consists of 863 amino acids, spanning from position 26 to 649, with a molecular weight of 96.6 kDa. The protein is engineered with a C-terminal fusion of a 239 amino acid hIgG-His-Tag and purified using proprietary chromatographic techniques.
Sterile filtered, colorless solution.
The ErbB4 solution is provided at a concentration of 0.25 mg/ml in phosphate buffered saline (pH 7.4) containing 10% glycerol.
For short-term storage (2-4 weeks), the product should be stored at 4°C. For extended periods, storage at -20°C in a frozen state is recommended. To enhance long-term stability, the addition of a carrier protein (0.1% HSA or BSA) is advised. Repeated freezing and thawing should be avoided.
Purity is determined to be greater than 90.0% using SDS-PAGE analysis.
The biological activity of the protein is evaluated based on its binding affinity to Human NRG1 (CAT# cyt-1186) in a functional ELISA assay. The ED50 value is determined to be less than or equal to 0.2 ug/ml.
Receptor tyrosine-protein kinase erbB-4, Proto-oncogene-like protein c-ErbB-4, Tyrosine kinase-type cell surface receptor HER4, p180erbB4, 4ICD, E4ICD, s80HER4, ERBB4, HER4, receptor tyrosine-protein kinase erbB-4 isoform JM-a/CVT-1, receptor tyrosine-protein kinase erbB-4, tyrosine kinase-type cell surface receptor HER4, avian erythroblastic leukemia viral (v-erb-b2) oncogene homolog 4, proto-oncogene-like protein c-ErbB-4, ALS19, v-erb-a erythroblastic leukemia viral oncogene homolog 4, v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 4, human epidermal growth factor receptor 4.
HEK293 Cells.
QSVCAGTENK LSSLSDLEQQ YRALRKYYEN CEVVMGNLEI TSIEHNRDLS FLRSVREVTG YVLVALNQFR YLPLENLRII RGTKLYEDRY ALAIFLNYRK DGNFGLQELG LKNLTEILNG GVYVDQNKFL CYADTIHWQD IVRNPWPSNL TLVSTNGSSG CGRCHKSCTG RCWGPTENHC QTLTRTVCAE QCDGRCYGPY VSDCCHRECA GGCSGPKDTD CFACMNFNDS GACVTQCPQT FVYNPTTFQL EHNFNAKYTY GAFCVKKCPH NFVVDSSSCV RACPSSKMEV EENGIKMCKP CTDICPKACD GIGTGSLMSA QTVDSSNIDK FINCTKINGN LIFLVTGIHG DPYNAIEAID PEKLNVFRTV REITGFLNIQ SWPPNMTDFS VFSNLVTIGG RVLYSGLSLL ILKQQGITSL QFQSLKEISA GNIYITDNSN LCYYHTINWT TLFSTINQRI VIRDNRKAEN CTAEGMVCNH LCSSDGCWGP GPDQCLSCRR FSRGRICIES CNLYDGEFRE FENGSICVEC DPQCEKMEDG LLTCHGPGPD NCTKCSHFKD GPNCVEKCPD GLQGANSFIF KYADPDRECH PCHPNCTQGC NGPTSHDCIY YPWTGHSTLP QHARLEPKSC DKTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHED PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLH QDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSRDELTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS LSLSPGKHHH HHH
ErbB4, also known as HER4 (Human Epidermal growth factor Receptor 4), is a 180-kDa type I membrane glycoprotein belonging to the ErbB family of tyrosine kinase receptors. It functions as a receptor for members of the epidermal growth factor (EGF) family of growth factors. Unlike ErbB3, which contains a defective kinase domain, ErbB4 possesses a fully functional tyrosine kinase domain essential for signal transduction. The receptor spans from amino acids Gln26 to Arg649 (Accession # Q15303) and contains multiple domains including an extracellular ligand-binding region, a transmembrane domain, and an intracellular kinase domain .
When comparing the four ErbB family members (ErbB1/EGFR, ErbB2/HER2, ErbB3/HER3, and ErbB4/HER4), ErbB4 is unique in that it undergoes regulated intramembrane proteolysis, producing a soluble intracellular domain capable of nuclear translocation. This mechanism allows ErbB4 to function as both a traditional cell surface receptor and a transcriptional co-regulator, distinguishing it functionally from other family members.
ErbB4 exhibits a tissue-specific expression pattern that researchers must consider when designing experimental models. The receptor is predominantly expressed in:
Normal skeletal muscle
Heart tissue (particularly important for cardiac development and function)
Pituitary gland
Brain (especially in regions involved in synaptic plasticity)
Breast tissue (and several breast carcinomas)
Colon (including detection in smooth muscle cells of human colon cancer tissue)
This tissue distribution has significant implications for experimental design. When studying ErbB4 in cardiovascular contexts, researchers should utilize cardiac-specific conditional knockout models rather than global knockouts, which are embryonically lethal due to cardiac defects. The ErbB4−/−HER4+/heart mouse model, where human ErbB4 expression is driven in the heart by the α-myosin heavy chain promoter, demonstrates this approach by rescuing an otherwise lethal cardiac defect while allowing study of ErbB4 deletion in other tissues .
Additionally, when investigating ErbB4 in cancer models, researchers should verify endogenous expression levels in their chosen cell lines. MCF-7 breast cancer cells and certain colon cancer cell lines have been validated for ErbB4 expression studies .
ErbB4 can be activated by multiple ligands with varying effects on receptor dimerization and downstream signaling:
Primary ErbB4 ligands include:
Dimerization patterns:
Ligand-specific effects:
When designing experiments to study ErbB4 activation, researchers should carefully select specific ligands based on the biological context being studied. For cardiac protection studies, neuregulin-1 is often the preferred ligand due to its established role in the neuregulin-1/ERBB4 cardioprotective pathway .
Detecting and quantifying ErbB4 requires selecting appropriate methods based on your experimental goals. The following validated techniques offer different advantages:
Immunocytochemistry (ICC) for cellular localization:
ErbB4/Her4 can be detected in immersion-fixed cells (e.g., MCF-7 breast cancer line)
Optimal antibody concentration: 10 μg/mL for 3 hours at room temperature
Visualization with fluorescent-conjugated secondary antibodies
Counterstaining with DAPI allows nuclear visualization
ErbB4 typically shows specific staining localized to plasma membrane
Immunohistochemistry (IHC) for tissue sections:
Western blotting for protein expression levels:
Direct ELISA for quantitative assessment:
When designing experiments to detect ErbB4, consider the following technical recommendations:
Store antibodies at -20 to -70°C for up to 12 months
After reconstitution, store at 2-8°C for up to 1 month under sterile conditions
For longer storage after reconstitution, maintain at -20 to -70°C for up to 6 months
Use manual defrost freezers and avoid repeated freeze-thaw cycles
Four structurally different ErbB4 isoforms exist due to alternative splicing, making isoform-specific detection crucial for accurate experimental interpretation. The following methodological approaches enable effective isoform discrimination:
RT-PCR and qPCR analysis:
Design primers spanning the alternative splice junctions
For juxtamembrane region: distinguish between JM-a (cleavable) and JM-b (non-cleavable) variants
For cytoplasmic region: differentiate between CYT-1 (contains PI3K binding site) and CYT-2 (lacks this binding site)
Use quantitative PCR to determine relative isoform expression levels
Western blot analysis:
Resolve proteins on high-percentage gels to separate the slightly different molecular weights
Use antibodies that recognize specific isoforms when available
For JM-a isoforms, look for the cleaved intracellular domain fragment (~80 kDa)
Functional assays to verify isoform activity:
JM-a isoforms: assess nuclear localization following ligand stimulation
CYT-1 isoforms: examine PI3K pathway activation (Akt phosphorylation)
Use appropriate positive controls with known isoform expression
Isoform-specific knockdown:
Design siRNAs or shRNAs targeting isoform-specific sequences
Validate knockdown efficiency with isoform-specific qPCR
Confirm functional effects align with known isoform activities
This isoform heterogeneity has significant implications for ErbB4 research, as different isoforms may mediate distinct biological processes. For example, the JM-a isoforms that undergo proteolytic processing may be more relevant for studies of nuclear signaling, while CYT-1 isoforms might be more important in PI3K-dependent cardioprotection mechanisms .
Rigorous controls and validation steps are critical for generating reliable data when investigating ErbB4 signaling. Implement these essential measures:
Receptor specificity controls:
Ligand validation:
Include both positive controls (known ErbB4 ligands like NRG-1β) and negative controls (non-binding growth factors)
Determine dose-response relationships with increasing ligand concentrations
Confirm ligand bioactivity using established assays
Phosphorylation assessment:
Monitor receptor autophosphorylation as evidence of activation
Examine both ErbB4 phosphorylation and downstream effector activation
Use phospho-specific antibodies for key signaling nodes
Pharmacological validation:
Sex-specific considerations:
A comprehensive validation approach used in recent studies demonstrates the importance of these controls: small molecule EF-1's cardioprotective effects were validated as ErbB4-dependent by showing that these effects were absent in Erbb4-null mice. This approach confirms that observed effects are specifically mediated through ErbB4 rather than off-target mechanisms .
ErbB4 has emerged as a critical regulator of synaptic plasticity with significant implications for schizophrenia research. The following evidence establishes this connection:
Genetic associations:
Synaptic plasticity regulation:
Long-term potentiation (LTP) at Schaffer-collateral CA1 synapses is markedly enhanced in mutant mice lacking ErbB4
Acutely blocking ErbB4 with PD158780 in wild-type animals similarly enhances LTP
These findings indicate that ErbB4 activity constitutively suppresses LTP in the adult hippocampus
Conversely, increasing ErbB4 signaling with NRG-1β further suppresses LTP
Pathway specificity:
Molecular interactions:
The functional significance of these findings for schizophrenia research is substantial: cognitive deficits in schizophrenia may result from hyperfunction of ErbB4 signaling leading to suppressed glutamatergic synaptic plasticity. This aligns with the glutamate hypothesis of schizophrenia, which proposes NMDA receptor hypofunction as a contributing factor. These findings open new therapeutic approaches focused on modulating ErbB4 signaling to enhance synaptic plasticity and potentially improve cognitive function in schizophrenia .
Investigating ErbB4's role in synaptic plasticity requires specialized methodological approaches that can detect subtle changes in synaptic function. The following techniques have proven effective:
Electrophysiological approaches:
Experimental preparation:
Acute hippocampal slices (300-400 μm thick) from adult animals
Artificial cerebrospinal fluid (ACSF) with specific composition:
132 mM NaCl, 3 mM KCl, 1.25 mM NaH₂PO₄, 2 mM MgCl₂
11 mM D-glucose, 20 mM NaHCO₃, 2 mM CaCl₂
Saturated with 95% O₂/5% CO₂ at 28 ± 2°C (pH 7.40; 315-325 mOsm)
Supplementation with bicuculline methiodide (5 μM) to block GABA₂ receptors
Stimulation and recording parameters:
Pharmacological manipulations:
Data analysis:
Using these approaches, researchers have demonstrated that ErbB4 deletion enhances LTP: in ErbB4-deficient mice, fEPSP slope was 196 ± 13% of baseline 60 minutes after theta-burst stimulation, compared to 150 ± 7% in wild-type controls. Similar enhancement occurred with pharmacological inhibition, where PD158780-treated slices showed 189 ± 11% potentiation versus 149 ± 4% in untreated controls .
Understanding the distinctions between genetic deletion and acute pharmacological inhibition of ErbB4 provides important insights into the receptor's role in neurological function. These approaches differ in several key aspects:
ErbB4 plays critical roles in both cardiac development and adult heart function, making it an important target for cardiovascular research:
Developmental importance:
The ERBB system regulates cardiac embryonic development
Mice with global deletion of Erbb4 die during embryonic development due to cardiac defects
To study non-cardiac functions, researchers developed ErbB4−/−HER4heart mice, where human ErbB4 expression is restricted to cardiac tissue via the α-myosin heavy chain promoter
Adult cardiac function:
ErbB4 preserves normal cardiac function in adult hearts
The neuregulin-1/ERBB4 pathway provides cardioprotective benefits
Mice with conditional post-natal deletion of Erbb4 develop cardiomyopathy
Humans treated with anti-ERBB2 antibodies for breast cancer can develop cardiotoxicity, partly due to disruption of ErbB signaling
Stress response mechanisms:
The ERBB system is activated during cardiac overload or injury as a compensatory mechanism
ErbB4 signaling provides protection through multiple mechanisms:
Cellular targets:
The essential nature of ErbB4 in cardiac function is highlighted by the fact that researchers must use cardiac-specific rescue models to study ErbB4 function in other tissues. Without this cardiac expression, global ErbB4 knockout is embryonically lethal due to cardiac developmental defects .
Recent research has demonstrated that small-molecule activation of ErbB4 represents a promising therapeutic approach for heart failure. The mechanisms and evidence include:
Small-molecule activator identification:
Screening of 10,240 compounds identified eight structurally similar molecules (EF-1 to EF-8)
These compounds induce ErbB4 dimerization, with EF-1 being the most effective
This approach overcomes limitations of recombinant neuregulin-1 therapy, which requires intravenous delivery and lacks receptor specificity
Cellular protective mechanisms:
In vivo cardioprotective effects:
Sex-specific considerations:
This research demonstrates that direct, small-molecule-mediated activation of ErbB4 is feasible and effective for cardioprotection. The approach offers advantages over protein-based therapies, including oral bioavailability, greater specificity, and potentially reduced cost. The discovery of the EF compounds represents a significant step toward developing a novel class of drugs for treating heart failure .
Designing rigorous studies to investigate ErbB4's role in heart failure requires careful methodological planning. Key considerations include:
Model selection:
In vitro models:
In vivo models:
Sex considerations:
Validation of ErbB4 dependency:
Screening methodologies for identifying activators:
Outcome measures:
Following these methodological principles, researchers identified EF-1 as a promising ErbB4 activator that reduced heart damage through multiple mechanisms. The comprehensive validation approach - showing effects were absent in Erbb4-null mice - provides strong evidence for target specificity and mechanism of action .
ErbB4 heterodimerization represents a complex mechanism for diversifying signaling outcomes. The following aspects are critical for understanding this complexity:
Dimerization partners:
Ligand influence on dimer formation:
Signaling consequences:
Different dimer combinations activate distinct downstream pathways
ErbB4-ErbB2 dimers typically activate MAPK/ERK pathways strongly
ErbB4-ErbB3 dimers may preferentially activate PI3K/Akt signaling
ErbB4 homodimers have distinct signaling properties from heterodimers
Methodological approaches to study heterodimer specificity:
Co-immunoprecipitation to identify dimer partners
Proximity ligation assays to visualize dimers in situ
FRET/BRET approaches to measure dimer formation in live cells
Selective knockout/knockdown of potential partners
Therapeutic implications:
Understanding the rules governing ErbB4 dimerization patterns is essential for developing targeted therapeutic approaches. The identification of small molecules that specifically induce ErbB4 dimerization represents an important advance in this direction, offering greater specificity than natural ligands that may activate multiple ErbB receptors .
The molecular mechanisms underlying ErbB4's suppression of hippocampal long-term potentiation (LTP) involve complex interactions with glutamatergic signaling machinery:
Constitutive suppression of LTP:
NMDA receptor interactions:
Pathway specificity:
Pharmacological evidence:
Quantitative effects:
Understanding these mechanisms has significant implications for schizophrenia research, as cognitive deficits in this disorder may result from hyperfunction of ErbB4 signaling, leading to suppressed glutamatergic synaptic plasticity. Pharmacological targeting of ErbB4 to enhance LTP might therefore represent a novel therapeutic approach for cognitive symptoms of schizophrenia .
Contradictory findings in ErbB4 research literature can arise from multiple sources. Researchers should employ the following approaches to reconcile these inconsistencies:
By systematically addressing these factors, researchers can better understand apparent contradictions in the literature. The weight of evidence from multiple approaches (genetic deletion, pharmacological inhibition, and ligand application) consistently supports ErbB4's role as a suppressor of LTP in adult hippocampus, despite some contradictory findings in other experimental systems .
ErbB4 research has revealed several promising therapeutic applications across different disease areas:
Heart failure treatment:
Small-molecule ErbB4 activators represent a novel therapeutic class
Compounds like EF-1 show cardioprotective effects through multiple mechanisms:
Advantages over protein-based therapies include oral bioavailability and receptor specificity
Schizophrenia cognitive symptom management:
Cancer treatment adjuvants:
Chemotherapy-induced cardiotoxicity prevention:
The most advanced application appears to be in heart failure, where small-molecule ErbB4 activators have demonstrated efficacy in multiple models. The identification of compounds like EF-1 through EF-8 provides promising leads for further drug development in this area .
Advancing ErbB4 research requires several key methodological improvements:
Isoform-specific tools:
Improved in vivo imaging:
Methods to visualize ErbB4 activation and trafficking in living tissues
Techniques to monitor dimerization patterns in vivo
Real-time assessment of downstream signaling
These approaches would bridge the gap between in vitro and in vivo findings
Single-cell analysis techniques:
Methods to assess ErbB4 function in individual cells within complex tissues
Single-cell transcriptomics to identify cell-specific responses
Spatial transcriptomics to map ErbB4 activity in tissue context
These techniques would provide higher resolution understanding of ErbB4 biology
Sex-specific research frameworks:
Translational models:
Development of humanized models that better recapitulate human ErbB4 biology
Patient-derived systems to study disease-specific alterations
These approaches would enhance the predictive value of preclinical research
Addressing these methodological needs would enable researchers to resolve current contradictions in the literature, better understand the complex biology of ErbB4, and accelerate the development of therapeutic applications in cardiovascular disease, neuropsychiatric disorders, and beyond.
Integrating ErbB4 research with other signaling pathways presents significant opportunities for revealing new biological insights and therapeutic approaches:
Interaction with glutamatergic signaling:
Cross-talk with angiotensin-II pathways:
Integration with sex hormone signaling:
Connections with cancer signaling networks:
Integration with inflammatory pathways:
ErbB4 has anti-inflammatory actions in cardiac contexts
Understanding how ErbB4 modulates inflammatory signaling
This could extend therapeutic applications to inflammatory conditions
By studying ErbB4 not in isolation but as part of an integrated signaling network, researchers can develop more comprehensive models of its function and design more effective therapeutic strategies. The recent success in identifying small-molecule ErbB4 activators for heart failure treatment demonstrates the potential of this approach .
Tyrosine kinase ErbB-4, also known as HER4, is a member of the epidermal growth factor receptor (EGFR) family of receptor tyrosine kinases. This family includes four closely related receptors: EGFR (ErbB-1), ErbB-2 (HER2/neu), ErbB-3 (HER3), and ErbB-4 (HER4). These receptors play crucial roles in the regulation of cell growth, survival, differentiation, and migration.
ErbB-4 is a single-pass type I transmembrane protein with multiple functional domains . These include:
ErbB-4 is activated by binding to specific ligands, including neuregulins (NRG-2, NRG-3, and NRG-4), heparin-binding EGF-like growth factor, and betacellulin . Upon ligand binding, ErbB-4 undergoes dimerization and autophosphorylation, leading to the activation of downstream signaling pathways that regulate various cellular processes, including mitogenesis and differentiation .
ErbB-4 plays a critical role in the development and function of various tissues, including the heart, nervous system, and mammary glands. It is involved in the regulation of cell proliferation, differentiation, and survival. Additionally, ErbB-4 signaling has been implicated in the development of the central nervous system and the regulation of synaptic plasticity .
Mutations and dysregulation of ErbB-4 have been associated with various diseases, including cancer and neurological disorders . For example:
Human recombinant ErbB-4 is produced using recombinant DNA technology, which involves the insertion of the ERBB4 gene into a suitable expression system, such as bacteria, yeast, or mammalian cells. This allows for the production of large quantities of the protein for research and therapeutic purposes. Recombinant ErbB-4 is used in various studies to investigate its structure, function, and role in disease, as well as to develop potential therapeutic interventions targeting ErbB-4 signaling pathways.