RAMP1 Human

Receptor Activity-Modifying Protein 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to RAMP1 Human

Receptor Activity-Modifying Protein 1 (RAMP1) is a single-transmembrane protein critical for modulating the function of G-protein-coupled receptors (GPCRs). In humans, RAMP1 partners with calcitonin receptor-like receptor (CRLR) to form a heterodimer that functions as the receptor for calcitonin gene-related peptide (CGRP) . This complex is essential for CGRP-mediated signaling, which regulates vasodilation, neuronal transmission, and hematopoiesis . Structurally, RAMP1’s extracellular domain adopts a three-helix bundle stabilized by three disulfide bonds (Cys27–Cys82, Cys40–Cys72, Cys57–Cys104) .

Functional Interactions and Signaling Pathways

RAMP1’s primary role is to enable CRLR’s trafficking to the plasma membrane and dictate ligand specificity:

RAMP IsoformPartner ReceptorLigandBiological Function
RAMP1CRLRCGRPVasodilation, neuronal signaling, hematopoiesis
RAMP2CRLRAdrenomedullin (AM)Vascular relaxation, anti-inflammatory effects
RAMP3CRLRAM (lower affinity)Modulates AM signaling in specific tissues

CGRP binding to CRLR·RAMP1 activates adenylate cyclase, increasing intracellular cAMP and triggering downstream pathways (e.g., PKA activation, ERK phosphorylation) . In hematopoiesis, this signaling promotes stress-induced proliferation of hematopoietic stem/progenitor cells (HSPCs) .

Cluster Headache and Migraine

Genetic and expression studies link RAMP1 to cluster headache (CH):

  • Genetic association: The rs3754701 SNP in RAMP1 shows significant linkage with CH in Swedish cohorts .

  • Enhanced expression: RAMP1 mRNA levels are elevated in fibroblasts of CH patients compared to controls (p = 0.0073) .

  • Mouse models: Overexpression of human RAMP1 in mice increases sensitivity to CGRP-induced nausea and postural instability, mimicking vestibular migraine symptoms .

Hematopoietic Regulation

CGRP-CRLR/RAMP1 signaling supports hematopoiesis under stress:

  • CGRP stimulation: Enhances cAMP production in LSK (Lin⁻ Sca1⁺ c-Kit⁺) progenitor cells .

  • Proliferative stress: Ramp1 knockout mice show reduced repopulation capacity in bone marrow transplantation assays .

Mouse Models

ModelPhenotypeRelevance
Global RAMP1 KOImpaired CGRP receptor function; reduced hematopoietic repopulationValidates RAMP1’s role in CGRP signaling
Human RAMP1 overexpressionHyperresponsiveness to CGRP; vestibular dysfunction and nauseaMimics CGRP-mediated migraine pathophysiology

Antibody Validation

Commercial antibodies against RAMP1 (e.g., #844, #3158) show variable specificity:

  • Western blot: Detect multiple bands in transfected cells (e.g., HEK293) and rat brain .

  • Immunohistochemistry: Localize RAMP1 to cerebral vasculature and trigeminal ganglia .

Therapeutic Relevance

RAMP1 is a target for migraine therapies:

  • CGRP inhibitors: Monoclonal antibodies (e.g., erenumab) block CGRP binding to CRLR·RAMP1 .

  • Small-molecule antagonists: BIBN4096BS (gepants) inhibit CGRP binding via Trp74 in RAMP1 .

  • Limitations: Overexpression models show resistance to CGRP inhibitors, suggesting therapeutic challenges .

Product Specs

Introduction
RAMP, a member of the type I transmembrane protein family known as RAMP (receptor/calcitonin activity modifying proteins), possesses an extracellular N terminus and a cytoplasmic C terminus. Essential for CRLR transport to the plasma membrane, RAMPs interact with CRLR (calcitonin-receptor-like receptor), a protein with seven transmembrane domains. The functionality of the CRLR receptor, whether as an adrenomedullin receptor or a calcitonin-gene-related peptide (CGRP) receptor, is determined by the specific RAMP family members expressed. In the presence of RAMP1, CRLR acts as a CGRP receptor. RAMP1 plays a crucial role in the terminal glycosylation, maturation, and cell surface presentation of the CGRP receptor.
Description
Recombinant Human RAMP1, produced in E.coli, is a single, non-glycosylated polypeptide chain comprising 114 amino acids (27-117) with a molecular mass of 12.9 kDa. A 23 amino acid His-tag is fused to the N-terminus of RAMP1.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
The RAMP1 solution, at a concentration of 0.25mg/ml, contains 20mM Tris-HCl buffer at a pH of 8.0, 0.4M Urea, and 10% glycerol.
Stability
For optimal storage, keep at 4°C if the entire vial will be used within 2-4 weeks. For extended storage, freeze at -20°C. It is highly recommended to add a carrier protein such as 0.1% HSA or BSA for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity levels exceed 80% as determined by SDS-PAGE analysis.
Synonyms
Receptor (G Protein-Coupled) Activity Modifying Protein 1, Receptor (Calcitonin) Activity Modifying Protein 1, Calcitonin-Receptor-Like Receptor Activity-Modifying Protein 1, CRLR Activity-Modifying Protein 1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSCQEANYG ALLRELCLTQ FQVDMEAVGE TLWCDWGRTI RSYRELADCT WHMAEKLGCF WPNAEVDRFF LAVHGRYFRS CPISGRAVRD PPGS

Q&A

What is the basic structure and molecular weight of human RAMP1?

Human RAMP1 is a 14-18 kDa member of the RAMP family of proteins. The variability in molecular weight results from the presence or absence of intramolecular disulfide bonds that form in the endoplasmic reticulum and Golgi apparatus. Mature human RAMP1 is a 122 amino acid non-glycosylated type I transmembrane protein with a 91 amino acid extracellular domain (ECD, amino acids 27-117) and a ten amino acid cytoplasmic tail. Within the ECD, specific regions have specialized functions: residues 78-90 bind adrenomedullin (AM), while residues 91-103 bind calcitonin gene-related peptide (CGRP) .

Where is RAMP1 primarily expressed in human tissues?

RAMP1 shows a diverse expression pattern across human tissues. It is expressed in neurons, vascular endothelial cells, visceral and vascular smooth muscle cells, mammary epithelium, osteoblasts, and cardiomyocytes . In the human trigeminal ganglion, approximately 36% of neurons express RAMP1, as determined by immunohistochemical studies . RAMP1 has also been detected in human brain tissue, particularly in the hypothalamus, where it localizes to neuronal and glial processes .

What are the primary receptor complexes formed by human RAMP1?

RAMP1 forms heterodimeric receptor complexes through interactions with two primary partners:

  • With calcitonin receptor-like receptor (CLR): Forms an 84 kDa non-covalent receptor complex that serves as the canonical CGRP receptor

  • With calcitonin receptor (CT receptor): Forms a 76 kDa receptor complex that functions as an amylin receptor (AMY₁ receptor)

These protein-protein interactions are critical for determining receptor pharmacology and signaling specificity in different tissues.

What are the validated methods for detecting human RAMP1 in tissue samples?

Several validated detection methods have been established for human RAMP1:

  • Western Blotting: Human RAMP1 can be detected in tissue lysates (e.g., heart tissue) using specific antibodies. The protein appears as a band at approximately 17 kDa under reducing conditions using appropriate immunoblot buffers .

  • Immunohistochemistry: RAMP1 can be visualized in fixed, paraffin-embedded tissue sections using antigen retrieval techniques and specific antibodies. For optimal results, heat-induced epitope retrieval with basic antigen retrieval reagents is recommended, followed by staining with appropriate detection systems such as HRP-DAB .

  • Direct ELISA: Recombinant or native RAMP1 can be detected using direct ELISA methods, though some cross-reactivity (approximately 20%) with mouse RAMP1 may occur with certain antibodies .

When selecting detection methods, researchers should consider tissue type, fixation method, and potential cross-reactivity with other RAMP family members.

How can researchers effectively differentiate between endogenous RAMP1 and transfected human RAMP1 in experimental models?

Differentiating between endogenous and transfected human RAMP1 requires strategic approaches:

  • Species-specific antibodies: Use antibodies that specifically recognize human but not rodent RAMP1, allowing detection of only the transfected protein in transgenic models .

  • Epitope tagging: Engineer expression constructs with epitope tags (e.g., FLAG, HA, or His) that can be specifically detected without cross-reactivity with endogenous proteins.

  • Functional assays: Compare CGRP-induced cAMP responses between transgenic and non-transgenic samples. Tissues expressing human RAMP1 show distinct pharmacological profiles, including different maximal responses (Rmax) or EC₅₀ values, depending on the tissue type .

  • Antagonist sensitivity: Human RAMP1 demonstrates sensitivity to human-selective antagonists like telcagepant, which can be used to discriminate human from rodent RAMP1 receptors .

What are the optimal experimental conditions for studying RAMP1-CGRP receptor interactions?

When investigating RAMP1-CGRP receptor interactions, consider these methodological approaches:

  • Cell culture systems: Primary cultures of relevant tissues (vascular smooth muscle, trigeminal ganglia neurons) or clonal cell lines expressing defined combinations of CLR or CT receptors with RAMP1 provide controlled systems for interaction studies .

  • Signaling assays: cAMP production assays are well-established for measuring CGRP receptor functionality, as this is a primary signaling pathway downstream of receptor activation .

  • Binding studies: Radioligand binding assays using labeled CGRP can directly measure receptor-ligand interactions and determine binding parameters.

  • Immunoprecipitation: Co-immunoprecipitation of RAMP1 with CLR or CT receptor can confirm physical interactions between the proteins.

For functional studies, it's important to note that CGRP receptor properties may vary by tissue type. In transgenic mice expressing human RAMP1, smooth muscle cells show increased maximal response (Rmax) to CGRP with similar EC₅₀, while trigeminal ganglia show shifted EC₅₀ values with similar Rmax compared to non-transgenic controls .

How does human RAMP1 expression modify CGRP receptor pharmacology in transgenic models?

The expression of human RAMP1 in transgenic models produces tissue-specific modifications in CGRP receptor pharmacology:

  • Vascular smooth muscle: In cultured aortic smooth muscle from global hRAMP1 mice, CGRP-induced cAMP response shows a 10-fold greater maximal response (Rmax) compared to non-transgenic littermates, while maintaining similar EC₅₀ values .

  • Neuronal tissue: In cultured trigeminal ganglia neurons from global hRAMP1 mice, CGRP response exhibits a 40-fold leftward shift in EC₅₀ (increased potency) with similar maximal responses compared to non-transgenic controls .

  • Antagonist sensitivity: Tissues from hRAMP1 transgenic mice show sensitivity to the human-selective CGRP receptor antagonist telcagepant, which is ineffective in non-transgenic mouse tissues .

These differential effects suggest that the impact of RAMP1 on receptor function depends on the cellular context and possibly the relative expression levels of CLR versus CT receptor in different tissues.

What are the implications of RAMP1-expressing satellite glial cells in trigeminal ganglion for migraine research?

The discovery that satellite glial cells in the trigeminal ganglion express CGRP receptor components (CLR and RAMP1) but not CGRP itself has significant implications for migraine research:

  • Neuron-glia signaling: This expression pattern suggests a possible paracrine signaling mechanism where CGRP released from neurons may act on adjacent glial cells, establishing a neuron-glia communication network within the ganglion .

  • Amplification of signals: Activation of glial cells by neuronal CGRP could lead to release of inflammatory mediators or other signaling molecules that may amplify nociceptive signals.

  • Therapeutic targets: Satellite glial cells expressing CGRP receptors represent potential targets for CGRP receptor antagonists in migraine therapy, expanding our understanding of how these drugs might work beyond simply blocking neuronal receptors .

  • Migraine chronification: Persistent activation of glial CGRP receptors could contribute to sensitization processes and possibly play a role in the transition from episodic to chronic migraine.

This finding highlights the importance of considering both neuronal and glial components when investigating trigeminal pain mechanisms and developing targeted migraine therapies.

How do RAMP1-based CGRP and AMY₁ receptors differ in signaling properties and antagonist sensitivity?

RAMP1 forms functional receptors with both CLR (canonical CGRP receptor) and CT receptor (AMY₁ receptor), which exhibit distinct pharmacological profiles:

  • Signaling efficiency: In functional studies with transgenic hRAMP1 mice, the IC₅₀ values for telcagepant inhibition were closer to those observed for CT receptor/hRAMP1 than CLR/hRAMP1 in clonal cell lines, suggesting potential differences in signaling efficiency between these receptor types .

  • Antagonist sensitivity: While both receptor types can be blocked by CGRP receptor antagonists, their relative sensitivity differs. The partial effectiveness of telcagepant in transgenic hRAMP1 mice suggests either incomplete humanization or the presence of both canonical CGRP and AMY₁ receptors in the tissues studied .

  • Physiological relevance: Understanding the relative contributions of these different receptor complexes to CGRP signaling in various tissues is crucial for developing more selective therapeutic approaches.

These differences underscore the complexity of CGRP signaling and highlight the importance of characterizing both receptor subtypes when evaluating potential therapeutic compounds.

What controls should be included when studying RAMP1 expression and function?

Rigorous experimental design for RAMP1 research should include:

For expression studies:

  • Antibody validation: Confirm specificity using recombinant proteins and knockout/knockdown samples. Note that some antibodies may show cross-reactivity (e.g., ~20% cross-reactivity with mouse RAMP1 has been observed with certain human RAMP1 antibodies) .

  • Multiple detection methods: Complement immunohistochemistry with Western blotting or PCR to confirm expression patterns.

  • Negative controls: Include tissues known not to express RAMP1 and perform staining with non-immune serum or isotype control antibodies.

For functional studies:

  • Positive controls: Include cell lines with defined expression of RAMP1 and its partners (CLR or CT receptor).

  • Pharmacological validation: Use selective agonists and antagonists to confirm receptor identity.

  • Signal transduction controls: Include controls for cAMP assays such as forskolin stimulation to confirm cell viability and response capability.

  • Genetic controls: Compare responses in wild-type, knockout, or transgenic models as appropriate .

How should researchers address the challenge of RAMP1's heterodimeric nature in experimental design?

RAMP1's functional dependence on heterodimer formation presents unique experimental challenges:

  • Co-expression systems: Ensure co-expression of both RAMP1 and its partner (CLR or CT receptor) at appropriate ratios in recombinant systems.

  • Bicistronic expression constructs: Consider using bicistronic vectors to ensure stoichiometric expression of both components.

  • Sequential immunoprecipitation: Use sequential immunoprecipitation to isolate and characterize specific heterodimeric complexes.

  • Proximity ligation assays: Employ proximity ligation assays to visualize and quantify protein-protein interactions in situ.

  • Functional readouts: Complement biochemical assays with functional readouts (e.g., cAMP production, Ca²⁺ mobilization) to confirm the formation of functional receptor complexes .

When interpreting results, consider that changes in RAMP1 expression may affect multiple receptor systems (CGRP and amylin) simultaneously, potentially complicating the interpretation of phenotypic changes.

What are the methodological considerations for studying RAMP1 distribution in human trigeminal ganglia?

Studying RAMP1 in human trigeminal ganglia requires specific methodological considerations:

  • Tissue acquisition and processing: Human trigeminal ganglia obtained at autopsy must be carefully processed to preserve both morphology and antigenicity. Proper fixation and embedding procedures are critical .

  • Antigen retrieval: Heat-induced epitope retrieval using appropriate buffers is necessary for optimal immunodetection of RAMP1 in paraffin-embedded sections .

  • Quantification approach: For quantitative studies, establish clear criteria for counting positive cells. In previous studies, approximately 36% of human trigeminal ganglion neurons expressed RAMP1 .

  • Co-localization studies: To understand RAMP1's relationship with other proteins (CGRP, CLR), perform double or triple immunofluorescence labeling with appropriate controls for each antibody .

  • Neuronal vs. glial expression: Use specific markers to differentiate between neuronal and glial expression of RAMP1, as both cell types have been shown to express this protein in trigeminal ganglia .

  • Species comparison: Include parallel studies in experimental animals (e.g., rat) to validate findings and enable translation between human and preclinical models .

How should researchers interpret discrepancies between RAMP1 mRNA and protein expression?

When facing discrepancies between RAMP1 mRNA and protein levels:

  • Post-transcriptional regulation: Consider that RAMP1 may be subject to microRNA regulation or other post-transcriptional mechanisms that affect translation efficiency.

  • Protein stability: RAMP1 protein stability may vary depending on its association with receptor partners, as proper trafficking and expression often require heterodimer formation.

  • Detection sensitivity: The sensitivity limits of protein detection methods (Western blot, immunohistochemistry) versus mRNA detection (qPCR, RNA-seq) may contribute to apparent discrepancies.

  • Temporal dynamics: mRNA and protein expression may follow different temporal patterns, with mRNA changes preceding protein changes.

  • Cellular heterogeneity: In mixed cell populations, bulk measurements may mask cell-type-specific expression patterns. Consider single-cell approaches when feasible.

To address these challenges, researchers should employ multiple complementary techniques and include appropriate time-course experiments when investigating RAMP1 expression dynamics.

What factors affect the pharmacological profile of RAMP1-containing receptors in different experimental systems?

Several factors can influence the pharmacological properties of RAMP1-containing receptors:

  • Receptor partner ratio: The relative expression levels of RAMP1 versus CLR or CT receptor can affect the proportion of canonical CGRP versus AMY₁ receptors formed .

  • Species differences: Human and rodent RAMP1 confer different pharmacological properties, particularly regarding antagonist sensitivity. Human RAMP1 receptors are sensitive to telcagepant, while rodent receptors show significantly lower sensitivity .

  • Tissue-specific factors: As observed in transgenic hRAMP1 mice, the same genetic modification produces different functional effects in vascular smooth muscle (increased Rmax) versus trigeminal neurons (decreased EC₅₀) .

  • Receptor reserve: Differences in receptor expression levels between systems may create variable receptor reserve, affecting apparent potency measurements.

  • Signal transduction machinery: Variation in the expression of downstream signaling components between cell types or experimental systems may influence the measured response.

Researchers should carefully characterize their experimental system and avoid direct comparisons of absolute pharmacological parameters between different model systems without appropriate calibration.

How can researchers reconcile the conflicting data on RAMP1 and CGRP co-localization in trigeminal neurons?

Addressing discrepancies in RAMP1 and CGRP co-localization findings:

  • Technical considerations: Different fixation methods, antibody specificities, and detection sensitivities can significantly impact co-localization results.

  • Species differences: Patterns observed in human tissues may differ from those in rodent models. In human trigeminal ganglia, co-localization of CGRP and RAMP1 is rarely found, with these markers predominantly expressed in separate neuronal populations .

  • Regional variability: Co-localization patterns may vary across different regions of the trigeminal ganglion or in different branches of the trigeminal nerve.

  • Functional interpretation: Limited co-localization suggests paracrine rather than autocrine signaling, with CGRP released from one neuronal population acting on receptors on nearby neurons or glial cells .

  • Developmental changes: Expression patterns may change during development or in response to pathological conditions such as inflammation or injury.

To resolve these discrepancies, researchers should:

  • Clearly specify the exact regions examined

  • Use multiple antibodies targeting different epitopes

  • Employ highly sensitive detection methods

  • Consider quantitative approaches to co-localization analysis

  • Compare findings across species and experimental conditions

What are the most promising research applications for RAMP1 studies in human disease?

RAMP1 research holds significant promise for several human disease applications:

  • Migraine therapies: Understanding RAMP1's role in trigeminal pain pathways has already led to the development of CGRP receptor antagonists. Further characterization of neuronal versus glial RAMP1 expression may refine these approaches .

  • Cardiovascular disorders: Given RAMP1's expression in vascular smooth muscle and cardiomyocytes, targeting RAMP1-containing receptors may offer therapeutic avenues for hypertension, heart failure, or other cardiovascular conditions .

  • Inflammatory disorders: CGRP's involvement in neurogenic inflammation suggests potential applications for RAMP1-targeted approaches in inflammatory diseases.

  • Metabolic conditions: RAMP1's participation in forming amylin receptors (with CT receptor) places it at the intersection of metabolic regulation and potential approaches to diabetes and obesity.

Future research should focus on developing tools to selectively target specific RAMP1-containing receptor complexes to achieve greater therapeutic specificity.

What methodological advances would most benefit RAMP1 research?

Several methodological advances would significantly advance RAMP1 research:

  • Selective antibodies and ligands: Development of tools that can distinguish between canonical CGRP receptors (CLR/RAMP1) and AMY₁ receptors (CT receptor/RAMP1) would enable more precise characterization of these signaling systems.

  • Advanced imaging techniques: Super-resolution microscopy and in vivo imaging approaches would enhance our understanding of RAMP1 trafficking, localization, and dynamics at the cellular and tissue levels.

  • Single-cell analysis: Application of single-cell transcriptomics and proteomics to RAMP1-expressing tissues would provide insight into cellular heterogeneity and potentially identify new RAMP1-expressing cell populations.

  • Conditional genetic models: Development of cell-type-specific and inducible RAMP1 transgenic or knockout models would enable more precise dissection of RAMP1's role in specific tissues and physiological processes.

  • Structural biology approaches: Detailed structural characterization of RAMP1 in complex with its partners would facilitate structure-based drug design targeting specific receptor interfaces.

These methodological advances would address current limitations in RAMP1 research and accelerate translation of basic findings into clinical applications.

Product Science Overview

Introduction

Receptor Activity-Modifying Protein 1 (RAMP1) is a member of the RAMP family of single-transmembrane-domain proteins. These proteins play a crucial role in the functional expression of certain receptors, particularly the calcitonin gene-related peptide (CGRP) receptors. RAMP1 is essential for the transport and proper functioning of these receptors on the cell surface.

Structure and Function

RAMP1 is a type I transmembrane protein with an extracellular N-terminus and a cytoplasmic C-terminus. It is involved in the terminal glycosylation, maturation, and presentation of the CGRP receptor to the cell surface. The CGRP receptor, which has seven transmembrane domains, can function as either a CGRP receptor or an adrenomedullin receptor, depending on which members of the RAMP family are expressed .

Role in Receptor Expression

RAMP1 is required to transport the calcitonin-receptor-like receptor (CRLR) to the plasma membrane. In the presence of RAMP1, CRLR functions as a CGRP receptor. This interaction is crucial for the receptor’s ability to respond to its ligands and mediate downstream signaling pathways .

Interaction with Other Receptors

RAMP1 is not limited to interacting with CRLR. It has been shown to interact with other Class II G protein-coupled receptors (GPCRs), including the VPAC1 vasoactive intestinal polypeptide/pituitary adenylate cyclase-activating peptide receptor, glucagon receptor, and parathyroid hormone receptors. These interactions suggest that RAMP1 plays a broader role in modulating cell signaling through various GPCRs .

Recombinant Human RAMP1

Recombinant human RAMP1 is produced using various expression systems, including yeast, E. coli, baculovirus, and mammalian cells. This recombinant protein is used in research to study the functional aspects of RAMP1 and its interactions with different receptors. It is also utilized in drug discovery and development to identify potential therapeutic targets for diseases associated with CGRP and other related receptors .

Clinical Significance

RAMP1 has been implicated in various physiological and pathological processes. It is highly expressed in the cardiovascular system, suggesting a role in vascular function and regulation. Studies have shown that RAMP1 is a key regulator of angiogenesis in endothelial cells, indicating its potential involvement in vascular diseases . Additionally, RAMP1 has been associated with certain cancers, such as pineal dysgerminoma and malignant pineal area germ cell neoplasm .

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.