NRG1 Human

Heregulin-B2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Isoforms

NRG1 exists in over 14 isoforms generated via alternative splicing or promoter usage. Key structural features include:

DomainFunctionPresent in Isoforms
Ig-like domainCell adhesion and receptor bindingType I (HRG, ARIA), Type II (GGF2)
EGF-like domainErbB3/ErbB4 receptor activationAll isoforms
Cysteine-rich domainNeuronal development and myelinationType III (SMDF)
Transmembrane domainAnchoring to cell membraneType I, III, IV
  • Type I (Heregulin/ARIA): Contains Ig-like and EGF-like domains; regulates synaptic plasticity and acetylcholine receptor clustering .

  • Type II (GGF2): Involved in glial cell development .

  • Type III (SMDF): Lacks Ig domain but critical for Schwann cell myelination .

Recombinant human NRG1 (rhNRG1) used in research typically corresponds to the EGF-like domain (e.g., residues 177–244), essential for receptor binding .

Nervous System

  • Synaptic Plasticity: Regulates excitatory-inhibitory balance; disruptions linked to schizophrenia .

  • Myelination: Promotes oligodendrocyte differentiation and CNS axon myelination via ErbB4 .

  • Neuroprotection: Reduces infarct size by 90% in rodent stroke models and inhibits neuroinflammation .

Cardiovascular System

  • Cardiac Development: Essential for myocardial trabeculation and endothelial-mesenchymal signaling .

  • Heart Failure Therapy: Recombinant NRG1 (Neucardin®) improves ejection fraction in clinical trials (Phase II) by enhancing cardiomyocyte survival and calcium homeostasis .

Pulmonary System

  • Lung Development: NRG1/HER2-HER3 signaling increases epithelial cell proliferation and reduces surfactant protein-A (SP-A) in fetal lungs .

Schizophrenia

  • Genetic Link: NRG1 missense mutations (e.g., Val→Leu in transmembrane domain) reduce γ-secretase cleavage, elevating pro-inflammatory cytokines (IL-6, TNF-α) .

  • Animal Models: Transgenic mice show ventricular enlargement, hyperlocomotion, and synaptic deficits mimicking schizophrenia .

Heart Failure

  • Biomarker: Circulating NRG1β levels correlate with disease severity (median 6.2 ng/mL in NYHA class IV vs. 4.4 ng/mL in class I) and predict mortality (HR=1.58 for highest quartile) .

Hirschsprung’s Disease

  • GWAS Findings: NRG1 SNPs (rs16879552, rs7835688) are risk factors (OR=1.8–2.4) by disrupting enteric nervous system development .

Recombinant NRG1 in Clinical Trials

ConditionStudy PhaseDose (μg/kg/day)OutcomeSource
Chronic Heart FailureII0.3–1.2↑ Cardiac output by 30%; safe
Ischemic StrokePreclinical100 (rodents)↓ Infarct size, ↑ functional recovery

Drug Development Challenges

  • Blood-Brain Barrier Penetration: NRG1 crosses intact BBB in rodents, enabling CNS applications .

  • Safety: No hemorrhagic complications observed in stroke models .

Research Reagents

  • Recombinant NRG1-β2: 7.0 kDa peptide (61 aa), expressed in E. coli; used in neuroprotection assays .

  • Storage: Lyophilized powder stable at -20°C; reconstitute in PBS (≥100 μg/mL) .

Product Specs

Introduction
Neuregulin is a signaling protein that plays a crucial role in cardiac muscle cell function and structure. It acts by binding to ErbB2/ErbB4 receptor heterodimers, leading to their phosphorylation and promoting cardiomyocyte differentiation. Studies have shown that recombinant neuregulin can improve the organization of damaged myocardial cells and strengthen cell-to-cell connections. In animal models, neuregulin (NRG1) recombinant has demonstrated a protective effect against myocardial cell damage induced by ischemia, hypoxia, and viral infections.
Description
Recombinant Human Neuregulin-1 beta 2, produced in E. coli, is a single, non-glycosylated polypeptide chain with a molecular weight of 7.0kDa, comprising 61 amino acids. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
White, lyophilized (freeze-dried) powder, sterile-filtered.
Formulation
Lyophilized from a 0.2µm filtered solution in phosphate-buffered saline (PBS) at pH 7.4.
Solubility
Reconstitute the lyophilized NRG1 in sterile 18MΩ-cm H2O to a concentration of at least 100µg/ml. Further dilutions can be prepared in other aqueous solutions.
Stability
Lyophilized NRG1 remains stable at room temperature for up to 3 weeks. For long-term storage, store desiccated below -18°C. Reconstituted Heregulin should be stored at 4°C for 2-7 days. For future use, store below -18°C. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 96.0% as determined by: (a) Reverse-phase high-performance liquid chromatography (RP-HPLC) analysis. (b) Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis.
Biological Activity
The half-maximal effective concentration (ED₅₀), assessed through a cell proliferation assay using serum-free human MCF-7 cells, is less than 5ng/ml. This corresponds to a specific activity greater than 2.0 × 10⁵ U/mg.
Synonyms
Neuregulin-1, NRG1, GGF, HGL, HRGA, NDF, SMDF, HRG, ARIA, GGF2, HRG1.
Source
Escherichia Coli.
Amino Acid Sequence
SHLVKCAEKEKTFCVNGGECFMVKDLSNPSRYLCKCPNEFTGDRCQNYVMASFYKAEELYQ.

Q&A

What is the molecular structure and classification of NRG1 isoforms in humans?

NRG1 in humans comprises more than 30 isoforms generated through alternative splicing and promoter usage of a single gene. These isoforms are classified into six types (I-VI), each with a distinct N-terminal region . The NRG1 protein structure typically includes:

  • NH₂-terminal extracellular domains (ECD)

  • Transmembrane structural domains

  • Highly conserved COOH-terminal intracellular domains (ICD)

The enormous diversity of NRG1 isoforms results from alternative splicing of more than 30 exons combined with the use of different 5' promoters . To effectively differentiate between these isoforms in research settings, design type-specific primers targeting unique N-terminal sequences, using identical annealing temperatures (60°C) across all types and keeping PCR products under 200bp for optimal quantification efficiency .

How does the expression pattern of NRG1 isoforms vary in the human brain during development?

NRG1 isoform expression follows distinct developmental patterns in the human brain:

NRG1 TypeRelative AbundanceExpression Pattern During Development
Type III~73% of total NRG1Dominant isoform that increases proportionally with age
Type II~21% of total NRG1Second most abundant, decreases proportionally with age
Type I~5% of total NRG1Relatively stable expression throughout development
Type V~1% of total NRG1Higher expression in early development that decreases with age
Type IV<1% of total NRG1Low but stable expression
Type VI<0.1% of total NRG1Lowest abundance across all developmental stages

What mechanisms regulate NRG1 expression and activation in the nervous system?

NRG1 expression and activation are regulated through multiple mechanisms:

  • Transcriptional regulation: Different promoters control expression of specific NRG1 types, with genetic variations like rs6994992 affecting transcription efficiency .

  • Activity-dependent regulation: Neuronal activity modulates NRG1 expression in complex ways:

    • Depolarization with KCl increases expression of types I, II, and III NRG1

    • NMDA receptor activation selectively increases type III NRG1

    • GABA-A receptor activation decreases expression of types I and III NRG1

  • Post-translational processing: NRG1 requires proteolytic processing for activation:

    • ADAM17 (a disintegrin and metalloproteinase) is the primary protease that cleaves pro-NRG1 to release the active form

    • γ-secretase further processes the C-terminal fragment to generate NRG1-ICD, which can translocate to the nucleus

  • Regulatory feedback loops: NRG1 expression is also modulated by neurohormones (angiotensin II, phenylephrine, endothelin 1) and mechanical pressure, which can stimulate mRNA expression of NRG1 .

For accurate assessment of these regulatory mechanisms, use multiple complementary approaches including RT-qPCR, Western blotting with phospho-specific antibodies, and promoter activity assays.

How is NRG1 implicated in schizophrenia pathophysiology?

NRG1 has been consistently implicated in schizophrenia through several converging lines of evidence:

  • Genetic associations: Polymorphisms in the NRG1 gene, particularly rs6994992 in the type IV promoter region, are associated with schizophrenia risk. The T allele has been identified as a risk variant .

  • Expression alterations: The schizophrenia risk allele (T) at rs6994992 correlates with increased NRG1-IV expression in the hippocampus and dorsolateral prefrontal cortex . Experimental validation using luciferase promoter assays and site-directed mutagenesis confirms that rs6994992 is a functional cis-regulatory element affecting NRG1 expression .

  • Neurodevelopmental effects: rs6994992 is associated with altered cortical and subcortical neuroanatomical structure in human neonates, suggesting NRG1's role in early brain development contributes to schizophrenia vulnerability .

  • Functional impacts: The risk allele correlates with:

    • Impaired neurocognitive functioning

    • Abnormal sensory processing

    • Reduced neural connectivity

    • Abnormalities in brain morphology

To effectively study these associations, employ an integrated approach combining genotyping, quantitative expression analysis in postmortem tissue, functional assays of promoter activity, and structural/functional neuroimaging correlated with genotype.

What evidence supports NRG1 as a therapeutic target for Alzheimer's disease?

Compelling evidence supports NRG1 as a potential therapeutic target for Alzheimer's disease:

  • Cognitive enhancement: Overexpression of either type I or type III NRG1 via lentiviral vectors in the hippocampus of line 41 AD mouse models significantly improves performance in the Morris water maze task, demonstrating rescue of learning and memory deficits .

  • Neuroprotection: NRG1 overexpression significantly reverses the decreased expression of the neuronal marker MAP2 and synaptic markers PSD95 and synaptophysin in AD mice, indicating preservation of neuronal integrity and synaptic function .

  • Amyloid reduction: NRG1 treatment markedly reduces Aβ peptides and plaque load:

    • Type I NRG1 decreases amyloid deposition by 37%

    • Type III NRG1 decreases amyloid deposition by 66%

    • This occurs without altering APP levels, suggesting effects on processing or clearance rather than production

  • Enhanced Aβ clearance mechanism: Soluble ectodomains of both type I and type III NRG1 significantly increase expression of neprilysin (NEP), a major Aβ-degrading enzyme, in primary neuronal cultures. Consistent with this, NEP immunoreactivity increases in the hippocampus of NRG1-treated AD mice .

These findings suggest NRG1-based therapies could address multiple aspects of AD pathology simultaneously, making it a promising therapeutic target worthy of further clinical investigation.

How do different NRG1 isoforms contribute to distinct neurological functions and pathologies?

Different NRG1 isoforms contribute to distinct neurological functions through their structural diversity and spatiotemporal expression patterns:

  • Type III NRG1:

    • Primary function in myelination in the peripheral nervous system

    • Most abundant isoform in human brain (~73% of total NRG1)

    • Remains membrane-bound after initial cleavage, enabling sustained juxtacrine signaling

    • Particularly effective at reducing amyloid load (66% reduction) in AD models

  • Type I NRG1:

    • Important for acetylcholine receptor expression at neuromuscular junctions

    • Becomes fully soluble after cleavage, enabling paracrine signaling

    • Moderately effective at reducing amyloid load (37% reduction) in AD models

  • Type II NRG1:

    • Second most abundant in human brain (~21% of total)

    • Decreases proportionally with age

    • Roles in synaptogenesis and synaptic plasticity

  • Type IV NRG1:

    • Low abundance (<1%) but genetically linked to schizophrenia

    • The schizophrenia risk allele rs6994992 specifically affects type IV expression

  • Type V NRG1:

    • Higher expression in early development

    • Suggests particular importance during critical periods of brain development

The distinct functions of these isoforms explain why global NRG1 manipulation may yield contradictory results and why isoform-specific approaches are essential for both research and therapeutic development. To effectively study isoform-specific effects, use selective overexpression or knockdown approaches targeting specific variants rather than global NRG1 manipulation.

What are the most effective techniques for quantifying different NRG1 isoforms in human tissue samples?

Effective quantification of NRG1 isoforms in human tissue requires specialized techniques to distinguish between highly similar variants:

  • RT-qPCR with isoform-specific primers:

    • Design forward primers targeting unique N-terminal sequences of each type

    • Use reverse primers against common domains (Ig domain for types I, II, IV, V; EGF domain for types III and VI)

    • Ensure identical annealing temperatures (60°C) across all primer pairs

    • Keep PCR products <200bp for optimal efficiency

  • Digital droplet PCR (ddPCR):

    • Provides absolute quantification without standard curves

    • Superior sensitivity for low-abundance isoforms (types IV, VI)

    • Reduces variability in postmortem tissue with varying RNA quality

  • RNAscope in situ hybridization:

    • Enables visualization of isoform-specific expression in intact tissue

    • Allows cellular resolution of expression patterns

    • Can be combined with immunohistochemistry for cell type identification

  • Western blotting with domain-specific antibodies:

    • Use antibodies targeting unique epitopes in different NRG1 types

    • Include proper controls (recombinant proteins, knockout tissue)

    • Quantify using normalization to housekeeping proteins

For most accurate results, normalize expression to multiple reference genes validated for stability in your specific tissue type, and include appropriate positive controls for each isoform being measured.

How can researchers effectively manipulate NRG1 expression in experimental models of neurological disorders?

Researchers can manipulate NRG1 expression through several complementary approaches:

  • Viral vector-mediated overexpression:

    • Lentiviral vectors encoding specific NRG1 isoforms (type I, III) can be stereotaxically injected into target brain regions

    • This approach has successfully improved cognitive function and reduced neuropathology in AD mouse models

    • Allows spatial specificity and temporal control in adult animals

  • CRISPR/Cas9 genome editing:

    • Enables precise modification of specific NRG1 isoforms

    • Can introduce human disease-associated variants (e.g., rs6994992) into animal models

    • Useful for creating isoform-specific knockouts or knock-ins

  • Conditional genetic approaches:

    • Cre-loxP systems for temporal and cell type-specific manipulation

    • Tet-On/Off systems for reversible expression control

    • Allows separation of developmental vs. adult functions

  • Pharmacological interventions:

    • Recombinant NRG1 protein administration

    • Small molecules targeting NRG1 processing (ADAM17 modulators)

    • Compounds affecting downstream signaling pathways

  • Ex vivo models:

    • Organotypic slice cultures allow manipulation and analysis in a system retaining tissue architecture

    • Primary neuronal cultures enable mechanistic studies of NRG1 signaling

    • iPSC-derived neurons from patients with NRG1 variants can model human-specific effects

When designing NRG1 manipulation experiments, consider isoform specificity, developmental timing, cell-type specificity, and potential compensatory mechanisms that may obscure phenotypes.

What analytical approaches are recommended for interpreting NRG1 expression data in complex neuropsychiatric disorders?

Interpreting NRG1 expression data in neuropsychiatric disorders requires sophisticated analytical approaches:

  • Isoform-specific analysis:

    • Always analyze each NRG1 isoform separately rather than total NRG1

    • Calculate relative proportions of isoforms to detect composition shifts

    • Compare with developmental reference data to identify deviations from normal trajectories

  • Cell type deconvolution:

    • NRG1 is expressed in multiple cell types (neurons, glia, endothelial cells)

    • Use single-cell reference datasets to estimate cell-type contributions

    • Consider cell-type specific markers as covariates in analyses

  • Integration with genetic information:

    • Incorporate genotype data (particularly for rs6994992 and other risk variants)

    • Perform expression quantitative trait loci (eQTL) analyses

    • Consider allele-specific expression analyses for heterozygous individuals

  • Pathway analysis:

    • Examine downstream signaling components (ErbB receptors, PI3K/Akt, MAPK/ERK)

    • Include analysis of related neurotrophic factor pathways that may interact with NRG1

    • Use systems biology approaches to identify regulatory networks

  • Statistical considerations:

    • Account for multiple testing across isoforms

    • Include relevant covariates (age, sex, PMI, RNA quality, medication history)

    • Consider Bayesian approaches to incorporate prior biological knowledge

For example, when examining NRG1 in schizophrenia, stratify analyses by rs6994992 genotype, examine isoform-specific expression (particularly type IV), and correlate with ErbB receptor expression to identify potential compensatory changes in signaling pathways .

How do "forward" and "reverse" signaling mechanisms differ in NRG1 function?

NRG1 uniquely functions through both "forward" and "reverse" signaling mechanisms:

  • Forward signaling:

    • Canonical pathway where NRG1 acts as a ligand signaling to ErbB-expressing cells

    • NRG1's EGF-like domain binds to ErbB3 and ErbB4 receptors

    • This induces receptor dimerization (homodimers of ErbB4 or heterodimers of ErbB2/3, ErbB2/4, ErbB3/4)

    • Activates downstream pathways including PI3K/Akt, MAPK/ERK, and PLCγ

    • Mediates effects on differentiation, proliferation, survival, and synaptic plasticity

  • Reverse signaling:

    • Non-canonical pathway where NRG1 acts as a receptor rather than a ligand

    • After extracellular cleavage of pro-NRG1 by ADAM17, the C-terminal fragment undergoes further processing by γ-secretase

    • This generates an intracellular domain (NRG1-ICD) that translocates to the nucleus

    • NRG1-ICD functions as a transcriptional regulator, inhibiting apoptotic pathways

    • Particularly important for type III NRG1 due to its membrane tethering

The balance between these mechanisms varies by isoform type, developmental stage, and physiological context. To effectively study these distinct signaling modes, use domain-specific mutations that selectively disrupt forward versus reverse signaling, and employ subcellular fractionation to track nuclear translocation of NRG1-ICD.

What are the key downstream signaling pathways activated by different NRG1 isoforms?

Different NRG1 isoforms activate distinct patterns of downstream signaling:

Signaling PathwayPrimary FunctionsIsoform Specificity
PI3K/AktSurvival, metabolism, protein synthesisStrongly activated by types I and III
MAPK/ERKProliferation, differentiation, migrationDifferentially activated based on receptor dimerization patterns
PLCγ/PKCCalcium signaling, neuronal excitabilityMore strongly activated by type I than type III
JAK/STATGene transcription, inflammatory responsesActivated in specific cellular contexts
NF-κBImmune response, cell survivalActivated in response to stress conditions

The signaling specificity arises from:

  • Receptor selectivity: Different isoforms preferentially activate different ErbB receptor combinations:

    • Type III primarily signals through ErbB2/ErbB3 heterodimers

    • Type I effectively activates ErbB4 homodimers or ErbB2/ErbB4 heterodimers

  • Spatial localization:

    • Type III remains membrane-tethered after cleavage, creating signaling microdomains

    • Types I and II become fully soluble, enabling diffusion and more distant signaling

  • Temporal dynamics:

    • Different isoforms show distinct patterns of processing and degradation

    • This leads to varying durations of pathway activation

To effectively study these pathway differences, use phospho-specific antibodies for key signaling nodes, conduct time-course analyses, and employ pathway-specific inhibitors to dissect the contribution of each cascade to specific cellular outcomes.

How does proteolytic processing regulate NRG1 function in the nervous system?

Proteolytic processing is crucial for regulating NRG1 function through multiple mechanisms:

  • Activation of forward signaling:

    • NRG1 is initially synthesized as a pro-NRG1 transmembrane protein

    • ADAM17 (and potentially other metalloproteases) cleaves the extracellular domain

    • This releases the EGF-like domain-containing ectodomain that can activate ErbB receptors

    • For type III NRG1, the initial cleavage still leaves the N-terminus membrane-attached due to its cysteine-rich domain

  • Initiation of reverse signaling:

    • Following ADAM17 cleavage, the C-terminal fragment of NRG1 becomes a substrate for γ-secretase

    • γ-secretase cleaves within the transmembrane domain, releasing NRG1-ICD

    • NRG1-ICD translocates to the nucleus to regulate gene expression

  • Regulation by neuronal activity:

    • Depolarization and NMDA receptor activation enhance proteolytic processing

    • This provides a mechanism for activity-dependent regulation of NRG1 signaling

  • Disease relevance:

    • Altered proteolytic processing of NRG1 may contribute to pathological conditions

    • In Alzheimer's disease, NRG1 processing may compete with APP for γ-secretase, potentially affecting Aβ production

This sequential proteolytic processing creates multiple bioactive fragments from a single NRG1 molecule, enabling complex signaling outcomes. To effectively study these processes, use protease inhibitors (ADAM17 inhibitors, γ-secretase inhibitors), generate cleavage-resistant mutants, and employ antibodies specific to different processing products.

What are the therapeutic implications of NRG1's role in cardiac repair and regeneration?

NRG1 shows significant promise as a therapeutic target for cardiac conditions:

  • Cardiomyocyte proliferation stimulation:

    • NRG1 is a powerful cardiovascular proliferation stimulant

    • It activates multiple signaling pathways to stimulate cell cycle reentry in adult cardiomyocytes

    • This proliferative effect is crucial for myocardial regeneration

  • Differentiation regulation:

    • NRG1 promotes expression of genes related to working-type cardiomyocytes

    • It induces cardiomyocyte differentiation into cardiac conduction system cells

    • NRG1 promotes stem cell differentiation into working-type cardiomyocytes

  • Protective mechanisms:

    • The NRG1-ErbB pathway functions as a compensatory protective mechanism for cardiac injury

    • It serves as a critical mediator in the cross-talk between microvascular endothelium and myocytes

    • NRG1 may help regulate excessive inflammation in endangered myocardial regions following infarction

  • Clinical development status:

    • Recombinant NRG1 (specifically the EGF-like domain) has advanced to clinical trials for heart failure

    • Animal models show improvement in cardiac function after pathological injury

These findings suggest therapeutic potential for recombinant NRG1 administration in conditions including myocardial infarction, heart failure, and cardiomyopathy. The therapeutic approach requires careful consideration of dosing, timing relative to injury, and delivery method to maximize cardiac regenerative potential while minimizing potential off-target effects.

What challenges must be overcome in developing NRG1-based therapies for neurological disorders?

Developing NRG1-based therapies faces several significant challenges:

  • Isoform complexity and specificity:

    • With >30 isoforms having distinct functions, targeting specific isoforms is crucial

    • Different disorders may require isoform-specific approaches (e.g., type I vs. type III for AD)

    • Delivery of full-length proteins is challenging due to size and complexity

  • Blood-brain barrier penetration:

    • Large proteins like NRG1 have limited BBB penetration

    • Potential solutions include intranasal delivery, BBB-penetrating peptides, or viral vector-mediated expression

    • The EGF-like domain alone may have better penetration characteristics

  • Temporal and spatial targeting:

    • Developmental timing of NRG1 intervention is critical

    • Overactivation in inappropriate contexts could have adverse effects

    • Regional specificity may be necessary (e.g., hippocampal targeting for AD)

  • Potential oncogenic concerns:

    • NRG1-ErbB signaling is implicated in some cancers

    • Long-term safety requires careful evaluation

    • Tissue-restricted expression systems might mitigate this risk

  • Translational challenges:

    • Human NRG1 isoform composition differs from animal models

    • Functional validation in human cellular models is essential

    • Disease heterogeneity may require personalized approaches based on genetic background

Research approaches to address these challenges include development of small molecule mimetics with better BBB penetration, isoform-specific targeting strategies, controlled-release delivery systems, and validation in human iPSC-derived neuronal models before advancing to clinical trials.

What emerging research directions are most promising for advancing NRG1 understanding and applications?

Several promising research directions are emerging in the NRG1 field:

  • Single-cell resolution analysis:

    • Single-cell transcriptomics to map isoform expression in specific cell populations

    • Spatial transcriptomics to understand regional distribution of NRG1 isoforms

    • These approaches will clarify cell type-specific functions of different NRG1 variants

  • Human stem cell-based models:

    • iPSC-derived neurons from patients with NRG1 variants

    • Brain organoids to study NRG1 in human neurodevelopment

    • CRISPR-engineered isogenic lines to isolate effects of specific variants

  • Novel delivery technologies:

    • Nanoparticle-mediated delivery of NRG1 proteins or mRNA

    • AAV vectors optimized for specific cell type targeting

    • Optogenetic or chemogenetic control of NRG1 expression for precise temporal regulation

  • Multimodal therapeutic approaches:

    • Combining NRG1-based therapies with complementary approaches

    • For AD: pairing NRG1 with amyloid-targeting strategies

    • For schizophrenia: combining with glutamatergic or dopaminergic modulators

  • Biomarker development:

    • Circulating NRG1 fragments as potential diagnostic/prognostic markers

    • Imaging ligands to assess ErbB receptor engagement

    • Genetic signatures that predict response to NRG1-targeted therapies

  • Cross-disorder investigations:

    • Examining NRG1's role across neuropsychiatric conditions (schizophrenia, bipolar disorder, autism)

    • Investigating shared mechanisms between neurological and cardiac pathologies

    • Understanding how NRG1 dysfunction contributes to comorbidities

These emerging directions will require interdisciplinary collaboration between molecular neuroscientists, geneticists, clinicians, and bioengineers. The most promising advances will likely come from integrating insights across multiple levels—from molecular mechanisms to circuit function to clinical outcomes.

Product Science Overview

Introduction

Heregulin-B2, also known as Neuregulin-1 (NRG1), is a member of the neuregulin family of proteins. These proteins play a crucial role in cell signaling, particularly in the development and function of the nervous system and heart. Heregulin-B2 is a signaling protein that interacts with the ErbB family of receptors, specifically ErbB2 and ErbB4, to mediate various cellular processes .

Structure and Expression

Heregulin-B2 is a recombinant protein produced in Escherichia coli (E. coli). It is a single, non-glycosylated polypeptide chain containing 61 amino acids, with a molecular mass of approximately 7055 Daltons . The protein is typically lyophilized (freeze-dried) and can be reconstituted for use in various experimental applications.

Biological Functions

Heregulin-B2 plays a significant role in the development and maintenance of the nervous system and cardiac muscle cells. It induces the phosphorylation of ErbB2/ErbB4 receptor heterodimers, which leads to cardiomyocyte differentiation and improved heart structure and function . Additionally, Heregulin-B2 has been shown to stimulate the proliferation of human MCF-7 cells, a breast cancer cell line, under serum-free conditions .

Mechanism of Action

The interaction of Heregulin-B2 with ErbB receptors triggers a cascade of intracellular signaling pathways. These pathways involve the activation of various kinases and scaffold proteins, leading to changes in cell morphology, migration, and proliferation . In breast cancer cells, Heregulin-B2 signaling has been implicated in the development of an aggressive phenotype and resistance to anti-HER2 therapies such as trastuzumab and trastuzumab-emtansine .

Clinical Implications

Heregulin-B2’s role in cell signaling and proliferation makes it a potential target for therapeutic interventions in diseases such as cancer and heart disease. Research has shown that Heregulin-B2 can reduce the damage to myocardial cells caused by ischemia, hypoxia, and viral infections . In breast cancer, understanding the molecular mechanisms of Heregulin-B2 signaling can help develop more effective treatments to control cell motility and drug resistance .

Storage and Stability

Lyophilized Heregulin-B2 is stable at room temperature for up to three weeks but should be stored desiccated below -18°C for long-term storage. Upon reconstitution, it should be stored at 4°C for short-term use (2-7 days) and below -18°C for future use. It is important to avoid repeated freeze-thaw cycles to maintain the protein’s activity .

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.