ECSIT Human

ECSIT homolog Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to ECSIT Human

Evolutionarily Conserved Signaling Intermediate in Toll Pathway (ECSIT) is a multifunctional protein encoded by the ECSIT gene in humans. It serves as a cytosolic adaptor protein involved in inflammatory signaling, mitochondrial complex I assembly, and embryonic development . The human ECSIT protein (UniProt ID: Q9BQ95) exists in multiple isoforms, including a 49 kDa full-length cytoplasmic form and a 45 kDa mitochondrial variant . Recombinant human ECSIT (rhECSIT) is widely used in research, produced as a 24.6 kDa polypeptide (amino acids 19–217) with an N-terminal His-tag for purification .

Gene and Protein Structure

  • Gene location: Chromosome 19p13.2, spanning 9 exons .

  • Protein domains:

    • N-terminal mitochondrial targeting sequence (residues 1–48) .

    • Pentatricopeptide repeat (PPR) motif (residues 90–266) .

    • Pleckstrin homology (PH) domain (residues 275–380) .

  • Molecular weight:

    • Full-length: 49 kDa (431 amino acids) .

    • Recombinant truncated form: 24.6 kDa (222 amino acids) .

Post-Translational Modifications

  • Lysine 372 ubiquitination is critical for interactions with NF-κB proteins (p65/p50) .

Inflammatory and Immune Signaling

  • TLR/NF-κB pathway: Scaffolds TRAF6 and MAP3K1 to activate NF-κB, essential for innate immunity .

  • Antiviral responses: Bridges RIG-I/MDA5 receptors to VISA, enhancing interferon production .

Mitochondrial Complex I Assembly

  • Stabilizes assembly chaperone NDUFAF1, ensuring proper complex I (NADH:ubiquinone oxidoreductase) formation .

  • Depletion disrupts oxidative phosphorylation, reducing ATP production .

Cardiac Function

  • Key finding: Humanized mice (hECSIT+/+) exhibit age-dependent cardiac hypertrophy, mitochondrial fission, and heart failure due to reduced complex I activity .

  • Clinical correlation: Low ECSIT expression in human hearts correlates with fibrosis and cardiomyopathy .

Humanized Mouse Model

ObservationhECSIT+/+ Mice vs. Wild-Type
Complex I activity↓ 60% (reduced subunit assembly)
ATP production↓ 45%
Mitochondrial dynamicsImpaired fusion, excessive fission
Survival at 12 months<20% vs. >80% (wild-type)

Human Cardiac Disease

  • Study cohort: Patients with low cardiac ECSIT levels showed:

    • ↑ Fibrosis (2.5-fold increase) .

    • ↑ Left ventricular mass index (LVMI) .

Research Tools and Antibodies

  • Antibody validation: Rabbit anti-ECSIT (Proteintech #83295-1-RR) detects 49 kDa bands in HeLa, Jurkat, and A549 cells .

  • Applications: Western blot (1:5,000–1:50,000), IHC (1:50–1:500) .

Pathological and Therapeutic Insights

  • Therapeutic target: Enhancing ECSIT stability could mitigate mitochondrial dysfunction in heart failure .

  • Current limitations: Human ECSIT’s intrinsic lability complicates therapeutic strategies .

Product Specs

Introduction
ECSIT homolog (ECSIT) is a ubiquitously expressed protein that plays a crucial role as an adaptor protein in the cytoplasmic signal transduction cascade initiated by Toll receptor activation. ECSIT facilitates the proteolytic activation of MAP3K1. Moreover, ECSIT participates in the BMP signaling pathway. It is essential for normal embryonic development. Initially categorized as a cytoplasmic protein, ECSIT specifically interacts with TNF receptor-associated factor (TRAF)-6 within the TLR pathway. Notably, ECSIT gene knockdown leads to severely compromised complex I assembly and impaired mitochondrial function.
Description
Recombinant human ECSIT, produced in E. coli, is a single polypeptide chain consisting of 222 amino acids (19-217) with a molecular weight of 24.6 kDa. This protein is fused to a 23 amino acid His-tag at its N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A sterile, colorless solution.
Formulation
The ECSIT solution is prepared in a buffer containing 20mM Tris-HCl (pH 8.0), 0.1M NaCl, 1mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), store at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Exceeds 90% purity as determined by SDS-PAGE analysis.
Synonyms
ECSIT Homolog (Drosophila), Evolutionarily Conserved Signaling Intermediate In Toll Pathway Mitochondrial, Likely Ortholog Of Mouse Signaling Intermediate In Toll Pathway Evolutionarily Conserved, Protein SITPEC.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSGTCGAAL TGTSISQVPL PKDSTGAADP PQPHIVGIQS PDQQAALARH NPARPVFVEG PFSLWLRNKC VYYHILRADL LPPEEREVEE TPEEWNLYYP MQLDLEYVRS GWDNYEFDIN EVEEGPVFAM CMAGAHDQAT MAKWIQGLQE TNPTLAQIPV VFRLAGSTRE LQTSSAGLEE PPLPEDHQEE DDNLQRQQQG QS

Q&A

What is ECSIT and what are its major functions in human cells?

ECSIT (Evolutionarily Conserved Signalling Intermediate in Toll pathway) is a 431 amino acid adapter protein in humans with multiple identifiable isoforms (50/33kDa), with a potential third isoform (24kDa) based on splice prediction . The protein serves dual critical functions: it acts as an essential assembly factor for mitochondrial complex I in the electron transport chain, and it participates in the Toll/IL-1 pathway as part of innate immune signaling . Human ECSIT contains three recognizable domains: an N-terminal mitochondrial targeting sequence (amino acids 1-48), a highly ordered pentatricopeptide repeat region (PPR) (amino acids 90-266), and a less ordered C-terminal domain resembling pleckstrin homology domains (amino acids 275-380) . Through these structural components, ECSIT enables vital cellular processes including energy production and inflammatory response coordination.

How does ECSIT structure relate to its functional roles?

The human ECSIT protein's structure directly informs its dual functionality in mitochondrial and immune pathways. Its N-terminal mitochondrial targeting sequence (amino acids 1-48) directs the protein to mitochondria, supporting its role in complex I assembly of the electron transport chain . The pentatricopeptide repeat region (amino acids 90-266) likely facilitates protein-protein interactions essential for both pathways. In the toll-like receptor pathway, ECSIT's structure enables it to bind specifically to TRAF6 (Tumor Necrosis Factor Receptor Associated Factor 6), which then facilitates the phosphorylation of MEKK1 (MAP3K1) . This structural arrangement allows ECSIT to function as an adapter protein, bridging different signaling components in both mitochondrial assembly and immune response pathways, demonstrating how protein architecture directly determines biological functionality.

What are the key considerations for designing ECSIT knockdown or knockout experiments?

When designing ECSIT knockdown or knockout experiments, researchers must carefully consider several methodological factors. First, the selection between transient (siRNA/shRNA) versus stable (CRISPR-Cas9) approaches should be based on the specific research question—transient methods allow examination of immediate effects while stable modifications enable long-term studies. Second, tissue-specificity is crucial; ECSIT plays different roles across tissues, so researchers should validate knockdown efficiency in the specific cell types under investigation . Third, appropriate controls must be implemented, including non-targeting constructs and rescue experiments to confirm phenotype specificity.

The experimental design should include:

Design ElementConsiderationsImplementation
Control GroupsRandom assignment to eliminate biasUse both wild-type and scramble/non-targeting controls
VariablesClear definition of independent (ECSIT levels) and dependent (phenotypic outcomes) variablesMeasure multiple dependent variables including complex I activity, mitochondrial function, and TLR pathway activity
Hypothesis TestingFocused, testable hypothesisDevelop specific predictions about the effects of ECSIT manipulation
Measurement MethodsReliable quantification techniquesWestern blotting for protein levels, RT-PCR for transcript levels, functional assays for pathway activities

Additionally, researchers should include parallel assessment of both mitochondrial complex I assembly and TLR pathway function to distinguish between ECSIT's dual roles .

How should researchers design experiments to differentiate between ECSIT's roles in TLR signaling versus mitochondrial functions?

To effectively differentiate between ECSIT's roles in TLR signaling and mitochondrial functions, researchers should implement a multi-faceted experimental approach. First, compartment-specific ECSIT variants should be generated—constructs lacking the mitochondrial targeting sequence would preferentially affect TLR signaling, while variants with mutations in the C-terminal domain might predominantly impact mitochondrial function . Second, selective pathway stimulation can be performed; researchers can activate TLR pathways using specific ligands like lipopolysaccharide (LPS) while separately assessing mitochondrial function through respirometry analyses.

A comprehensive experimental design would include:

  • Domain-specific mutations targeting either mitochondrial or TLR pathway functions

  • Subcellular fractionation to determine protein localization changes

  • Parallel assessment of:

    • Mitochondrial parameters: Complex I assembly (BN-PAGE), oxygen consumption rates, mitochondrial membrane potential

    • TLR pathway markers: NF-κB activation, cytokine production, TRAF6 and MEKK1 interactions

The research should employ specialized techniques such as proximity ligation assays to visualize protein-protein interactions in different cellular compartments . Time-course experiments are particularly valuable, as they can reveal whether effects on one pathway precede effects on the other, potentially indicating a hierarchical relationship between ECSIT's dual functions.

What control measures are essential when studying ECSIT mutations in human cell models?

When studying ECSIT mutations in human cell models, implementing rigorous control measures is essential for generating reliable and interpretable data. The experimental design should include multiple layers of controls addressing genetic, cellular, and environmental variables.

Key control measures include:

Control TypePurposeImplementation Strategy
Genetic ControlsEnsure specificity of observed effectsInclude isogenic cell lines differing only in the ECSIT mutation of interest
Expression ControlsAccount for expression level variationsEstablish stable cell lines with comparable ECSIT expression levels
Functional ControlsValidate phenotypic specificityPerform rescue experiments with wild-type ECSIT
Environmental ControlsMinimize experimental variabilityStandardize culture conditions, passage numbers, and experimental timing
Technical ControlsEnsure methodological consistencyInclude standard curves, internal controls for protein loading, and technical replicates

Researchers should additionally implement domain-specific controls, such as mutations affecting only the mitochondrial targeting sequence or only the TRAF6-binding domain, to parse out pathway-specific effects . Statistical analysis should employ appropriate tests based on experimental design, with control for multiple comparisons when assessing complex phenotypes . Finally, researchers should validate key findings using complementary methodologies—for example, confirming protein interaction changes identified by co-immunoprecipitation with alternative techniques like proximity ligation assays.

What methodological approaches best identify the pathogenic pathways connecting ECSIT to hypertrophic cardiomyopathy?

Identifying pathogenic pathways connecting ECSIT to hypertrophic cardiomyopathy (HCM) requires a multi-modal methodological approach that integrates molecular, cellular, and physiological analyses. Research on ECSIT N209I/N209I mouse models has demonstrated development of HCM with signs including vacuolation, mineralization, and myocyte disorganization beginning at 6 weeks of age, with progression to myocyte hypertrophy by 8-12 weeks . To translate these findings to human contexts, researchers should employ:

  • Systems biology approaches integrating:

    • Transcriptomic profiling of cardiac tissue to identify dysregulated gene networks

    • Proteomic analysis to detect altered protein interactions and post-translational modifications

    • Metabolomic assessment to identify energy metabolism perturbations

  • Functional assays examining:

    • Mitochondrial complex I assembly and activity in cardiac tissue samples

    • Calcium handling in cardiomyocytes derived from patient iPSCs

    • Contractile properties of engineered heart tissues with ECSIT mutations

  • Temporal analyses tracking:

    • Disease progression markers at defined intervals (similar to the mouse model time course)

    • Compensatory mechanism activation during early disease stages

    • Structural changes using advanced imaging modalities

The methodological framework should incorporate both hypothesis-testing and hypothesis-generating components, allowing for directed investigation of known pathways (mitochondrial dysfunction, TLR signaling) while remaining open to discovering novel mechanisms . Statistical approaches should include multivariate analyses capable of detecting complex interactions between pathways. Researchers should validate findings across multiple experimental systems, from cell lines to animal models to human samples when available, to establish robust causal relationships.

How can researchers effectively trace the cause-effect relationship between ECSIT mutations and complex I deficiencies?

Tracing cause-effect relationships between ECSIT mutations and complex I deficiencies requires systematic methodologies that establish temporal sequences and mechanistic links. Researchers should implement a multi-level approach that combines genetic manipulation with detailed biochemical and functional analyses.

A comprehensive methodology would include:

  • Genetic confirmation using:

    • Inducible expression systems (e.g., Tet-On/Off) to demonstrate temporal correlation between ECSIT mutation expression and complex I deficiency onset

    • Rescue experiments showing restoration of complex I function upon wild-type ECSIT reintroduction

    • Dose-dependent studies correlating mutation "dosage" with severity of complex I deficiency

  • Assembly pathway analysis through:

    • Time-course studies of complex I assembly intermediates using Blue Native PAGE

    • Pulse-chase experiments tracking the incorporation of newly synthesized subunits

    • Proximity labeling approaches (BioID, APEX) to identify altered interaction partners in the assembly process

  • Functional consequence assessment via:

    • High-resolution respirometry measuring oxygen consumption rates

    • Mitochondrial membrane potential measurements in live cells

    • Reactive oxygen species production quantification

    • ATP synthesis rate determination

Research on ECSIT N209I/N209I mouse models demonstrated a 98% reduction in complex I protein levels in cardiac tissue, establishing a strong correlation between ECSIT mutation and complex I deficiency . To establish causality rather than mere correlation, researchers should employ difference-in-differences approaches to control for confounding variables and use statistical mediation analysis to test whether specific assembly defects mediate the relationship between mutations and functional outcomes. Process tracing and Bayesian updating methodologies can further strengthen causal inferences in complex biological systems .

What are the most effective techniques for investigating tissue-specific ECSIT requirements in humans?

Investigating tissue-specific ECSIT requirements in humans presents unique challenges that necessitate specialized methodological approaches. Since direct genetic manipulation in human subjects is not ethically permissible, researchers must employ alternative strategies that leverage available human samples and model systems.

Effective techniques include:

  • Human tissue analysis approaches:

    • Quantitative immunohistochemistry of post-mortem or biopsy tissues to assess ECSIT expression patterns across tissue types

    • Single-cell RNA sequencing to determine cell-specific expression profiles within heterogeneous tissues

    • Proteomic analysis of tissue-specific ECSIT interaction networks

  • Human cellular models:

    • Tissue-specific differentiation of induced pluripotent stem cells (iPSCs) harboring ECSIT mutations

    • 3D organoid cultures to recapitulate tissue-specific microenvironments

    • Co-culture systems to examine cell-cell interactions mediated by ECSIT

  • Comparative methodologies:

    • Analysis of tissue-specific phenotypes in patients with naturally occurring ECSIT variants

    • Parallel assessment of multiple tissues from the same donor to control for genetic background

    • Cross-species comparative studies to identify conserved tissue-specific requirements

The research approach should implement rigorous experimental design principles with appropriate control groups and randomization where possible . Statistical analysis should account for inter-individual variability in human samples through appropriate mixed-effects modeling. Researchers should consider employing public value mapping frameworks to assess the broader implications of tissue-specific findings . Additionally, integration of data from multiple methodological approaches through systematic review techniques such as meta-narrative synthesis can strengthen the evidence base for tissue-specific ECSIT requirements .

What statistical methods are most appropriate for analyzing ECSIT expression data across different human tissues?

Analyzing ECSIT expression data across different human tissues requires sophisticated statistical approaches that account for biological variability, tissue heterogeneity, and potential confounding factors. When examining tissue-specific expression patterns, researchers should implement a multi-layered statistical framework.

Recommended statistical approaches include:

  • Descriptive statistics:

    • Normalization methods appropriate for the specific data type (microarray, RNA-seq, protein quantification)

    • Visualization techniques including heatmaps, violin plots, and principal component analysis to identify patterns

  • Inferential statistics:

    • Mixed-effects models to account for within-subject correlations when multiple tissues come from the same donors

    • ANOVA or non-parametric alternatives (Kruskal-Wallis) with appropriate post-hoc tests for multi-tissue comparisons

    • Careful consideration of multiple testing correction (e.g., Benjamini-Hochberg FDR) to control false discovery rates

  • Advanced analytical approaches:

    • Network analysis to identify tissue-specific co-expression partners

    • Machine learning methods (e.g., random forests) to identify tissue-specific patterns

    • Bayesian hierarchical models to integrate prior biological knowledge with observed data

When comparing tissues that might have different baseline characteristics, difference-in-differences approaches can be particularly valuable . For longitudinal expression studies, time-series analyses should be employed to characterize expression dynamics. Publication bias should be considered when integrating data from multiple studies; researchers should consider methods like funnel plot analysis to detect potential reporting biases . Statistical power calculations should be performed a priori to ensure adequate sample sizes for detecting biologically meaningful differences across tissues.

How should researchers approach conflicting results between mitochondrial and immunological phenotypes in ECSIT studies?

When confronting conflicting results between mitochondrial and immunological phenotypes in ECSIT studies, researchers should implement a systematic approach to resolve these apparent contradictions. Such conflicts often reflect the biological complexity of ECSIT's dual functionality rather than experimental error.

A methodical approach to conflicting results includes:

  • Comprehensive experimental validation:

    • Replicate experiments using alternative methodologies to confirm observations

    • Vary experimental conditions to identify context-dependent effects

    • Employ multiple cell lines or tissue types to determine generalizability

  • Mechanistic integration analysis:

    • Perform time-course studies to establish temporal relationships between phenotypes

    • Utilize pathway inhibitors to dissect potential crosstalk between mitochondrial and immune functions

    • Conduct epistasis experiments with other pathway components to map hierarchical relationships

  • Contextual interpretation frameworks:

    • Consider cell type-specific requirements for ECSIT function

    • Evaluate energy demand differences across experimental conditions

    • Assess the activation state of both pathways in experimental systems

When analyzing conflicting results, researchers should implement critical interpretive synthesis methods that allow integration of diverse data types and experimental approaches . Qualitative comparative analysis can help identify necessary and sufficient conditions for specific phenotypes. Research on ECSIT has demonstrated that its roles in mitochondrial complex I assembly and TLR signaling can be experimentally distinguished , suggesting that experimental design factors may contribute to apparent conflicts. Researchers should apply systematic review techniques like meta-narrative approaches to place conflicting results within broader theoretical frameworks .

What frameworks exist for integrating multi-omics data in ECSIT functional studies?

Integrating multi-omics data in ECSIT functional studies requires sophisticated computational frameworks that can synthesize diverse molecular information into cohesive biological insights. As ECSIT operates at the intersection of multiple pathways, multi-omics approaches are particularly valuable for comprehensively characterizing its functions.

Effective integration frameworks include:

  • Data-driven integration approaches:

    • Joint dimension reduction methods (e.g., multi-omics factor analysis)

    • Network-based integration (e.g., similarity network fusion)

    • Matrix factorization techniques for identifying latent patterns across data types

  • Knowledge-based integration strategies:

    • Pathway enrichment analyses across multiple omics layers

    • Causal reasoning frameworks incorporating prior knowledge

    • Biological process-centered integration focusing on ECSIT-related functions

  • Advanced computational methodologies:

    • Machine learning models trained on multi-omics signatures

    • Bayesian integrative models incorporating uncertainty

    • Systems biology approaches using ordinary differential equations to model dynamic processes

Implementation should follow a structured process with quality control at each stage. Researchers should first independently analyze each omics dataset, then perform pairwise integrations before attempting full multi-omics synthesis. Statistical approaches should account for different noise characteristics and dynamic ranges across data types. Visualization techniques like Sankey diagrams or multi-layer network visualizations can help communicate complex relationships .

When applying these frameworks specifically to ECSIT studies, researchers should focus on integrating data that illuminates both mitochondrial and immunological functions . Process tracing methodologies can help establish causal relationships across different omics layers . Researchers should also consider implementing storyline approaches to multi-omics interpretation, which can facilitate communication of complex findings to diverse stakeholders .

How can researchers effectively measure the clinical impact of ECSIT-related discoveries?

Measuring the clinical impact of ECSIT-related discoveries requires a comprehensive evaluation framework that tracks progress from basic science findings to patient outcomes. Given ECSIT's emerging role in conditions like hypertrophic cardiomyopathy , evaluating clinical impact involves both immediate translational outcomes and long-term health benefits.

A structured approach for impact assessment includes:

  • Translational metrics tracking:

    • Development of diagnostic biomarkers based on ECSIT pathway dysregulation

    • Progression of therapeutic candidates targeting ECSIT or downstream effectors

    • Clinical trial initiation and progression milestones

  • Healthcare implementation measures:

    • Adoption of ECSIT-based diagnostic approaches in clinical practice

    • Changes in clinical guidelines incorporating ECSIT-related mechanistic insights

    • Diffusion of knowledge among clinicians treating related conditions

  • Patient outcome evaluations:

    • Disease-specific quality of life improvements

    • Changes in disease progression rates following intervention

    • Economic impact analyses including cost-effectiveness of new interventions

Researchers should implement methodological frameworks such as the Payback Framework to systematically evaluate multiple impact dimensions . Time-lag analysis is particularly important, as translation of basic findings to clinical impact typically spans 17 years on average . Process evaluation methodologies can help identify barriers and facilitators in the translational pathway. Public Value Mapping can complement traditional impact metrics by assessing how ECSIT research contributes to broader societal values beyond economic returns .

Impact assessment should incorporate both quantitative metrics (citation analysis, patenting activity) and qualitative approaches (stakeholder interviews, case studies). Researchers should recognize that impact pathways are rarely linear and often involve productive interactions among multiple stakeholders .

What methodological approaches are most appropriate for evaluating the translational potential of ECSIT research?

Evaluating the translational potential of ECSIT research requires methodological approaches that can assess both scientific merit and practical applicability in clinical contexts. As ECSIT research spans basic molecular mechanisms to potential therapeutic applications in conditions like hypertrophic cardiomyopathy , comprehensive evaluation frameworks are essential.

Appropriate methodological approaches include:

  • Systematic evidence assessment:

    • Rapid realist review to identify contexts, mechanisms, and outcomes in existing literature

    • Cross-discipline evidence principles to evaluate credibility across research disciplines

    • Gap analysis to identify missing translational elements

  • Stakeholder engagement methods:

    • Participatory Impact Pathways Analysis to map potential routes to impact

    • Productive interactions mapping to identify key relationships for translation

    • Patient and clinician involvement in research priority setting

  • Translational readiness evaluation:

    • Technology Readiness Level (TRL) assessment adapted for biological discoveries

    • Assessment of intellectual property landscape and commercialization potential

    • Regulatory pathway analysis for diagnostic or therapeutic applications

Implementation should follow a structured process beginning with comprehensive literature synthesis using systematic review methodologies . Researchers should employ impact evaluation methodologies such as ASIRPA (a comprehensive theory-based approach to assessing the societal impacts of a research organization) to map potential pathways from discovery to application. Difference-in-differences approaches can help isolate the contribution of specific ECSIT findings to broader translational progress .

When evaluating translational potential specifically for ECSIT research, investigators should consider the dual pathway involvement (mitochondrial and immunological) as potentially offering multiple intervention points. The assessment should acknowledge the time required for translation, which averages 17 years from discovery to implementation , and implement creative, participatory methods for stakeholder engagement in evaluation processes .

How should researchers design longitudinal studies to assess long-term implications of ECSIT variations?

Designing longitudinal studies to assess long-term implications of ECSIT variations requires careful methodological planning to capture meaningful temporal patterns while maintaining study feasibility. Given ECSIT's role in fundamental cellular processes and its implications for conditions like hypertrophic cardiomyopathy , long-term studies are essential for understanding disease progression and intervention effects.

Key methodological considerations include:

  • Cohort design elements:

    • Prospective recruitment strategies targeting populations with ECSIT variations

    • Sample size calculations accounting for anticipated attrition over extended timeframes

    • Nested case-control components for efficient resource utilization

  • Temporal assessment frameworks:

    • Interval determination based on expected progression rates of relevant phenotypes

    • Age-stratified analysis plans to distinguish developmental from progressive effects

    • Event-triggered additional assessments to capture critical transitions

  • Methodological sustainability approaches:

    • Biobanking protocols for future analysis with emerging technologies

    • Remote assessment capabilities to maintain participant engagement

    • Flexible protocol design allowing for technology evolution without compromising data continuity

The longitudinal design should implement true experimental design principles where ethical and practical , or quasi-experimental approaches with appropriate controls when randomization is not possible. Difference-in-differences methodologies can be particularly valuable for isolating ECSIT-specific effects from general temporal trends . Statistical approaches should include mixed-effects modeling to account for repeated measures and missing data handling strategies appropriate for longitudinal designs.

Mouse model studies of ECSIT N209I/N209I mutations have demonstrated phenotype progression from 6 weeks to 12 weeks , which can inform human study timeframes while accounting for species differences. Researchers should consider implementing ideal-type impact pathways and creativity-based research methods for long-term engagement with participants . Additionally, researchers should establish data and biospecimen collection protocols that enable future analysis with emerging technologies, maximizing the long-term value of the longitudinal cohort.

What emerging technologies show promise for advancing ECSIT functional studies in human tissues?

Several cutting-edge technologies are poised to revolutionize ECSIT functional studies in human tissues, offering unprecedented resolution and manipulation capabilities. These emerging approaches promise to overcome current limitations in studying tissue-specific ECSIT requirements and pathway interactions.

Promising emerging technologies include:

  • Advanced genetic engineering approaches:

    • Base editing and prime editing for precise ECSIT mutations without double-strand breaks

    • Inducible CRISPR interference/activation systems for temporal control of ECSIT expression

    • RNA editing technologies for transient manipulation of ECSIT transcripts

  • Advanced imaging and structural analysis:

    • Cryo-electron microscopy for high-resolution ECSIT complex structures

    • Live-cell super-resolution microscopy tracking ECSIT dynamics

    • Spatial transcriptomics/proteomics to map ECSIT pathway components in tissue contexts

  • Multicellular human model systems:

    • Organ-on-chip technologies incorporating multiple cell types relevant to ECSIT function

    • Patient-derived organoids for disease modeling

    • Engineered 3D tissue constructs with controlled microenvironments

The implementation of these technologies should follow systematic validation processes, with careful comparison to established methodologies . Researchers should consider implementing arts-based research methods for creative exploration of complex ECSIT pathway dynamics . Analytical frameworks should incorporate recent advances in Google Scholar and database coverage to ensure comprehensive literature incorporation .

For ECSIT-specific applications, technologies enabling simultaneous monitoring of both mitochondrial function and immune pathway activation would be particularly valuable given ECSIT's dual functionality . Researchers should consider implementing ideal-type impact pathways to map how these technologies might accelerate translation of ECSIT findings to clinical applications . Statistical approaches for analyzing data from these emerging technologies should account for their unique characteristics, including potential biases and error structures.

How might interdisciplinary collaboration enhance methodological approaches to ECSIT research?

Interdisciplinary collaboration offers transformative potential for enhancing methodological approaches to ECSIT research by integrating diverse expertise, technologies, and analytical frameworks. Given ECSIT's complex role at the intersection of mitochondrial function and immune signaling , collaborative approaches are particularly valuable for comprehensive characterization.

Key interdisciplinary collaboration opportunities include:

  • Cross-disciplinary methodology integration:

    • Computational biology approaches for modeling ECSIT pathway dynamics

    • Systems biology frameworks for integrating diverse experimental data

    • Bioengineering techniques for creating advanced model systems

  • Clinical-basic science partnerships:

    • Translational collaborations linking molecular mechanisms to clinical phenotypes

    • Biomarker development bridging laboratory and clinical assessments

    • Patient stratification approaches based on ECSIT pathway characteristics

  • Methodological innovation through discipline convergence:

    • Data science approaches for analyzing complex multi-dimensional datasets

    • Network science methods for mapping ECSIT interaction landscapes

    • Implementation science frameworks for translating ECSIT findings to practice

Effective implementation requires structured collaboration models with clear communication channels and shared conceptual frameworks. Researchers should consider employing guide frameworks for non-specialists to understand methods from other disciplines . Participatory research methods can enhance stakeholder engagement across disciplinary boundaries .

For ECSIT-specific interdisciplinary approaches, collaborations between mitochondrial biologists and immunologists are particularly valuable given ECSIT's dual functionality . Statistical methods should be selected to accommodate the diverse data types generated through interdisciplinary work, potentially employing meta-analytic techniques for data integration . Researchers should implement process evaluation methodologies to continuously refine collaborative approaches and identify barriers to effective interdisciplinary work .

What are the key methodological challenges in translating ECSIT findings from model systems to human applications?

Translating ECSIT findings from model systems to human applications presents several methodological challenges that require systematic approaches to overcome. While model systems like the ECSIT N209I/N209I mouse have provided valuable insights into phenotypes such as hypertrophic cardiomyopathy , bridging the species gap involves complex methodological considerations.

Key challenges and approaches include:

  • Species differences assessment:

    • Systematic comparison of ECSIT sequence, structure, and interaction partners across species

    • Cross-species functional assays to identify conserved versus divergent mechanisms

    • Evolutionary analysis to contextualize functional differences

  • Human model system development:

    • iPSC-derived cell types expressing human ECSIT variants

    • Humanized animal models incorporating human ECSIT

    • Validation frameworks for assessing physiological relevance of engineered systems

  • Translational framework implementation:

    • Biomarker identification connecting model findings to human disease indicators

    • Intervention testing pipelines with appropriate translational endpoints

    • Systematic approaches for evaluating therapeutic candidate relevance to human pathology

Implementation requires careful experimental design with appropriate control groups that account for species-specific differences . Researchers should consider employing multiple model systems in parallel to triangulate findings. Statistical approaches should include formal methods for evaluating concordance between animal and human data, potentially employing Bayesian frameworks to incorporate varying levels of evidence quality .

For ECSIT-specific translation, particular attention should be paid to potential differences in tissue-specific expression patterns between model organisms and humans . Methodological frameworks such as the Payback Framework can help prioritize translational efforts based on potential impact . Researchers should implement narrative approaches for communicating complex translational findings to diverse stakeholders, potentially employing arts-based methods for enhancing engagement .

Product Science Overview

Structure and Expression

The ECSIT protein is encoded by the ECSIT gene located on chromosome 19 in humans. The recombinant form of ECSIT is typically expressed in Escherichia coli (E. coli) and is often tagged with a polyhistidine (His) tag to facilitate purification. The recombinant human ECSIT protein consists of 201 amino acids and has a predicted molecular mass of approximately 22.9 kDa .

Function and Significance

ECSIT is an adapter protein involved in the Toll-like receptor (TLR) and interleukin-1 receptor (IL-1R) signaling pathways. These pathways are essential for the activation of NF-kappa-B (NF-κB), a transcription factor that regulates the expression of genes involved in immune and inflammatory responses . ECSIT specifically interacts with TRAF6 (TNF receptor-associated factor 6) and MEKK-1 (MAP3K1), facilitating the activation of NF-κB .

In addition to its role in immune signaling, ECSIT is also involved in mitochondrial function. It is required for the efficient assembly of mitochondrial NADH:ubiquinone oxidoreductase (complex I), which is a critical component of the mitochondrial respiratory chain . Knockdown of ECSIT results in impaired complex I assembly and disturbed mitochondrial function .

Applications and Research

Recombinant human ECSIT protein is widely used in research to study its role in various cellular processes. It is particularly valuable in investigating the mechanisms of immune response and mitochondrial function. The protein is also used in the development of therapeutic strategies targeting immune and inflammatory diseases.

Stability and Storage

Recombinant human ECSIT protein is typically provided as a lyophilized powder and is stable for up to twelve months when stored at -20°C to -80°C under sterile conditions. It is recommended to aliquot the protein to avoid repeated freeze-thaw cycles .

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.