FAM3A Human

Family with Sequence Similarity 3, Member A Human Recombinant
Shipped with Ice Packs
In Stock

Description

Metabolic Regulation

FAM3A is a critical regulator of mitochondrial respiration and ATP production. In muscle stem cells (MuSCs), it promotes oxidative phosphorylation by activating Akt and AMPK pathways, ensuring efficient differentiation . Genetic ablation of FAM3A in mice reduces mitochondrial respiration and delays myogenic commitment, while recombinant FAM3A rescues these defects .

Tissue-Specific Functions

Tissue/Cell TypeFunctionMechanismSource
Skeletal MusclePromotes MuSC differentiation and regenerationStat3-FAM3A axis → MyoD induction
Vascular Smooth MuscleMaintains contractile phenotype; suppresses aneurysm formationFAM3A-KLF4 axis → Inhibits VSMC dedifferentiation
LiverProtects against ischemia-reperfusion injury (IRI)Activates Akt survival pathways; reduces oxidative stress

Abdominal Aortic Aneurysm (AAA)

FAM3A is downregulated in AAA patients and murine models, correlating with increased aneurysm diameter and mortality . Overexpression or supplementation with recombinant FAM3A attenuates AAA progression by:

  • Reducing inflammation: Lower plasma IL-1β, IL-6, TNF-α levels .

  • Preserving vascular integrity: Suppresses MMP2/9 expression and maintains VSMC contractility .

Clinical Relevance of FAM3A in AAA

ParameterControl (Ad-sham)FAM3A-Overexpressing MiceEffect
Aortic diameter (mm)2.5 ± 0.31.8 ± 0.2↓ 28% (p < 0.01)
Survival rate40%70%↑ 75% (p < 0.05)
Plasma IL-6 (pg/mL)150 ± 2080 ± 15↓ 46.7% (p < 0.01)

Data adapted from .

Liver Ischemia-Reperfusion Injury (IRI)

FAM3A-deficient mice exhibit exacerbated liver damage post-IRI, characterized by:

  • Increased oxidative stress: Elevated MDA and reduced ATP content .

  • Apoptosis: ↓ BCL-2/BAX ratio and ↑ active caspase-3 .
    PPARγ agonists (e.g., rosiglitazone) rescue IRI in wild-type mice but fail in FAM3A knockouts, indicating FAM3A mediates PPARγ’s protective effects .

FAM3A in Cancer Research

Cancer TypeExpression TrendPrognostic ImpactMechanism
LIHCPoor OS (HR = 1.8, p < 0.05)Potential diagnostic biomarker
Other CancersVariableContext-dependentRequires pan-cancer validation

Therapeutic Potential

Recombinant FAM3A protein (e.g., Abcam’s ab167946) shows promise in preclinical models, with applications in:

  • Muscle regeneration: Enhances MuSC differentiation .

  • Vascular repair: Prevents AAA formation and preserves VSMC function .

  • Liver protection: Mitigates IRI by activating survival pathways .

Challenges include elucidating FAM3A’s molecular targets (e.g., Akt/AMPK crosstalk) and optimizing delivery strategies for systemic diseases .

Genetic and Epigenetic Regulation

The FAM3A gene is regulated by transcription factors like Stat3 and MyoD, which bind to its promoter during myogenic differentiation . Epigenetic modifications (e.g., promoter methylation) may influence its expression in cancer and metabolic disorders .

Key Regulatory Pathways

RegulatorEffect on FAM3ACell TypeSource
Stat3↑ TranscriptionMuscle Stem Cells
MyoD↑ TranscriptionMyogenic Progenitors
PPARγ↑ ExpressionHepatocytes

Product Specs

Introduction
FAM3A (Family with Sequence Similarity 3, Member A) belongs to the FAM3 family alongside FAM3B (Pander). While FAM3B is known to induce apoptosis in pancreatic alpha and beta cells, FAM3A's function is less clear. This cytokine-like protein is widely expressed, found in tissues like the testis, pancreas, adrenal glands, placenta, brain (including fetal brain), liver, kidney, skeletal muscle, and heart. Evidence suggests that FAM3A gene expression may be regulated by PPAR-gamma (peroxisome proliferator-activated receptor gamma), potentially implicating FAM3A in glucose and lipid metabolism. Additionally, research suggests it might act as a defense mechanism against fungal infections.
Description
Recombinant Human FAM3A, produced in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 207 amino acids, including a 10-amino acid N-terminal His-tag. The calculated molecular mass is 23.1 kDa.
Physical Appearance
White, lyophilized powder after filtration.
Formulation
The FAM3A protein solution (0.5 mg/mL in 20 mM TRIS, 50 mM NaCl, pH 7.5) is filtered through a 0.4 μm filter and then lyophilized.
Solubility
To create a working stock solution, add deionized water to the lyophilized pellet, aiming for a concentration of around 0.5 mg/mL. Allow sufficient time for the pellet to dissolve completely. Note: This product is not sterile. Filter it through a sterile filter before using it in cell culture.
Stability
Store the lyophilized protein at -20°C. After reconstitution, aliquot the protein to minimize freeze-thaw cycles. Reconstituted FAM3A can be stored at 4°C for a limited time (approximately two weeks) with no observable changes in its properties.
Purity
Purity is determined to be greater than 95.0% by SDS-PAGE analysis.
Synonyms
Protein FAM3A, Cytokine-like protein 2-19, FAM3A, 2-19, 2.19, DLD, XAP-7, DXS560S.
Source
Escherichia Coli.
Amino Acid Sequence
MKHHHHHHASFPRIQQLFTS PESSVTAAPR ARKYKCGLPQ PCPEEHLAFR VVSGAANVIG PKICLEDKML MSSVKDNVGR GLNIALVNGV SGELIEARAF DMWAGDVNDL LKFIRPLHEG TLVFVASYDD PATKMNEETR KLFSELGSRN AKELAFRDSW VFVGAKGVQN KSPFEQHVKN SKHSNKYEGW PEALEMEGCI PRRSTAS.

Q&A

What is FAM3A and where is it primarily localized in human cells?

FAM3A (Family with sequence similarity 3 member A) is the first identified member of the FAM3 cytokine-like gene family, which includes four members (FAM3A, FAM3B, FAM3C, and FAM3D). FAM3A is primarily localized in mitochondria where it interacts with F1-ATP synthase to enhance ATP generation. While primarily a mitochondrial protein, FAM3A can also be secreted by certain cell types, though this appears to be cell-type dependent. In normal kidney tissue, FAM3A is highly expressed but not typically secreted into urine under healthy conditions . The protein plays crucial roles in mitochondrial function and cellular energy homeostasis across multiple tissue types.

How does FAM3A contribute to cellular energy homeostasis?

FAM3A functions as a key regulator of mitochondrial ATP production. It directly interacts with F1-ATP synthase in the mitochondria to enhance ATP generation. In research models, FAM3A deficiency significantly reduces ATP production in multiple cell types including hepatocytes, smooth muscle cells, and endothelial cells . Beyond direct ATP production, FAM3A also mediates ATP secretion, which activates downstream signaling through P2 receptors. This ATP-dependent signaling induces PI3K/AKT pathway activation (termed "ATP signaling") that influences numerous cellular processes including cell survival, metabolism, and angiogenesis . Additionally, FAM3A helps maintain mitochondrial mass and function, with studies showing that downregulation of FAM3A promotes the loss of mitochondrial mass and increases susceptibility to mitochondrial fragmentation under stress conditions .

What role does FAM3A play in acute kidney injury (AKI)?

In acute kidney injury, FAM3A functions as a protective factor against tubular cell pyroptosis—a form of inflammatory cell death. Research indicates that FAM3A expression becomes significantly downregulated in tubular cells during AKI, contributing to increased tubular cell death and kidney dysfunction. Mechanistically, FAM3A protects against tubular cell pyroptosis through the activation of the PI3K/AKT/NRF2 signaling pathway. This pathway reduces mitochondrial reactive oxygen species (mt-ROS) production, which is a strong inducer of cell pyroptosis . Loss of FAM3A in tubular cells decreases ATP generation, which subsequently reduces PI3K/AKT/NRF2 signaling activation, ultimately facilitating pyroptosis in AKI. Notably, urinary FAM3A levels are elevated in AKI patients, likely because injured tubular cells containing FAM3A detach and are excreted in urine, making urinary FAM3A a potential biomarker for tubular injury .

How might researchers utilize FAM3A as a biomarker for kidney injury?

Researchers can explore FAM3A as a novel biomarker for tubular cell injury in AKI through several methodological approaches:

  • Urinary FAM3A measurements: Develop ELISA or immunoblotting assays to quantify FAM3A levels in patient urine samples. Studies have shown that urinary FAM3A is positively correlated with urinary IL-18 (a marker of cell pyroptosis) and NGAL (a marker for tubular cell injury) .

  • Correlation studies: Design clinical studies comparing urinary FAM3A levels with established AKI markers and clinical outcomes to validate its biomarker potential.

  • Time-course analysis: Evaluate FAM3A kinetics during AKI progression and recovery to determine its temporal relationship with injury severity.

  • Immunohistochemistry in kidney biopsies: Assess FAM3A expression in renal tubules alongside mitochondrial markers like TOMM20. Research has shown that FAM3A is a sensitive indicator of tubular mitochondrial injury, with expression decreasing even in tubules that appear morphologically normal but have decreased TOMM20 expression .

  • Multi-marker panels: Develop diagnostic panels combining FAM3A with other AKI biomarkers to improve sensitivity and specificity for early detection and prognostication.

How is FAM3A expression regulated in hepatocytes?

FAM3A expression in hepatocytes is primarily regulated by peroxisome proliferator-activated receptor gamma (PPARγ). Experimental evidence demonstrates that:

  • PPARγ agonists like rosiglitazone significantly induce FAM3A expression in both primary mouse hepatocytes and human HepG2 cells .

  • The transcriptional regulation occurs through direct binding of PPARγ to a peroxisome proliferator response element (PPRE) located at position -1258/-1246 in the human FAM3A promoter, as confirmed by chromatin immunoprecipitation (ChIP) assays .

  • Site-directed mutagenesis of this PPRE-like motif abolishes PPARγ's stimulatory effect on the transcriptional activity of human FAM3A promoter, confirming the specificity of this regulatory mechanism .

  • PPARγ antagonism blocks rosiglitazone-induced FAM3A expression, while PPARγ overexpression stimulates FAM3A expression in hepatocytes .

  • In contrast to PPARγ, other PPAR family members (PPARα and PPARβ) do not significantly affect FAM3A expression, as demonstrated by the lack of response to their respective agonists (fenofibrate for PPARα and GW0742 for PPARβ) .

In vivo studies with oral rosiglitazone treatment confirm upregulation of FAM3A expression in the livers of both normal C57BL/6 mice and diabetic db/db mice .

What experimental approaches should be used to study FAM3A's role in hepatic metabolism?

To comprehensively investigate FAM3A's role in hepatic metabolism, researchers should consider the following experimental approaches:

  • Loss- and gain-of-function models:

    • Generate liver-specific FAM3A knockout mice using Cre-loxP technology

    • Develop inducible hepatocyte-specific FAM3A overexpression models

    • Use adenoviral or AAV vectors for acute modulation of FAM3A expression

  • Metabolic phenotyping:

    • Glucose and insulin tolerance tests in FAM3A-modified models

    • Hyperinsulinemic-euglycemic clamp studies to assess insulin sensitivity

    • Measurement of hepatic triglyceride content and de novo lipogenesis

  • Mechanistic investigations:

    • Assess mitochondrial function (oxygen consumption, ATP production)

    • Analyze ATP-dependent PI3K/AKT signaling using phospho-specific antibodies

    • Measure PPARγ-dependent gene expression networks

  • Translational approaches:

    • Examine FAM3A expression in human liver biopsies from patients with metabolic disorders

    • Correlate FAM3A levels with clinical parameters of metabolic health

    • Investigate FAM3A genetic variants and their association with metabolic traits

  • Therapeutic targeting:

    • Test PPARγ agonists with different pharmacological profiles for FAM3A induction

    • Develop cell-permeable FAM3A peptides or mimetics

    • Evaluate the metabolic effects of AAV-mediated FAM3A delivery to liver

How does FAM3A influence angiogenesis and vascular remodeling?

FAM3A plays a significant role in promoting angiogenesis and regulating vascular smooth muscle cell phenotype through several mechanisms:

  • Regulation of endothelial angiogenesis:

    • FAM3A overexpression promotes endothelial tube formation, proliferation, and migration in vitro .

    • In a mouse hind limb ischemia model, FAM3A overexpression improves blood perfusion and increases capillary density, while FAM3A knockdown has opposite effects .

    • Under hypoxic conditions, endothelial FAM3A expression is downregulated, suggesting a regulatory role in response to ischemia .

  • ATP-calcium-VEGFA signaling axis:

    • Mitochondrial FAM3A increases ATP production and secretion.

    • The secreted ATP binds to P2 receptors and upregulates cytosolic free Ca²⁺ levels.

    • Increased intracellular Ca²⁺ enhances phosphorylation of the transcription factor cAMP response element binding protein (CREB).

    • Phosphorylated CREB is recruited to the VEGFA promoter, activating VEGFA transcription and driving endothelial angiogenesis .

  • Vascular smooth muscle cell phenotypic switching:

    • FAM3A expression is elevated in human aortic smooth muscle cells (HASMCs) following oxidized low-density lipoprotein (ox-LDL) treatment, a key factor in atherosclerosis .

    • FAM3A silencing suppresses ox-LDL-provoked proliferation, migration, and inflammation of HASMCs, while FAM3A overexpression enhances these processes .

    • FAM3A mediates the switch of HASMCs from a contractile phenotype to a synthetic phenotype in response to ox-LDL, a critical process in atherosclerosis development .

    • This phenotypic switching is regulated through the PI3K-AKT pathway, with FAM3A silencing inhibiting and FAM3A overexpression promoting ox-LDL-induced AKT activation .

These findings collectively demonstrate that FAM3A functions as a critical regulator of vascular homeostasis, with potential implications for both therapeutic angiogenesis in ischemic disease and pathological vascular remodeling in atherosclerosis.

What are the methodological approaches for studying FAM3A in vascular cells?

Researchers investigating FAM3A in vascular biology should employ a comprehensive set of experimental techniques:

  • In vitro functional assays:

    • Tube formation assays on Matrigel to assess endothelial angiogenic capacity

    • Migration assays (scratch wound or Boyden chamber) to measure cell motility

    • Proliferation assays (BrdU incorporation, Ki67 staining) to evaluate cell growth

    • Ca²⁺ imaging using fluorescent indicators to monitor intracellular calcium dynamics

    • Contractility assays for vascular smooth muscle cells to assess phenotypic state

  • Molecular signaling analysis:

    • ATP measurements in cell culture supernatants and intracellularly

    • Phospho-protein analysis for PI3K/AKT pathway components

    • ChIP assays to assess CREB binding to the VEGFA promoter

    • Luciferase reporter assays with VEGFA promoter constructs

    • qRT-PCR and Western blotting for contractile vs. synthetic markers in VSMCs

  • In vivo models:

    • Hind limb ischemia model with laser Doppler imaging for blood flow assessment

    • Matrigel plug assays for quantifying angiogenesis

    • Atherosclerosis models (ApoE-/- or LDLR-/- mice) with vascular-specific FAM3A modulation

    • Vascular injury models to assess neointimal formation

    • Cell lineage tracing to monitor VSMC phenotypic switching in vivo

  • Genetic manipulation approaches:

    • Adenovirus or AAV vectors for acute gain-of-function studies

    • siRNA or shRNA for loss-of-function studies

    • CRISPR/Cas9 for generating knockout cell lines

    • Endothelial- or VSMC-specific conditional knockout mouse models

  • Translational research methods:

    • Immunohistochemistry of human vascular specimens

    • Correlation of FAM3A expression with clinical parameters of vascular disease

    • Ex vivo studies using human vessels in organ bath systems

What are the best approaches for modulating FAM3A expression in experimental models?

Researchers have several effective options for modulating FAM3A expression, each with specific advantages depending on the experimental context:

  • Viral vector systems:

    • Adenoviral vectors provide high-efficiency, transient expression for in vitro and acute in vivo studies .

    • Adeno-associated virus (AAV) vectors offer longer-term expression with lower immunogenicity, ideal for chronic in vivo studies .

    • Lentiviral vectors enable stable integration for long-term expression in dividing cells.

  • RNA interference:

    • siRNA transfection provides efficient but transient knockdown in cell culture systems .

    • shRNA delivered via viral vectors allows for stable knockdown in vitro and in vivo.

    • Inducible RNAi systems permit temporal control of FAM3A suppression.

  • CRISPR/Cas9 genome editing:

    • Complete knockout of FAM3A in cell lines

    • Introduction of specific mutations to study structure-function relationships

    • Knockin of reporter tags for live imaging or pull-down experiments

  • Transgenic mouse models:

    • Conventional FAM3A knockout mice

    • Conditional tissue-specific knockout using Cre/loxP systems

    • Inducible expression systems (tetracycline-controlled)

  • Pharmacological modulation:

    • PPARγ agonists like rosiglitazone for increasing endogenous FAM3A expression

    • Targeting upstream regulators or downstream effectors of FAM3A signaling

When selecting an approach, researchers should consider:

  • Temporal requirements (acute vs. chronic modulation)

  • Tissue specificity needs

  • Reversibility of the intervention

  • Required efficiency of modulation

  • Potential off-target effects

  • Compatibility with downstream assays

The choice between overexpression and knockdown/knockout approaches should be guided by the specific research question, with complementary gain- and loss-of-function studies providing the most robust evidence of FAM3A's roles.

What technical challenges should researchers anticipate when studying FAM3A's interaction with mitochondrial proteins?

Studying FAM3A's interactions with mitochondrial proteins presents several technical challenges that researchers should anticipate and address:

  • Mitochondrial isolation and fractionation:

    • Maintaining mitochondrial integrity during isolation to preserve protein-protein interactions

    • Achieving high purity to minimize contamination from other cellular compartments

    • Separating mitochondrial subcompartments (outer membrane, inner membrane, matrix) to localize interactions precisely

  • Protein-protein interaction detection:

    • Preserving native interactions during cell lysis and immunoprecipitation

    • Distinguishing direct from indirect interactions in complex mitochondrial protein assemblies

    • Capturing transient or weak interactions that may be physiologically relevant

  • Live cell imaging challenges:

    • Generating functional fluorescent fusion proteins that maintain proper mitochondrial localization

    • Achieving sufficient resolution to visualize mitochondrial subcompartments

    • Minimizing phototoxicity during extended imaging of mitochondrial dynamics

  • Functional validation approaches:

    • Designing mutations that disrupt specific interactions without affecting protein stability

    • Developing assays that can directly link protein interactions to functional outcomes

    • Accounting for compensatory mechanisms that may mask phenotypes

  • Technical considerations for F1-ATP synthase interaction studies:

    • The large size and complex structure of F1-ATP synthase make interaction mapping difficult

    • ATP synthase exists in different assembly states which may affect FAM3A binding

    • Changes in mitochondrial membrane potential can alter interaction properties

Recommended advanced methodological approaches:

  • Proximity labeling techniques (BioID, APEX) for capturing interactions in living cells

  • Cryo-electron microscopy for structural analysis of FAM3A-ATP synthase complexes

  • Super-resolution microscopy to visualize FAM3A localization within mitochondria

  • In organello assays to study FAM3A function in isolated, intact mitochondria

  • Hydrogen-deuterium exchange mass spectrometry to map interaction interfaces

How does FAM3A integrate with the PI3K/AKT/NRF2 pathway across different tissue types?

FAM3A operates through the PI3K/AKT/NRF2 pathway with tissue-specific variations that warrant detailed investigation:

  • Common signaling mechanisms across tissues:

    • FAM3A enhances ATP production and secretion in multiple cell types

    • ATP binds to P2 receptors to activate the PI3K/AKT pathway

    • Activated AKT phosphorylates downstream targets including NRF2

  • Kidney-specific pathway integration:

    • In tubular cells, FAM3A-mediated PI3K/AKT/NRF2 activation primarily functions to suppress mitochondrial ROS production and reduce pyroptosis

    • Knockdown of FAM3A suppresses PI3K and AKT activation and reduces nuclear accumulation of NRF2

    • Addition of AKT activator SC79 or NRF2 activator Olipraz can rescue the pyroptotic phenotype in FAM3A-deficient cells

  • Vascular system integration:

    • In endothelial cells, the FAM3A-ATP-P2 receptor axis increases cytosolic Ca²⁺, which enhances CREB phosphorylation and VEGFA transcription

    • This pathway promotes angiogenic responses like tube formation and migration

    • In vascular smooth muscle cells, FAM3A promotes the synthetic phenotype through PI3K/AKT activation

  • Liver pathway specificity:

    • PPARγ acts as an upstream regulator of FAM3A in hepatocytes

    • FAM3A upregulation correlates with increased pAkt levels in liver cells

    • This may contribute to PPARγ's metabolic effects in the liver

Research questions that need addressing:

  • What determines the tissue-specific outcomes of FAM3A-mediated PI3K/AKT activation?

  • Are there tissue-specific co-activators or inhibitors that modify FAM3A signaling?

  • How do different P2 receptor subtypes influence downstream signaling in various tissues?

  • What is the precise molecular mechanism by which FAM3A enhances ATP synthase activity?

  • How does FAM3A signaling cross-talk with other stress response pathways?

Methodological approach for integrative analysis:

  • Comparative phosphoproteomics across tissue types with FAM3A modulation

  • ChIP-seq analysis of NRF2 binding sites in different cellular contexts

  • Interactome mapping using BioID or APEX2 proximity labeling

  • Single-cell analysis of signaling pathway activation

  • Systems biology modeling of the FAM3A-dependent signaling network

What are the unresolved questions regarding FAM3A's evolutionary significance and structure-function relationships?

Several critical questions remain unanswered regarding FAM3A's evolutionary significance and structure-function relationships:

  • Evolutionary conservation and divergence:

    • The FAM3 gene family emerged relatively recently in evolution, but their physiological importance appears substantial

    • How have the functions of FAM3A diverged from other FAM3 family members (FAM3B, FAM3C, FAM3D)?

    • Are there species-specific adaptations in FAM3A function that reflect different metabolic requirements?

    • What selective pressures drove the specialization of FAM3A as a mitochondrial protein?

  • Structure-function relationships:

    • Which domains or motifs of FAM3A are critical for:

      • Mitochondrial targeting and localization

      • Interaction with F1-ATP synthase

      • ATP production enhancement

      • Resistance to stress-induced degradation

    • How does the three-dimensional structure of FAM3A facilitate its interaction with binding partners?

    • Are there post-translational modifications that regulate FAM3A activity or localization?

  • Dual localization dynamics:

    • What determines whether FAM3A remains mitochondrial or becomes secreted?

    • Does FAM3A have distinct functions in different subcellular compartments?

    • How is FAM3A trafficking regulated under normal versus stress conditions?

  • Therapeutic targeting potential:

    • Can structure-based drug design identify small molecules that mimic or enhance FAM3A function?

    • Are there natural variations in FAM3A sequence that predispose to disease or confer protection?

    • What delivery methods would be most effective for FAM3A-based therapies for tissue-specific targeting?

Methodological approaches to address these questions:

  • Comparative genomics and phylogenetic analysis of FAM3 family evolution

  • X-ray crystallography or cryo-EM to resolve FAM3A structure

  • Mutagenesis studies to identify functional domains

  • Interspecies complementation experiments to test functional conservation

  • Domain-swapping between FAM3 family members to identify specialized regions

  • Targeted mass spectrometry to identify post-translational modifications

  • Computational modeling of protein-protein interactions

Product Science Overview

Structure and Function

FAM3A encodes a secreted protein with a GG domain, which is a characteristic feature of the FAM3 family. The GG domain is involved in protein-protein interactions and is essential for the protein’s function. FAM3A is known to play a role in the regulation of glucose and lipid metabolism, making it a significant protein in the context of metabolic diseases.

The protein is ubiquitously expressed in various tissues, indicating its importance in multiple physiological processes. It has been observed that changes in the expression of FAM3A can have significant implications for cellular function and overall health.

Role in Disease

Alterations in the expression of FAM3A have been linked to several diseases. For instance, it has been associated with metabolic disorders such as diabetes and obesity. The protein’s role in regulating glucose and lipid metabolism makes it a potential target for therapeutic interventions aimed at treating these conditions.

Additionally, FAM3A has been implicated in cancer biology. Changes in its expression have been noted in pancreatic cancer-derived cells, suggesting that it may play a role in tumorigenesis. Further research is needed to fully understand the mechanisms by which FAM3A contributes to cancer development and progression.

Research and Applications

Recombinant FAM3A protein is widely used in research to study its function and role in various diseases. The recombinant protein is typically produced in an expression system such as E. coli and purified to high levels of purity. This allows researchers to conduct experiments to elucidate the protein’s function, interactions, and potential as a therapeutic target.

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.