GFER Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method or location. For specific delivery times, please consult your local distributor.
Synonyms
ALR_HUMAN antibody; Augmenter of liver regeneration antibody; ERV1 antibody; ERV1 homolog antibody; Erv1 like growth factor antibody; FAD-linked sulfhydryl oxidase ALR antibody; GFER antibody; Growth factor augmenter of liver regeneration antibody; Growth factor erv1 like antibody; Hepatic regenerative stimulation substance antibody; Hepatopoietin antibody; Hepatopoietin protein antibody; hERV1 antibody; HPO antibody; HPO1 antibody; HPO2 antibody; HSS antibody
Target Names
Uniprot No.

Target Background

Function
GFER (Growth Factor-Regulating ERV1) is a FAD-dependent sulfhydryl oxidase that plays a crucial role in the mitochondrial intermembrane space. It regenerates the redox-active disulfide bonds in CHCHD4/MIA40, a chaperone essential for disulfide bond formation and protein folding. The reduced form of CHCHD4/MIA40 forms a transient intermolecular disulfide bridge with GFER/ERV1, leading to the regeneration of essential disulfide bonds in CHCHD4/MIA40, while GFER/ERV1 becomes re-oxidized by donating electrons to cytochrome c or molecular oxygen. Additionally, GFER may act as an autocrine hepatotrophic growth factor, promoting liver regeneration.
Gene References Into Functions
  • Data, including studies on knockout mice, suggest that KIBRA plays a significant role in regulating HPO activity, YAP signaling, and actin cytoskeletal dynamics in podocytes. Expression of KIBRA and YAP, along with YAP phosphorylation, is upregulated in glomeruli of patients with focal segmental glomerulosclerosis. (KIBRA = kidney/brain protein-KIBRA; HPO = hepatopoietin protein; YAP = Yes associated protein-1) PMID: 28982981
  • Loss of DLG5 expression has been linked to breast cancer progression by inactivating the Hippo signaling pathway and increasing nuclear YAP. PMID: 28169360
  • WWC2 functions as a tumor suppressor by negatively regulating the Hippo signaling pathway and may serve as a prognostic marker in hepatocellular carcinoma. PMID: 28815883
  • Studies have demonstrated that targeted inhibition of ALR expression in Jurkat cells triggers cell growth inhibition and sensitizes cells to VCR by promoting apoptosis and regulating the cell cycle. PMID: 29048676
  • IKBKE plays a crucial role in regulating cell proliferation, invasion, and epithelial-mesenchymal transition of malignant glioma cells both in vitro and in vivo by impacting the Hippo pathway. PMID: 28548934
  • Analysis of four unrelated families with patients carrying compound heterozygous variants in GFER revealed phenotypic similarities. These findings contribute to a better understanding of this apparently homogeneous disorder, characterized by clinical and biological hallmarks of mitochondrial disease. PMID: 28155230
  • Research indicates that ALR negatively regulates the autophagy process through an association with the AMPK/mTOR signaling pathway. Autophagy inhibits apoptosis and plays a protective role under conditions of oxidative stress. PMID: 28466106
  • The interaction between HPO and MOB1 is not essential for development and tissue growth control. PMID: 28947795
  • Overexpression of augmenter of liver regeneration (ALR) in liver cancer has been found to improve sensitivity to antitumor drugs by increasing the retention of intracellular drugs, potentially through modulation of the ABCB1 and ABCG2 signaling pathway. PMID: 28825695
  • ALR is involved in regulating the mitochondrial fission machinery. PMID: 28646508
  • Research provides evidence demonstrating Hippo-independent regulation of TEADs and its potential implications for new cancer therapeutics. PMID: 28964625
  • ALR protects steatotic hepatocytes from ischemia reperfusion injury by attenuating oxidative stress and mitochondrial dysfunction. PMID: 28704337
  • Findings indicate that MARK4 acts as a negative regulator of the Hippo kinase cassette to promote YAP/TAZ activity. Loss of MARK4 restrains the tumorigenic properties of breast cancer cells. PMID: 28183853
  • ALR, dependent on its localization, influences the acute-phase response (APR), at least in part, by modifying STAT3 activation. This dual signaling of ALR suggests its pivotal role in the regulation of APR, a crucial event in liver injury and regeneration. PMID: 28506765
  • Research investigates the mechanistic regulation and linkage of the ROR1-HER3 and Hippo-YAP pathway in a cancer-specific context. PMID: 28114269
  • Induction of S100A7 by the Hippo-YAP pathway in cervical and glossopharyngeal squamous cell carcinoma has been described. PMID: 27907036
  • ALR protects cells from apoptosis, partly through increased autophagy in HepG2 cells. PMID: 25954098
  • Knockdown of GFER exhibits anti-inflammatory actions via suppression of the mitogen-activated protein kinase signaling pathway. PMID: 25929436
  • ALR plays a protective role against hydrogen peroxide-induced oxidative stress in renal proximal tubule cells. PMID: 25633409
  • Data indicate that overexpression of 23 kDa augmenter of liver regeneration (ALR) in hepatic cell line LO2 cells promotes cell proliferation and enhances cell resistance to hydrogen peroxide. PMID: 26271971
  • Upregulation of miR-130b enhances stem cell-like phenotype in glioblastoma by inactivating the Hippo signaling pathway. PMID: 26241672
  • This review summarizes current findings on the regulation of Hippo signaling in liver regeneration and tumorigenesis, focusing on how the loss of tumor suppressor components of the Hippo pathway results in liver cancers. [review] PMID: 25476204
  • Here we provide an overview of its roles in regulating stem cells in epithelial tissues and its potential implications in related cancers. [review] PMID: 25476205
  • The purpose of this review is to summarize recent findings and discuss how the Hippo pathway responds to cellular stress and regulates early development events, tissue homeostasis, as well as tumorigenesis. [review] PMID: 25476206
  • In this review, we briefly describe the components of the Hippo pathway and focus on recent progress with respect to the regulation of the Hippo pathway by GPCRs and G proteins in cancer cells. [review] PMID: 25491506
  • Control of growth and beyond: a special issue on Hippo signaling PMID: 25467756
  • This review will discuss and summarize the roles of several core components of the Hippo pathway in mammary gland development and breast cancer. [review] PMID: 25467757
  • In this review, we discuss the roles of non-canonical Hippo/Mst signaling pathways in lymphocyte development and functions. [review] PMID: 25487919
  • ALR is involved in the progression of renal fibrosis. Administration of rhALR protects the kidney against renal fibrosis by inhibiting TGF-beta/Smad activity. PMID: 24844766
  • Enhanced ALR gene expression was negatively correlated with advanced histopathological grade and stage in both colon cancer cell lines and human tissue samples. PMID: 25778301
  • A model for the functional defect in Erv1 R182H has been proposed, which could potentially be extended to human ALR R194H and provides insights into the molecular basis of autosomal recessive myopathy. PMID: 25269795
  • ALR, Bcl-2 protein, clusterin, and reactive oxygen species expression in muscle tissue biopsies from mitochondrial myopathy-affected patients was determined. PMID: 23916837
  • ALR protects Jurkat T leukemia cells from vincristine-induced cell death. PMID: 23810409
  • The protective effect of hepatic stimulator substance against endoplasmic reticulum stress may be associated with the removal of reactive oxygen species to restore the activity of the sarco-endoplasmic reticulum Ca(2+)-ATPase. PMID: 24284796
  • These results collectively suggest that the Hippo pathway negatively regulates the actin-binding activity of Amot family members through direct phosphorylation. PMID: 24225952
  • Import and oxidative folding of proteins are kinetically and functionally coupled and depend on the expression of Mia40, ALR, and the intracellular glutathione pool. PMID: 23676665
  • This work studies the catalytic mechanism of the short, cytokine form of augmenter of liver regeneration using model thiol substrates of the enzyme. PMID: 24147449
  • Nrf2 activates ALR via antioxidant response element and links oxidative stress to liver regeneration. PMID: 23887691
  • Small molecule MitoBloCK-6 inhibits Erv1/ALR and thus mitochondrial protein import in human embryonic stem cells. PMID: 23597483
  • Overexpression of hALR results in influencing sperm morphology and quantity, ultimately reducing male fertility. PMID: 22863717
  • A refined 2.4 A resolution of ALR structure is in good agreement with both the X-ray data (R(cryst) of 0.165, R(free) of 0.211). PMID: 22948913
  • The unstructured domain performs a dual function in two cellular compartments: it acts as (i) a mitochondrial targeting signal in the cytosol and (ii) a crucial recognition site in the disulfide relay system of the intermembrane space. PMID: 23207295
  • ALR level in serum may indicate hepatocyte proliferation or liver regeneration. High ALR levels in serum in the early stage of acute-on-chronic liver failure may suggest a good prognosis. PMID: 22246190
  • The role of ALR is investigated with regards to activated caspase-3, ROS, apoptotic cell number, and mitochondrial degeneration. PMID: 22476097
  • The study elucidates the mechanism of the electron flux within ALR, characterizing at the atomic level the ALR intermediates that allow electrons to rapidly flow to cytochrome c. PMID: 22224850
  • The role of Gfer is the restriction of unwarranted proliferation in HSCs through the inhibition of Jab1 and subsequent stabilization and nuclear retention of p27kip1. PMID: 21636978
  • Data indicate that cytosolic ALR reduces hepatoma cell migration, augments epithelial growth, and therefore, may act as an antimetastatic and EMT reversing protein. PMID: 21152698
  • Molecular recognition and substrate mimicry drive the electron-transfer process between MIA40 and ALR. PMID: 21383138
  • A mutation in the GFER gene causes an infantile mitochondrial disorder. GFER mutation in a patient's muscle leads to: a reduction in complex I, II, and IV activity; abnormal morphology of the mitochondria; and mtDNA multiple deletions. PMID: 19409522
  • [review] Chronic lack of action by hepatic insulin-sensitizing substance (HISS) results in a progressive and predictable series of homeostatic dysfunctions typical of type 2 diabetes. PMID: 20393596
Database Links

HGNC: 4236

OMIM: 600924

KEGG: hsa:2671

STRING: 9606.ENSP00000248114

UniGene: Hs.27184

Involvement In Disease
Myopathy, mitochondrial progressive, with congenital cataract, hearing loss and developmental delay (MPMCHD)
Subcellular Location
[Isoform 1]: Mitochondrion intermembrane space. Mitochondrion.; [Isoform 2]: Cytoplasm. Secreted.
Tissue Specificity
Ubiquitously expressed. Highest expression in the testis and liver and low expression in the muscle.

Q&A

What is GFER and why is it an important research target?

GFER, also known as Augmenter of Liver Regeneration (ALR), is an approximately 30 kDa mitochondrial sulfhydryl oxidase that plays critical roles in multiple cellular processes . In humans, the canonical protein consists of 205 amino acid residues with a molecular mass of 23.4 kDa . GFER is ubiquitously expressed across many tissue types and has multiple subcellular localizations including the mitochondrial intermembrane space, cytoplasm, and is also secreted .

The protein is involved in cellular responses to lipopolysaccharides and TNF-mediated signaling pathways . GFER's significance as a research target stems from its critical roles in liver regeneration, mitochondrial function, and hematopoietic stem cell regulation. Mutations in GFER have been associated with mitochondrial myopathy, highlighting its clinical relevance .

What applications are most commonly used with GFER antibodies?

GFER antibodies have been validated for multiple experimental applications, with Western blotting (WB) being the most widely utilized technique . Other common applications include:

  • Immunohistochemistry (IHC) - For detection of GFER in tissue sections, with validated protocols for both paraffin-embedded and frozen tissues

  • Enzyme-Linked Immunosorbent Assay (ELISA) - For quantitative detection of GFER in solution

  • Immunocytochemistry (ICC) - For subcellular localization studies in cultured cells

  • Flow cytometry (FACS) - For quantitative analysis of GFER expression in cell populations

When designing experiments, researchers should consider that GFER antibodies have been successfully used to detect the protein in various human tissues, with specific validation in liver samples and hepatocellular carcinoma cell lines such as HepG2 and Hep3B .

How should I optimize Western blot protocols for GFER detection?

For optimal Western blot detection of GFER, consider the following methodological approach:

  • Sample preparation: Use reducing conditions as demonstrated in validated protocols

  • Gel selection: Since GFER has a molecular weight of approximately 20-24 kDa, 12-15% SDS-PAGE gels are recommended for optimal resolution

  • Transfer: PVDF membranes are preferred over nitrocellulose for GFER detection

  • Antibody concentration: Start with 2 μg/mL for monoclonal antibodies as a baseline and adjust as needed

  • Secondary antibody: HRP-conjugated secondary antibodies work well with established detection systems

In HepG2 and Hep3B human hepatocellular carcinoma cell lines, GFER typically appears as specific bands at approximately 20-24 kDa when analyzed under reducing conditions using Immunoblot Buffer Group 1 . If multiple bands appear, this may represent different isoforms, as up to two different isoforms have been reported for this protein .

What controls should I include when working with GFER antibodies?

When designing experiments using GFER antibodies, include the following controls:

  • Positive tissue/cell controls:

    • Human liver tissue for immunohistochemistry

    • HepG2 and Hep3B cell lysates for Western blotting

    • For gene expression studies, human tissues from the Human Total Master Panel II have been validated

  • Loading/housekeeping controls:

    • GAPDH has been validated as a housekeeping gene for qPCR normalization in GFER mRNA quantification

    • For Western blotting, standard loading controls such as β-actin or GAPDH are appropriate

  • Negative controls:

    • Primary antibody omission

    • Non-specific IgG of the same species and isotype as the GFER antibody

  • Recombinant protein controls:

    • E. coli-derived recombinant human GFER/ALR (Arg82-Asp205) has been used as a positive control in antibody validation

How does GFER regulate hematopoietic stem cell (HSC) function and quiescence?

GFER plays a critical role in regulating HSC proliferation and maintaining stem cell quiescence through a complex molecular mechanism:

  • GFER promotes quiescence and maintains the functional integrity of HSCs by restricting unwarranted proliferation

  • Mechanistically, GFER binds to the COP9 signalosome subunit Jab1, preventing its association with p27^kip1^

  • This interaction leads to stabilization and nuclear retention of p27^kip1^, a key cell cycle inhibitor

  • Knockdown (KD) of Gfer in cKit+Sca1+Flk2-CD34-Lineage- (KLS) cells results in:

    • Loss of cell quiescence

    • Compromised ability to engraft in the bone marrow of lethally irradiated recipient mice

    • Significant reduction in p27^kip1^ levels

  • Conversely, overexpression of Gfer elevates the level and nuclear retention of p27^kip1^

This Gfer-mediated pro-quiescence mechanism could potentially be therapeutically exploited in the treatment of hematological malignancies associated with elevated Jab1 and reduced p27^kip1^ levels .

What methods are most effective for studying GFER mutations in relation to mitochondrial diseases?

When investigating GFER mutations in relation to mitochondrial diseases, researchers should employ a multifaceted methodological approach:

  • Genetic analysis:

    • Retrotranscribe wild-type GFER cDNA from control muscle tissue for comparative studies

    • Clone the cDNA into expression vectors (e.g., pAcGFP1-N2) for functional studies

    • Propagate recombinant vectors in ultracompetent E. coli cells for stability

  • Cell-based functional studies:

    • Use patient and control primary fibroblasts for transfection experiments

    • Consider electroporation with the Human Dermal Fibroblast kit for efficient transfection

    • Establish stable cell lines (e.g., HEK293) expressing wild-type and mutated GFER cDNA for comparative studies

    • Evaluate transfection rates and expression using fluorescence microscopy and FACS analysis

  • mRNA quantification:

    • Reverse transcribe total RNA and quantify by qPCR using the ΔΔCt method

    • Use appropriate gene expression assays (e.g., Taqman Hs00193365_m1 for GFER)

    • Include GAPDH as a control housekeeping gene (Hs99999905_m1)

    • Compare expression in patient tissues with control human tissues

  • Mitochondrial function assessment:

    • Evaluate complex IV activity in GFER mutants

    • Assess genetic stability of mtDNA

    • Examine mitochondrial morphological changes using appropriate imaging techniques

These methods have successfully established GFER's role in the human Disulfide Relay System (DRS) and enhanced understanding of the pathogenesis of a novel mitochondrial disease .

What are the key considerations for selecting the optimal GFER antibody for specific research applications?

Selecting the optimal GFER antibody requires careful consideration of several factors based on your specific research application:

ApplicationHostClonalityEpitope RegionValidated SamplesRecommended Dilution
Western BlotRabbitPolyclonalFull length or C-terminalHepG2, Hep3B cells1:500-1:2000
Western BlotMouseMonoclonalArg82-Asp205HepG2, Hep3B cells2 μg/mL
IHCRabbitPolyclonalVariousHuman liver, thyroid cancer1:50-1:200
IHCMouseMonoclonalArg82-Asp205Human liver15 μg/mL

Key selection considerations include:

  • Target region specificity:

    • For full-length GFER detection, antibodies targeting amino acids 1-198 are available

    • For specific isoform detection, choose antibodies targeting unique regions (e.g., C-terminal epitopes)

    • For functional studies, consider antibodies targeting the functional domains (e.g., AA 81-205)

  • Host species and cross-reactivity:

    • Rabbit polyclonal antibodies typically offer reactivity across human, mouse, and rat samples

    • Mouse monoclonal antibodies may offer higher specificity but potentially more limited cross-reactivity

  • Validation evidence:

    • Prioritize antibodies with documented validation in your specific application and cell/tissue type

    • Consider antibodies with multiple validation methods (WB, IHC, ICC)

  • Buffer and storage requirements:

    • Most GFER antibodies are supplied in phosphate buffered solutions (pH 7.4) with glycerol and require -20°C storage

    • Avoid repeated freeze/thaw cycles to maintain antibody performance

How can I troubleshoot common issues when using GFER antibodies for mitochondrial localization studies?

When conducting mitochondrial localization studies with GFER antibodies, researchers may encounter several challenges. Here are methodological approaches to troubleshoot common issues:

  • Poor mitochondrial signal specificity:

    • Use heat-induced epitope retrieval with Antigen Retrieval Reagent-Basic before antibody incubation

    • Perform overnight incubation at 4°C to enhance specific binding

    • Co-stain with established mitochondrial markers to confirm localization

    • Use super-resolution microscopy techniques for better resolution of mitochondrial structures

  • High background in immunohistochemistry:

    • Optimize blocking conditions (5% BSA or serum from the same species as the secondary antibody)

    • Reduce primary antibody concentration (start with 15 μg/mL for monoclonal antibodies)

    • Use chromogenic detection systems optimized for mitochondrial proteins (e.g., HRP-DAB Cell & Tissue Staining Kit)

    • Counter-stain with hematoxylin for better visualization of cellular structures

  • Multiple bands in Western blot:

    • Verify if bands represent different isoforms (GFER has up to two reported isoforms)

    • Use reducing conditions to eliminate non-specific disulfide interactions

    • Increase washing stringency to reduce non-specific binding

    • Consider using more specific monoclonal antibodies if polyclonal antibodies show excessive cross-reactivity

  • Inconsistent results across different cell lines:

    • Be aware that GFER expression varies across tissue types

    • Validate antibody performance in each new cell line before conducting full experiments

    • Use positive control cell lines like HepG2 or Hep3B where GFER detection has been validated

What methodologies can be used to study GFER's role in the mitochondrial disulfide relay system?

Investigating GFER's role in the mitochondrial disulfide relay system (DRS) requires specialized techniques targeting protein-protein interactions and redox biochemistry:

  • Protein-protein interaction studies:

    • Identify interaction partners such as Jab1 using co-immunoprecipitation techniques

    • Use fluorescence resonance energy transfer (FRET) to visualize interactions in live cells

    • Apply proximity ligation assays to detect and quantify protein associations in fixed cells

  • Functional rescue experiments:

    • Transfect wild-type GFER cDNA into cells with GFER mutations or knockdown

    • Evaluate mitochondrial function recovery through:

      • Complex IV activity measurements

      • mtDNA stability assessments

      • Mitochondrial morphology analysis

  • Redox state analysis:

    • Employ redox proteomics to identify substrates of GFER's sulfhydryl oxidase activity

    • Use redox-sensitive fluorescent proteins to monitor real-time changes in mitochondrial redox state

    • Apply mass spectrometry to identify oxidation states of GFER substrates

  • Genetic manipulation approaches:

    • Create stable cell lines overexpressing wild-type or mutated GFER for comparative studies

    • Use CRISPR-Cas9 to generate GFER knockout models

    • Apply inducible expression systems to study temporal effects of GFER activity

  • Mitochondrial function assessment:

    • Measure oxidative phosphorylation efficiency

    • Assess maintenance of mitochondrial genomes

    • Evaluate effects on the cell division cycle

These methodologies have successfully established that mutations in GFER can reproduce complex IV activity defects, cause genetic instability of mtDNA, and lead to mitochondrial morphological defects, contributing to our understanding of novel mitochondrial diseases .

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.