HLA-G Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to HLA-G Recombinant Monoclonal Antibodies

HLA-G recombinant monoclonal antibodies are engineered immunoglobulins designed to target the human leukocyte antigen G (HLA-G), a non-classical MHC-I molecule critical for immune tolerance. These antibodies enable precise detection, functional blocking, and therapeutic modulation of HLA-G, which is expressed in placental trophoblasts, regulatory immune cells, and pathologically in cancers and autoimmune diseases . Unlike classical HLA molecules, HLA-G exhibits restricted tissue expression and immunosuppressive properties, making it a key target for studying immune evasion and tolerance mechanisms .

Functional Blocking Assays

HLA-G monoclonal antibodies are used to disrupt immune modulation in vitro:

  • NK Cell Activity: 87G blocks HLA-G1 on melanoma cells, restoring NK-mediated lysis .

  • Dendritic Cell (DC) Maturation: Anti-HLA-G antibodies prevent DC-induced Treg expansion, enhancing T cell responses .

  • Cancer Immunotherapy: IDO-1 upregulation in triple-negative breast cancer (TNBC) correlates with HLA-G expression, which is neutralized by anti-HLA-G antibodies to restore NK function .

Detection and Quantification

MethodAntibodyKey FindingsSource
Flow Cytometry87G, 17A5Detects HLA-G+ T cells in HIV patients; identifies placental trophoblasts .
ImmunohistochemistryHLAG/8344RLocalizes HLA-G in placental syncytiotrophoblasts and bladder carcinoma .
ELISAMEM-G9/G233Measures soluble HLA-G (sHLA-G) in autoimmune disease and cancer sera; correlates with clinical outcomes .

Transplantation and Autoimmune Research

  • Graft Survival: B2M-HLA-G5 recombinant protein (detected via anti-HLA-G antibodies) delays allograft rejection in murine models .

  • Pre-eclampsia: Reduced CD4+HLA-G+ T cells in pre-eclamptic patients are identified using flow cytometry with HLA-G-specific antibodies .

Immune Evasion in Cancer

HLA-G expression on tumor cells (e.g., melanoma, breast cancer) suppresses NK and T cell responses. Antibodies like 87G and G233 reverse this inhibition, enhancing antitumor immunity . For example:

  • HER2+ Breast Cancer: HLA-G upregulation in trastuzumab-resistant tumors is linked to NK receptor KIR2DL4 engagement, which is disrupted by anti-HLA-G blockade .

Receptor Specificity and Isoform Differences

  • LILRB1 vs. LILRB2: MEM-G9/G233 bind HLA-G1 without overlapping LILRB1’s binding site, while MEM-G1 competes with LILRB2 for HLA-G2 .

  • Therapeutic Limitations: Current antibodies (e.g., MEM-G9) lack reactivity to HLA-G2/G3/G4, limiting their utility in detecting all isoforms .

Challenges and Future Directions

ChallengeSolution/Research FocusSource
Isoform-Specific DetectionDeveloping antibodies targeting α3 domain (G1/G2/G3/G4) or α1 domain (G1/G5/G6) .
Clinical TranslationOptimizing recombinant antibodies for therapeutic use (e.g., blocking HLA-G in cancer) .
Epitope MappingStructural studies to identify conformation-specific epitopes for improved targeting .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The HLA-G recombinant monoclonal antibody is meticulously produced through a well-defined process. It begins with the in vitro cloning of HLA-G antibody genes, seamlessly integrated into expression vectors. These vectors are then transfected into host cells, providing an environment conducive to the expression of the recombinant antibody within a cell culture setting. Subsequently, the HLA-G recombinant monoclonal antibody undergoes affinity chromatography purification.

This antibody exhibits specific binding to the human HLA-G protein in ELISA and FC assays. HLA-G plays a crucial role as an immunomodulatory molecule, involved in immune tolerance, immune regulation, and immune evasion. Its functions are particularly significant during pregnancy, in cancer biology, and in various pathological conditions where the regulation or suppression of immune responses is essential.

Form
Liquid
Lead Time
Generally, we can dispatch the products within 1-3 business days after receiving your orders. Delivery timelines may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
Synonyms
HLA class I histocompatibility antigen, alpha chain G (HLA G antigen) (MHC class I antigen G), HLA-G, HLA-6.0 HLAG
Target Names
Uniprot No.

Target Background

Function

HLA-G is a non-classical major histocompatibility class Ib molecule that plays a critical role in immune regulatory processes at the maternal-fetal interface. It forms a complex with B2M (beta-2 microglobulin) and binds a limited repertoire of nonamer self-peptides derived from intracellular proteins, including histones and ribosomal proteins.

This peptide-bound HLA-G-B2M complex acts as a ligand for inhibitory/activating KIR2DL4, LILRB1, and LILRB2 receptors on uterine immune cells, contributing to fetal development while maintaining maternal-fetal tolerance. The interaction with KIR2DL4 and LILRB1 receptors on decidual NK cells triggers NK cell senescence-associated secretory phenotype, acting as a molecular switch to promote vascular remodeling and fetal growth in early pregnancy. Interaction with KIR2DL4 receptors on decidual macrophages induces proinflammatory cytokine production, primarily associated with tissue remodeling.

Furthermore, interaction with LILRB2 receptors triggers the differentiation of type 1 regulatory T cells and myeloid-derived suppressor cells, both of which actively maintain maternal-fetal tolerance. HLA-G reprograms B cells towards an immune suppressive phenotype via LILRB1. It may induce immune activation/suppression via intercellular membrane transfer (trogocytosis), potentially enabling interaction with KIR2DL4, which predominantly resides in endosomes.

Through interaction with the inhibitory receptor CD160 on endothelial cells, HLA-G may regulate angiogenesis in immune-privileged sites. While it likely does not bind B2M and presents peptides, it negatively regulates NK cell- and CD8+ T cell-mediated cytotoxicity. It may play a role in balancing tolerance and antiviral-immunity at the maternal-fetal interface by controlling the effector functions of NK, CD8+ T cells, and B cells.

Gene References Into Functions
  1. HLA-G, NRP1, and PD-1 may be involved in the immune response in psoriatic patients. PMID: 29790686
  2. Given that other 14-bp associations were inconclusive and that other variation sites observed at HLA-G 3'UTR exhibit a proven role in post-transcriptional regulation of HLA-G expression, the complete 3'UTR segment should be analyzed in terms of disease susceptibility, instead of a single polymorphism. PMID: 30102938
  3. A study demonstrated that HLA-G 14-bp Ins/Del polymorphism may exert no influence on susceptibility to viruses - Meta-analysis. PMID: 30235670
  4. The findings indicate that the GPER/miR148a/HLAG signaling pathway may mediate the development of ovarian endometriosis and could be a potential therapeutic target for the treatment of endometriosis. PMID: 29845209
  5. Significantly different distribution of HLA-G polymorphism (rs1611715), but not the serum level of sHLA-G, were found between multiple sclerosis patients and healthy individuals. PMID: 29924453
  6. These results highlight the importance of HLA-G promoter SNPs in pregnancy outcomes. However, to reach a more definite conclusion, subsequent studies on 3' UTR and other positions with polymorphism in the 5' UTR regions with larger sample sizes are necessary. PMID: 29797531
  7. HLA-G expression is associated with poor survival in stage III gastric cancer patients and represents a possible immunoescape mechanism of cancer cells. PMID: 29845360
  8. This study shows that HLA-G molecules have a role in predicting the newborn's likelihood of oral HPV infection at birth. PMID: 29544814
  9. HLA-G is highly represented in ovarian carcinoma, suggesting a potential association with progressive disease mechanisms. PMID: 29499226
  10. This study shows that HLA-G is expressed in intestinal samples of ulcerative colitis and Crohn's disease patients. PMID: 29588183
  11. This study shows that HLA-G mediated immune regulation is impaired by a single amino acid exchange in the alpha 2 domain. PMID: 29605689
  12. No significant differences in single nucleotide polymorphisms in the 3' untranslated region of HLA-G in placentas between spontaneous preterm birth and preeclampsia. PMID: 29540242
  13. The HLA-G 14bp ins/del gene polymorphism is an important risk factor for the incidence and poor outcome of NHL cases. PMID: 29388862
  14. These findings showed that HLA-G overexpression in tumor tissue correlated with poor prognosis in pancreatic adenocarcinoma. PMID: 29549230
  15. This meta-analysis suggested that the HLA-G 14-bp insertion allele may increase the risk of recurrent implantation failure in Caucasians. PMID: 28707147
  16. A study shows that HLA-G alleles are skewed in autism spectrum disorder (ASD) children. HLA-G alleles seen in ASD result in immune activation. The skewing of HLA-G alleles seen in ASD might play a pathogenic role. PMID: 28923404
  17. These data confirm an important role of HLA-G 3'UTR polymorphisms in the development of severe forms of sepsis in Brazil. PMID: 28941746
  18. A 14bp indel in HLA-G associated with increased flow-mediated dilatation of the brachial artery in renal transplant recipients. PMID: 28987961
  19. The maternal HLA-G 14-bp Ins/Del polymorphism is not associated with preeclampsia risk. PMID: 29105301
  20. The HLA-G 14-bp Ins/Del polymorphism may be a marker for genetic susceptibility to chronic hepatitis B infection. PMID: 28929613
  21. The presence of the HLA-G +3142 CC genotype was associated with higher susceptibility to CMV infection after kidney transplantation. PMID: 29113092
  22. The results from the study suggest a possible involvement of HLA-G in the risk modulation toward hepatitis C virus infection. PMID: 28083985
  23. Our results suggest that elevated levels of HLA-G expression, specifically sHLA-G1 homodimers, are indeed associated with persistent infection with HBV. sHLA-G (sHLA-G1 and HLA-G5 both) levels may be a prognostic indicator for spontaneous recovery. PMID: 28429836
  24. Results show that HLA-G rs1233334:CT protected against progression of endometriosis. PMID: 29234882
  25. The results suggest that HLA-G polymorphism has a small effect on systemic lupus erythematosus susceptibility and that soluble HLA-G may be involved in the pathogenesis of the disease. PMID: 28695673
  26. sHLA-G levels is an independent prognostic factor and improves the prognostic stratification offered by traditional prognosticators in CRC patients. PMID: 28415627
  27. We found a significant association between HLA-G rs1063320 (thorn3142G>C) and 14-bp ins/del variants and risk of recurrent spontaneous abortion. PMID: 28600033
  28. HLA-G del/del genotypes were more frequent among mothers with gestational diabetes than in the control group. Babies carrying HLA-G del/del showed the highest sHLA-G levels. PMID: 28655969
  29. No statistically significant differences in either HLA-G allele or genotype frequencies between pre-eclampsia cases and the control group have been observed. PMID: 29061057
  30. Overexpressed HLA-G revealed its ability to inhibit the cell proliferation and cytotoxic activity of NK-92MI cells, and reduce the secretion of IFN-gamma and TNF-alpha through immunoglobulin-like transcript 2. PMID: 29224003
  31. Soluble HLA-G was significantly higher in the persistent wheezing (positive modified asthma predictive index) infant group compared with the transient wheezing (negative modified asthma predictive index) group (P = 0.008). PMID: 27880031
  32. This study shows associations between fetal HLA-G genotype and birth weight and placental weight in a large cohort of pregnant women. PMID: 28267635
  33. Evidence is presented that HLA-G1 monomer exhibits therapeutic effects on atopic dermatitis-like skin lesions in mice. PMID: 28675838
  34. This study shows that HLA-G is expressed by ARPE-19 retinal pigment epithelium cells and is upregulated as a response to pro-inflammatory cytokines. PMID: 28442288
  35. Cellular activation and pathological processes are associated with an aberrant or a neo-expression of HLA-G/soluble HLA-G. (Review). PMID: 27440734
  36. The +3142 C>G, but not the 14bp INS/DEL polymorphism may constitute a genetic susceptibility factor to multiple sclerosis in the Tunisian population. PMID: 27771469
  37. Novel HLA-G-regulating miRs, miR-628-5p and miR-548q, were identified in renal cell carcinoma. PMID: 27057628
  38. High expression of total soluble HLA-G and vesicular soluble HLA-G, together with the presence of the 14-bp deletion allele, might be involved in implantation failure. PMID: 26959830
  39. The presented data suggest that the investigated HLA-G*0105N allele is potentially associated with recurrent spontaneous abortions through linkage disequilibrium with other genetic elements. PMID: 26754761
  40. These findings demonstrate the association of HLA-G polymorphism with breast cancer susceptibility in the Tunisian population. PMID: 26754763
  41. Results strongly suggest the involvement of HLA-G in human African trypanosomiasis disease progression. PMID: 27470243
  42. Women with preterm premature rupture of membranes had significantly higher serum and vaginal sHLA-G concentrations compared to controls. PMID: 27232355
  43. Serum levels of soluble HLA-G were similar in CMV- and CMV+ subjects, but levels in culture supernatants were significantly higher in cells from CMV+ than from CMV- subjects stimulated with CMV antigens. The HLA-G ligand KIR2DL4 was mainly expressed on NK cells and CD56+ T cells with no differences between CMV+ and CMV- subjects. PMID: 28817184
  44. Our study is the first to suggest that HLA-G expression may influence the clinical outcome of chronic myeloid leukemia. PMID: 28841441
  45. SV40 immune-inhibitory direct effect and the presence of high levels of the immune-inhibitory HLA-G molecules could cooperate in impairing B lymphocyte activation towards SV40-specific peptides. PMID: 27443345
  46. HLA-G 3'UTR polymorphisms associated with a greater magnitude of HLA-G production were associated with differentiated thyroid tumors and with variables implicated in poor prognosis. PMID: 27914217
  47. The inhibitory properties of bronchial endothelial cells are dysregulated in stable lung transplant recipients via TGF-beta, IL-10, and HLA-G signaling pathway. PMID: 27820781
  48. HLA-G expression was upregulated in chondrocyte-differentiated mesenchymal stem cells under hypoxia context and could be boosted in allogenic settings. PMID: 27465875
  49. This review provides new insights into the mechanisms controlling the expression and function of HLA-G at the maternal-fetal interface, and discusses their relevance for fetal tolerance. PMID: 28279591
  50. We propose a massively parallel sequencing (NGS) with a bioinformatics strategy to evaluate the entire HLA-G regulatory and coding segments. PMID: 28135606

Show More

Hide All

Database Links

HGNC: 4964

OMIM: 142871

KEGG: hsa:3135

STRING: 9606.ENSP00000353472

UniGene: Hs.512152

Protein Families
MHC class I family
Subcellular Location
[Isoform 1]: Cell membrane; Single-pass type I membrane protein. Endoplasmic reticulum membrane. Early endosome membrane.; [Soluble HLA class I histocompatibility antigen, alpha chain G]: Secreted.; [Isoform 2]: Cell membrane; Single-pass type I membrane protein.; [Isoform 3]: Cell membrane; Single-pass type I membrane protein.; [Isoform 4]: Cell membrane; Single-pass type I membrane protein.; [Isoform 5]: Secreted. Early endosome.; [Isoform 6]: Secreted.; [Isoform 7]: Secreted.; Cell projection, filopodium membrane.
Tissue Specificity
Expressed in adult eye. Expressed in immune cell subsets including monocytes, myeloid and plasmacytoid dendritic cells and regulatory T cells (Tr1)(at protein level). Secreted by follicular dendritic cell and follicular helper T cells. Isoform 5: Detected

Q&A

What is HLA-G and why is it significant in immunology research?

HLA-G is a non-classical MHC class I molecule characterized by restricted tissue expression, low polymorphism, and multiple immunoregulatory properties. Unlike classical HLA molecules, HLA-G plays a specialized role in immune tolerance rather than antigen presentation. Its significance stems from its capacity to inhibit allogeneic proliferation of CD4+ T cells, suppress natural killer (NK) and CD8+ T cell cytotoxicity, prevent dendritic cell maturation, and inhibit B cell activation . HLA-G can also trigger apoptosis in antigen-specific CD8+ T lymphocytes, representing a crucial mechanism in immune regulation . These properties have established HLA-G as an emerging "immune checkpoint" molecule with significant implications for reproductive immunology, transplantation, tumor immunology, and autoimmune disease research .

What are the known isoforms of HLA-G and how are they detected?

HLA-G exists in seven documented isoforms (HLA-G1 through HLA-G7), with variable expression patterns and functional properties. Different monoclonal antibodies recognize specific isoforms, which is crucial for experimental design:

Antibody CloneRecognized IsoformsApplicationsSpecial Characteristics
BFL.1HLA-G1 (full-length)Flow cytometryDistinguishes between classical HLA-A/B and non-classical HLA-G
MEM-G/9HLA-G1Flow cytometrySuitable for transfected cell lines, PE-conjugated
MEM-G/1Denatured soluble and membrane-bound HLA-GWestern blotting, IHCRecognizes HLA-G under reducing conditions
87GHLA-G1 and soluble HLA-G5Flow cytometryPre-titrated for flow cytometric analysis

Most commercially available antibodies detect the HLA-G heavy chain, which has a molecular weight of approximately 39 kDa for the full-length HLA-G1 isoform . When designing experiments to detect specific isoforms, researchers must carefully select the appropriate antibody based on its recognition properties and the experimental methodology.

How do recombinant monoclonal antibodies to HLA-G differ from traditional monoclonal antibodies?

Recombinant monoclonal antibodies to HLA-G offer several advantages over traditional hybridoma-derived antibodies. Traditional HLA-G monoclonal antibodies like BFL.1 are produced through hybridoma technology, involving immunization of transgenic mice (such as HLA-B27/human beta 2-microglobulin double-transgenic mice) with transfected cells expressing HLA-G . In contrast, recombinant antibodies are generated using molecular cloning techniques where antibody genes are isolated, sequenced, and expressed in controlled expression systems.

The recombinant approach offers several research advantages:

  • Reduced batch-to-batch variability, ensuring consistent experimental results

  • Precise engineering of antibody properties, including affinity, specificity, and Fc region functionality

  • Greater reproducibility in binding characteristics

  • Potential for humanization, reducing background in human tissue studies

  • Ability to create fusion proteins with reporter molecules or therapeutic agents

When selecting between traditional and recombinant HLA-G antibodies, researchers should consider these differences in relation to their specific experimental requirements.

What are the primary research applications of HLA-G monoclonal antibodies?

HLA-G monoclonal antibodies serve diverse research applications across multiple fields:

Research AreaApplicationMethodologyKey Considerations
Reproductive ImmunologyStudying maternal-fetal toleranceIHC of placental tissues, Flow cytometry of trophoblastsFocus on extravillous cytotrophoblast cells
Tumor ImmunologyInvestigating immune escape mechanismsIHC of tumor tissues, Flow cytometry of tumor cellsCompare with normal tissue controls
TransplantationMonitoring graft toleranceFlow cytometry, IHCCorrelation with clinical outcomes
Autoimmune DiseaseExamining regulatory mechanismsFlow cytometry of immune cell subsetsAnalyze HLA-G+ CD4+ and CD8+ T cells
Basic ResearchCharacterizing isoform expressionWestern blotting, Flow cytometrySelect antibodies recognizing specific isoforms

Each application requires careful selection of the appropriate antibody clone and detection method based on the specific research question and sample type.

Which HLA-G monoclonal antibody clone should I select for specific experimental applications?

Selecting the appropriate HLA-G monoclonal antibody depends critically on your experimental technique and research question. Based on validated applications, consider the following methodological recommendations:

For flow cytometry:

  • MEM-G/9 (ab24384): Optimal for detecting native HLA-G1 on cell surfaces. Particularly effective with transfected cell lines and has been validated in 12 publications .

  • 87G: Pre-titrated (5 μL/0.25 μg per test) for flow cytometric analysis of stimulated U937 cells. Recognizes both HLA-G1 and soluble HLA-G5 isoforms .

  • BFL.1: Specifically recognizes membrane-bound HLA-G on trophoblasts and HLA-G-expressing cell lines while avoiding cross-reactivity with classical HLA molecules .

For Western blotting:

  • MEM-G/1: Specifically designed for detecting denatured HLA-G heavy chain under reducing conditions. Optimal dilution for Western blotting is 1:60-1:100 .

  • Proteintech 16913-1-AP: Detects HLA-G in Western blotting with recommended dilutions of 1:1000-1:4000 .

For immunohistochemistry (paraffin sections):

  • MEM-G/1: Requires heat-mediated antigen retrieval using sodium citrate buffer (pH 6.0) with dilutions of 1:60-1:100. Optimal for detecting HLA-G in placental tissue, particularly in extravillous cytotrophoblast cells .

  • Proteintech 16913-1-AP: Validated for immunohistochemistry applications with human samples .

What are the optimal protocols for using HLA-G antibodies in flow cytometry?

To achieve optimal results when using HLA-G antibodies in flow cytometry, follow this methodological framework:

  • Sample preparation:

    • For cell lines: Harvest cells in logarithmic growth phase

    • For primary cells: Isolate cells using density gradient centrifugation

    • Wash cells twice in PBS containing 2% FBS to reduce background

  • Staining protocol:

    • For MEM-G/9 PE-conjugated antibody: Use 5-10 μL per 1×10^6 cells in 100 μL staining buffer

    • For 87G PE-conjugated antibody: Use precisely 5 μL (0.25 μg) per test as pre-titrated for flow cytometric analysis

    • Incubate for 30 minutes at 4°C in the dark

    • Wash twice with staining buffer

    • Resuspend in 300-500 μL of staining buffer for acquisition

  • Crucial controls:

    • Isotype control (matching the antibody's isotype - typically IgG1 for anti-HLA-G clones)

    • FMO (Fluorescence Minus One) controls

    • Positive control: JEG-3 or HLA-G-transfected cell lines like LCL-HLA-G1

    • Negative control: Cell lines known to be HLA-G negative, such as untransfected L cells

  • Data analysis considerations:

    • Gate on live, single cells

    • Report both percentage of positive cells and mean fluorescence intensity

    • For HLA-G expression on specific immune subsets, use appropriate lineage markers in combination with HLA-G staining

Validation experiments have demonstrated successful surface staining of HLA-G1 transfectants (LCL-HLA-G1) using anti-HLA-G (MEM-G/9) PE, confirming the efficacy of this methodological approach .

What are the recommended approaches for validating HLA-G antibody specificity?

Validating HLA-G antibody specificity is crucial for experimental rigor and requires multiple complementary approaches:

  • Positive and negative control cell lines:

    • Positive controls: JEG-3 and HLA-G-transfected JAR human choriocarcinoma cell lines naturally express HLA-G

    • Negative controls: Parental untransfected L cells and HLA-B7/HLA-A3-transfected L cells should not show reactivity

    • Human cell lines known to express classical HLA class I proteins but not HLA-G serve as critical specificity controls

  • Biochemical validation:

    • Immunoprecipitation of biotinylated membrane lysates from HLA-G-expressing cell lines should yield a 39-kDa protein (full-length HLA-G1)

    • Compare results with W6/32 mAb, which immunoprecipitates both classical and non-classical HLA class I heavy chains

    • Verify protein size matches predicted molecular weight: 38 kDa calculated, typically observed at 33-45 kDa range due to post-translational modifications

  • Blocking experiments:

    • Pre-incubate antibody with recombinant HLA-G to demonstrate specific binding inhibition

    • Include peptide competition assays where relevant

  • Cross-reactivity assessment:

    • Test against cells expressing other HLA class I molecules to confirm absence of binding

    • Verify antibody doesn't recognize classical HLA-A and HLA-B molecules

  • Genetic validation:

    • Use cells with confirmed HLA-G expression through RT-PCR or RNA-seq as additional controls

    • Consider using HLA-G knockout or knockdown systems where available

Following these comprehensive validation steps ensures that experimental findings using HLA-G antibodies truly reflect HLA-G biology rather than non-specific interactions.

How should immunoprecipitation with HLA-G antibodies be performed?

For successful immunoprecipitation (IP) of HLA-G proteins, follow this optimized protocol based on established methodologies:

  • Cell preparation and lysis:

    • Culture HLA-G-expressing cells (JEG-3, transfected cell lines) to 80-90% confluence

    • For membrane protein analysis, perform surface biotinylation before lysis

    • Lyse cells in buffer containing 1% NP-40 or equivalent detergent, supplemented with protease inhibitors

    • Centrifuge lysate at 14,000g for 15 minutes at 4°C to remove debris

  • Antibody selection and pre-clearing:

    • For full-length HLA-G1 isoform: Use BFL.1 or W6/32 antibodies

    • For denatured HLA-G: Use MEM-G/1 antibody

    • Pre-clear lysate with Protein A/G beads for 1 hour at 4°C to reduce non-specific binding

  • Immunoprecipitation procedure:

    • Add 2-5 μg of HLA-G antibody to 500 μL of cleared lysate

    • Incubate overnight at 4°C with gentle rotation

    • Add 50 μL of Protein A/G beads and incubate for 2-4 hours at 4°C

    • Wash beads 4-5 times with lysis buffer

    • Elute proteins by boiling in SDS sample buffer for 5 minutes

  • Analysis of immunoprecipitated products:

    • Resolve proteins by SDS-PAGE

    • For biotinylated surface proteins, detect with streptavidin-HRP

    • For total HLA-G detection, perform Western blotting with a different HLA-G antibody clone

    • Expected molecular weight for full-length HLA-G1: 39 kDa

This approach has successfully demonstrated that BFL.1 antibody, like W6/32, immunoprecipitates a 39-kDa protein from HLA-G-expressing cell lines, corresponding to the full-length HLA-G1 isoform, while crucially not recognizing classical HLA class I products .

How can HLA-G antibodies be used to investigate maternal-fetal immune tolerance mechanisms?

HLA-G plays a pivotal role at the maternal-fetal interface, creating immune tolerance essential for successful pregnancy. Research approaches using HLA-G antibodies include:

  • Placental tissue analysis:

    • Immunohistochemistry using MEM-G/1 antibody (1:60-1:100 dilution) with heat-mediated antigen retrieval (sodium citrate buffer, pH 6.0) can detect HLA-G in extravillous cytotrophoblast cells

    • Compare HLA-G expression patterns between normal pregnancies and those with complications (preeclampsia, recurrent miscarriage)

    • Correlate HLA-G expression with immune cell infiltration patterns

  • Cytotrophoblast isolation and characterization:

    • Use BFL.1 antibody for flow cytometric analysis of first-trimester placental cytotrophoblast cells

    • Quantify the percentage of HLA-G-positive cytotrophoblasts and correlation with invasion capacity

    • Analyze HLA-G expression during trophoblast differentiation using multiple antibody clones to detect different isoforms

  • Maternal-fetal interface immune profiling:

    • Apply 87G antibody to identify HLA-G-expressing cellular subsets at the decidua

    • Correlate HLA-G expression with presence of regulatory T cells and decidual NK cells

    • Investigate interactions between HLA-G+ trophoblasts and maternal immune cells using co-culture systems

  • Soluble HLA-G quantification:

    • Measure sHLA-G levels in maternal serum and correlate with pregnancy outcomes

    • Examine the relationship between membrane-bound and soluble HLA-G forms

    • Investigate the immunomodulatory effects of sHLA-G on maternal immune cells

The key methodological consideration is selecting appropriate antibodies that recognize specific HLA-G isoforms relevant to the maternal-fetal interface, as different isoforms may have distinct roles in creating the immunotolerant microenvironment necessary for successful pregnancy.

What methodological considerations are essential when studying HLA-G expression in tumor immunology?

When investigating HLA-G expression in tumor contexts, several methodological considerations are critical for robust and reproducible results:

  • Tissue sample selection and processing:

    • Include both tumor center and invasive margin samples

    • Process tissues consistently to avoid variable fixation effects on epitope preservation

    • For paraffin sections, standardize antigen retrieval methods (sodium citrate buffer pH 6.0 recommended for MEM-G/1)

    • Include adjacent normal tissue as internal controls

  • Antibody selection strategy:

    • For tissue sections: Use MEM-G/1 (1:60-1:100) with appropriate antigen retrieval

    • For flow cytometry of cell suspensions: Use MEM-G/9 or 87G to detect native conformations

    • For Western blotting: Employ Proteintech 16913-1-AP (1:1000-1:4000) to detect denatured HLA-G

    • Consider using multiple antibody clones to validate findings

  • Interpretation and quantification protocols:

    • Score both percentage of positive cells and staining intensity

    • Evaluate membrane and cytoplasmic staining separately

    • Correlate with immune cell infiltration patterns (CD8+ T cells, NK cells)

    • Assess relationship with other immune checkpoint molecules

  • Functional validation approaches:

    • Co-culture HLA-G+ tumor cells with immune effector cells

    • Measure cytotoxicity inhibition and cytokine production modulation

    • Perform HLA-G blocking experiments to confirm functional relevance

    • Consider HLA-G knockdown/knockout studies to demonstrate causality

The aberrant expression of HLA-G has been documented in various human neoplastic diseases including melanoma, breast carcinoma, renal carcinoma, chronic lymphocytic leukemia (CLL), acute myeloid leukemia (AML), and B-CLL . HLA-G plays a significant role in tumor immune escape mechanisms, potentially allowing cancer cells to evade immune surveillance . This understanding underscores the importance of methodologically rigorous approaches to studying HLA-G in tumor contexts.

How can the interaction between HLA-G and its receptors be investigated using recombinant antibodies?

Investigating HLA-G interactions with its receptors (CD85j/ILT2, CD85d/ILT4, and CD158) requires sophisticated methodological approaches:

  • Co-immunoprecipitation studies:

    • Use anti-HLA-G antibodies (BFL.1, MEM-G/9) to pull down HLA-G complexes from cells expressing both HLA-G and its receptors

    • Perform reverse co-IP with receptor-specific antibodies

    • Western blot to detect interacting partners

    • Include appropriate controls (isotype antibodies, HLA-G-negative cells)

  • Flow cytometry-based binding assays:

    • Generate recombinant soluble forms of HLA-G

    • Label with fluorescent dyes or biotin

    • Measure binding to receptor-expressing cells by flow cytometry

    • Perform competition assays with unlabeled HLA-G or anti-receptor antibodies

  • Microscopy approaches for visualizing interactions:

    • Immunofluorescence co-localization of HLA-G and receptors

    • Proximity ligation assay (PLA) to detect protein-protein interactions in situ

    • Live-cell imaging with fluorescently tagged proteins

  • Functional assays:

    • Measure inhibition of NK cell cytotoxicity or T cell proliferation

    • Assess the impact on dendritic cell maturation

    • Evaluate cytokine production shifts toward anti-inflammatory profiles

    • Use receptor blocking antibodies or HLA-G blocking antibodies to confirm specificity

HLA-G exhibits immunomodulatory effects through its interaction with these receptors, including inhibition of allogeneic proliferation of CD4+ T cells, NK and CD8+ T cell cytotoxicity, maturation of dendritic cells, and activation of B cells . Understanding these interactions mechanistically is crucial for developing potential therapeutic approaches targeting the HLA-G/receptor axis.

What approaches are most effective for studying HLA-G+ immune cells in autoimmune and inflammatory disorders?

To effectively study HLA-G-expressing immune cells in autoimmune and inflammatory contexts, researchers should implement the following methodological approaches:

  • Multi-parameter flow cytometry:

    • Combine HLA-G antibodies (87G or MEM-G/9) with lineage markers for T cells, B cells, NK cells, monocytes, and dendritic cells

    • Include functional markers (activation, exhaustion, regulatory phenotype)

    • Use optimal panel design to minimize spectral overlap

    • Apply standardized gating strategies across patient cohorts

  • Single-cell analysis techniques:

    • Single-cell RNA sequencing of sorted HLA-G+ immune cells

    • Spatial transcriptomics to map HLA-G+ cells within tissue microenvironments

    • Cytometry by time of flight (CyTOF) for high-dimensional phenotyping

  • Functional characterization protocols:

    • Isolate HLA-G+ CD4+ and HLA-G+ CD8+ T cells from peripheral blood

    • Assess proliferative capacity, cytokine production profiles

    • Evaluate suppressive functions on effector immune cells

    • Compare with conventional regulatory T cells

  • Longitudinal clinical correlations:

    • Monitor HLA-G+ immune cell frequencies during disease progression

    • Track changes in response to therapeutic interventions

    • Correlate with clinical parameters and biomarkers of disease activity

HLA-G+ CD4+ or CD8+ T cells identified in normal human peripheral blood are thought to function as regulatory cells, exhibiting hypoproliferative characteristics with a unique cytokine profile distinct from conventional Tregs . These cells have been reported in various autoimmune/inflammatory disorders, suggesting they play important immunoregulatory roles in these conditions . The presence and frequency of these cells may provide insights into disease mechanisms and potential therapeutic targets.

Why might I observe discrepancies in HLA-G detection between different antibody clones?

Discrepancies in HLA-G detection between antibody clones are common and can arise from several methodological factors:

  • Epitope recognition differences:

    • BFL.1 specifically recognizes conformational epitopes on HLA-G that distinguish it from classical HLA-A and -B molecules

    • MEM-G/1 recognizes denatured HLA-G epitopes, making it suitable for Western blotting but not native protein detection

    • MEM-G/9 binds native conformational HLA-G epitopes, ideal for flow cytometry of non-denatured samples

    • 87G recognizes both HLA-G1 and soluble HLA-G5 isoforms, potentially giving broader reactivity patterns

  • Isoform specificity variations:

    • HLA-G has seven isoforms (HLA-G1 to HLA-G7) with different structures and tissue distribution

    • BFL.1 and W6/32 immunoprecipitate a 39-kDa protein in HLA-G-expressing cell lines, corresponding to full-length HLA-G1

    • Some antibodies may not recognize truncated isoforms or soluble forms

    • Alternative splicing may affect epitope accessibility

  • Technical and sample preparation influences:

    • Fixation can alter epitope accessibility (critical for IHC/ICC applications)

    • Reducing conditions in Western blotting affect epitope recognition (MEM-G/1 specifically recognizes HLA-G under reducing conditions)

    • Different flow cytometry buffers may affect antibody binding efficiency

    • Freeze-thaw cycles can impact protein conformation and antibody recognition

  • Control-related considerations:

    • Inconsistent positive controls between experiments (JEG-3, transfected cell lines, primary trophoblasts)

    • Variable HLA-G expression levels in supposedly positive samples

    • Background staining differences between antibody clones and detection systems

When encountering discrepancies, validate findings using multiple detection methods and antibody clones, always including appropriate positive controls (JEG-3 or HLA-G-transfected cell lines) and negative controls (untransfected L cells or HLA-A/B-transfected L cells) .

How can I distinguish between membrane-bound and soluble forms of HLA-G?

Distinguishing between membrane-bound and soluble HLA-G forms requires specialized methodological approaches:

  • Antibody selection strategy:

    • 87G antibody recognizes both HLA-G1 (membrane-bound) and HLA-G5 (soluble)

    • Use antibodies specific to unique regions in soluble forms (absent transmembrane domain)

    • For comprehensive analysis, employ antibodies recognizing shared epitopes to detect total HLA-G

  • Cell surface versus intracellular staining:

    • Perform non-permeabilized staining to detect only membrane-bound forms

    • Follow with permeabilization and staining to detect intracellular/soluble forms

    • Calculate the difference to estimate relative distribution

  • Biochemical separation techniques:

    • Ultracentrifugation to separate membrane fractions from soluble proteins

    • Immunoprecipitation from different cellular fractions

    • Western blotting to detect size differences (HLA-G1: 39 kDa vs. smaller soluble isoforms)

  • ELISA-based approaches for soluble HLA-G:

    • Use capture antibodies recognizing shared epitopes

    • Detection antibodies specific for soluble forms

    • Compare with total HLA-G levels

    • Include appropriate standards and controls

The biological significance of different HLA-G forms varies: membrane-bound HLA-G1 on trophoblasts directly interacts with maternal immune cells to establish immune tolerance at the maternal-fetal interface , while soluble HLA-G5 can trigger apoptosis in antigen-specific CD8+ T lymphocytes and create an anti-inflammatory environment through IL-10 release . Understanding which form predominates provides crucial insights into the immunomodulatory mechanisms at play in different biological contexts.

What controls should I include when studying HLA-G expression in clinical samples?

When investigating HLA-G expression in clinical samples, a comprehensive control strategy is essential for valid interpretation:

  • Positive tissue controls:

    • Placental extravillous cytotrophoblast: The gold standard positive control for HLA-G expression

    • JEG-3 or HLA-G-transfected JAR human choriocarcinoma cell lines for cell-based assays

    • Specifically document subsections like extravillous trophoblast in placental samples

  • Negative tissue controls:

    • Adjacent normal tissue (for tumor studies)

    • Non-HLA-G-expressing cell lines: Untransfected L cells, HLA-B7 and HLA-A3-transfected L cells

    • Human cell lines known to express classical HLA class I proteins but not HLA-G

  • Methodological controls:

    • Isotype control antibodies matched to primary antibody class and concentration

    • Secondary antibody-only controls (for indirect detection methods)

    • Antibody dilution series to establish optimal signal-to-noise ratio

    • For IHC: Omit primary antibody, substitute non-relevant primary antibody

  • Analytical validation controls:

    • Multiple antibody clones targeting different HLA-G epitopes

    • Complementary detection methods (IHC, flow cytometry, Western blot)

    • mRNA detection (RT-PCR, in situ hybridization) to correlate with protein findings

    • Quantitative standards for consistent scoring across samples

When analyzing clinical samples, report both the percentage of positive cells and staining intensity. For immunohistochemistry of paraffin sections using MEM-G/1, implement heat-mediated antigen retrieval with sodium citrate buffer (pH 6.0) at dilutions of 1:60-1:100 . Document all control results alongside experimental findings to demonstrate technical validity.

How can I address potential cross-reactivity with classical HLA class I molecules?

Cross-reactivity with classical HLA class I molecules represents a significant challenge when studying HLA-G. Implement these methodological approaches to ensure specificity:

  • Antibody selection for specificity:

    • BFL.1 antibody specifically distinguishes between classical HLA-A and -B and non-classical HLA-G molecules, as demonstrated by its selective binding to HLA-G-expressing cells but not to parental untransfected or HLA-B7/HLA-A3-transfected L cells

    • Validate antibody specificity using cells with known HLA expression patterns

    • Consider using antibodies raised against specific HLA-G peptide sequences absent in classical HLA molecules

  • Comprehensive validation testing:

    • Test antibodies against panels of cells expressing different classical HLA molecules

    • Include blocking experiments with purified classical HLA proteins

    • Perform peptide competition assays with HLA-G-specific and shared peptides

    • Use Western blotting to confirm molecular weight differences

  • Technical approaches to minimize cross-reactivity:

    • Optimize antibody concentrations to maximize specific signal while minimizing background

    • For flow cytometry: Include blocking steps with human serum or Fc block

    • For IHC: Implement stringent washing protocols and appropriate blocking

    • Consider differential fixation methods that may preserve HLA-G-specific epitopes

  • Genetic and molecular validation:

    • Correlate protein detection with HLA-G mRNA expression

    • Use HLA-G knockout/knockdown systems as negative controls

    • Compare results with pan-HLA antibodies (like W6/32) to distinguish patterns

    • Consider mass spectrometry-based approaches for definitive identification

The key experimental finding supporting BFL.1 specificity is its lack of reactivity with human cell lines known to express classical HLA class I proteins, while it successfully immunoprecipitates a 39-kDa protein from HLA-G-expressing cell lines . This contrasts with W6/32, which immunoprecipitates both classical and non-classical HLA class I heavy chains .

How are HLA-G antibodies being used to investigate immune checkpoint pathways?

HLA-G is increasingly recognized as an important "immune checkpoint" molecule with distinct properties from classical immune checkpoints like PD-1/PD-L1 . Researchers are employing HLA-G antibodies in several innovative approaches:

  • Comparative immune checkpoint profiling:

    • Multi-parameter flow cytometry with HLA-G antibodies (MEM-G/9, 87G) alongside other checkpoint molecules (PD-1, PD-L1, CTLA-4)

    • Correlation of HLA-G expression with established checkpoint molecules in tissue sections

    • Functional studies comparing T cell exhaustion markers between HLA-G+ and HLA-G- populations

    • Analysis of synergistic effects between HLA-G and other checkpoint pathways

  • Mechanistic signaling studies:

    • Investigation of shared and distinct signaling pathways downstream of HLA-G receptor engagement

    • Phosphoproteomic analysis of changes induced by HLA-G receptor binding

    • Comparison with canonical checkpoint molecule signaling

    • Identification of potential combinatorial targeting strategies

  • Therapeutic targeting approaches:

    • Development of blocking antibodies against HLA-G for potential checkpoint inhibition therapy

    • Testing combination approaches with established checkpoint inhibitors

    • Assessment of HLA-G+ immune cell depletion strategies

    • Correlation of HLA-G expression with response to existing checkpoint inhibitors

  • Clinical translation applications:

    • Biomarker development for patient stratification in immunotherapy trials

    • Monitoring changes in HLA-G expression during immunotherapy treatment

    • Identification of resistance mechanisms involving HLA-G upregulation

    • Exploration of HLA-G in non-responders to current checkpoint inhibitors

HLA-G creates an immunosuppressive microenvironment through multiple mechanisms, including inhibition of NK and CD8+ T cell cytotoxicity, allogeneic CD4+ T cell proliferation, and dendritic cell maturation . Its role in creating an anti-inflammatory environment through IL-10 release further establishes its importance in immune regulation . This multifaceted immunomodulatory profile positions HLA-G as a compelling target for next-generation immunotherapy approaches.

What role does HLA-G play in autoimmune and inflammatory disorders?

HLA-G has emerged as a critical immunoregulatory molecule in autoimmune and inflammatory contexts, with researchers using specific antibodies to elucidate its role:

  • Cellular distribution and phenotype:

    • HLA-G+ CD4+ and CD8+ T cells have been identified in peripheral blood as potential regulatory cells

    • These cells exhibit a hypoproliferative phenotype with a unique cytokine profile distinct from conventional Tregs

    • Flow cytometric identification using 87G or MEM-G/9 antibodies allows comprehensive phenotyping of these populations

  • Functional characterization:

    • HLA-G+ T cells may represent specialized regulatory populations that differ from classical Foxp3+ Tregs

    • These cells can inhibit proliferation and cytotoxicity of effector immune cells

    • They participate in creating an anti-inflammatory environment through cytokine modulation

    • The balance between membrane-bound and soluble HLA-G may influence disease progression

  • Disease associations and mechanisms:

    • HLA-G expressing immune cells have been reported in various autoimmune disorders

    • Altered frequencies of HLA-G+ cells may correlate with disease activity

    • Genetic polymorphisms affecting HLA-G expression may influence disease susceptibility

    • Therapeutic modulation of HLA-G could represent a novel treatment approach

  • Therapeutic implications:

    • Monitoring HLA-G+ regulatory cells during conventional immunotherapy

    • Development of strategies to expand or activate HLA-G+ regulatory populations

    • Potential for recombinant HLA-G as a therapeutic agent

    • Targeting HLA-G pathways to restore immune tolerance

The presence of HLA-G+ immune cells in autoimmune conditions suggests they may represent an adaptive regulatory mechanism attempting to control inflammation . Understanding their functional characteristics and how they differ from conventional regulatory populations could provide insights into disease pathogenesis and potential therapeutic targets.

What are the current approaches for measuring HLA-G-mediated immunosuppression in vitro?

Researchers employ several standardized approaches to quantify HLA-G-mediated immunosuppression, each requiring specific antibodies:

  • T cell proliferation inhibition assays:

    • Co-culture purified T cells with HLA-G-expressing cells or soluble HLA-G

    • Measure proliferation via CFSE dilution, 3H-thymidine incorporation, or Ki-67 expression

    • Include blocking anti-HLA-G antibodies to confirm specificity

    • Quantify percentage inhibition compared to control conditions

  • NK and CD8+ T cell cytotoxicity suppression:

    • Standard chromium release assays with HLA-G+ target cells

    • Flow cytometry-based killing assays (caspase activation, membrane permeability)

    • Real-time impedance-based cytotoxicity measurements

    • Include HLA-G blocking with specific antibodies (MEM-G/9, 87G) to validate HLA-G-dependence

  • Dendritic cell maturation inhibition:

    • Generate immature DCs from monocytes

    • Expose to maturation stimuli with/without HLA-G

    • Measure surface maturation markers (CD80, CD83, CD86, HLA-DR)

    • Assess cytokine production and T cell stimulatory capacity

  • Apoptosis induction in antigen-specific CD8+ T cells:

    • Co-culture antigen-specific CD8+ T cells with soluble HLA-G

    • Measure apoptosis via Annexin V/PI staining, TUNEL, or caspase activation

    • Perform blocking experiments with anti-HLA-G antibodies

    • Quantify dose-dependent effects

  • Cytokine modulation assays:

    • Measure shifts in cytokine profiles (Th1/Th2 balance)

    • Quantify IL-10 production in response to HLA-G exposure

    • Assess changes in inflammatory vs. anti-inflammatory mediators

    • Use HLA-G blocking antibodies as controls

These functional assays reveal that HLA-G inhibits allogeneic proliferation of CD4+ T cells, NK and CD8+ T cell cytotoxicity, maturation of dendritic cells, and activation of B cells . Soluble HLA-G can trigger apoptosis in antigen-specific CD8+ T lymphocytes and create an anti-inflammatory environment through IL-10 release . Using standardized assays allows for comparison of results across different experimental systems and laboratories.

How might HLA-G antibodies contribute to the development of new immunotherapeutic strategies?

HLA-G antibodies are opening new possibilities for immunotherapeutic approaches across multiple disease contexts:

  • Blocking antibodies for cancer immunotherapy:

    • Development of therapeutic antibodies targeting HLA-G to prevent tumor immune escape

    • Combination approaches with established checkpoint inhibitors (anti-PD-1/PD-L1, anti-CTLA-4)

    • Patient stratification based on HLA-G expression profiles

    • Monitoring therapy response using flow cytometry with MEM-G/9 or 87G antibodies

  • Agonistic approaches for autoimmunity and transplantation:

    • Recombinant HLA-G fusion proteins to promote immune tolerance

    • Expansion of HLA-G+ regulatory T cells for adoptive cell therapy

    • HLA-G-based tolerance induction protocols for transplantation

    • Monitoring therapeutic efficacy using validated antibodies and standardized assays

  • Diagnostic and prognostic applications:

    • Development of standardized HLA-G detection systems for patient stratification

    • Identification of responder populations for immunotherapy

    • Monitoring disease progression and treatment response

    • Correlation of HLA-G expression patterns with clinical outcomes

  • Novel therapeutic formats:

    • Bispecific antibodies linking HLA-G with other immune targets

    • HLA-G-based chimeric antigen receptors for regulatory cell therapy

    • Nanoparticle delivery of HLA-G to specific tissue sites

    • Gene therapy approaches to modulate HLA-G expression

The development of well-characterized monoclonal antibodies against HLA-G, such as BFL.1, MEM-G/9, MEM-G/1, and 87G, has been instrumental in advancing our understanding of HLA-G biology . These antibodies not only serve as research tools but also provide the foundation for developing therapeutic antibodies targeting the HLA-G pathway. As our understanding of HLA-G's role as an "immune checkpoint" molecule continues to evolve , antibody-based approaches targeting this pathway represent a promising frontier in immunotherapy development.

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.