Pressure Overload-Induced Heart Failure:
Mydgf−/− mice exhibited exacerbated left ventricular (LV) hypertrophy, reduced capillary density, and interstitial fibrosis compared to wild-type (WT) mice after transverse aortic constriction (TAC) .
Mechanistically, MYDGF enhances SERCA2a (sarco/endoplasmic reticulum Ca²⁺-ATPase) expression and PIM1 kinase activity, improving Ca²⁺ cycling and sarcomere function .
Myocardial Infarction (MI):
Bone Loss Models:
Endothelial Injury:
Apical Resection Model:
4. Mechanistic Insights: Signaling Pathways and Molecular Targets
MYDGF exerts its effects through distinct pathways depending on tissue context:
Recombinant MYDGF: Subcutaneous administration improved survival and LV function in pressure-overloaded mice .
Gene Therapy: Adenoviral SERCA2a delivery rescued cardiac function in Mydgf−/− mice, suggesting combinatorial therapeutic potential .
Bone Marrow Targeting: AAV-MYDGF delivery to bone marrow reversed osteoporosis and atherosclerosis, highlighting its endocrine/paracrine role .
D17Wsu104e, Il25, Ly6elg, MYDGF, Interleukin-25, IL-25, Stromal cell-derived growth factor SF20.
MGSSHHHHHH SSGLVPRGSH MGSVSEPTTV PFDVRPGGVV HSFSQDVGPG NKFTCTFTYA SQGGTNEQWQ MSLGTSEDSQ HFTCTIWRPQ GKSYLYFTQF KAELRGAEIE YAMAYSKAAF ERESDVPLKS EEFEVTKTAV SHRPGAFKAE LSKLVIVAKA ARSEL.
To accurately characterize MYDGF expression sources, researchers should consider employing multiple detection methods, including single-cell RNA sequencing approaches to identify specific cellular contributions to MYDGF production under various physiological and pathological conditions.
Several important genetic mouse models have been developed for MYDGF research:
Global MYDGF knockout (MYDGF-KO) mice - These mice have complete deletion of the MYDGF gene throughout all tissues, allowing for assessment of systemic effects of MYDGF deficiency .
MYDGF-EGFP reporter mice - These transgenic mice express enhanced green fluorescent protein under the control of the MYDGF promoter, enabling visualization of MYDGF expression patterns in various tissues .
Myeloid cell-specific MYDGF knockout mice - Generated by breeding MYDGF-floxed mice with LysMCre+ mice, these models allow for specific deletion of MYDGF in myeloid cells, particularly useful for investigating the contribution of myeloid-derived MYDGF in disease models .
Double knockout models - Researchers have created MYDGF and ApoE double knockout mice (DKO) to study the role of MYDGF in atherosclerosis .
When selecting an appropriate mouse model, researchers should consider the specific research question and design experiments that account for potential compensatory mechanisms in knockout models. Background strain differences can significantly affect phenotypes, so proper controls and backcrossing strategies are essential for robust results.
MYDGF expression undergoes significant dynamic changes following cardiac injury in mouse models. In neonatal mouse hearts, MYDGF expression is significantly induced after cardiac injury such as apical resection . This upregulation appears to be a protective response, as MYDGF deficiency impedes neonatal heart regeneration and injury-induced cardiomyocyte proliferation .
In adult mice with myocardial infarction (MI), the expression patterns of MYDGF also change, but the cellular sources may differ from neonatal hearts. For studying these expression changes, researchers should consider:
Temporal analysis: Examine MYDGF expression at multiple timepoints (6h, 24h, 72h, 7d, 14d, 28d) post-injury to capture the complete dynamics.
Spatial analysis: Use immunohistochemistry to map MYDGF expression across different regions of the heart (infarct zone, border zone, remote myocardium).
Cell-type specific analysis: Employ flow cytometry and single-cell approaches to identify which cell populations contribute to MYDGF production at different phases of cardiac repair.
Quantitative measurements of MYDGF protein can be performed using ELISA or targeted LC-MS assays on plasma samples from mice before and after cardiac injury .
MYDGF employs multiple molecular pathways to protect against endothelial injury in mice. The primary protective mechanisms include:
Inhibition of NF-κB signaling: MYDGF has been demonstrated to inhibit endothelial NF-κB signaling, evidenced by decreased endothelial inflammation and adhesion responses, reduced leukocyte homing, and decreased endothelial expression of phosphorylated IκBα and nuclear phosphorylated p65 .
Regulation of MAP4K4/NF-κB pathway: Research has shown that endothelial MAP4K4 is involved in the action of MYDGF on NF-κB signaling. When MAP4K4 was specifically knocked down in endothelial cells, the activation of NF-κB signaling was attenuated. MYDGF did not affect other signaling proteins including p38MAPK, ERK, JNK, and IKKβ .
PKCθ involvement: MYDGF regulates the phosphorylation of MAP4K4 in mouse aortic endothelial cells through PKCθ, revealing a PKCθ-MAP4K4-NF-κB signaling axis .
Anti-apoptotic effects: Recombinant MYDGF treatment attenuates endothelial apoptosis by decreasing apoptotic proteins (cleaved caspase-3 and bax) and increasing anti-apoptotic protein (bcl-2) expression .
Reduction of endothelial permeability: MYDGF treatment decreases endothelial permeability, which is crucial for maintaining vascular integrity .
For investigating these mechanisms, researchers should employ:
Phosphorylation-specific antibodies to monitor signaling pathway activation
Gene silencing approaches (siRNA, CRISPR/Cas9) to validate specific pathway components
Recombinant MYDGF protein at physiologically relevant concentrations (50 ng/ml has been shown to be optimal for in vitro studies)
In vitro models of endothelial stress, such as palmitic acid-induced inflammation and injury
MYDGF plays distinct roles in cardiac regeneration depending on the developmental stage:
In neonatal mice:
MYDGF deficiency impedes neonatal heart regeneration and injury-induced cardiomyocyte proliferation .
MYDGF predominantly originates from endothelial cells rather than macrophages in injured neonatal hearts .
MYDGF promotes cardiomyocyte proliferation by activating the c-Myc/FoxM1 pathway .
In adult mice:
MYDGF protects against cardiac injury following myocardial infarction .
Recombinant MYDGF protein can be used as a therapeutic approach for cardiac repair post MI .
The regenerative capacity appears more limited compared to neonatal mice, likely due to differences in cardiomyocyte cell cycle activity.
Methodological approaches to study these differences include:
Using 5-ethynyl-2'-deoxyuridine (EdU) incorporation assays to quantify cardiomyocyte proliferation
Immunofluorescence staining for proliferation markers (Ki67, phospho-histone H3) combined with cardiomyocyte markers (cardiac troponin T)
Cardiac-specific transcriptome analysis to identify age-dependent differences in MYDGF-responsive genes
Echocardiography to assess functional recovery in different age groups
Researchers should carefully control for age-dependent differences in baseline cardiomyocyte proliferation rates and regenerative capacity when comparing MYDGF effects between neonatal and adult mice.
MYDGF demonstrates significant atheroprotective properties in mouse models through multiple mechanisms:
MYDGF levels are decreased in atherosclerotic patients and mice, indicating a potential protective role. Plasma MYDGF is positively associated with vascular endothelium-dependent dilation in both patients and mice with atherosclerosis .
Myeloid cell-specific MYDGF deletion is associated with endothelial injury and inflammation in mice. This deletion itself is sufficient to induce endothelial injury and inflammation even under normal chow diet conditions .
MYDGF inhibits endothelial inflammation and adhesion responses, resulting in:
MYDGF improves plaque composition and stability:
MYDGF also improves metabolic parameters, including insulin resistance, lipid profiles, and decreased body weight gain, which indirectly contributes to its anti-atherosclerotic effects .
To study MYDGF in atherosclerosis, researchers should:
Use appropriate atherosclerotic mouse models (ApoE knockout, LDLR knockout)
Consider both prevention and intervention experimental designs
Employ comprehensive atherosclerosis assessment including lesion quantification, plaque composition analysis, and inflammatory marker profiling
Assess endothelial function using techniques such as wire myography or flow-mediated dilation measurements
Optimal methodologies for studying MYDGF function include:
For protein detection and quantification:
For tracking MYDGF distribution:
For functional studies:
Bone marrow transplantation to determine the contribution of bone marrow-derived MYDGF
AAV-mediated overexpression of MYDGF for therapeutic applications
Recombinant MYDGF protein administration (optimum concentration for in vitro studies: 50 ng/ml)
Coculture experiments with bone marrow cells and target tissues to study paracrine effects
For genetic manipulation:
For phenotyping:
When designing experiments, researchers should carefully consider the tissue-specific expression of MYDGF and select methods that best capture its function in the context of interest.
Designing rigorous comparative studies between systemic and tissue-specific MYDGF deficiency requires careful consideration of several factors:
Mouse model selection:
For systemic deficiency: Use global MYDGF knockout (MYDGF-KO) mice
For tissue-specific deficiency: Use conditional knockouts such as myeloid cell-specific MYDGF knockout mice (generated by breeding MYDGF-floxed mice with LysMCre+ mice)
For endothelial-specific studies: Consider generating endothelial-specific knockouts using VE-cadherin Cre transgenic mice crossed with MYDGF-floxed mice
Control selection:
Use littermate controls whenever possible to minimize genetic background variations
Include Cre-only controls to account for potential Cre toxicity effects
Consider including heterozygous mice to assess gene dosage effects
Experimental design:
Conduct parallel experiments with both models under identical conditions
Include time-course analyses to capture developmental or progressive effects
Use sufficient sample sizes based on power calculations (typically n=8-12 per group for most cardiovascular phenotyping)
Phenotyping approach:
Perform comprehensive phenotyping across multiple organ systems
Include both baseline and stress conditions (e.g., Western diet, cardiac injury models)
Assess compensatory mechanisms that may be upregulated in different knockout models
Data analysis considerations:
Use appropriate statistical methods for multiple comparisons
Consider interactions between genotype and environmental factors
Account for sex-based differences in phenotypes
This approach allows researchers to differentiate between direct effects of MYDGF deficiency in specific tissues versus secondary effects resulting from systemic absence of the protein.
When using recombinant MYDGF protein for experimental studies, researchers should consider:
For in vitro studies:
For in vivo studies:
Delivery method: Consider different administration routes (intravenous, intraperitoneal, local injection) depending on the target tissue.
Dosing regimen: Establish appropriate dosing based on pharmacokinetic studies. Previous studies have used IRB-NHS-MYDGF at 10 mg/kg body weight via tail vein injection .
Tracking: Consider labeling strategies such as IRB-NHS labeling for tracking MYDGF distribution in tissues .
Stability and half-life: Determine the half-life of recombinant MYDGF in circulation to establish appropriate dosing intervals.
Physiological relevance: Compare administered doses to endogenous levels measured in healthy and disease states.
For both applications:
Source validation: Confirm biological activity of the recombinant protein through functional assays.
Batch consistency: Use the same batch for an entire experiment to minimize variability.
Storage considerations: Follow manufacturer recommendations for reconstitution, aliquoting, and storage to maintain protein activity.
To effectively measure MYDGF-induced changes in inflammatory responses, researchers should employ a multi-faceted approach:
Systemic inflammation assessment:
Tissue-specific inflammation analysis:
Endothelial inflammation evaluation:
Molecular signaling pathways:
Functional assays:
Perform leukocyte adhesion assays to quantify the functional impact of MYDGF on inflammatory cell recruitment
Use thioglycolate-stimulated peritoneal exudate cells from GFP-expressing mice injected into recipient mice to assess leukocyte homing in vivo
Measure macrophage migration and inflammatory responses in response to stimuli like palmitic acid
When designing these experiments, researchers should include appropriate timing for measurements (acute vs. chronic responses) and consider both prevention and intervention paradigms to fully characterize MYDGF's effects on inflammation.
Common pitfalls in MYDGF mouse studies and their solutions include:
Antibody specificity issues:
Pitfall: Limited commercial antibody quality for MYDGF detection
Solution: Validate antibodies using MYDGF knockout tissues as negative controls; consider using epitope-tagged MYDGF for improved detection
Developmental compensation in global knockout models:
Pitfall: Global MYDGF knockout mice may develop compensatory mechanisms that mask phenotypes
Solution: Use inducible knockout systems or acute knockdown approaches (e.g., siRNA, antisense oligonucleotides) to avoid developmental compensation
Background strain effects:
Pitfall: Different mouse strains may show variable responses to MYDGF manipulation
Solution: Backcross knockout lines to a common genetic background (at least 10 generations); always use littermate controls
Inconsistent injury models:
Pitfall: Variability in cardiac injury models (MI, apical resection) can lead to inconsistent results
Solution: Standardize surgical procedures, use experienced surgeons, and perform sham operations as controls
Detection sensitivity for circulating MYDGF:
Cell type-specific effects:
Pitfall: MYDGF effects may vary by cell type, leading to contradictory results
Solution: Perform cell type-specific studies using isolated primary cells and conditional knockout models
Recombinant protein quality:
Pitfall: Variable quality of recombinant MYDGF can affect experimental outcomes
Solution: Rigorously test each batch for biological activity; consider producing in-house protein with consistent protocols
Timing of interventions:
Pitfall: Inappropriate timing of MYDGF administration may miss therapeutic windows
Solution: Conduct time-course studies to identify optimal intervention points in disease models
By anticipating these common pitfalls, researchers can design more robust studies and generate more reliable data on MYDGF functions in mouse models.
Reconciling contradictory findings about MYDGF cellular sources requires systematic investigation and careful experimental design:
Use complementary detection methods:
Combine RNA-based approaches (in situ hybridization, single-cell RNA-seq) with protein detection methods (immunohistochemistry, flow cytometry)
Verify findings with multiple antibodies targeting different epitopes of MYDGF
Employ lineage tracing strategies:
Consider context-dependency:
Systematically compare MYDGF expression across different:
Developmental stages (embryonic, neonatal, adult)
Disease states (baseline, acute injury, chronic disease)
Tissue microenvironments (normoxic vs. hypoxic)
Perform conditional deletion studies:
Delete MYDGF in specific cell populations (myeloid cells, endothelial cells, cardiomyocytes)
Assess the impact on MYDGF levels in different tissues to determine relative contributions
Use bone marrow chimeras:
Apply single-cell resolution techniques:
Use single-cell RNA sequencing to comprehensively map MYDGF expression across all cell types
Perform multiplexed immunofluorescence to simultaneously identify multiple cell markers alongside MYDGF
Consider technical factors:
Tissue processing methods may affect antigen preservation
Antibody sensitivity may vary between applications
Different promoter constructs in reporter mice may have varying fidelity to endogenous expression
An integrated approach that acknowledges tissue-specific and context-dependent expression patterns will help resolve apparent contradictions. For example, the finding that MYDGF is predominantly expressed by endothelial cells in injured neonatal hearts contrasts with its characterization as a myeloid-derived factor, highlighting the importance of context in MYDGF biology.
MYDGF shows considerable therapeutic potential in mouse models of cardiovascular disease, with several promising applications:
Myocardial infarction therapy:
Recombinant MYDGF protein injection has been shown to improve heart regeneration post-MI in adult mice
MYDGF promotes cardiomyocyte proliferation through activation of the c-Myc/FoxM1 pathway, suggesting it could enhance cardiac repair
Future studies should focus on optimizing delivery methods, timing, and dosing regimens for maximal therapeutic effect
Atherosclerosis treatment:
MYDGF inhibits endothelial inflammation and adhesion responses, blunts leukocyte homing, and protects against endothelial injury
Bone marrow-specific overexpression of MYDGF attenuates atherosclerosis in mouse models
Developing targeted MYDGF delivery to atherosclerotic plaques could offer new therapeutic approaches
Metabolic disease management:
MYDGF has been shown to improve insulin resistance, lipid profiles, and decrease body weight gain
These metabolic improvements indirectly contribute to cardiovascular protection
Investigating MYDGF's effects in mouse models of metabolic syndrome and type 2 diabetes represents an important future direction
Anti-inflammatory therapy:
Gene therapy approaches:
For translational relevance, researchers should focus on:
Developing stable formulations of recombinant MYDGF with extended half-life
Establishing pharmacokinetic and pharmacodynamic profiles in larger animal models
Identifying patient populations most likely to benefit from MYDGF-based therapies
Investigating potential synergies with standard-of-care treatments
Despite significant advances in MYDGF research, several important questions remain unanswered:
Receptor identification and signaling:
The specific receptor(s) that mediate MYDGF effects have not been identified
The complete signaling cascade downstream of MYDGF binding remains incompletely characterized
Understanding receptor distribution across tissues could explain tissue-specific effects
Developmental roles:
The role of MYDGF during embryonic and postnatal development is poorly understood
Whether MYDGF influences stem cell behavior and differentiation requires investigation
Potential compensatory mechanisms in MYDGF knockout mice need further exploration
Organ-specific functions:
While cardiac and vascular effects are well-studied, MYDGF's roles in other organs remain largely unknown
The function of MYDGF in the brain, liver, kidney, and other tissues warrants investigation
Organ-specific knockout models could help address these questions
Aging-related changes:
How MYDGF expression and function change with aging is not well-characterized
Whether MYDGF supplementation could mitigate age-related cardiovascular decline represents an important question
Comparative studies between young and aged mice would provide valuable insights
Sex-specific differences:
Most studies have not adequately addressed potential sex differences in MYDGF biology
Hormonal influences on MYDGF expression and function need exploration
Sex-stratified analyses in future studies would address this knowledge gap
Crosstalk with other growth factors:
How MYDGF interacts with other growth factors and cytokines remains largely unknown
Potential synergistic or antagonistic relationships could influence therapeutic applications
Combination studies with established growth factors would provide valuable insights
Translational aspects:
The translational relevance of mouse findings to human cardiovascular disease requires validation
Whether genetic variants in human MYDGF affect cardiovascular risk has not been thoroughly investigated
Human studies correlating MYDGF levels with cardiovascular outcomes would help bridge this gap
Addressing these questions will provide a more comprehensive understanding of MYDGF biology and accelerate the development of MYDGF-based therapeutic strategies.
MYDGF was first identified in bone marrow-derived monocytes and macrophages, where it was found to have cardioprotective and tissue-repairing properties, particularly after myocardial infarction . Due to its potent cardiomyocyte-protective and angiogenic activity, it was formally named Myeloid-Derived Growth Factor (MYDGF) .
MYDGF has been implicated in several diseases, including: