METRNL Human

Meteorin-Like Protein Human Recombinant
Shipped with Ice Packs
In Stock

Description

Glucose Homeostasis

METRNL enhances insulin sensitivity through:

  • AMPKα2/HDAC5/GLUT4 pathway: Increases glucose transporter expression in adipocytes/muscle

  • PPARδ activation: Upregulates fatty acid oxidation genes (CPT1, ACO, FABP3)

  • Inflammation suppression: Reduces TNFα (40%) and MCP-1 (35%) in palmitate-treated cells

In HFD-fed mice, METRNL administration:

  • Improved glucose tolerance by 28% (p<0.01)

  • Reduced fasting glucose by 18% (p<0.05)

Lipid Metabolism

EffectMechanismExperimental Outcome
Adipose browningEosinophil-mediated IL-4 production2-fold UCP1 increase in white fat
Hepatic lipid clearanceHDL-C elevation (15%), TG reduction (22%) Improved NASH markers in models
Fatty acid oxidationAMPK/PPARδ-dependent CPT1 induction40% increase in β-oxidation rates

Disease Associations

ConditionMETRNL Level ChangeClinical Implications
Type 2 Diabetes↓ 35% vs controls Inverse correlation with HbA1c (r=-0.42)
Coronary Artery Disease↓ 28% in patients Linked to endothelial dysfunction
Obesity↓ 40-50% in BMI >30 Predicts insulin resistance (OR=1.8)
Neonatal BAT Activity↑ 60% vs adults Correlates with thermogenesis (r=0.51)

In a longitudinal infant cohort, METRNL levels decreased progressively from birth (12.4±1.1 ng/mL) to 12 months (5.3±0.8 ng/mL), paralleling BAT activity reduction .

Therapeutic Potential

Preclinical Evidence:

  • Recombinant METRNL (2 μg/day) reduced HFD-induced weight gain by 15% in mice

  • Adenoviral overexpression improved glucose tolerance by 32% in diabetic models

  • Neuroprotection: Enhanced neurite outgrowth by 45% in vitro

Challenges:

  • No established dosing regimens for human trials

  • Unknown receptor identity limits targeted drug development

Research Frontiers

  1. Cardiometabolic Protection: METRNL attenuates endothelial apoptosis (50% reduction) via E2F1-TXNIP axis modulation

  2. Immunomodulation: Induces anti-inflammatory IL-10 (3-fold) and TGF-β (2.5-fold) in macrophages

  3. Neurological Effects: Correlates with BDNF levels (r=0.38) in aging models

Product Specs

Introduction

Known also as Meteorin-like protein, METRNL is a hormone that is secreted in response to physical activity (by skeletal muscle) or cold exposure (by adipose tissue). This hormone has been shown to increase energy expenditure, promote the browning of white fat, and improve glucose tolerance. METRNL achieves these effects by stimulating the migration of various immune cells into adipose tissue, where they initiate processes that enhance thermogenesis.

Description

Recombinant Human METRNL, produced in E. coli, is a single, non-glycosylated polypeptide chain. This chain comprises 276 amino acids (a.a 46-311), including a 10 a.a N-terminal His tag, resulting in a calculated molecular mass of 31.2kDa.

Physical Appearance
White powder, lyophilized after filtration.
Formulation

The METRNL protein undergoes filtration (0.4µm) and lyophilization from a solution of 0.5mg/ml in 50mM acetate buffer (pH 4) containing 5% (w/v) trehalose and 1mM DTT.

Solubility

To prepare a working stock solution of approximately 0.5mg/ml, it is recommended to reconstitute the lyophilized pellet in 0.1M acetate buffer (pH 4) and allow for complete dissolution.

Stability
Lyophilized protein should be stored at -20°C. After reconstitution, aliquot the protein to prevent repeated freeze-thaw cycles. Reconstituted METRNL remains stable at 4°C for a limited period and shows no significant change after two weeks at this temperature.
Purity

Purity is determined to be greater than 95.0% by SDS-PAGE analysis.

Synonyms

Meteorin-like protein, Subfatin, METRNL.

Source
Escherichia Coli.
Amino Acid Sequence

MKHHHHHHAS QYSSDRCSWK GSGLTHEAHR KEVEQVYLRC AAGAVEWMYP TGALIVNLRP NTFSPARHLT VCIRSFTDSS GANIYLEKTG ELRLLVPDGD GRPGRVQCFG LEQGGLFVEA TPQQDIGRRT TGFQYELVRR HRASDLHELS APCRPCSDTE VLLAVCTSDF AVRGSIQQVT HEPERQDSAI HLRVSRLYRQ KSRVFEPVPE GDGHWQGRVR TLLECGVRPG HGDFLFTGHM HFGEARLGCA PRFKDFQRMY RDAQERGLNP CEVGTD.

Q&A

What is the most reliable method to measure METRNL levels in human samples?

For accurate quantification of human METRNL, enzyme-linked immunosorbent assay (ELISA) is currently the gold standard methodology. When measuring METRNL in human samples, researchers should consider:

  • Sample type specificity: While ELISA kits are optimized for serum, plasma, and cell culture supernatants, expression levels vary significantly between these mediums .

  • Pre-analytical considerations: Fasting versus fed states can significantly influence circulating METRNL levels, as demonstrated in both pre-training and post-training exercise studies .

  • Cross-validation: Due to variability between commercial assays, researchers should validate findings using complementary approaches such as Western blotting or mass spectrometry when possible.

  • Reference ranges: Establishing appropriate normative values for your specific population is essential, as baseline METRNL levels have shown variations across different metabolic states .

Methodologically, researchers should implement rigorous control procedures including technical replicates, standard curves verification, and inclusion of both positive and negative controls to ensure reliable quantification.

How does METRNL expression differ across human tissues?

METRNL demonstrates distinct tissue-specific expression patterns that should inform experimental design and interpretation:

  • High expression: Adipose tissue, skin, and mucosal barrier tissues show predominant METRNL expression .

  • Moderate expression: Skeletal muscle exhibits notable expression, particularly following exercise stimulation .

  • Low expression: Central nervous system (CNS) tissues generally show lower basal expression compared to peripheral tissues, although METRNL can cross the blood-brain barrier .

  • Developmental differences: During early development, METRNL is exclusively expressed in dorsal root ganglions and inner ear, but this pattern changes in adult tissues .

When investigating tissue-specific functions, researchers should employ multiple methodologies including immunohistochemistry, in situ hybridization, and quantitative PCR to confirm expression patterns. The choice of experimental controls should account for these tissue-specific differences to avoid misinterpretation of results.

What experimental models are most appropriate for studying METRNL function?

The selection of experimental models for METRNL research should be guided by the specific aspect under investigation:

  • Knockout models: METRNL−/− mice exhibit B-cell immune system defects, including lower serum IgG levels (particularly IgG2b and IgG3), making them valuable for immunological studies .

  • Transgenic overexpression: While specific METRNL transgenic models are less common, PGC-1α4 transgenic mice show increased METRNL expression and can serve as indirect models .

  • Cell culture systems:

    • Adipocyte models are appropriate for metabolic studies

    • Macrophage cultures for immunological investigations

    • Endothelial cells for angiogenesis research

    • Myocyte cultures for exercise-related mechanisms

For exercise studies, electrical stimulation of rodent limbs has successfully simulated resistance exercise effects on METRNL expression . Researchers should carefully select models based on their specific research questions while acknowledging translational limitations between animal models and human physiology.

What methodological approaches should be used to investigate METRNL's role in metabolism?

Investigating METRNL's metabolic functions requires multi-faceted methodological approaches:

  • Glucose metabolism assessment: Employ glucose tolerance tests (GTT), insulin tolerance tests (ITT), and hyperinsulinemic-euglycemic clamp techniques to evaluate METRNL's effects on glucose homeostasis .

  • Thermogenesis analysis: Use indirect calorimetry, infrared thermography, and molecular markers (UCP1, PGC-1α) to assess METRNL's impact on energy expenditure .

  • Adipose tissue browning: Implement histological staining, gene expression analysis of browning markers, and mitochondrial respiration assays to examine METRNL's role in adipose tissue remodeling .

  • Lipid metabolism: Apply lipidomics approaches, fatty acid oxidation assays, and triglyceride accumulation measurements to comprehensively evaluate METRNL's effects on lipid handling .

When designing metabolic studies, researchers should carefully control for confounding factors including feeding status, circadian rhythms, sex differences, and environmental temperature, all of which can significantly influence METRNL-mediated metabolic processes.

How can researchers address contradictory findings regarding METRNL levels in obesity and diabetes?

The contradictory findings regarding METRNL levels in metabolic disorders require methodological rigor to resolve:

  • Stratification approach: Implement precise patient stratification based on:

    • BMI categories (normal, overweight, obese)

    • Diabetes status (normoglycemic, prediabetic, T2DM)

    • Insulin resistance measures (HOMA-IR values)

    • Duration of metabolic disease

    • Medication status and type

  • Confounding variable control: Researchers must methodologically address:

    • Physical activity levels (quantified objectively)

    • Dietary patterns (through validated food frequency questionnaires)

    • Age and sex influences on METRNL expression

    • Comorbidities that might influence METRNL regulation

  • Longitudinal designs: Employ within-subject repeated measures to track changes in METRNL over disease progression rather than relying solely on cross-sectional comparisons .

  • Multi-tissue assessment: Evaluate both circulating and tissue-specific METRNL levels, as discordant patterns have been observed between blood levels and tissue expression, particularly in cardiac conditions .

This methodological approach may help resolve the apparent contradictions where some studies report elevated METRNL in obese T2DM patients compared to non-obese T2DM individuals, while others suggest opposing patterns .

What experimental approaches can differentiate between local and systemic effects of METRNL?

Distinguishing between local tissue-specific and systemic effects of METRNL requires sophisticated experimental designs:

  • Tissue-specific genetic manipulation:

    • Implement Cre-lox recombination systems for adipose-specific or muscle-specific METRNL knockdown/overexpression

    • Compare with whole-body knockouts to identify tissue-autonomous functions

  • Parabiosis experiments: Surgical joining of circulation between METRNL-deficient and wild-type animals can help differentiate between endocrine and local paracrine effects

  • Ex vivo tissue culture systems: Culture specific tissues with recombinant METRNL or conditioned media from METRNL-expressing cells to assess direct tissue responses

  • In vivo tracking: Label recombinant METRNL with traceable markers to monitor tissue distribution and blood-brain barrier crossing

  • Secondary signal isolation: Implement selective receptor blockade in target tissues to identify which METRNL effects require intermediate signaling molecules versus direct action

This methodological framework can help resolve whether METRNL's effects on inflammation, adipose browning, and glucose metabolism are mediated through direct action or intermediate signals.

How should researchers design experiments to elucidate METRNL's role in exercise adaptations?

Exercise-induced METRNL regulation requires specific methodological considerations:

  • Exercise protocol specificity:

    • Different exercise modalities (aerobic, HIIT, resistance) should be compared within the same study design

    • Exercise intensity, duration, and frequency must be precisely controlled and reported

    • Acute versus chronic exercise effects should be differentiated through appropriate timing of measurements

  • Temporal sampling design:

    • Implement multiple timepoint sampling (pre-exercise, immediately post-exercise, recovery phases)

    • Consider both acute response and training adaptation timelines

    • Account for diurnal variations in METRNL expression

  • Mechanistic pathway validation:

    • Pharmacological inhibition of proposed pathways (PGC-1α, AMPK)

    • RNAi or CRISPR approaches to validate key mediators

    • Concurrent measurement of proposed downstream effectors

  • Population considerations:

    • Training status of subjects significantly impacts METRNL response

    • Metabolic health status alters exercise-induced METRNL dynamics

    • Age and sex should be controlled or specifically investigated as variables

The methodological framework should incorporate these elements while controlling for nutritional status, as both fed and fasting states influence METRNL secretion patterns following exercise interventions .

What techniques are optimal for investigating METRNL-KIT signaling in cardiac angiogenesis?

The investigation of METRNL-KIT signaling in cardiac angiogenesis requires specialized approaches:

  • Receptor binding assays:

    • Surface plasmon resonance to quantify METRNL-KIT binding kinetics

    • Co-immunoprecipitation to verify protein-protein interactions in cardiac tissue

    • Proximity ligation assays to visualize METRNL-KIT interaction in situ

  • Signaling cascade analysis:

    • Phosphoproteomic profiling to map KIT downstream effectors upon METRNL stimulation

    • Selective inhibition of pathway components to establish hierarchy

    • Live-cell imaging with fluorescent reporters to track signaling dynamics

  • Functional angiogenesis assessment:

    • Endothelial tube formation assays with KIT inhibition/knockdown

    • Ex vivo aortic ring sprouting assays under METRNL stimulation

    • In vivo Matrigel plug assays with METRNL and KIT modulation

  • Cardiac-specific modeling:

    • Myocardial infarction models with local METRNL delivery via infusion pump

    • Endothelial cell-specific KIT knockout to isolate angiogenic mechanism

    • Lineage tracing of KIT-expressing endothelial cells responding to METRNL

These methodological approaches can help delineate the specific role of METRNL as a ligand for KIT receptor tyrosine kinase in promoting angiogenesis after cardiac infarction, a process critical for limiting cardiac damage .

What are the critical methodological challenges in measuring METRNL in clinical studies?

Clinical studies of METRNL face several methodological challenges that must be addressed:

  • Pre-analytical variables:

    • Time of day (circadian variation)

    • Fasting/fed status significantly impacts measurements

    • Exercise history (acute exercise can elevate METRNL for hours)

    • Sample processing time and temperature stability

  • Population heterogeneity:

    • Metabolic health status dramatically affects baseline levels

    • Medication use (particularly those affecting metabolism)

    • Comorbidities with inflammatory components

    • Age and sex differences in METRNL regulation

  • Statistical approach:

    • Sample size calculation should account for high inter-individual variability

    • Appropriate statistical methods for non-normally distributed data

    • Multivariate analysis to account for confounding factors

    • Correction for multiple testing when exploring multiple correlations

  • Standardization issues:

    • Different commercial ELISA kits may yield varying absolute values

    • Lack of standardized reference materials

    • Inconsistent reporting of units and normalization approaches

Researchers should implement robust quality control procedures, detailed reporting of methodological factors, and consideration of these variables in study design and analysis to improve reproducibility in clinical METRNL research.

How can researchers investigate METRNL's role in inflammatory processes?

Investigating METRNL's immunomodulatory functions requires specific methodological approaches:

  • Macrophage polarization assessment:

    • Flow cytometry to quantify M1/M2 polarization markers

    • Gene expression profiling of polarization signatures

    • Functional assays (phagocytosis, cytokine production)

    • Co-culture systems with METRNL stimulation or neutralization

  • In vivo inflammation models:

    • Allergic asthma models (shown to respond to METRNL intervention)

    • Sterile inflammation resolution assays

    • Autoimmune disease models with METRNL administration

    • Tissue-specific conditional knockout approaches

  • Pathway analysis:

    • NF-κB activation measurement through reporter systems

    • Inflammasome activity assessment (NLRP3)

    • Chemokine production profiles (CCL3, CCL4)

    • Quantification of Type 2 inflammatory responses

  • Translational approaches:

    • Ex vivo stimulation of human immune cells with recombinant METRNL

    • Analysis of immune cell populations in METRNL-high versus METRNL-low patients

    • Correlation of METRNL levels with inflammatory biomarkers in clinical samples

These methodological approaches can help clarify METRNL's complex role in immunological processes, as evidence suggests both pro-inflammatory and anti-inflammatory functions depending on context and tissue .

What experimental design is recommended for studying METRNL in neurodevelopmental contexts?

Given METRNL's emerging role in neurodevelopment, particularly in inner ear development and cognitive function, specialized experimental designs are warranted:

  • Developmental timeline analysis:

    • Stage-specific expression profiling during embryonic development

    • Conditional knockout models with temporal control

    • In situ hybridization to map spatiotemporal expression patterns in neural tissues

  • Functional assessment:

    • Auditory brainstem response (ABR) testing for inner ear function

    • Neurite outgrowth assays with primary neuronal cultures

    • Migration assays to assess METRNL's effect on neural precursor movement

    • Cognitive testing in METRNL-deficient models, particularly in aging paradigms

  • Mechanistic investigation:

    • Analysis of BDNF, TrkB, and GFAP levels in hippocampus following METRNL manipulation

    • Assessment of PAX2/5/8 pathway activity, as METRNL appears to be a downstream target

    • Blood-brain barrier crossing studies using labeled METRNL

  • Clinical correlation:

    • Analysis of METRNL variants/levels in patients with inner ear disorders

    • Investigation of METRNL in Mild Ring 17 Syndrome, given its chromosomal location

    • Cerebrospinal fluid measurement correlated with cognitive performance

These methodological considerations can guide researchers investigating METRNL's neurological roles, which appear distinct from its metabolic functions and may have relevance for aging-related cognitive dysfunction .

How should researchers approach studying METRNL in cancer biology?

The emerging role of METRNL in cancer biology requires specific methodological considerations:

  • Expression analysis:

    • Comprehensive tissue microarray screening across cancer types

    • Correlation with clinicopathological features and prognosis

    • Single-cell RNA sequencing to identify METRNL-producing cells within tumor microenvironment

  • Functional characterization:

    • Stable overexpression and knockdown in cancer cell lines

    • Assessment of proliferation, migration, invasion, and apoptosis

    • Evaluation of angiogenic potential given METRNL's role in cardiac angiogenesis

  • Tumor microenvironment investigation:

    • Analysis of tumor-associated macrophage polarization in response to METRNL

    • Assessment of tumor-infiltrating lymphocyte profiles

    • Evaluation of METRNL's effect on immune checkpoint molecules

  • Epigenetic regulation:

    • DNA methylation analysis of METRNL promoter in cancer tissues

    • Investigation of miRNA regulation, as suggested in bladder cancer studies

    • Chromatin immunoprecipitation to identify transcription factors regulating METRNL in tumors

What are the recommended methodological approaches for resolving contradictory data on METRNL and obesity?

The literature contains contradictory findings regarding METRNL levels in obesity, necessitating specific methodological approaches:

  • Comprehensive phenotyping:

    • Beyond BMI, implement body composition analysis (DEXA, MRI)

    • Distinguish between subcutaneous and visceral adiposity

    • Assess adipose tissue inflammation and fibrosis

    • Measure fat distribution patterns and ectopic fat deposition

  • Temporal dynamics:

    • Longitudinal measurements during weight gain/loss

    • Intervention studies with multiple timepoints

    • Assessment during different metabolic challenges (meal tests, exercise)

  • Source identification:

    • Tissue-specific contribution analysis

    • Adipose tissue depot-specific expression patterns

    • Muscle biopsy correlation with circulating levels

    • Assessment of METRNL clearance rates in different metabolic states

  • Integrative multi-omics:

    • Correlation with metabolomic profiles

    • Lipidomic signatures associated with METRNL levels

    • Transcriptomic analysis of target tissues

    • Proteomic assessment of METRNL-associated signaling networks

Studies have reported that METRNL plasma levels were elevated more in obese T2DM patients (BMI > 30 kg/m²) than in non-obese T2DM patients (20 kg/m² ≤ BMI ≤ 30 kg/m²) . This apparent contradiction with METRNL's beneficial metabolic effects may represent a compensatory response to metabolic stress or reflect resistance to METRNL action in obesity, similar to patterns observed with other beneficial adipokines.

What experimental approaches best elucidate METRNL signaling pathways?

Investigating METRNL's molecular signaling requires systematic methodological approaches:

  • Receptor identification:

    • Affinity purification coupled with mass spectrometry

    • Cell surface binding assays with labeled METRNL

    • Receptor knockout validation in responding cells

    • Verification of KIT as a METRNL receptor in different tissues

  • Pathway mapping:

    • Phosphoproteomic profiling following METRNL stimulation

    • Time-course analysis of signaling cascades

    • Selective inhibitor approach to establish pathway hierarchy

    • CRISPR screening to identify essential signaling components

  • Downstream target validation:

    • ChIP-seq analysis to identify transcriptional targets

    • Validation of key nodes through genetic manipulation

    • Metabolic flux analysis to confirm functional outcomes

    • Assessment of AMPK and PPAR-γ activation

  • Integration with other pathways:

    • Interaction with inflammatory signaling (NF-κB, inflammasome)

    • Cross-talk with insulin signaling pathway

    • Relationship with PGC-1α signaling cascade

    • Interaction with thyroid hormone signaling

The main signaling pathways identified include AMPK activation, PPAR-γ signaling, and effects on intracellular calcium and ROS levels, as depicted in Figure 2 from the literature :

![Figure 2: Main signal pathways of Metrnl in myocytes. Metrnl activates AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor-δ (PPAR-γ) signaling by increasing intracellular calcium ion, reactive oxygen species (ROS), or AMP/ATP ratio levels in skeletal muscle cells.]

Understanding these signaling mechanisms is critical for developing targeted interventions based on METRNL biology.

How can researchers effectively study the relationship between METRNL and specific biomarkers?

The exploration of relationships between METRNL and various biomarkers requires specific methodological considerations:

  • Correlation analysis framework:

    • Multivariate regression models adjusting for confounders

    • Mediation analysis to identify indirect relationships

    • Principal component analysis to handle multiple correlated biomarkers

    • Network analysis to visualize complex relationships

  • Biomarker selection strategy:

    • Inflammatory markers (hs-CRP, IL-6, TNF-α)

    • Metabolic indicators (HOMA-IR, HbA1c, lipid profiles)

    • Tissue-specific markers (adipokines, myokines)

    • Disease-specific biomarkers (cardiac, renal, hepatic function)

  • Temporal relationship assessment:

    • Longitudinal designs with repeated measurements

    • Cross-lagged panel analysis to establish directionality

    • Intervention studies with biomarker monitoring

    • Time-series analysis for dynamic relationships

  • Biological validation:

    • In vitro mechanistic studies to confirm direct relationships

    • Animal models with METRNL manipulation

    • Ex vivo human tissue experiments

    • Genetic approaches (Mendelian randomization where feasible)

Current evidence indicates significant relationships between METRNL and several biomarkers, including HOMA-IR for insulin resistance , eosinophil numbers in exercise studies , and BDNF and TrkB levels in cognitive studies .

These methodological approaches can help establish whether METRNL is simply a biomarker or a mechanistically relevant factor in various physiological and pathological processes.

Product Science Overview

Introduction

Meteorin-like protein (METRNL), also known as Subfatin, Cometin, and Interleukin 39, is a secreted protein that has garnered significant interest due to its diverse roles in various physiological processes. This protein is encoded by the METRNL gene located on human chromosome 17 (17q25.3) and consists of 311 amino acids, including a 45 amino acid signal peptide .

Structural Characteristics

METRNL shares approximately 58% similarity with another protein called Meteorin . The mature form of METRNL is a 30 kDa protein, which is secreted after the cleavage of its signal peptide . The protein contains two structural domains, CUB and NTR, which are crucial for its function .

Expression and Distribution

Unlike Meteorin, which is predominantly expressed in the central nervous system, METRNL has a broader distribution. It is highly expressed in adipocytes, endothelial cells, activated monocytes, and myocytes . Additionally, METRNL is found in barrier tissues such as the skin and mucosal tissues of the digestive and respiratory tracts . The protein is secreted mainly by the endothelium, and its levels in circulation are significantly reduced when endothelial-specific knockout of METRNL is performed .

Biological Functions

METRNL plays a crucial role in regulating glucose and lipid metabolism, maintaining cardiovascular system homeostasis, and promoting anti-inflammatory responses . It is involved in various physiological processes, including thermogenesis in brown/beige adipocytes and the production of anti-inflammatory molecules . METRNL also induces the phosphorylation of STAT3, a transcription factor involved in various cellular processes .

Clinical Significance

METRNL has emerged as a promising biomarker and therapeutic target for cardiovascular and metabolic diseases. Its predictive and therapeutic roles have been studied in conditions such as atherosclerosis, ischemic heart disease, cardiac remodeling, heart failure, hypertension, chemotherapy-induced myocardial injury, diabetes mellitus, and obesity . The decline in circulating METRNL levels is closely associated with the occurrence of these diseases .

Recombinant Human METRNL

Recombinant human METRNL is produced using Chinese Hamster Ovary (CHO) cell lines and is available in carrier-free formulations . The recombinant protein is used in various research applications, including studies on its bioactivity, such as inducing STAT3 phosphorylation in embryonic rat cortical neurons . The protein is typically lyophilized and reconstituted in PBS for experimental use .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.