GDF15 Mouse

Growth and Differentiation factor 15 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Cardioprotective Effects

  • Pressure Overload Hypertrophy:
    Transgenic mice overexpressing cardiac-specific GDF15 showed resistance to pressure overload-induced hypertrophy, while Gdf15⁻/⁻ mice exhibited exacerbated hypertrophy and ventricular dysfunction .

    • Mechanism: GDF15 activates SMAD2/3 signaling, which inhibits hypertrophic pathways. SMAD6/7 overexpression reverses this effect .

    • Functional Outcome: Intravenous delivery of recombinant GDF15 attenuated heart failure in muscle lim protein-deficient mice .

  • Myocardial Infarction:
    Gdf15⁻/⁻ mice developed larger infarct sizes post-coronary ligation compared to wild-type controls, highlighting its role in limiting ischemia-reperfusion injury .

Metabolic Regulation

  • Obesity and Appetite Control:

    • Recombinant GDF15 (rhGDF15) reduced high-fat diet (HFD) preference in mice, promoting weight loss via GFRAL-dependent pathways .

    • Gdf15⁻/⁻ mice on HFDs gained more weight than wild-type littermates .

  • Nonalcoholic Steatohepatitis (NASH):

    • Gdf15⁻/⁻ mice developed worsened hepatic steatosis, inflammation, and fibrosis on NASH-inducing diets, while GDF15-transgenic mice showed reduced liver injury and improved metabolic parameters .

    • GDF15 suppressed fibrotic gene expression in hepatic stellate cells (HSCs) in vitro .

Neuroprotection and Cancer

  • Neuronal Survival:
    Hypothalamic GDF15 modulates appetite via neuropeptide Y and pro-opiomelanocortin pathways .

  • Tumor Suppression:
    Orthotopic tumor models (e.g., MCF-7 breast cancer cells) revealed that GDF15 overexpression reduced tumor growth by ~50% .

Cardiovascular Diseases

  • Recombinant GDF15 attenuated ventricular dilation in cardiomyopathy models .

  • Conflicting roles in atherosclerosis: GDF15 deletion increased macrophage accumulation in LDL receptor⁻/⁻ mice but reduced early plaque formation in another study .

Kidney Protection

  • Diabetic Gdf15⁻/⁻ mice exhibited increased renal damage (e.g., elevated KIM-1, α-SMA) compared to wild-type controls. Recombinant GDF15 reduced fibrosis in unilateral ureter obstruction models .

Exercise Adaptation

  • Gdf15 ablation did not impair exercise capacity in mice but increased skeletal muscle stress response (e.g., elevated Fgf21 expression post-exercise) .

Research Applications of Recombinant Mouse GDF15

  • In Vitro Studies: Used to assess SMAD2/3 phosphorylation in cardiomyocytes .

  • In Vivo Delivery: Administered intravenously or intraperitoneally to study endocrine effects on heart, liver, and kidneys .

Limitations and Controversies

  • Tissue-Specific Effects: GDF15 is undetectable in normal mouse hearts but induced during hypertrophy , complicating baseline studies.

  • Dose-Dependent Outcomes: Anti-fibrotic effects in HSCs required supraphysiological GDF15 doses (>1 ng/mL) .

Product Specs

Introduction
Growth Differentiation Factor 15 (GDF15) is a protein belonging to the Transforming Growth Factor Beta (TGF-β) superfamily. This family plays a crucial role in regulating inflammation and programmed cell death (apoptosis) in response to tissue injury and during disease progression. GDF15 is primarily produced by the liver, and its expression significantly increases upon damage to organs like the liver, kidneys, heart, and lungs. GDF15 exhibits diverse effects on cells, either stimulating proliferation or promoting growth arrest and differentiation, depending on the cell type and its developmental stage. In cerebellar granule neurons, GDF15 has been shown to prevent apoptosis by activating the Akt pathway and suppressing the activity of the ERK pathway. Notably, GDF15 acts as a local and systemic factor that counteracts the heart's thickening response to stress and prevents the decline in the heart's pumping ability.
Description
Recombinant Mouse GDF15, produced in E. coli, is a single-chain polypeptide that lacks glycosylation. It consists of 138 amino acids (spanning positions 189 to 303), resulting in a molecular weight of 14.9 kDa. A 23-amino acid His-tag is attached to the N-terminus of GDF15 to facilitate purification using specialized chromatographic methods.
Physical Appearance
A clear and sterile solution.
Formulation
GDF15 protein solution is provided at a concentration of 1.0 mg/ml. The solution is buffered with 20mM Phosphate buffer at a pH of 8.0 and contains 10% glycerol.
Stability
For short-term storage (up to 4 weeks), keep at 4°C. For longer storage, freeze at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is recommended for long-term storage. It's important to minimize repeated freezing and thawing.
Purity
The purity is determined to be greater than 90% using SDS-PAGE analysis.
Synonyms
Growth/differentiation factor 15, GDF-15.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSSAHAHPR DSCPLGPGRC CHLETVQATL EDLGWSDWVL SPRQLQLSMC VGECPHLYRS ANTHAQIKAR LHGLQPDKVP APCCVPSSYT PVVLMHRTDS GVSLQTYDDL VARGCHCA.

Q&A

What is GDF15 and what are its primary biological functions in mice?

GDF15, also known as NSAID activated gene-1 (NAG-1), is a stress-responsive cytokine associated with multiple biological processes including energy homeostasis, body weight regulation, and responses to cellular stress. In mice, GDF15 has been shown to regulate body weight and fat content through mechanisms affecting both food intake and metabolic processes. Transgenic mice expressing human GDF15 (hGDF15) demonstrate significant reductions in body weight and fat content despite unchanged food consumption in some models . The primary biological function appears to be signaling nutritional or inflammatory stress through its specific receptor GFRAL, which is expressed in the hindbrain, particularly in the nucleus tractus solitarius (NTS) and area postrema (AP) . GDF15 serves as an endocrine signal triggered by various cellular stressors, distinguishing it from classical intestinally-derived satiety hormones by its modest response to routine caloric fluctuations but robust induction during sustained metabolic stress .

How do GDF15 knockout mice differ from wild-type mice in metabolic parameters?

GDF15 knockout (KO) mice exhibit notable differences from wild-type littermates in several key metabolic parameters:

  • Body weight: GDF15 KO mice are approximately 6-10% heavier than wild-type mice on standard chow diet, with the difference becoming more pronounced when fed a high-fat diet .

  • Fat accumulation: These mice demonstrate increased adipose tissue accumulation, particularly when challenged with obesogenic diets .

  • Liver pathology: In models of non-alcoholic steatohepatitis (NASH), GDF15 KO mice show enhanced NASH phenotypes including increased fibrosis and inflammatory markers .

  • Glucose metabolism: KO mice typically show reduced glucose tolerance and increased insulin resistance compared to wild-type counterparts .

  • Inflammatory responses: The absence of GDF15 affects inflammatory processes, though interestingly, GDF15 appears dispensable for some aspects of LPS-induced anorexia as GDF15 KO mice still show reduced food intake following LPS administration .

These differences suggest that endogenous GDF15 plays an important physiological role in maintaining metabolic homeostasis, particularly during metabolic stress conditions.

What is the receptor for GDF15 in mice and where is it expressed?

GDF15 signals through a specific receptor called GDNF family receptor alpha-like (GFRAL). This receptor forms a heterodimer with the co-receptor RET (rearranged during transfection) to transduce GDF15 signaling . Key characteristics of GFRAL include:

  • Tissue distribution: GFRAL expression is remarkably restricted to specific regions of the hindbrain, particularly the nucleus tractus solitarius (NTS) and area postrema (AP) .

  • Neuroanatomical significance: These hindbrain regions are critical for processing various physiological signals related to feeding, nausea, and aversive responses. Selective lesioning of these regions renders mice unresponsive to the anorexigenic effects of GDF15 .

  • Downstream signaling: Activation of GFRAL by GDF15 leads to subsequent cFos activation in the parabrachial nucleus (PBN), which has been linked to appetite suppression and aversive responses .

  • Genetic necessity: GFRAL knockout mice are resistant to the weight-reducing and anorexigenic effects of recombinant GDF15 administration, confirming that this receptor is essential for GDF15's metabolic effects .

The highly specific expression pattern of GFRAL explains why GDF15's effects are predominantly on food intake and energy homeostasis, rather than having broader systemic actions that might be expected from a circulating factor.

What are the key considerations when designing experiments with transgenic GDF15 mouse models?

When designing experiments using transgenic GDF15 mouse models, researchers should consider several critical factors to ensure robust and interpretable results:

  • Promoter selection: Different promoters used to drive GDF15 expression result in varying phenotypes. Studies show that:

    • Ubiquitous expression promoters lead to widespread GDF15 production with consistent body weight reduction but without appetite suppression

    • Tissue-specific promoters (e.g., liver-specific or macrophage-specific) can produce different phenotypes - macrophage-specific expression shows lower food intake while liver-specific expression mimics ubiquitous expression

  • Expression levels: The serum concentration of GDF15 is crucial to phenotypic manifestation:

    • Circulating levels between 3-50 ng/mL are common in transgenic models

    • Higher expression levels may trigger more pronounced phenotypes

    • Physiological vs. pharmacological levels should be clearly distinguished in result interpretation

  • Duration of expression: Chronic vs. acute expression produces different metabolic effects:

    • Chronic expression in transgenic models often leads to metabolic adaptation and altered oxidative metabolism

    • Acute administration typically affects food intake without immediate changes in energy expenditure

  • Control selection: Appropriate littermate controls are essential as strain backgrounds can significantly influence metabolic parameters and responses to GDF15.

  • Feeding conditions: Testing mice under different nutritional challenges is important:

    • Standard chow vs. high-fat diet responses may differ significantly

    • Food intake patterns should be carefully monitored (continuous vs. meal frequency analysis)

  • Age and sex considerations: Both factors influence GDF15 responses and baseline metabolic parameters.

These considerations are crucial for designing experiments that yield reproducible and physiologically relevant results when studying GDF15 biology in transgenic mouse models.

How should recombinant GDF15 administration experiments be designed to distinguish between acute and chronic effects?

Designing recombinant GDF15 (rGDF15) administration experiments requires careful consideration to properly distinguish between acute and chronic effects:

Acute administration design:

  • Dosing strategy:

    • Single-dose experiments typically use 0.01-0.1 mg/kg subcutaneously

    • Include dose-response curve to establish minimum effective dose

    • Time course measurements at 1, 4, 8, and 24 hours post-administration to capture peak effects

  • Parameters to measure:

    • Food intake at regular intervals (1, 2, 4, 8, 24 hours)

    • Behavioral assessments for potential aversive responses

    • Metabolic parameters (glucose, insulin) at key timepoints

    • cFos activation in brain regions of interest (NTS, AP, PBN) using immunohistochemistry

  • Control considerations:

    • Vehicle-treated groups with identical handling

    • Pair-fed controls to distinguish direct metabolic effects from those secondary to reduced food intake

    • Include positive controls for anorexia (e.g., LPS, lithium chloride) for comparison

Chronic administration design:

  • Dosing regimen:

    • Every-other-day or daily administration for 3+ weeks

    • Consistent timing of administration relative to feeding cycles

    • Gradual dose escalation to account for potential adaptation

  • Parameters to track:

    • Daily body weight and food intake

    • Body composition analysis (before, during, and after treatment)

    • Energy expenditure and respiratory exchange ratio via metabolic chambers

    • Longitudinal glucose tolerance and insulin sensitivity tests

    • Tissue-specific gene expression changes in metabolic pathways

  • Mechanistic considerations:

    • Include GFRAL knockout controls to confirm receptor-dependent effects

    • Measure adaptation through changes in receptor expression or downstream signaling

    • Assess compensatory mechanisms that might emerge during chronic treatment

  • Post-treatment analysis:

    • Recovery period assessment to determine persistence of effects

    • Tissue collection for molecular analysis of metabolic adaptation

The primary differences observed between acute and chronic rGDF15 administration in mice include changes in oxidative metabolism, which become apparent only after sustained treatment, while anorexigenic effects are typically evident immediately . Designing experiments that can distinguish these temporal effects is essential for understanding GDF15's physiological versus pharmacological actions.

What controls are necessary when investigating GDF15's role in inflammatory responses to LPS in mice?

  • Genetic controls:

    • Include both wild-type and GDF15 knockout littermates

    • GFRAL knockout mice to distinguish receptor-dependent effects

    • Consider heterozygotes to assess gene dosage effects

  • Dosage controls:

    • Careful LPS dose selection is crucial - studies show 600 μg/kg provides moderate illness while ensuring survival

    • Perform dose-response curves (e.g., 50-1000 μg/kg) to determine appropriate dosage

    • Include sublethal and moderate doses to capture different response thresholds

  • Timing controls:

    • Measure parameters at multiple timepoints (1, 2, 4, 8, 24 hours post-LPS)

    • GDF15 elevations occur within 2 hours of LPS administration in multiple species

    • Include pre-treatment measurements as baseline

  • Alternative inflammatory mediator controls:

    • Measure other cytokines (TNF-α, IL-6, IL-1β) that may compensate for GDF15 absence

    • Assess prostaglandin production (especially PGE2) which may drive anorexic responses

    • Include COX2 inhibitors to distinguish prostaglandin-dependent responses

  • Environmental controls:

    • Housing temperature significantly impacts metabolic responses to inflammation

    • Control for time of day due to circadian influences on inflammatory responses

    • Food accessibility standardization (ad libitum vs. restricted)

  • Behavioral controls:

    • Pair-fed controls to distinguish direct effects from those secondary to anorexia

    • Include non-inflammatory anorexigenic controls (e.g., GLP-1 agonists)

    • Monitor multiple sickness behaviors beyond food intake (locomotor activity, temperature, etc.)

  • Pharmacological controls:

    • Anti-GDF15 neutralizing antibodies in wild-type mice

    • Recombinant GDF15 administration to determine if it can potentiate or substitute for LPS effects

The research indicates that while LPS robustly increases GDF15 levels, the anorexic response to LPS appears to be preserved in both GDF15 and GFRAL knockout mice, suggesting compensatory mechanisms . These comprehensive controls help distinguish between necessary and contributory roles of GDF15 in the inflammatory response.

How can researchers resolve contradictory findings regarding GDF15's effects on food intake in different mouse models?

Resolving contradictory findings regarding GDF15's effects on food intake across different mouse models requires systematic consideration of several experimental variables:

  • Model-specific differences:

    • Transgenic expression vs. recombinant administration: Transgenic mice expressing GDF15 ubiquitously often show unchanged food intake despite lower body weight, while acute rGDF15 administration consistently reduces food intake .

    • Expression localization: Macrophage-specific GDF15 expression (controlled by c-fms promoter) shows appetite suppression, while ubiquitous or liver-specific expression does not .

    • Adaptive compensations: Chronic GDF15 exposure in transgenic models may trigger neural circuit adaptations that normalize food intake over time .

  • Methodological considerations:

    • Measurement timing: Food intake should be assessed at multiple timepoints post-intervention

    • Measurement methodology: Manual weighing vs. automated systems can yield different results

    • Housing conditions: Single vs. group-housed mice may show different feeding behaviors

    • Circadian considerations: Time of day for measurements significantly impacts feeding results

  • Comparative analysis approach:

    Model TypeFood Intake EffectBody Weight EffectPossible Explanation
    Ubiquitous GDF15 transgenicUnchangedDecreasedMetabolic adaptation/increased energy expenditure
    Macrophage-specific GDF15 transgenicDecreasedDecreasedDifferent signaling or delivery to brain regions
    Acute rGDF15 administrationDecreasedDecreasedDirect GFRAL activation without adaptation
    Chronic rGDF15 administrationInitially decreased, then variableDecreasedProgressive adaptation of neural circuits
  • Reconciliation strategies:

    • Perform side-by-side comparisons of multiple models under identical conditions

    • Time-course experiments to distinguish initial from adapted responses

    • Gene expression analysis of GFRAL and downstream signaling components in different models

    • Consider developmental timing - embryonic expression vs. adult-onset expression

    • Examine tissue-specific metabolic changes that might compensate for unchanged food intake

  • Mechanistic investigations:

    • Neural pathway activation analysis using cFos or other immediate early genes

    • Electrophysiological recording from GFRAL-expressing neurons in different models

    • Assessment of leptin, ghrelin, and other feeding-related hormones that might compensate

The most likely explanation for these disparate findings is that GDF15 acts through both appetite suppression and altered energy metabolism, with the balance between these effects depending on exposure pattern, timing, and duration . The development of conditional and inducible models would help resolve these contradictions by allowing temporal control of GDF15 expression.

How should energy expenditure data be properly analyzed in GDF15 mouse studies to avoid misinterpretation?

Energy expenditure data in GDF15 mouse studies requires careful analysis to avoid misinterpretation, as several methodological challenges exist:

  • Body weight normalization considerations:

    • GDF15 interventions often change body weight and composition, complicating interpretation

    • Multiple normalization approaches should be employed:

      • Per animal (absolute values)

      • Per total body weight

      • Per lean body mass (preferred)

      • ANCOVA analysis with body weight as covariate

  • Data collection protocols:

    • Acclimation period: Minimum 24-48 hours of acclimation to metabolic chambers

    • Measurement duration: Collect data over multiple 24-hour cycles

    • Temporal resolution: Analyze both aggregate data and time-specific measurements

    • Activity correlation: Concurrent measurement of physical activity to distinguish activity-dependent and independent energy expenditure

  • Respiratory exchange ratio (RER) analysis:

    • GDF15 consistently decreases RER, indicating a shift from carbohydrate to lipid metabolism

    • RER should be analyzed both:

      • Across 24-hour cycles

      • During specific metabolic states (fed, fasted, active, rest phases)

    • Changes in RER often precede measurable changes in total energy expenditure

  • Interpreting conflicting findings:

    • Short-term vs. long-term effects: Research shows changes in oxidative metabolism are more evident after chronic GDF15 elevation

    • Acute treatment studies often report no changes in energy expenditure

    • Chronic elevations (transgenic or extended administration) typically show increased energy expenditure and metabolic shifts

  • Comprehensive approach to resolve discrepancies:

    MeasurementAnalysis ApproachCommon PitfallsRecommended Solution
    Total energy expenditureMultiple normalization methodsSingle normalization methodUse both per animal and per lean mass; include ANCOVA
    RER24-hour and phase-specificAggregate data onlyAnalyze circadian patterns and responses to feeding
    Tissue metabolismEx vivo tissue analysisRelying solely on indirect calorimetryInclude tissue-specific metabolic gene expression
  • Complementary measurements:

    • Gene/protein expression of metabolic markers (UCP1, PGC1α, CPT1)

    • Tissue-specific substrate utilization

    • Cold challenge tests to assess thermogenic capacity

    • Pair-feeding experiments to distinguish food intake effects from direct metabolic effects

As noted in the literature, "The difficulties and complexity in evaluation of energy metabolism in mice may provide an explanation for the inconsistent findings on energy expenditure" . Researchers should be aware that the most consistent metabolic finding across GDF15 studies is the shift from carbohydrate to lipid metabolism as indicated by RER changes .

What explains the differential effects of GDF15 in acute versus chronic exposure models?

The differential effects of GDF15 in acute versus chronic exposure models can be explained by several key mechanisms:

  • Temporal adaptation of neural circuitry:

    • Acute exposure: Robust activation of GFRAL-positive neurons in the NTS and AP leads to immediate anorexigenic effects and potential aversive responses

    • Chronic exposure: Adaptive changes in receptor sensitivity, signaling efficiency, or downstream neural circuit function may develop over time, potentially explaining why transgenic mice with constitutive GDF15 expression often show normal food intake

  • Metabolic adaptation timeline:

    • Immediate effects (0-24 hours): Primarily focused on reduced food intake without significant changes in energy expenditure

    • Intermediate effects (days): Beginning of metabolic substrate utilization shift (carbohydrate to lipid)

    • Long-term effects (weeks): Comprehensive metabolic reprogramming affecting:

      • Oxidative metabolism gene expression

      • Mitochondrial function and biogenesis

      • Tissue-specific substrate utilization pathways

  • Molecular mechanisms explaining differential effects:

    • Receptor desensitization: Potential downregulation of GFRAL or associated signaling components

    • Compensatory hormone production: Changes in leptin, ghrelin, or other energy-regulating hormones

    • Tissue remodeling: Long-term exposure induces adipose tissue browning and liver metabolic reprogramming

    • Inflammatory adaptation: Initial inflammatory responses to GDF15 may resolve with chronic exposure

  • Experimental evidence supporting adaptation:

    • Transgenic mice expressing GDF15 since embryonic development show metabolic rather than appetite-based weight differences

    • Extended recombinant GDF15 administration studies (3+ weeks) demonstrate shifts toward increased oxygen consumption and altered energy utilization

    • Gene expression changes in lipolysis, thermogenesis, and oxidative metabolism markers occur primarily after prolonged GDF15 exposure

This differential response pattern suggests that GDF15 initially functions as an acute stress signal triggering aversive and anorexigenic responses, but with sustained elevation, it promotes metabolic adaptation that prioritizes fat utilization and increased energy expenditure. This biphasic response pattern may reflect GDF15's evolutionary role in adaptating to nutritional stress conditions .

How does the integrated stress response regulate GDF15 expression in different mouse tissues?

The integrated stress response (ISR) is a central cellular pathway that regulates GDF15 expression across various mouse tissues in response to different stressors:

  • Molecular mechanisms of ISR activation of GDF15:

    • Various cellular stressors (nutrient deprivation, ER stress, oxidative stress) activate distinct kinases (PERK, GCN2, PKR, HRI)

    • These kinases phosphorylate eIF2α (eukaryotic translation initiation factor 2α)

    • Phosphorylated eIF2α leads to preferential translation of specific mRNAs, including ATF4 (activating transcription factor 4)

    • ATF4 directly regulates GDF15 gene expression by binding to its promoter region

  • Tissue-specific ISR activation patterns:

    • Liver: Primary site of GDF15 induction during amino acid imbalance and protein restriction

    • Adipose tissue: Produces GDF15 in response to high-fat feeding and thermogenic stress

    • Muscle: Induces GDF15 during exercise and mitochondrial stress

    • Immune cells: Express GDF15 during inflammatory activation (e.g., LPS stimulation)

    • Brain: Limited GDF15 production due to blood-brain barrier constraints

  • Nutritional stress triggers:

    • Amino acid imbalance: Lysine-deficient diets rapidly induce GDF15 via GCN2-mediated ISR activation

    • Chronic high-fat feeding: Activates the ISR in multiple tissues, particularly adipose and liver

    • Protein restriction: Triggers hepatic GDF15 production via similar mechanisms to amino acid imbalance

  • Experimental approaches to study ISR regulation of GDF15:

    • Genetic models: Tissue-specific deletion of ISR components (e.g., liver-specific ATF4 knockout)

    • Pharmacological tools: ISR inhibitors (ISRIB) or activators (Tunicamycin, Thapsigargin)

    • Nutritional models: Custom diets with specific nutrient imbalances

    • Time-course analysis: Monitoring phospho-eIF2α, ATF4, and GDF15 induction patterns

  • Therapeutic implications:

    • Targeting the ISR-GDF15 axis may be beneficial in conditions where GDF15 is protective (obesity, diabetes)

    • Inhibiting excessive GDF15 production might mitigate unwanted anorexia in conditions like cancer cachexia

Understanding the ISR regulation of GDF15 helps explain why GDF15 levels increase during various forms of nutritional stress but remain relatively stable during simple caloric restriction or excess, distinguishing it from classical appetite-regulating hormones . This stress-responsive pattern supports GDF15's role as an endocrine signal of nutritional or cellular stress rather than a routine satiety factor.

What is the relationship between GDF15 and conditioned taste aversion in mice, and how does this inform therapeutic applications?

The relationship between GDF15 and conditioned taste aversion (CTA) in mice has important implications for understanding both its physiological role and potential therapeutic applications:

  • Experimental evidence linking GDF15 to CTA:

    • Subcutaneous GDF15 administration (0.01-0.1 mg/kg) reduces preference for saccharin in two-bottle preference tests, similar to lithium chloride (a known aversive stimulus)

    • GDF15 administration activates brain regions associated with aversive responses, particularly the parabrachial nucleus (PBN)

    • The dose response for CTA closely parallels the dose response for anorexia, suggesting these effects may be mechanistically linked

  • Neuroanatomical basis for GDF15-induced aversion:

    • GDF15 receptor (GFRAL) is expressed in hindbrain regions (NTS, AP) that detect noxious stimuli and toxins

    • These regions project to the PBN, which mediates both aversive learning and appetite suppression

    • Similar neural circuits are activated by other stimuli that cause nausea or malaise (LPS, lithium chloride)

    • The restricted expression of GFRAL to these specific brain regions suggests evolutionary adaptation for detecting harmful substances

  • Physiological significance:

    • GDF15-induced aversion may represent an adaptive response to nutritional stress or toxin exposure

    • Elevated GDF15 during prolonged high-fat feeding or amino acid imbalance could signal dietary inadequacy

    • This aversive component might help animals avoid nutritionally imbalanced food sources

  • Implications for therapeutic applications:

    Potential ApplicationConsiderationMitigation Strategy
    Anti-obesity therapyGDF15-induced aversion/nausea may limit tolerabilityDevelop derivatives with reduced aversive potential
    Appetite regulationAversive effects may contribute to weight lossTarget downstream pathways that mediate metabolic but not aversive effects
    Cancer cachexiaGDF15 may contribute to tumor-induced anorexiaGDF15 antagonists might reduce cancer-associated appetite suppression
    Diabetes therapyChronic dosing may lead to adaptation of aversive responseIntermittent dosing schedules or combination therapies
  • Experimental approaches to distinguish anorexia from aversion:

    • Conditioned place preference/aversion tests to quantify motivational aspects

    • Operant responding for food to assess hunger versus aversion

    • Meal pattern analysis (meal size vs. meal frequency) to distinguish satiety from aversion

    • Concurrent monitoring of nausea biomarkers (e.g., kaolin consumption in rodents)

  • Adaptation considerations:

    • Chronic exposure in transgenic models appears to normalize feeding behavior, suggesting potential adaptation to aversive effects

    • This adaptation might offer a therapeutic window where beneficial metabolic effects persist after aversive effects subside

The dual nature of GDF15 as both a metabolic regulator and an aversive stimulus presents both challenges and opportunities for therapeutic development . Understanding the molecular mechanisms that differentiate these effects could enable the development of more targeted approaches that preserve beneficial metabolic actions while minimizing aversive side effects.

How do different mouse strains respond to GDF15 manipulation, and what does this reveal about genetic modifiers of GDF15 signaling?

Different mouse strains demonstrate variable responses to GDF15 manipulation, providing valuable insights into genetic modifiers of GDF15 signaling:

  • Strain-specific response patterns:

    • C57BL/6J: Standard reference strain showing robust anorexia and weight loss with GDF15 administration

    • FVB: Similar metabolic response profile to C57BL/6J when expressing GDF15 transgenically

    • DBA/2J: Often shows more pronounced metabolic phenotypes in response to GDF15

    • BALB/c: May exhibit altered behavioral responses to GDF15-mediated aversive effects

    • 129Sv: Background may influence GFRAL expression patterns and signaling efficiency

  • Key phenotypic variations observed across strains:

    • Magnitude of weight loss in response to equivalent GDF15 doses

    • Duration of anorexigenic effects before adaptation occurs

    • Balance between food intake reduction versus metabolic adjustments

    • Susceptibility to GDF15-induced taste aversion

    • Baseline metabolic parameters that may influence response sensitivity

  • Genetic factors potentially modifying GDF15 effects:

    • GFRAL expression levels and patterns

    • RET co-receptor variants affecting signaling efficiency

    • Downstream signaling component polymorphisms

    • Compensatory hormonal system differences (leptin, ghrelin)

    • Neuroanatomical variations in regions processing GDF15 signals

  • Experimental approaches to identify modifiers:

    • Quantitative trait locus (QTL) mapping using response to GDF15 as a phenotype

    • Recombinant inbred strain panels to isolate genetic contributors

    • Congenic strain development to confirm candidate modifier genes

    • Cross-fostering experiments to distinguish developmental from genetic factors

    • Transcriptomic analysis of response variability between strains

  • Translational relevance for human therapeutics:

    • Strain variations may model population heterogeneity in human GDF15 responses

    • Identification of favorable response predictors could enable patient stratification

    • Understanding compensatory mechanisms in resistant strains may reveal therapeutic targets

    • Strain differences in side effect profiles may guide development of better-tolerated GDF15-based therapies

The study of strain-specific responses to GDF15 manipulation is particularly important given the observation that "expression of hGDF15 has profound biological effects on body weight and fat content" , yet the magnitude of these effects may vary considerably between genetic backgrounds. This variation likely reflects the complex interactions between GDF15 signaling and other energy homeostasis pathways, potentially revealing novel therapeutic targets or biomarkers for predicting treatment response.

How does GDF15 affect metabolic parameters in mouse models of non-alcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH)?

GDF15 demonstrates significant protective effects in mouse models of non-alcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH), affecting multiple aspects of disease pathophysiology:

  • Effects on liver pathology:

    • GDF15 transgenic mice show protection against inflammatory steatosis when challenged with NASH-inducing diets

    • Liver-specific GDF15 expression (using liver-specific promoters) provides similar protection to ubiquitous expression

    • GDF15 knockout mice develop more severe NASH phenotypes, indicating an endogenous protective role

  • Molecular mechanisms of hepatoprotection:

    • Suppression of fibrosis-related gene expression:

      • Downregulation of TGFβ1 (Tgfb1)

      • Reduced collagen production (Col1a1)

      • Decreased tissue inhibitor of metalloproteinases (Timp1)

      • Lower α-smooth muscle actin expression (Acta2)

    • Inhibition of inflammatory pathways:

      • Reduced osteopontin expression in the liver

      • Decreased inflammatory cell infiltration

      • Attenuated hepatic inflammasome activation

  • Metabolic effects relevant to NAFLD/NASH:

    • Improved insulin sensitivity reduces hepatic de novo lipogenesis

    • Shift from carbohydrate to lipid metabolism (lower RER) promotes fat utilization

    • Reduced body weight and adiposity decrease lipid flux to the liver

    • Protection against diet-induced obesity limits hepatic steatosis development

  • Biomarker potential:

    • Serum GDF15 levels correlate with risk of advanced fibrosis in NAFLD patients

    • This association remains significant after adjusting for age, gender, BMI, insulin resistance and skeletal muscle mass

    • Suggesting GDF15 may serve as an independent biomarker for fibrosis progression

  • Therapeutic implications:

    Therapeutic StrategyPotential BenefitSupporting Evidence
    GDF15 administrationReduced steatosis and fibrosisProtection in mouse models
    Targeting GDF15 expressionPrevention of NASH progressionEnhanced GDF15 expression reduces steatohepatitis
    Combination with metabolic therapiesSynergistic hepatoprotectionGDF15's effects on metabolism complement direct hepatic actions
    Biomarker applicationPatient stratification for interventionCorrelation with fibrosis risk independent of other factors
  • Research considerations:

    • Dose-dependency of effects (physiological vs. pharmacological levels)

    • Timing of intervention (preventive vs. therapeutic application)

    • Direct hepatic effects vs. secondary benefits from systemic metabolic improvements

    • Long-term safety and efficacy with chronic administration

The evidence suggests that "GDF15's suppression of fibrosis may be a suitable candidate or target for drug development" in NAFLD/NASH. The dual action of GDF15 on both metabolic parameters and direct hepatic inflammatory/fibrotic pathways makes it particularly promising for addressing this complex metabolic liver disease.

What is the role of GDF15 in mouse models of infection and inflammation, and how does this compare to its metabolic functions?

GDF15's role in mouse models of infection and inflammation reveals a complex interplay with its metabolic functions:

  • Inflammatory induction of GDF15:

    • LPS administration rapidly increases circulating GDF15 levels in mice within 2 hours

    • This response is consistent across species (mice, rats, humans)

    • The magnitude of GDF15 elevation correlates with inflammatory intensity

    • Different housing temperatures do not affect LPS-induced GDF15 increases

  • GDF15's role in acute inflammation:

    • Despite robust GDF15 induction, genetic studies reveal surprising functional findings:

      • GDF15 knockout mice still exhibit anorexia following LPS administration

      • GFRAL knockout mice also maintain LPS-induced anorexic responses

      • These findings indicate that GDF15 is not necessary for inflammation-induced anorexia

    • Inflammatory cytokine production (TNF-α, IL-6) remains robustly elevated in both wild-type and GDF15 knockout mice following LPS administration

  • Comparative analysis of inflammatory versus metabolic roles:

    ParameterInflammatory ContextMetabolic ContextImplications
    NecessityNot required for LPS anorexia Essential for body weight regulation Context-dependent significance
    RedundancyHigh redundancy with other inflammatory mediators Less redundancy in metabolic pathwaysDifferent therapeutic potential
    Induction kineticsRapid (hours) Slower (days to weeks) for chronic HFD Acute vs. chronic signaling roles
    Dose-responseSubstantial elevation with moderate LPS Modest changes with routine caloric fluctuations Sensitivity to different stressors
  • Integration of inflammatory and metabolic functions:

    • GDF15 may represent a stress signal that connects inflammatory status to metabolic adaptation

    • In cancer cachexia models, GDF15 appears more critical for appetite suppression than in acute infection

    • The pronounced increase during inflammation may serve functions beyond appetite regulation:

      • Tissue protection from inflammatory damage

      • Metabolic adaptation to infectious challenge

      • Potential immunomodulatory effects

  • Experimental considerations for studying dual roles:

    • Use of multiple inflammatory models beyond LPS (bacterial/viral infection, sterile inflammation)

    • Careful dose selection to distinguish physiological from pathological responses

    • Temporal profiling to capture both acute and resolution phases

    • Tissue-specific deletion models to identify critical sources during inflammation

  • Therapeutic implications of dual inflammatory/metabolic roles:

    • Targeting GDF15 in inflammatory conditions may need to consider both beneficial and detrimental effects

    • Cancer cachexia interventions might benefit from GDF15 inhibition

    • Metabolic disease treatments may need to account for potential immunomodulatory effects

The evidence suggests that "GDF15 is not necessary to the anorexic responses to LPS" , which contrasts with its essential role in other contexts like cancer anorexia. This indicates context-dependent functions that may reflect "potentially adaptive or pathophysiological roles that the pronounced increase in circulating GDF15 plays in directing the physiological responses to systemic infections" .

How does GDF15 influence glucose metabolism and insulin sensitivity in mouse models of diabetes?

GDF15 exhibits significant beneficial effects on glucose metabolism and insulin sensitivity in mouse models of diabetes, through multiple mechanisms:

  • Effects on glucose homeostasis:

    • Transgenic mice expressing GDF15 show improved glucose tolerance

    • GDF15 administration enhances insulin sensitivity in both lean and obese mice

    • These beneficial effects are observed in conjunction with, but not solely dependent on, reductions in body weight and fat content

  • Molecular mechanisms affecting insulin sensitivity:

    • Reduction in inflammasome activity:

      • GDF15 expression reduces NLRP3 inflammasome activity, which contributes to insulin resistance in obesity

      • This effect occurs in various tissues including adipose tissue and liver

    • Altered tissue-specific metabolism:

      • Promotion of skeletal muscle glucose uptake

      • Reduction in hepatic gluconeogenesis

      • Enhanced adipose tissue insulin signaling

    • Shift in substrate utilization:

      • Decreased respiratory exchange ratio indicates preference for lipid metabolism

      • Reduced ectopic lipid accumulation in insulin-sensitive tissues

      • Improved mitochondrial function and metabolic flexibility

  • Comparative analysis of anti-diabetic effects:

    MechanismObservation in GDF15 ModelsRelation to Diabetes Pathophysiology
    Body weight reductionConsistent finding in most models Ameliorates insulin resistance through reduced adiposity
    Adipose tissue remodelingReduction in inflammatory markersDecreases systemic insulin resistance
    Liver metabolismSuppression of steatosis and inflammation Improves hepatic insulin sensitivity
    Inflammatory modulationReduction in NLRP3 activity Attenuates inflammation-driven insulin resistance
    Food intakeVariable effects depending on model May contribute to weight reduction in some contexts
  • Effects in different diabetes models:

    • Diet-induced obesity models: Consistent improvements in glucose tolerance and insulin sensitivity

    • Genetic obesity models (ob/ob, db/db): GDF15 administration improves metabolic parameters

    • Chemical diabetes induction (streptozotocin): Less explored but may show tissue-protective effects

  • Therapeutic implications for diabetes:

    • "These metabolic effects coupled with the reduction in body weight and fat content makes GDF15 as an attractive candidate for use in the treatment of type 2 diabetes"

    • Potential advantages over existing therapies:

      • Dual action on body weight and direct insulin-sensitizing effects

      • Novel mechanism distinct from current anti-diabetic agents

      • Potential for combination with existing therapies

  • Research considerations:

    • Dose-response relationships for metabolic versus weight-reducing effects

    • Duration of treatment needed for maximal benefit

    • Potential development of tolerance to metabolic effects

    • Optimal treatment regimens (continuous vs. intermittent)

    • Identification of patient subgroups most likely to benefit

The consistent finding that GDF15 improves glucose tolerance and insulin sensitivity makes it a promising candidate for type 2 diabetes treatment. The multiple mechanisms through which GDF15 acts on glucose metabolism—both weight-dependent and independent pathways—suggest it may offer advantages over single-mechanism anti-diabetic approaches.

What are the optimal methods for measuring GDF15 levels in mouse models?

Accurate measurement of GDF15 levels in mouse models requires careful methodological considerations:

  • Sample collection considerations:

    • Blood sampling timing:

      • Circadian variations affect GDF15 levels

      • Consistent collection time is crucial for comparative studies

      • Fasting status significantly impacts measurements

    • Sample processing:

      • Rapid separation of serum/plasma (within 30 minutes of collection)

      • Standardized centrifugation protocols (e.g., 2000g for 15 minutes at 4°C)

      • Storage at -80°C with minimal freeze-thaw cycles

    • Collection methods:

      • Cardiac puncture yields highest volume but is terminal

      • Submandibular bleeding allows for longitudinal measurements

      • Tail vein sampling provides smaller volumes but enables repeated measures

  • Quantification methods comparison:

    MethodAdvantagesLimitationsBest Applications
    ELISAWidely available, species-specificHigher CV%, labor intensiveStandard quantification
    Multiplex assaysMultiple analytes simultaneouslyPotential cross-reactivityComprehensive profiling
    Mass spectrometryHighest specificity, can detect variantsComplex sample prep, expensiveDetailed isoform analysis
    SomaScan platformHigh sensitivity, broad dynamic rangeLimited availability, costLarge-scale studies
    Olink proximity extensionHigh specificity and sensitivitySpecialized equipment neededMulti-biomarker studies
  • Species considerations:

    • Mouse GDF15 vs. human GDF15:

      • Critical to use species-appropriate antibodies/assays

      • Human GDF15 ELISA kits may not accurately detect mouse GDF15

      • For transgenic models expressing human GDF15, human-specific assays are required

    • Detection ranges:

      • Baseline mouse serum GDF15: 50-300 pg/mL

      • Transgenic models: 3-50 ng/mL

      • Post-LPS or stress: 1-10 ng/mL

  • Tissue expression analysis:

    • Quantitative PCR:

      • Appropriate for relative expression changes

      • Requires careful reference gene selection (β-actin, GAPDH)

    • Western blotting:

      • Can distinguish between pro-GDF15 and mature GDF15

      • Requires validated antibodies for specific detection

    • Immunohistochemistry:

      • Allows cellular localization of expression

      • Critical antigen retrieval and antibody validation needed

  • Technical validation recommendations:

    • Standard curve range should encompass physiological and pathological concentrations

    • Spike-and-recovery experiments to verify accuracy in biological matrices

    • Multiple dilutions to confirm linearity

    • Inter- and intra-assay coefficient of variation determination

    • Comparison with reference method for select samples

  • Data reporting standards:

    • Clear description of fasting status and collection timing

    • Detailed assay characteristics (sensitivity, specificity, CV%)

    • Raw values alongside normalized data where appropriate

    • Transparent discussion of outliers and handling methods

Implementing these methodological considerations ensures reliable and reproducible GDF15 measurements across different experimental conditions and models, facilitating meaningful comparisons between studies and accurate interpretation of GDF15's biological roles.

What are the key considerations for designing GFRAL knockout validation experiments in GDF15 research?

Designing rigorous GFRAL knockout validation experiments is critical for establishing the receptor-dependence of GDF15 effects:

  • Genetic knockout design considerations:

    • Global vs. conditional knockouts:

      • Global GFRAL knockout eliminates all receptor signaling

      • Conditional (e.g., inducible) knockouts avoid developmental compensations

      • Region-specific deletion can pinpoint critical neuroanatomical sites

    • Knockout verification:

      • Genomic verification via PCR genotyping

      • mRNA expression analysis in hindbrain regions

      • Protein verification by immunohistochemistry or Western blotting

      • Functional validation using GDF15 administration challenges

  • Comprehensive phenotyping parameters:

    • Baseline characterization:

      • Body weight and composition throughout development

      • Food intake patterns under standard conditions

      • Energy expenditure and RER measurements

      • Glucose homeostasis parameters

    • Response to GDF15 administration:

      • Food intake at multiple timepoints post-administration

      • Body weight changes (acute and chronic)

      • Neural activation (cFos) in relevant brain regions

      • Metabolic responses (glucose tolerance, energy expenditure)

  • Control groups and experimental design:

    Control TypePurposeDesign Considerations
    Wild-type littermatesDirect comparison accounting for genetic backgroundUse heterozygous breeding to generate littermates
    Heterozygous miceAssess gene dosage effectsInclude in all experiments as intermediate phenotype
    GDF15 knockoutCompare receptor vs. ligand deletionParallel experiments with identical protocols
    Vehicle-treated knockoutsControl for injection effectsSame volume/vehicle composition as GDF15
  • Validation experiments for GFRAL specificity:

    • Dose-response curves to GDF15 in wild-type vs. knockout mice

    • Testing alternative ligands that might signal through GFRAL

    • Cross-administration studies with other GDNF-family ligands

    • Evaluation of compensatory receptor expression in knockouts

  • Molecular and cellular validation approaches:

    • Ex vivo electrophysiology of NTS/AP neurons from wild-type vs. knockout mice

    • Primary neuron cultures from hindbrain regions

    • RET signaling pathway activation assessment

    • Transcriptomic profiling of hindbrain regions following GDF15 administration

  • Response to physiological challenges:

    • High-fat diet challenge to assess obesity susceptibility

    • LPS administration to evaluate inflammatory anorexia responses

    • Cancer models to assess cachexia development

    • Food restriction to measure compensatory hyperphagia

  • Rescue experiments:

    • Viral re-expression of GFRAL in knockout background

    • Region-specific rescue to map functional domains

    • Dose-dependent rescue to establish minimum receptor threshold

Multiple studies have confirmed that "GFRAL (−/−) mice" are resistant to the effects of administered GDF15 , underscoring the critical importance of properly validated GFRAL knockout models. These validation experiments establish that GFRAL is indeed the obligate receptor for GDF15's metabolic effects, though importantly, some GDF15 responses during inflammation appear to persist in GFRAL knockout mice , suggesting potential alternative signaling mechanisms in specific contexts.

How should conditioned taste aversion experiments be properly designed and interpreted in GDF15 research?

Properly designing and interpreting conditioned taste aversion (CTA) experiments in GDF15 research requires careful methodological considerations:

  • Experimental design fundamentals:

    • Protocol standardization:

      • Two-bottle preference test is standard for CTA assessment

      • Saccharin (0.1-0.2%) as novel tastant due to its distinct flavor profile and negligible caloric value

      • Water as alternative choice to measure relative preference

      • Precise timing between tastant exposure and GDF15 administration

    • Control groups:

      • Vehicle-treated positive controls

      • Lithium chloride (LiCl) as positive control (standard dose: 0.15M, 2% body weight IP)

      • Saline controls for injection stress effects

      • Non-conditioned controls to establish baseline preference

  • Critical experimental parameters:

    ParameterRecommendationRationale
    GDF15 dosingMultiple doses (0.001-0.1 mg/kg) Establish dose-response relationship
    Administration timingImmediately after tastant exposureEnsures temporal association
    Preference testing24-48h post-conditioningAllows sufficient memory consolidation
    Testing durationMinimum 24h with regular monitoringCaptures both initial and sustained aversion
    Pre-exposureFamiliarization with testing apparatusReduces novelty effects on consumption
  • Interpretation framework:

    • Quantitative analysis:

      • Preference ratio calculation: (saccharin consumption)/(total fluid consumption)

      • Consumption normalized to body weight

      • Statistical comparison against vehicle control and positive control

      • Analysis of dose-dependence and alignment with anorexigenic dose-response

    • Qualitative assessment:

      • Observation of orofacial responses during tastant re-exposure

      • Assessment of approach-avoidance behaviors

      • Monitoring for physical signs of malaise

  • Validating CTA versus general anorexia:

    • Cross-substance generalization testing:

      • Test whether aversion transfers to other novel flavors

      • Evaluate whether familiar foods are also avoided

    • Hunger challenge experiments:

      • Test CTA following food deprivation

      • Compare suppression of preferred versus standard chow intake

    • Palatability testing:

      • Brief-access lickometer tests to assess immediate hedonic responses

      • Progressive ratio operant testing to measure motivation

  • Mechanistic investigations:

    • Receptor dependence:

      • Compare CTA in wild-type versus GFRAL knockout mice

      • Test whether CTA correlates with brain regions expressing GFRAL

    • Neural circuit mapping:

      • Assess cFos activation patterns following GDF15 administration

      • Compare to patterns induced by established aversive stimuli (LiCl)

      • Chemogenetic or optogenetic manipulation of candidate pathways

  • Translation to therapeutic implications:

    • Tolerance development assessment:

      • Test whether repeated GDF15 administration leads to CTA extinction

      • Evaluate if intermittent dosing reduces aversive conditioning

    • Separation of therapeutic from aversive effects:

      • Correlation analysis between weight loss efficacy and CTA strength

      • Testing of structural analogs for differential effects

      • Combined administration with anti-emetic agents

The evidence that "GDF15 treatment at 0.01 mg/kg and 0.1 mg/kg also reduced saccharin consumption and increased water consumption compared to vehicle control" suggests a robust CTA effect. This finding has significant implications for therapeutic applications, as it suggests the anorexigenic effects of GDF15 may be partially mediated through aversive mechanisms rather than purely homeostatic appetite regulation.

Product Science Overview

Gene and Protein Structure

GDF-15 is encoded by the GDF15 gene. In mice, this gene is located on chromosome 8 . The protein is initially synthesized as a precursor, which undergoes proteolytic cleavage to produce the mature, biologically active form. The mature GDF-15 protein forms a disulfide-linked homodimer .

Expression and Regulation

Under normal physiological conditions, GDF-15 is expressed at low levels in most tissues. However, its expression is significantly upregulated in response to various stress conditions, such as tissue injury, inflammation, and cancer . In mice, high levels of GDF-15 are observed in the placenta, bone marrow stroma, liver, and kidney .

Biological Functions

GDF-15 plays a multifaceted role in various biological processes:

  1. Regulation of Inflammatory Pathways: GDF-15 has been shown to inhibit the production of tumor necrosis factor-alpha (TNF-α) from lipopolysaccharide-stimulated macrophages .
  2. Apoptosis and Cell Survival: It prevents apoptosis in cerebellar granule neurons by activating the Akt pathway and inhibiting the ERK pathway .
  3. Cell Growth and Differentiation: GDF-15 can promote either proliferation or growth arrest and differentiation, depending on the cellular context .
  4. Metabolic Regulation: GDF-15 is involved in the regulation of food intake and body weight, particularly under conditions of dietary excess .
Clinical Significance

Due to its diverse roles, GDF-15 has been extensively studied as a biomarker for various pathological conditions, including:

  • Cardiovascular Diseases: Elevated levels of GDF-15 are associated with myocardial infarction and heart failure .
  • Cancer: GDF-15 is often upregulated in various cancers and is being explored as a target for cancer immunotherapy .
  • Metabolic Disorders: Its role in regulating appetite and body weight makes it a potential target for treating obesity and related metabolic disorders .
Recombinant GDF-15

Recombinant GDF-15, particularly the mouse variant, is widely used in research to study its biological functions and potential therapeutic applications. The recombinant protein is typically produced in Escherichia coli or mammalian expression systems and purified to ensure biological 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.