FAIM Human

Fas Apoptotic Inhibitory Molecule Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to FAIM Human

FAIM Human (Fas Apoptotic Inhibitory Molecule) is a conserved, intracellular protein that regulates apoptosis and cellular stress responses. Originally identified in Fas-resistant B lymphocytes, it functions as an anti-apoptotic molecule by modulating death receptor signaling and protein aggregation pathways . The recombinant form of FAIM Human is produced in E. coli as a non-glycosylated polypeptide (26.4 kDa) containing 236 amino acids (1–213 aa with a 23-residue His-tag) . Two isoforms exist: FAIM-S (ubiquitously expressed) and FAIM-L (neuron-specific with 22 additional N-terminal residues) .

Anti-Apoptotic Activity

  • B Cells: FAIM-S inhibits Fas-mediated apoptosis by enhancing NF-κB and ERK signaling, promoting survival during B-cell activation .

  • Neurons: FAIM-L prevents TNFα- and Fas-induced apoptosis by stabilizing XIAP and suppressing caspase-3 activation .

  • Stem Cells: Upregulation of FAIM protects aged human mesenchymal stem cells (hMSCs) from oxidative stress, mediated by SRT1720 (a SIRT1 activator) .

Metabolic Regulation

DiseasePhenotypeFAIM’s Role
Obesity/HepatosteatosisNon-hyperphagic obesity, adipocyte hypertrophyDeficiency impairs insulin signaling (Akt/IRS-1 pathways) .
HyperglycaemiaInsulin resistance, elevated glucose levelsFAIM-KO mice show reduced glucose uptake in hepatocytes .

Key Findings:

  • FAIM knockout (KO) mice exhibit spontaneous obesity, hepatosteatosis, and dyslipidemia due to disrupted insulin/Akt signaling .

  • FAIM regulates lipid synthesis genes (SREBP-1a, FAS) and glucose metabolism in hepatocytes .

Neuroprotection and Neurodegeneration

  • Parkinson’s Disease (PD): FAIM prevents α-synuclein aggregation, a hallmark of PD pathology. FAIM-deficient neurons show increased cytotoxic α-synuclein aggregates .

  • Alzheimer’s Disease (AD): FAIM inhibits Aβ and tau aggregation in vitro, suggesting potential therapeutic applications .

  • Axon Pruning: FAIM-L regulates caspase-3-dependent axon pruning and long-term depression in neurons .

Disease Associations and Clinical Relevance

DiseaseTissue/CellFAIM’s RoleMechanism
Multiple MyelomaMyeloma cell linesUpregulated in IRF4-expressing cells; poor prognosisIRF4-FAIM-Akt feedback loop promotes tumorigenesis .
Prostate CancerProstate tissuesmiR-133b target; contributes to tumorigenesisModulates tissue homeostasis .
Intellectual DisabilityNeuronsDownregulated in patientsUnknown (linked to synaptic dysfunction) .

Therapeutic Implications:

  • PD/AD: Enhancing FAIM activity may dissolve protein aggregates, offering disease-modifying therapy .

  • Cancer: Targeting FAIM in multiple myeloma could improve survival outcomes .

Emerging Applications and Future Directions

  • Stem Cell Therapy: SRT1720/SIRT1 activation upregulates FAIM, enhancing hMSC survival and therapeutic efficacy in myocardial infarction models .

  • Protein Aggregation: FAIM’s intrinsic chaperone activity (distinct from HSPs) enables solubilization of pre-formed aggregates, positioning it as a novel therapeutic target .

  • Diagnostic Biomarkers: FAIM expression levels correlate with disease progression in multiple myeloma and obesity, warranting further clinical validation .

Product Specs

Introduction
Fas apoptotic inhibitory molecule, also known as FAIM, functions as an inducible effector molecule that mediates Fas resistance produced by surface Ig engagement in B cells. Additionally, FAIM protects against death receptor-triggered apoptosis and regulates B-cell signaling and differentiation. FAIM is associated with diseases such as hemorrhagic thrombocythemia and food allergy.
Description
Recombinant human FAIM protein, produced in E. coli, is a single, non-glycosylated polypeptide chain containing 236 amino acids (residues 1-213) with a molecular mass of 26.4 kDa. The FAIM protein is fused to a 23 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Sterile, clear, and colorless solution.
Formulation
FAIM protein solution (0.5 mg/mL) in 20 mM Tris-HCl buffer (pH 8.0), 0.4 M urea, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Greater than 85% purity as determined by SDS-PAGE.
Synonyms
FAIM1, Fas Apoptotic Inhibitory Molecule, FAIM2, LFG, NMP35, Fas Apoptotic Inhibitory Molecule 1, FAIM.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMLLPFIR TLPLLCYNHL LVSPDSATLS PPYSLEKMTD LVAVWDVALS DGVHKIEFEH GTTSGKRVVY VDGKEEIRKE WMFKLVGKET FYVGAAKTKA TINIDAISGF AYEYTLEING KSLKKYMEDR SKTTNTWVLH MDGENFRIVL EKDAMDVWCN GKKLETAGEF VDDGTETHFS IGNHDCYIKA VSSGKRKEGI IHTLIVDNRE IPEIAS.

Q&A

What is FAIM and what are its primary functions in human cells?

FAIM (Fas Apoptosis Inhibitory Molecule) is a protein that confers resistance to Fas-induced apoptosis in various cell types, including lymphocytes, hepatocytes, and neurons. It serves as a critical regulator of programmed cell death pathways by inhibiting apoptotic signals. In T cells, FAIM is upregulated upon T cell receptor (TCR) engagement and protects thymocytes from TCR-mediated apoptosis by influencing the activation of caspase-8 and caspase-9 . In neurons, FAIM has been shown to protect against stress-induced death by preventing or reversing the aggregation of alpha-synuclein, making it potentially relevant to neurodegenerative conditions such as Parkinson's disease .

How does FAIM expression vary across different human tissues?

FAIM expression patterns vary significantly across human tissue types, with notable presence in lymphoid tissues, the central nervous system, and hepatic tissues. The protein demonstrates both constitutive and inducible expression depending on cell type and environmental signals. In the thymus, FAIM expression is dynamically regulated, with increased expression following T cell receptor engagement . This tissue-specific expression pattern suggests specialized roles for FAIM in different cellular contexts, particularly in tissues where regulated cell death is crucial for homeostasis.

What are the key structural and functional domains of the FAIM protein?

FAIM protein contains several functional domains that contribute to its anti-apoptotic activity. The protein interacts with multiple signaling pathways, including those related to Akt activation. Studies have shown that FAIM influences the localization of Akt to lipid rafts during TCR signaling . The protein's structure enables it to interact with apoptotic machinery components while also affecting protein degradation pathways, as evidenced by its impact on Nur77 ubiquitination and degradation in thymocytes .

What evidence supports FAIM's role in preventing neurodegeneration?

Recent research has demonstrated that FAIM can protect neurons from stress-induced death specifically by preventing or reversing the aggregation of alpha-synuclein, which is the major pathological hallmark of Parkinson's disease (PD). Experimental evidence indicates that FAIM-deficient cells accumulate more cytotoxic alpha-synuclein . This protective function positions FAIM as a potential therapeutic target for neurodegenerative conditions characterized by protein aggregation. The capacity of FAIM to modulate alpha-synuclein aggregation suggests a direct mechanistic link between FAIM activity and neuronal survival in the context of proteinopathies.

How might FAIM be utilized in developing treatments for Parkinson's disease?

FAIM's ability to prevent or reverse alpha-synuclein aggregation positions it as a promising candidate for disease-modifying therapies for Parkinson's disease. Research approaches include: (1) screening drugs that enhance FAIM activity, (2) identifying more active FAIM variants, and (3) developing combination therapies that couple FAIM's aggregate-dissolving activity with other therapeutic agents targeting alpha-synuclein . The development of delivery systems for FAIM to dopaminergic neurons represents a critical step in translating these findings to clinical applications. Using genome engineering technologies, researchers can generate neurons from cells of individuals with SNCA mutations to test FAIM supplementation's efficacy in preventing pathological changes associated with PD .

What experimental models are most appropriate for studying FAIM's neuroprotective effects?

Optimal experimental models for investigating FAIM's neuroprotective functions include:

  • Human iPSC-derived neurons from individuals with SNCA mutations that lead to alpha-synuclein aggregation

  • FAIM knockout and overexpression neuronal models to compare neuronal survival and alpha-synuclein aggregation

  • In vivo models with selective FAIM modulation in dopaminergic neurons

These approaches allow researchers to assess whether increasing FAIM levels can reverse PD pathology and determine the extent to which FAIM deficiency contributes to disease progression . Comparing neurons that lack FAIM with those containing normal amounts provides critical insights into the protein's role in preventing neurodegenerative processes.

How does FAIM regulate T cell receptor-mediated apoptosis?

FAIM regulates T cell receptor (TCR)-mediated apoptosis through multiple mechanisms. Upon TCR engagement, FAIM is upregulated in thymocytes, where it inhibits the activation of caspase-8 and caspase-9, critical executioners of the apoptotic cascade. In FAIM-deficient thymocytes, TCR cross-linking leads to elevated levels of the orphan nuclear receptor Nur77, which plays a significant role in thymocyte apoptosis . FAIM influences the ubiquitination and degradation of Nur77 protein through the activation of Akt, a kinase required for Nur77 ubiquitination. Studies using FAIM-deficient primary thymocytes and FAIM-overexpressing DO-11.10 T cells demonstrate that FAIM acts upstream of Akt during TCR signaling and affects Akt localization to lipid rafts .

What are the consequences of FAIM deficiency on thymocyte development?

FAIM deficiency significantly impacts thymocyte development and survival. In vivo studies show that injection of anti-CD3 antibodies leads to augmented depletion of CD4+CD8+ T cells in the thymus of faim−/− mice compared to wild-type controls . This increased susceptibility to apoptosis is characterized by:

  • Enhanced activation of caspase-8 and caspase-9 following TCR engagement

  • Elevated Nur77 protein levels due to reduced ubiquitination and degradation

  • Defective TCR-induced activation of Akt

These findings suggest that FAIM plays a crucial role in regulating the negative selection of thymocytes during T cell development, potentially influencing the establishment of central tolerance .

What methodologies are most effective for studying FAIM's role in T cell signaling?

The most effective methodologies for investigating FAIM's function in T cell signaling include:

  • Generation of FAIM-deficient and FAIM-overexpressing T cell lines using CRISPR-Cas9 or lentiviral transduction

  • Assessment of TCR-mediated apoptosis through flow cytometry with Annexin V and propidium iodide staining

  • Analysis of signaling cascades using western blotting for phosphorylated Akt and other pathway components

  • Evaluation of protein-protein interactions through co-immunoprecipitation and proximity ligation assays

  • Determination of subcellular localization via confocal microscopy of fluorescently tagged proteins

These approaches have successfully revealed FAIM's impact on critical signaling events, including Akt activation and Nur77 regulation .

What is the FAIM framework in the context of healthcare machine learning?

In healthcare machine learning, FAIM (Fairness-aware Interpretable Modeling) is an interpretable framework designed to improve model fairness without compromising performance. The framework features an interactive interface that helps identify "fairer" models from a set of high-performing options. FAIM promotes the integration of data-driven evidence with clinical expertise to enhance contextualized fairness in medical decision-making . The framework is particularly valuable in high-stakes medical applications where biased predictions can lead to healthcare disparities and inequitable treatment decisions.

How does FAIM address intersectional biases in clinical prediction models?

FAIM addresses intersectional biases arising from factors such as race and sex through several mechanisms:

  • It leverages varying degrees of model reliance on variables (including sensitive variables) to identify alternative model formulations that improve fairness without significantly impairing performance

  • It utilizes models' diverse fairness profiles and near-optimal performance to facilitate informed discussions between clinicians and model developers

  • It examines the impact of excluding some or all sensitive variables on model fairness

  • It visualizes findings to help clinicians select fairness-enhanced models with reasonable interpretation

FAIM has demonstrated significant reductions in intersectional biases when predicting hospital admission in emergency department settings, with improvements of 53.5%-57.6% in fairness metrics for the MIMIC-IV-ED case and 17.7%-21.7% for the SGH-ED case compared to baseline models .

What fairness metrics are used to evaluate FAIM models?

FAIM models are evaluated using established fairness metrics to assess the reduction of biases. The evaluation includes:

The statistical significance of these improvements is consistently high (p < 0.001), demonstrating FAIM's effectiveness in mitigating biases while maintaining predictive performance .

What experimental designs are most appropriate for investigating FAIM's role in disease models?

The most appropriate experimental designs for studying FAIM's role in disease include:

  • Genetic manipulation studies: Using CRISPR-Cas9 to create FAIM knockout models or overexpression systems to evaluate phenotypic changes

  • Patient-derived models: Generating neurons from cells of individuals with relevant mutations (e.g., SNCA for Parkinson's) to test FAIM's effects

  • Comparative analysis: Assessing survival and protein aggregation in cells with varying FAIM expression levels

  • In vivo models: Utilizing targeted approaches to modulate FAIM in specific tissues or cell types

For Parkinson's disease research specifically, studies involving dopamine-producing neurons derived from individuals with PD-associated mutations allow researchers to assess whether FAIM supplementation can reverse pathological features .

How can researchers effectively measure FAIM's impact on protein aggregation?

To effectively measure FAIM's impact on protein aggregation, researchers can employ multiple complementary techniques:

  • Fluorescence microscopy: Using fluorescently-tagged proteins to visualize aggregation dynamics in live cells

  • Biochemical fractionation: Separating soluble and insoluble protein fractions to quantify aggregate formation

  • Thioflavin T assays: Measuring amyloid-like aggregate formation through fluorescent dye binding

  • FRET-based sensors: Detecting conformational changes and protein-protein interactions in real-time

  • Electron microscopy: Visualizing aggregate morphology at ultrastructural resolution

These methodologies can be applied to models with varying FAIM expression levels to determine the protein's capacity to prevent or reverse aggregation of alpha-synuclein or other aggregation-prone proteins .

What analytical approaches best assess fairness in FAIM-based machine learning models?

The most effective analytical approaches for assessing fairness in FAIM-based machine learning models include:

  • Subgroup analysis: Evaluating model performance across different demographic subgroups defined by sensitive attributes

  • Fairness metrics calculation: Computing statistical measures such as disparities in true positive and true negative rates

  • Shapley additive explanations (SHAP): Analyzing variable importance and contributions to model predictions

  • Ablation studies: Comparing model performance with and without sensitive variables to isolate their effects

  • Comparison with established bias mitigation methods: Benchmarking FAIM against approaches like reweighting, reductions, and adversarial debiasing

These analytical approaches have revealed that FAIM models significantly outperform both fairness-unaware baseline models and other bias mitigation methods in terms of fairness, while maintaining comparable discrimination performance .

How might FAIM's dual role in apoptosis and protein aggregation be mechanistically linked?

The dual functionality of FAIM in regulating apoptosis and preventing protein aggregation suggests intriguing mechanistic connections between these processes. One hypothesis proposes that FAIM may influence cellular proteostasis networks that simultaneously regulate both protein quality control and cell death decisions. Since protein aggregation often triggers apoptotic signaling through unfolded protein responses or mitochondrial dysfunction, FAIM could act as a central modulator that prevents cell death by targeting upstream aggregation events .

Research approaches to explore this connection include:

  • Identifying shared interaction partners between FAIM's anti-apoptotic and anti-aggregation functions

  • Investigating FAIM's subcellular localization during various cellular stresses

  • Examining whether FAIM-mediated Akt activation influences protein degradation pathways beyond Nur77 regulation

The elucidation of these mechanistic links could provide novel insights into fundamental cellular processes and identify new therapeutic targets for conditions involving both aberrant cell death and protein aggregation.

How can FAIM models be integrated with other explainable AI approaches for clinical decision support?

Integration of FAIM with other explainable AI approaches can create more comprehensive clinical decision support systems through:

  • Hierarchical interpretability frameworks: Combining FAIM's fairness-aware model selection with local explanation methods like LIME or SHAP

  • Interactive visualization dashboards: Developing interfaces that display both fairness metrics and feature importance

  • Ensemble approaches: Leveraging multiple interpretable models with FAIM's fairness evaluation to provide consensus predictions

  • Domain-specific knowledge graphs: Mapping model decisions to clinical knowledge bases to contextualize predictions

The FAIM framework already employs explainable AI (XAI) methods to clarify variable importance changes due to fairness enhancement, which improves interpretation . Further integration with complementary explainable AI approaches would enable clinicians to understand not only why a prediction was made but also how that prediction might differentially impact various patient subgroups.

What emerging technologies might enhance the study of FAIM's role in human disease?

Several cutting-edge technologies show promise for advancing FAIM research:

  • Single-cell multi-omics: Combining transcriptomics, proteomics, and metabolomics at single-cell resolution to trace FAIM's impact on cellular pathways

  • Spatially-resolved proteomics: Mapping FAIM interactions and effects within specific subcellular compartments

  • Cryo-electron microscopy: Determining high-resolution structures of FAIM in complex with its interacting partners

  • Protein engineering approaches: Developing FAIM variants with enhanced stability or function for therapeutic applications

  • Organ-on-chip technologies: Testing FAIM interventions in physiologically relevant microenvironments

For computational FAIM applications, federated learning approaches could enable model development across multiple healthcare institutions while preserving data privacy, and reinforcement learning could optimize fairness-performance trade-offs in real-time clinical applications .

Product Science Overview

Introduction

The Fas Apoptotic Inhibitory Molecule (FAIM) is a protein that plays a crucial role in regulating apoptosis, or programmed cell death. This protein is particularly significant in the context of neurodegenerative diseases and immune responses. FAIM was originally discovered in 1999 and has since been the subject of extensive research due to its potential therapeutic applications .

Structure and Classification

FAIM is a 20-kDa cytosolic protein composed of 179 amino acids . It is highly conserved across mammalian species, indicating its essential role in cellular processes. There are two main isoforms of FAIM: FAIM1 and FAIM2. FAIM1 is predominantly expressed in immune cells, while FAIM2 is mainly found in neuronal cells .

Biological Properties and Functions

FAIM functions as an inhibitor of the Fas signaling pathway, which is a critical pathway for inducing apoptosis. By interfering with this pathway, FAIM helps to prevent unnecessary cell death, thereby contributing to cell survival and homeostasis . In neurons, FAIM2 has been shown to protect against stress-induced apoptosis, particularly in the retina .

Mode of Action

FAIM inhibits apoptosis by interacting with components of the Fas signaling pathway. Specifically, it binds to the Fas receptor and prevents the activation of caspase-8, a key enzyme in the apoptotic process . This interaction blocks the downstream signaling events that lead to cell death, thereby promoting cell survival.

Regulatory Mechanisms

The expression and activity of FAIM are regulated by various factors, including stress signals and cellular conditions. For instance, FAIM2 levels increase in response to retinal detachment, suggesting a role in protecting photoreceptor cells under stress . Additionally, FAIM interacts with other proteins such as p53 and HSP90, which further modulate its activity and stability .

Therapeutic Potential

Given its role in inhibiting apoptosis, FAIM has significant therapeutic potential, particularly in the treatment of neurodegenerative diseases and conditions involving excessive cell death. For example, recombinant human FAIM has been shown to dissolve pathological amyloid-β species, which are implicated in Alzheimer’s disease . This suggests that FAIM could be a valuable target for developing treatments for such conditions.

Quick Inquiry

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