NRG1 A Human

Neuregulin-1/Heregulin Alpha (EGF Domain) Human Recombinant
Shipped with Ice Packs
In Stock

Description

Nervous System Development and Synaptic Plasticity

  • Synaptic Regulation: NRG1 modulates excitatory-inhibitory balance in cortical neurons. Dysregulation is linked to schizophrenia, with type I isoforms showing elevated hippocampal expression in patients .

  • Neuroprotection: In ischemic stroke models, NRG1 reduces infarct size by 90%, enhances blood-brain barrier integrity, and suppresses microglial activation . Its therapeutic window extends up to 12 hours post-stroke .

  • Enteric Nervous System (ENS): NRG1 promotes survival of enteric neurons, with receptor (ErbB2/ErbB3) deficiencies causing colonic aganglionosis .

Cardiac Maintenance and Ischemia Response

  • Development: Essential for myocardial differentiation and sarcomeric organization .

  • Ischemia Protection: Recombinant NRG1 (rhNRG-1) improves cardiac function in heart failure by activating cMLCK, SERCA2, and FAK pathways .

  • Cell Adhesion: Restores intercellular contacts in isolated myocytes via ErbB2-FAK-p130(CAS) complex formation .

Anti-Inflammatory and Metabolic Effects

  • Stroke: Attenuates pro-inflammatory cytokines (IL-1β, MCP-1) by modulating NFκB pathways .

  • Hemorrhagic Stroke: Preserves tight junction proteins (Occludin, Claudin-5) and reduces BBB permeability .

Cardiovascular Therapies

ApplicationMechanismOutcome
Heart FailureEnhances cardiac myocyte survival, sarcomeric integrity, Ca²⁺ homeostasisImproves ejection fraction in phase II trials
Ischemia/ReperfusionReduces infarct expansion via ErbB4-mediated signaling90% reduction in rodent models

Neurological Disorders

  • Schizophrenia: Genetic variants in the 5′ NRG1 region correlate with elevated type I isoform expression in the hippocampus .

  • Stroke Recovery: NRG1 administration 3 days post-stroke improves functional outcomes in rodents .

Recombinant NRG1 Production

ParameterValue
SourceE. coli (non-glycosylated, 61 amino acids)
Purity>95% (SDS-PAGE, RP-HPLC)
BioactivityED₅₀ < 5 ng/ml (MCF-7 cell proliferation assay)

Key Research Insights and Challenges

  • Therapeutic Potential: rhNRG-1’s safety in heart failure trials and preclinical stroke efficacy position it as a candidate for CNS disorders .

  • Genetic Complexity: Noncoding SNPs in NRG1 (e.g., deCODE haplotype) influence isoform expression and schizophrenia risk .

  • Limitations: Isoform-specific roles remain poorly understood, necessitating targeted therapies .

Product Specs

Introduction
The Neuregulin/Heregulin family encompasses structurally related polypeptide growth factors originating from alternatively spliced genes (NRG1, NRG2, NRG3, and NRG4). Currently, over 14 soluble and transmembrane proteins are derived from the NRG1 gene. These isoforms undergo proteolytic processing of their extracellular domain, leading to the release of soluble growth factors. Among these isoforms are the heregulins (HRGs), glial growth factors (GGFs), and sensory and motor neuron-derived factor (SMDF). Notably, all these factors share Ig and EGF-like domains, enabling them to bind to ErbB3 and ErbB4 receptor tyrosine kinases. This binding triggers the heterodimerization of ErbB3 and ErbB4 with ErbB2, consequently activating intrinsic kinase activity and resulting in tyrosine phosphorylation. The NRG1 isoforms play a crucial role in inducing growth and differentiation in various cell types, including epithelial, neuronal, glial, and others.
Description
Recombinant Human Neuregulin-1/Heregulin Alpha (EGF Domain), produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 65 amino acids. It has a molecular mass of 7.4 kDa. The purification of NRG1-A is achieved through proprietary chromatographic techniques.
Physical Appearance
White, sterile-filtered powder obtained through lyophilization (freeze-drying).
Formulation
Lyophilized from a 0.2 µm filtered solution in 20 mM phosphate buffer (PB), with a pH of 6.0 and 150 mM sodium chloride (NaCl).
Solubility
For reconstitution, it is advised to dissolve the lyophilized NRG1-A in sterile 18 MΩ-cm H₂O at a concentration of at least 100 µg/ml. Further dilutions can be made using other aqueous solutions.
Stability
While lyophilized NRG1-A remains stable at room temperature for up to 3 weeks, it's recommended to store it desiccated below -18°C. After reconstitution, NRG1-A should be stored at 4°C for 2-7 days. For long-term storage, keep it below -18°C. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 97.0%, 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 ED₅₀, determined through dose-dependent stimulation of human MCF-7 cell proliferation, is less than 40 ng/ml. This corresponds to a specific activity greater than 2.5 × 10⁴ units/mg.
Synonyms
Neuregulin-1, Heregulin Alpha, NRG1-A, NRG1 A.
Source
Escherichia Coli.
Amino Acid Sequence
SHLVKCAEKE KTFCVNGGEC FMVKDLSNPS RYLCKCQPGF TGARCTENVP MKVQNQEKAE ELYQK.

Q&A

What is NRG1 and what are its main isoforms in humans?

NRG1 belongs to a family of structurally related glycoproteins encoded by four distinct but related genes (Nrg1, Nrg2, Nrg3, and Nrg4) . It functions as a key developmental growth factor that binds to and activates the ErbB class of receptor tyrosine kinases. Human NRG1 undergoes extensive alternative splicing and differential promoter usage, generating several distinct isoforms, classified as types I-VI. These isoforms differ in their temporal expression patterns and functional properties throughout neurodevelopment and aging. The structural variations determine specific signaling mechanisms, with each isoform contributing differently to processes including cell migration, synaptic formation and plasticity, and myelination .

How are NRG1 isoforms expressed during human brain development?

NRG1 isoform expression is temporally regulated in the human prefrontal cortex during development and throughout the lifespan. Research has revealed distinct expression patterns:

  • NRG1 type I: Most abundant during the first two weeks of the second trimester and declines with fetal gestational age, becoming stable at birth

  • NRG1 type III: Increases significantly with fetal gestational age and decreases dramatically after birth, reaching stable levels in early adolescence

  • NRG1 type IV-NV: Uniquely regulated, being expressed after gestational age 15 weeks through birth until 3 years of age, after which it becomes transcriptionally undetectable

These highly regulated expression patterns suggest specific roles for each isoform during critical developmental windows of human brain formation and maturation.

What cellular mechanisms regulate NRG1 function?

NRG1 undergoes complex post-translational processing that significantly impacts its biological activity. Research has identified that NRG1-IVNV represents a novel nuclear-enriched, truncated NRG1 protein that is resistant to proteolytic processing . For membrane-bound NRG1 forms, proteolytic cleavage releases the ectodomain, which can then activate ErbB receptors. This activation triggers downstream signaling cascades including the MAPK and PI3K/Akt pathways. The spatial presentation of NRG1 is also crucial, as demonstrated by studies showing that immobilized NRG1 can accelerate neural crest-like cell differentiation toward functional Schwann cells through sustained Erk1/2 activation and YAP/TAZ nuclear translocation . This signaling differs substantially from soluble NRG1, highlighting the importance of protein presentation in determining functional outcomes.

How is NRG1 linked to schizophrenia risk?

Polymorphisms in the NRG1 gene have been associated with schizophrenia risk across multiple populations. The original risk haplotype (HapICE) was first identified in the Icelandic population and includes SNP8NRG243177 (rs6994992) located in the 5' end of the NRG1 gene. This association has been subsequently validated in Scottish, English, Irish, and Northern Indian populations . The rs6994992 genotype is functionally significant, as homozygosity for the schizophrenia-risk allele (T) results in lower cortical NRG1-IVNV expression levels during development. This molecular mechanism represents a potential pathway of early developmental risk for schizophrenia at the NRG1 locus . While NRG1 polymorphisms have not been consistently identified in large genome-wide association studies of schizophrenia, meta-analyses of published data and three GWA schizophrenia datasets have provided additional support for association of the NRG1 HapICE region .

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

Multiple lines of evidence suggest NRG1 signaling may influence cognitive function and neuropathology in Alzheimer's disease (AD). Experimental studies demonstrate that overexpression of either type I or type III NRG1 in the hippocampus of AD mouse models improves deficits in the Morris water maze behavioral task and significantly ameliorates neuropathology . Both NRG1 types effectively reverse the decreased expression of neuronal marker MAP2 and synaptic markers PSD95 and synaptophysin observed in AD mice. Furthermore, levels of Aβ peptides and plaques were markedly reduced following NRG1 overexpression .

The mechanism underlying these beneficial effects appears to involve multiple pathways. Research shows that soluble ectodomains of both type I and type III NRG1 significantly increase expression of neprilysin (NEP), an Aβ-degrading enzyme, in primary neuronal cultures . This increase occurs partly through transcriptional activation of the NEP promoter. Additionally, NRG1 plays roles in synaptic differentiation and function, including increasing dendritic spine size, modulating long-term potentiation at CA1 synapses, and enhancing entorhinal-hippocampal synaptic transmission, all of which may contribute to cognitive improvement in AD models .

What methodological approaches should researchers use to study NRG1 function in neurological disorders?

When investigating NRG1 in neurological disorders, researchers should employ multifaceted approaches that reflect the complexity of NRG1 biology. For gene expression studies, quantitative PCR should be designed to distinguish between the various NRG1 isoforms, as each has distinct spatial and temporal expression patterns . In genotype-phenotype correlation studies, the rs6994992 polymorphism shows particular relevance to neuropsychiatric disorders and should be prioritized .

For functional studies in AD models, researchers should consider both behavioral assessments (such as the Morris water maze) and neuropathological analyses examining neuronal and synaptic markers (MAP2, PSD95, synaptophysin) alongside amyloid burden measurements . When manipulating NRG1 expression, both viral-mediated overexpression and recombinant protein administration approaches have demonstrated efficacy, with important distinctions between full-length proteins and soluble ectodomains . Cell-type specific analyses are critical, as ErbB4 (a key NRG1 receptor) is expressed primarily in interneurons, while ErbB2 and ErbB3 receptors predominate in glial cells, suggesting potential indirect regulation of neuronal function through glial intermediaries .

How does the spatial presentation of NRG1 affect its signaling outcomes?

The spatial presentation of NRG1 significantly alters its signaling dynamics and downstream biological effects. Research comparing soluble versus immobilized NRG1 (iNRG1) demonstrates that immobilization leads to sustained Erk1/2 activation and subsequent YAP/TAZ nuclear translocation in neural crest-like cells . This altered signaling profile accelerates their differentiation toward functional Schwann cells. The immobilized presentation resembles the natural presentation of NRG1 on neuronal axons, which interact with immature Schwann cells during development .

The mechanistic distinction between soluble and immobilized NRG1 appears to involve differences in receptor clustering, internalization kinetics, and the duration of downstream pathway activation. While soluble NRG1 typically induces rapid and transient signaling followed by receptor internalization and signal termination, immobilized NRG1 prevents receptor internalization, resulting in prolonged activation of downstream signaling cascades. This sustained signaling appears critical for the transcriptional changes necessary for cellular differentiation programs. Researchers investigating NRG1 signaling should therefore carefully consider the presentation method when designing experiments, as it may fundamentally alter biological outcomes .

What is the relationship between NRG1 expression and ErbB receptor activation in neural development?

During neural development, NRG1-ErbB signaling represents a complex, bidirectional relationship that regulates multiple aspects of cellular differentiation and function. NRG1 functions as a ligand for ErbB receptors, with different isoforms showing preferential binding to specific receptor combinations. NRG1 type III is particularly important in peripheral nervous system development, where it is expressed on axonal surfaces and signals to Schwann cells expressing ErbB2/ErbB3 receptors .

The spatial and temporal regulation of NRG1 expression patterns directly determines the activation profile of ErbB receptors in target cells. For instance, the developmental expression trajectory of NRG1 type III, which increases significantly during fetal gestation and decreases after birth, corresponds with critical periods of Schwann cell differentiation and myelination . This precisely regulated expression ensures appropriate timing and magnitude of ErbB receptor activation in target cells.

In mature neural systems, NRG1-ErbB signaling continues to regulate synaptic function through mechanisms involving modulation of neurotransmitter release and receptor trafficking. Disruptions to this signaling balance have been implicated in various neuropsychiatric disorders, including schizophrenia, where altered NRG1 expression due to genetic polymorphisms may lead to abnormal ErbB receptor activation during critical developmental windows .

How do NRG1 fusion proteins contribute to oncogenesis?

NRG1 gene fusions represent a significant oncogenic mechanism in multiple cancer types. These fusions typically result in excessive accumulation of the NRG1-fusion protein at the cell surface, causing persistent activation of ErbB receptor tyrosine kinases. This aberrant activation drives hyperactive signaling through the mTOR and MAPK pathways, promoting tumorigenesis .

Real-world clinical studies have identified NRG1 fusions across various cancer types, with the highest prevalence in non-small cell lung cancer (NSCLC, 39%), followed by pancreatic cancer (17%), cholangiocarcinoma (11%), colorectal cancer (11%), and several others at lower frequencies . These findings highlight the importance of screening for NRG1 fusions in clinical oncology practice, particularly in NSCLC patients.

The therapeutic targeting of NRG1 fusion-positive tumors represents an emerging precision medicine approach. Afatinib, an irreversible ErbB family blocker, has demonstrated clinical responses in patients with NRG1 fusion-positive tumors. Clinical management of these patients varies, with afatinib being administered in different treatment lines (frontline through fourth-line settings) . Ongoing research is needed to optimize detection methods for NRG1 fusions and to develop more specific therapeutic approaches targeting these oncogenic drivers.

What techniques are optimal for detecting different NRG1 isoforms?

Accurate detection and quantification of NRG1 isoforms require specialized molecular techniques due to their structural complexity and sequence similarities. For transcript analysis, quantitative PCR with isoform-specific primers targeting unique exon junctions has proven effective in developmental studies of human brain tissue . This approach allowed researchers to map the temporal expression patterns of NRG1 types I-IV during prenatal and postnatal neurodevelopment.

Advanced techniques for comprehensive isoform profiling include RNA sequencing with long-read technologies, which can characterize the full spectrum of alternative splicing events. Mass spectrometry-based proteomics can identify and quantify specific protein isoforms and their post-translational modifications. For functional studies, selective genetic manipulation (such as isoform-specific knockdown or overexpression) combined with phenotypic assays provides insights into isoform-specific functions .

How can NRG1 signaling be modulated for therapeutic applications?

Therapeutic modulation of NRG1 signaling can be achieved through several strategies, each with distinct advantages for specific conditions. For neurodegenerative disorders like Alzheimer's disease, direct administration of recombinant NRG1 or viral-mediated overexpression of NRG1 isoforms has shown promise in preclinical models. Both type I and type III NRG1 improved cognitive deficits and reduced neuropathology in AD mouse models when delivered via lentiviral vectors into the hippocampus .

The mechanism of administration significantly impacts therapeutic outcomes. Research demonstrates that soluble ectodomains of NRG1 increase expression of neprilysin (NEP), an Aβ-degrading enzyme, partly through transcriptional activation of the NEP promoter . This provides a potential therapeutic mechanism for reducing amyloid burden in AD. The choice between full-length NRG1 and soluble ectodomains should be guided by the specific therapeutic objective and target tissue.

For cancer therapy, the approach differs significantly. In NRG1 fusion-positive tumors, the goal is to inhibit aberrant NRG1 signaling. ErbB receptor tyrosine kinase inhibitors like afatinib have demonstrated clinical efficacy in patients with NRG1 fusion-positive tumors across multiple cancer types . Future therapeutic approaches may include specific targeting of the fusion proteins themselves or their downstream signaling pathways. Clinical data suggest that treatment timing is important, with varying efficacy observed when afatinib is administered in different treatment lines .

How might single-cell technologies advance our understanding of NRG1 biology?

Single-cell technologies offer unprecedented opportunities to unravel the complex biology of NRG1 across diverse cell types and developmental stages. Traditional bulk tissue analyses, while valuable, inevitably average expression across heterogeneous cell populations, potentially masking cell type-specific regulation patterns. Single-cell RNA sequencing (scRNA-seq) can reveal cell type-specific expression patterns of different NRG1 isoforms and their receptors across development and in disease states, providing a high-resolution map of NRG1 signaling networks .

This approach is particularly valuable given the known expression of NRG1 receptors in specific cell populations—ErbB4 in interneurons and ErbB2/ErbB3 in glial cells . Single-cell technologies could identify previously unrecognized cell populations that express or respond to specific NRG1 isoforms. Furthermore, spatial transcriptomics can preserve information about the physical location of cells expressing NRG1 or its receptors, allowing researchers to investigate signaling interactions within their native tissue architecture.

For functional studies, single-cell CRISPR screens could identify novel regulators of NRG1 expression or signaling, while single-cell proteomics and phosphoproteomics could map the downstream signaling cascades activated by different NRG1 isoforms with unprecedented resolution. These technologies have the potential to transform our understanding of how NRG1 signaling contributes to normal development and disease pathogenesis.

What is the potential of targeting NRG1 signaling in cancer therapy?

The emerging understanding of NRG1 fusions in cancer biology presents significant opportunities for targeted therapies. Real-world clinical data have confirmed that NRG1 fusions are detectable in meaningful numbers of patients with NSCLC and other cancer types . These fusions create oncogenic drivers by causing excess accumulation of the NRG1-fusion protein at the cell surface, leading to persistent activation of ErbB receptor tyrosine kinases and downstream oncogenic pathways .

Future therapeutic strategies may include more specific targeting of the fusion proteins themselves or development of degraders targeting NRG1 fusion proteins. Combination approaches targeting both NRG1 signaling and complementary pathways might overcome potential resistance mechanisms. As molecular profiling becomes more routine in clinical oncology, identification of patients with NRG1 fusions will likely increase, expanding the population who might benefit from targeted approaches. Larger retrospective and prospective studies assessing treatment outcomes in patients with NRG1 gene fusion-positive tumors are needed to optimize therapeutic strategies .

Product Science Overview

Introduction

Neuregulin-1 (NRG1), also known as Heregulin, is a member of the neuregulin family of structurally related glycoproteins. These proteins are known for their role in cell signaling and interaction with the ErbB family of receptor tyrosine kinases. NRG1 is particularly significant due to its involvement in various biological processes, including neural development, cardiac function, and cancer progression.

Structure and Isoforms

NRG1 is produced through alternative splicing of the NRG1 gene, resulting in multiple isoforms. These isoforms can be broadly categorized into two main types based on their EGF-like domain: NRG1-alpha and NRG1-beta. The EGF-like domain is crucial for binding to ErbB receptors and initiating downstream signaling pathways .

Biological Functions

NRG1 plays a pivotal role in the development and function of the nervous system and heart. It is involved in the differentiation and proliferation of Schwann cells, which are essential for myelination in the peripheral nervous system . Additionally, NRG1 is critical for the development of the neuromuscular junction and the regulation of neurotransmitter receptor expression .

In the heart, NRG1 is essential for cardiac development and function. It promotes the survival, growth, and differentiation of cardiac cells, and is involved in the response to cardiac injury .

Mechanism of Action

NRG1 exerts its effects by binding to the ErbB family of receptors, specifically ErbB3 and ErbB4. Upon binding, these receptors dimerize and undergo autophosphorylation, activating various intracellular signaling pathways such as the PI3K/Akt and MAPK pathways . These pathways are involved in cell survival, proliferation, and differentiation.

Recombinant NRG1

Recombinant human NRG1-alpha (EGF domain) is produced using recombinant DNA technology. It is typically expressed in bacterial or mammalian cell systems and purified for research and therapeutic applications. The recombinant protein retains the biological activity of the native protein and is used in various studies to understand its role in cell signaling and disease .

Applications in Research and Medicine

Recombinant NRG1 is widely used in research to study its role in neural and cardiac development, as well as its involvement in cancer. It is also being explored as a potential therapeutic agent for neurodegenerative diseases, heart failure, and certain types of cancer .

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.