TLR7 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
We are generally able to ship your orders within 1-3 working days after receiving them. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timelines.
Synonyms
PRO285 antibody; TLR 7 antibody; Tlr7 antibody; TLR7_HUMAN antibody; Toll like receptor 7 antibody; Toll-like receptor 7 antibody; UNQ248 antibody
Target Names
Uniprot No.

Target Background

Function
TLR7 is an endosomal receptor that plays a crucial role in both innate and adaptive immunity. It functions as a key regulator of the host immune response against pathogens by recognizing uridine-containing single-stranded RNAs (ssRNAs) of viral origin or guanosine analogs. Upon binding to these agonists, TLR7 undergoes dimerization, bringing the TIR domains from the two molecules into direct contact. This interaction facilitates the recruitment of the TIR-containing downstream adapter MYD88 through homotypic interaction. Subsequently, the Myddosome signaling complex is formed, involving IRAK4, IRAK1, TRAF6, and TRAF3, leading to the activation of downstream transcription factors NF-κB and IRF7. These activated transcription factors ultimately induce the production of proinflammatory cytokines and interferons, respectively.
Gene References Into Functions
  1. Enhanced TLR7 expression due to biallelism contributes to a higher risk of developing Systemic Lupus Erythematosus (SLE). PMID: 29374079
  2. Btk acts within the TLR7/8 pathway, mediating Ser-536 phosphorylation of p65 RelA and subsequent nuclear entry in primary human macrophages. PMID: 29567473
  3. The miRNA profile did not significantly differ between SLE and Antiphospholipid Syndrome (APS), but was driven by the presence or absence of an IFN signature. TLR7 stimulation induced a general downregulation of miRNAs, similar to the pattern observed in SLE and APS patients. PMID: 29873766
  4. TLR7 upregulation plays a role in liver cancer cell proliferation involving lipid rafts. PMID: 27588480
  5. Research demonstrates that Hepatitis C virus (HCV) genomic RNA harbors specific sequences that initiate an anti-HCV immune response through TLR7 and TLR8 in various antigen-presenting cells. PMID: 27385120
  6. TLR7, TLR9, and JAK2 genes are potential biomarkers for Systemic Sclerosis. High TLR7 expression positively correlated with the late form of the disease. Decreased levels of TLR9 and JAK2 mRNA were found in the patient cohort compared to non-SSc individuals. PMID: 29147913
  7. Studies indicate that TLR7 upregulation is associated with Respiratory Syncytial Virus infection and the induction of oxidative stress, mediated by the anti-inflammatory effects of Nrf2/ARE pathway inhibitors or agonists. PMID: 29392496
  8. TLR7 and TLR8 may modulate different immune responses in monocytes and macrophages. PMID: 29448098
  9. Positive rates of iNOS in cervical tissues were 72.1%, 28.2%, and 3.1% in the -HPV-positive patients with cervical cancer (CC group), HR-HPV group, and controls, respectively (P < 0.05). Levels of TLR3, TLR4, TLR7, TLR8, NF-κB p65, and iNOS in cervical epithelial cells were higher in the CC group than in other groups. PMID: 28626766
  10. Data suggest a central role of XBP1 in TLR7-induced IFNα production and identify XBP1 as a potential novel therapeutic target in IFNα-driven autoimmune and inflammatory diseases. PMID: 28408069
  11. These findings suggest that increased EV71 and CA16 replication mediated by autophagy in 16HBE cells might promote degradation of the endosome, leading to suppression of the TLR7-mediated IFN-I signaling pathway. PMID: 29052054
  12. The present study investigated the effects of vitamin D3 on the expression of TLR3, TLR7, and TLR9 in Systemic Lupus Erythematosus patients. PMID: 28544067
  13. Findings indicate that Hepatitis C virus induces CD4 T cell impairment via TLR7, which may contribute to failure of virus eradication, casting doubts on the use of TLR7 agonists to boost innate immunity in chronic RNA virus infections. PMID: 27876497
  14. TLR 5, 7, and 9 expression patterns differed between HPV-positive and -negative oropharyngeal squamous cell carcinoma patients. In HPV-positive tumors, the expression of TLR 5 and 7 correlated with tumor recurrence. PMID: 28856441
  15. Low TLR7 copy number is a risk factor for chronic hepatitis B virus infection but is not associated with later stages of disease progression. PMID: 28321161
  16. TLR7 and TLR8 genetic polymorphisms are associated with susceptibility to Mycobacterium tuberculosis infection, and the link is shaped by less effective MTB phagocytosis and impaired TLR signaling. PMID: 27156628
  17. A study evaluated innate immune profiles following TLR stimulation in HIV-1-infected mothers and newborns, finding significantly compromised cytokine responses upon extracellular and intracellular TLR activation. Myeloid dendritic cell (DC) responsiveness appeared to be less impaired than plasmacytoid DCs, and might be enhanced through TLR7/TLR8 activation. PMID: 23826189
  18. This is the first study illustrating certain genotypes of TLR-7 and TLR-8 single nucleotide polymorphisms viz. CT(p = 0.002)]; rs3853839[GC(p < 0.001), CC(p = 0.039)] and rs3764879[GC(p < 0.001)] were considerably associated with Chikungunya virus susceptibility. PMID: 28888110
  19. A study demonstrates that activation of TLR7 signaling in T cells can inhibit Th17 cell differentiation from naive T cells and IL-17 production in established Th17 cells. The study further reports that downregulation of STAT3 signaling is responsible for TLR7-mediated inhibition of Th17 cells due to induction of suppressor of cytokine signaling 3 and 5. PMID: 28652396
  20. Data indicate a mechanism in which Toll-like receptors TLR7/8 signaling, through shedding of FcgRIIA, shifts neutrophil function from phagocytosis to a programmed necrosis pathway, neutrophil extracellular trap formation (NETosis). PMID: 28606989
  21. TLR7 is abundantly expressed in human atherosclerotic plaques. TLR7 ligation elicits the secretion of pro-inflammatory and anti-inflammatory cytokines, and high TLR7 expression in plaques is associated with better patient outcome. PMID: 27864310
  22. This study identified TLR7 as a mediator of monocyte differentiation and M2 macrophage polarization during Hepatitis C virus infection. PMID: 28122964
  23. Increased susceptibility to autoimmune diseases is associated with single nucleotide polymorphisms and copy number variations of Toll-like receptor 7. [review] PMID: 28397237
  24. COPD lung tissue explants showed a greater pro-inflammatory response to TLR3 or TLR7/8 activation than control smokers. PMID: 27729782
  25. The activation of TLR7 increased CCND3 expression via the downregulation of miR-15b in B cells. PMID: 26144250
  26. TLR9 and TLR7 were upregulated both at the mRNA and protein levels in wounds of type 2 diabetes mellitus patients compared to the non-diabetic patients and may lead to an unresolved inflammatory response and non-healing chronic ulcers. PMID: 25586463
  27. This study suggests that deliberate control of TLR signaling is a key factor in the success of mRNA-driven cellular reprogramming. PMID: 27586271
  28. Changes in mRNA expression of selected toll-like receptors (TLR2, TLR4, TLR7), stress cytokine prolactin (PRL), and pro- and anti-inflammatory cytokines (TNF-α, IL-6, IL-12, IL-10) in peripheral blood monocytes of celiac disease patients, were investigated. PMID: 28028951
  29. The ER-stress inducing property of IMQ is possibly of importance for its efficacy in treating basal cell carcinoma, in situ melanoma, and squamous cell carcinoma. Our data could potentially be harnessed for rational design of even more potent ER-stress inducers and new anti-cancer drugs. PMID: 27003259
  30. TLR8 polymorphisms were associated with an increased risk and TLR7 polymorphisms with a decreased risk of recurrent rhinovirus infections. PMID: 28403045
  31. We found lower expression levels of TLR1, TLR3, TLR4, TLR7 and TLR9 in PBMCs from patients with ALL compared with those from control patients. We also observed that the PBMCs from patients with Pre-B and B ALL had lower TLR4 expression than controls. PMID: 27277333
  32. Interferon alpha (IFN-α) secretion induced by Toll-like receptor 7 and Toll-like receptor 8 (TLR7/TLR8) activation was observed in Common Variable Immunodeficiency (CVID), which was recovered with Toll-like receptor 9 (TLR9) signaling. PMID: 27392462
  33. This study shows that TLR7 gene polymorphism is associated with susceptibility to HIV and HCV co-infection. PMID: 28062211
  34. TLR7 gene rs2897827 may influence TLR7 mRNA expression and the plasma ApoA1 level in male ischemic stroke patients. PMID: 27427388
  35. Considering that chronic hepatitis B infection is not yet curable, it could be possible to activate TLR7-related immunological pathways as a therapy directed towards persistent HBV infection. PMID: 27373425
  36. Both TLR 5 and 7 are expressed in salivary adenoid cystic carcinoma on the cell membranes as well as in cytoplasm. PMID: 26888781
  37. Human Toll-like receptor 7 ligand binding domain. PMID: 25817271
  38. TLR7 mRNA expression was significantly elevated in patients with relapsing remitting multiple sclerosis. PMID: 26996115
  39. Our meta-analysis suggests that TLR7, TLR8, and TLR9 polymorphisms are associated with the development of systemic lupus erythematosus in Caucasian, Asian, and African populations. PMID: 26762473
  40. Activation of TLR7 upregulated the expression levels of IFN-λ1 and MMP-9 were increased by ~3 fold, whereas other genes (p53, PTEN, TIMP-1) were upregulated by ~2 fold, and VEGF was marginally upregulated after 10 min. PMID: 26718740
  41. Increased expression of TLR7 and TLR9 in Myasthenia Gravis thymus is accompanied by active Epstein-Barr virus infection. PMID: 26723518
  42. The low expression of TLR-7 in tumor and high expression of TLR-7 in stroma predict a good clinical outcome for oral squamous cell carcinoma patients. PMID: 25828894
  43. TLR7 and 8 polymorphisms may play a considerable role in the pathogenesis of asthma. PMID: 26725554
  44. NADPH oxidase-deficient patient-derived B cells also expressed enhanced levels of TLR7 and TLR9 mRNA and protein compared to the same cells reconstituted to restore oxidase activity. PMID: 26340429
  45. Data show that differences in codon bias limit Toll-like receptor 7 (TLR7) expression relative to Toll-like receptor 9 (TLR9). PMID: 26903634
  46. Activation of TLR7 suppresses the progression of pancreatic cancer. PMID: 26238718
  47. Results show that TLR7 and TLR8 on trophoblastic cells play an important role in the prevention of intrauterine HBV transmission by inhibiting HBV translocation across trophoblasts. PMID: 26315138
  48. In females, the incidence of the X-linked TLR7 rs179008/Gln11Leu polymorphism was significantly lower in sarcoidosis patients. PMID: 24071890
  49. A key regulatory cytokine found increased upon TLR7 stimulation. PMID: 25923141
  50. This study established a correlation between miR-155 and TLR7 during hepatitis B virus infection and also demonstrated in vitro that increased miR-155 level could help to reduce viral load by targeting C/EBP-β. PMID: 25720442

Show More

Hide All

Database Links

HGNC: 15631

OMIM: 300365

KEGG: hsa:51284

STRING: 9606.ENSP00000370034

UniGene: Hs.659215

Protein Families
Toll-like receptor family
Subcellular Location
Endoplasmic reticulum membrane; Single-pass type I membrane protein. Endosome. Lysosome. Cytoplasmic vesicle, phagosome.
Tissue Specificity
Detected in brain, placenta, spleen, stomach, small intestine, lung and in plasmacytoid pre-dendritic cells. Expressed in peripheral mononuclear blood cells.

Q&A

What is TLR7 and what are its key structural and functional characteristics?

TLR7 (Toll-like receptor 7) is an endosomal pattern recognition receptor that plays a crucial role in innate and adaptive immunity. In humans, the canonical protein consists of 1049 amino acid residues with a molecular mass of approximately 120.9 kDa. TLR7 is predominantly localized in the endoplasmic reticulum, lysosomes, and cytoplasmic vesicles . This receptor functions as a sensor of viral RNA and specifically binds to guanosine and guanosine-containing compounds .

Methodologically, when studying TLR7, researchers should consider its subcellular localization when designing fractionation protocols and immunostaining experiments. Most experimental approaches require cell permeabilization, as TLR7 is not expressed on the cell surface but rather in intracellular compartments.

Where is TLR7 expressed in the human body and which cell types are important for TLR7 research?

TLR7 exhibits notable expression in several tissues including the brain, placenta, spleen, stomach, small intestine, and lung. At the cellular level, TLR7 is prominently expressed in plasmacytoid pre-dendritic cells, which are major producers of type I interferons in response to viral infections . TLR7 is also expressed in B cells and various myeloid cell populations, making these cell types important targets for TLR7-related research .

For experimental approaches, researchers should select appropriate cell types based on their research questions. Plasmacytoid dendritic cells are ideal for studying TLR7-mediated interferon responses, while B cells are crucial for investigating autoimmunity mechanisms. Recent methodological advances include CRISPR-Cas9 knockout protocols optimized for primary human plasmacytoid dendritic cells, which have revealed new insights into TLR7 biology .

How do TLR7 antibodies differ from TLR7 agonists in research applications?

TLR7 antibodies are immunological tools used primarily for detection and quantification of the TLR7 protein in various research applications such as Western blotting, immunohistochemistry, flow cytometry, and immunocytochemistry . They bind specifically to TLR7 protein epitopes and do not typically activate the receptor's signaling pathway.

In contrast, TLR7 agonists are compounds designed to bind to the active site of TLR7 and trigger its signaling cascade, mimicking viral RNA interactions. These agonists stimulate immune responses including cytokine production and cell activation. Recent advances include the development of nucleotide-modified oligoribonucleotides that selectively activate TLR7 while avoiding TLR8 activation through strategic introduction of 2' sugar-modified bases that prevent RNase-mediated degradation .

When designing experiments, researchers must clearly distinguish between using antibodies for TLR7 detection versus using agonists for TLR7 stimulation, as these represent fundamentally different experimental approaches with distinct outcomes and interpretations.

How can TLR7 antibodies be utilized in studying autoimmune diseases like lupus?

TLR7 antibodies serve as critical tools for investigating the mechanistic relationship between TLR7 and autoimmune pathology. In lupus research, these antibodies can be used to quantify TLR7 expression in patient samples, track receptor localization, and evaluate therapeutic interventions targeting the TLR7 pathway.

Recent research has demonstrated that anti-TLR7 monoclonal antibodies have therapeutic potential in lupus models. Studies in NZBWF1 mice, a lupus-prone strain, showed that administration of anti-TLR7 mAb, but not anti-TLR9 mAb, protected against nephritis development . The therapeutic mechanism involves inhibition of TLR7-dependent B cell activation and differentiation, resulting in reduced autoantibody production and decreased IgG deposition in glomeruli .

Methodologically, researchers investigating TLR7 in autoimmunity should employ flow cytometry to analyze relevant cell populations, including germinal center B cells, plasma cells, and memory T cells, all of which showed significant changes following anti-TLR7 antibody treatment in mouse models . Histological examination of kidney tissue coupled with immunofluorescence to detect immunoglobulin deposition provides crucial information about disease progression and therapeutic efficacy.

What is the significance of TLR7 gain-of-function variants in human lupus research?

Recent groundbreaking research has identified TLR7 gain-of-function variants as causative factors in human systemic lupus erythematosus. A de novo missense TLR7 variant (Y264H) was discovered in a child with severe lupus, establishing a direct genetic link between TLR7 hyperactivity and lupus pathogenesis .

This Y264H variant selectively increased sensitivity to guanosine and 2',3'-cGMP, demonstrating how subtle changes in ligand recognition can dramatically alter disease susceptibility . When introduced into mice, this variant was sufficient to cause lupus-like disease, providing powerful evidence of its pathogenicity.

For researchers studying autoimmunity, these findings highlight the importance of TLR7 genetic analysis in lupus patients and suggest new therapeutic approaches targeting TLR7 or its downstream adaptor MyD88. Methodologically, researchers should consider genetic screening for TLR7 variants in early-onset or severe lupus cases and develop functional assays to characterize the impact of identified variants on guanosine sensing and downstream signaling.

How can TLR7 agonist-antibody conjugates be designed for cancer immunotherapy research?

TLR7 agonist-antibody conjugates represent an innovative approach for targeted cancer immunotherapy. These conjugates combine the immune-stimulating properties of TLR7 agonists with the tumor-targeting specificity of antibodies, allowing for localized immune activation while minimizing systemic side effects.

The methodological approach involves generating antibody-drug conjugates (ADCs) that recognize tumor antigens expressed on cancer cell surfaces, with TLR7 agonists as the payload . These conjugates deliver TLR7 agonists specifically to the tumor microenvironment, where they can activate antigen-presenting cells and promote anti-tumor immunity.

In experimental systems, TLR7 agonist-ADCs have demonstrated robust activity in tumor antigen-presenting cell co-culture systems, indicated by dose-dependent upregulation of PD-L1 and CD86 on macrophages . In vivo studies have shown superior tumor growth control compared to intravenously administered free TLR7 agonists, with prolonged activation of myeloid cells in the tumor microenvironment and minimal peripheral immune activation .

Researchers developing such conjugates should incorporate pharmacokinetic and tissue distribution studies to confirm tumor-specific drug release, as well as comprehensive immune profiling to characterize the activation of relevant immune cell populations.

What validation steps are essential when using TLR7 antibodies in experimental systems?

Proper validation of TLR7 antibodies is crucial for generating reliable research data. Researchers should implement the following methodological steps:

  • Specificity testing: Compare staining patterns between wild-type and TLR7-knockout cells or tissues. Recent advances in CRISPR-Cas9 technology have enabled knockout generation in primary human plasmacytoid dendritic cells, providing valuable negative controls .

  • Cross-reactivity assessment: Test antibodies against related TLRs, particularly TLR8, which shares significant homology with TLR7. This is especially important when studying species differences or using the antibody in multiple applications.

  • Application-specific validation: Verify antibody performance in the specific application of interest (western blot, flow cytometry, IHC, etc.) as antibodies may perform differently across applications.

  • Concentration optimization: Titrate antibodies to determine the optimal concentration that maximizes specific signal while minimizing background.

  • Batch consistency verification: When changing antibody lots, perform side-by-side comparisons to ensure consistent performance.

These validation steps are essential for avoiding experimental artifacts and ensuring reproducible results in TLR7 research.

How can researchers distinguish between TLR7 and TLR8 activation in experimental systems?

Distinguishing between TLR7 and TLR8 activation presents a methodological challenge due to their high degree of homology. Researchers can implement the following approaches:

  • Use of selective ligands: Recent advances have enabled the rational design of TLR7-selective ligands by introducing precise 2' sugar-modified bases into oligoribonucleotides. These modifications prevent RNase-mediated degradation into monomeric uridine (required for TLR8 activation) while preserving TLR7 activation .

  • Receptor-specific knockout models: CRISPR-Cas9 technology can be used to generate TLR7 or TLR8 knockouts in relevant cell types. This allows for clean delineation of receptor-specific effects when using stimuli that might activate both receptors.

  • Cytokine profile analysis: TLR7 and TLR8 activation typically leads to distinctive cytokine signatures. TLR7 activation in plasmacytoid dendritic cells prominently induces type I interferons, while TLR8 activation in monocytes and myeloid dendritic cells primarily induces proinflammatory cytokines like TNF-α and IL-12.

  • Cell-type specific responses: Utilize cell types with differential expression of TLR7 and TLR8. For example, plasmacytoid dendritic cells predominantly express TLR7, while myeloid dendritic cells express both receptors.

  • Receptor-specific blocking antibodies: Apply validated TLR7 or TLR8 blocking antibodies to confirm receptor specificity of observed responses.

These approaches, often used in combination, enable researchers to attribute observed effects to the correct receptor pathway.

What experimental approaches are most effective for studying TLR7-dependent B cell responses?

TLR7-dependent B cell responses are central to both protective immunity and autoimmune pathology. The following methodological approaches have proven effective for their investigation:

  • Flow cytometric analysis of B cell subsets: Track changes in naïve, memory, germinal center, and plasma cell populations following TLR7 stimulation or blockade. Studies with anti-TLR7 mAb in lupus-prone mice showed significant decreases in germinal center B cells and plasma cells following treatment .

  • In vitro B cell stimulation assays: Culture purified B cells with TLR7 agonists alone or in combination with BCR stimuli to assess proliferation, survival, and differentiation. The combination of TLR7 and BCR signaling is particularly relevant for autoimmunity research, as enhanced TLR7 signaling drives aberrant survival of BCR-activated B cells .

  • Adoptive transfer experiments: Transfer wild-type or TLR7-deficient B cells into recipient mice to assess cell-intrinsic effects of TLR7 signaling. Research has shown that TLR7 signaling drives the accumulation of CD11c+ age-associated B cells and germinal center B cells in a cell-intrinsic manner .

  • Analysis of antibody production: Measure total and antigen-specific antibody levels using ELISA or multiplex assays following TLR7 stimulation or inhibition. Focus on autoantibody production when studying autoimmune contexts.

  • Single-cell transcriptomics: Apply this technology to identify gene expression signatures associated with TLR7 activation in different B cell subsets, providing insight into molecular mechanisms and potential therapeutic targets.

These methodologies provide complementary information about how TLR7 signaling influences B cell biology in both health and disease.

How should researchers interpret discrepancies between TLR7 mRNA expression and protein detection data?

Discrepancies between TLR7 mRNA and protein levels are common in research and require careful interpretation. These differences may reflect:

  • Post-transcriptional regulation: TLR7 expression may be controlled by microRNAs or RNA-binding proteins that affect translation efficiency without altering mRNA levels. Researchers should consider measuring these regulatory factors when discrepancies arise.

  • Protein stability and turnover: Differences in protein half-life compared to mRNA stability can lead to temporal disconnects between transcript and protein abundance. Pulse-chase experiments can help characterize TLR7 protein turnover rates.

  • Technical limitations: The sensitivity and specificity of detection methods differ between qPCR and protein detection techniques like Western blotting or flow cytometry. Verify antibody specificity using TLR7-knockout controls, and ensure appropriate loading controls for protein quantification .

  • Subcellular localization effects: TLR7 protein localization in endosomal compartments may affect extraction efficiency or antibody accessibility. Subcellular fractionation approaches can help resolve this issue.

  • Post-translational modifications: Glycosylation of TLR7 has been described , which may affect antibody recognition without altering mRNA levels. Consider using multiple antibodies targeting different epitopes when discrepancies are observed.

When facing such discrepancies, researchers should implement complementary methods (e.g., immunoprecipitation followed by mass spectrometry) to validate and reconcile differences between transcript and protein data.

What are the most informative experimental readouts for assessing TLR7 activation in different cell types?

Effective assessment of TLR7 activation requires selection of appropriate readouts based on cell type and research context:

  • Plasmacytoid dendritic cells:

    • Type I interferon production (measured by ELISA, bioassay, or intracellular cytokine staining)

    • Upregulation of activation markers (CD80, CD86, MHC-II) by flow cytometry

    • Phosphorylation of IRF7 by Western blotting or phospho-flow cytometry

  • B cells:

    • Proliferation (assessed by CFSE dilution or BrdU incorporation)

    • Survival mechanisms (by measuring Bcl-2 family proteins or annexin V staining)

    • Differentiation into germinal center B cells or plasma cells (using flow cytometry)

    • Antibody production and class switching (measured by ELISA)

  • Monocytes and Macrophages:

    • Proinflammatory cytokine production (TNF-α, IL-6, IL-12)

    • Upregulation of costimulatory molecules like PD-L1 and CD86

    • Phenotypic shifts between M1 and M2 polarization states

  • All cell types:

    • NF-κB pathway activation (measured by phospho-flow, Western blotting, or reporter assays)

    • MyD88 recruitment to TLR7 (assessed by co-immunoprecipitation or proximity ligation assay)

    • Transcriptional profiling of TLR7-responsive genes

Researchers should select readouts that align with their specific research questions while considering the temporal dynamics of different activation markers, which can vary considerably.

How can TLR7 activation be reliably measured in tissue samples from autoimmune disease models?

Measuring TLR7 activation in tissue samples presents unique challenges requiring specialized methodological approaches:

  • Histological assessment with key markers:

    • Perform multiplex immunofluorescence to simultaneously detect TLR7 expression and activation markers

    • Assess tissue infiltration by TLR7-expressing cells, particularly plasmacytoid dendritic cells and CD11c+ age-associated B cells

    • Quantify IgG deposition in tissues like kidney glomeruli in lupus models

  • Isolation of immune cells from tissues:

    • Optimize tissue digestion protocols that preserve surface markers and cell viability

    • Perform flow cytometry to quantify TLR7-expressing cell populations

    • Sort specific populations for downstream functional and molecular analyses

  • Transcriptional and protein analysis of tissue samples:

    • Measure expression of TLR7-dependent genes using qPCR or NanoString technology

    • Perform Western blotting to assess phosphorylation of signaling molecules downstream of TLR7

    • Apply spatial transcriptomics or single-cell RNA sequencing to identify cellular sources of TLR7-dependent responses

  • Ex vivo functional assays:

    • Culture isolated tissue cells with TLR7 inhibitors to assess reversibility of activation

    • Measure spontaneous cytokine production by tissue-derived cells

    • Assess responsiveness to additional TLR7 stimulation as a measure of receptor priming or tolerance

Implementing these approaches can provide complementary information about the status of TLR7 activation in complex tissue environments relevant to autoimmune pathology.

How do anti-TLR7 antibodies show therapeutic potential in lupus and other autoimmune disorders?

Anti-TLR7 antibodies have demonstrated significant therapeutic potential in autoimmune disease models through several mechanisms:

  • Inhibition of B cell activation and differentiation: Anti-TLR7 monoclonal antibodies decrease the frequencies of germinal center B cells and plasma cells in lupus-prone mice, leading to reduced autoantibody production . This directly addresses a key pathogenic process in lupus.

  • Reduction of pathogenic monocyte populations: In NZBWF1 lupus-prone mice, anti-TLR7 mAb abolished the lupus-associated increase in patrolling monocytes in circulation, spleen, and glomeruli. These monocytes typically express high levels of TLR7 and upregulate lupus-associated factors like IL-10, CD115, CD31, and TNFSF15 .

  • Prevention of kidney damage: Anti-TLR7 mAb treatment protected lupus-prone mice from nephritis development, as evidenced by decreased IgG deposition in glomeruli . This suggests potential efficacy against one of the most severe manifestations of lupus.

  • Reduction of splenomegaly: Treatment with anti-TLR7 mAb abolished splenomegaly in lupus-prone mice, normalizing spleen weight and splenocyte numbers . This represents a correction of a common physical manifestation of systemic autoimmunity.

  • Modulation of T cell responses: Anti-TLR7 mAb treatment significantly decreased CD4+ memory T cells in lupus-prone mice, possibly through inhibitory effects on dendritic cells that normally participate in T cell activation .

These findings suggest that therapeutic targeting of TLR7 with antibodies represents a promising approach for lupus and potentially other TLR7-driven autoimmune conditions, offering more specific intervention than current broad immunosuppressive strategies.

What considerations are important when designing TLR7 agonist-antibody conjugates for cancer immunotherapy?

The development of TLR7 agonist-antibody conjugates for cancer immunotherapy requires careful consideration of several key factors:

  • Target antigen selection: Choose tumor antigens that are highly expressed on cancer cells with minimal expression in normal tissues to maximize tumor specificity and minimize off-target effects. The antibody component should have high affinity and specificity for the selected antigen .

  • Conjugation chemistry optimization: The linkage between the TLR7 agonist and antibody must be stable in circulation but allow for appropriate release within the tumor microenvironment. Different linker chemistries can significantly impact pharmacokinetics, biodistribution, and efficacy .

  • Drug-to-antibody ratio (DAR) optimization: Determine the optimal number of TLR7 agonist molecules per antibody that maximizes efficacy without compromising the antibody's pharmacokinetic properties or increasing immunogenicity.

  • Selective activation of TLR7 vs. TLR8: Utilize molecular design strategies, such as nucleotide modifications, to create TLR7-selective agonists that avoid TLR8 activation if selective TLR7 stimulation is desired .

  • Evaluation metrics: Assess conjugate efficacy through multiple parameters:

    • Tumor growth control compared to free TLR7 agonist administration

    • Immune cell activation in the tumor microenvironment vs. periphery

    • Tumor-specific drug release confirmed by exposure studies

    • Development of tumor antigen-specific T cell responses

These considerations are essential for developing effective TLR7 agonist-ADCs that can generate localized immune activation within tumors while minimizing systemic immune-related adverse events.

What are the common technical challenges in detecting TLR7 protein expression and how can they be overcome?

Researchers frequently encounter technical challenges when detecting TLR7 protein expression. Here are common issues and methodological solutions:

  • Low expression levels: TLR7 is often expressed at relatively low levels, making detection challenging.

    • Solution: Implement signal amplification methods such as tyramide signal amplification for immunohistochemistry or use high-sensitivity detection systems for Western blotting.

    • Consider concentrating samples through immunoprecipitation before Western blotting.

  • Cross-reactivity with TLR8: Due to high homology between TLR7 and TLR8, antibodies may cross-react.

    • Solution: Validate antibody specificity using TLR7-knockout and TLR8-knockout controls.

    • Consider using multiple antibodies targeting different epitopes to confirm findings.

    • Implement CRISPR-Cas9 technology to generate specific knockouts for validation .

  • Subcellular localization challenges: TLR7's predominant localization in endosomal compartments can complicate detection.

    • Solution: Ensure adequate cell permeabilization for flow cytometry and immunocytochemistry.

    • Use subcellular fractionation to enrich for endosomal compartments when performing Western blotting.

    • Consider co-staining with endosomal markers to confirm appropriate localization.

  • Post-translational modifications: Glycosylation of TLR7 may affect antibody recognition.

    • Solution: Use deglycosylation enzymes to remove glycans before Western blotting if necessary.

    • Select antibodies that target epitopes known to be unaffected by glycosylation.

  • Fixation sensitivity: Some epitopes may be sensitive to certain fixation methods.

    • Solution: Compare multiple fixation approaches (paraformaldehyde, methanol, acetone) to identify optimal conditions.

    • Consider native conditions for flow cytometry when possible.

Addressing these technical challenges requires careful antibody selection, appropriate controls, and optimization of experimental protocols for specific applications.

How can researchers distinguish between direct and indirect effects when studying TLR7-mediated immune responses?

Differentiating direct TLR7-mediated effects from indirect downstream consequences represents a critical methodological challenge. Researchers can implement the following strategies:

  • Cell-specific genetic approaches:

    • Use conditional TLR7 knockout models with cell type-specific Cre recombinase expression.

    • Implement mixed bone marrow chimeras to determine cell-intrinsic versus cell-extrinsic effects.

    • Apply targeted siRNA or CRISPR approaches in specific cell populations.

    • Research using this approach revealed that TLR7-driven accumulation of CD11c+ age-associated B cells and germinal center B cells occurs in a cell-intrinsic manner, while increases in follicular and extrafollicular helper T cells represent cell-extrinsic effects .

  • Temporal analysis:

    • Establish detailed time courses to distinguish primary (direct) from secondary (indirect) effects.

    • Use rapid signaling events (e.g., phosphorylation cascades) as indicators of direct receptor engagement.

  • Pathway inhibition approaches:

    • Selectively block downstream mediators to identify their contribution to observed phenotypes.

    • For example, blocking cytokine receptors can help determine if effects are direct or cytokine-mediated.

    • Research showed that MyD88 deficiency (an adaptor protein downstream of TLR7) rescued autoimmunity and all cellular and serological phenotypes in TLR7 gain-of-function models .

  • Ex vivo and in vitro validation:

    • Isolate specific cell populations and stimulate them directly with TLR7 agonists.

    • Use transwell systems to separate cells while allowing soluble factor exchange.

  • Single-cell analyses:

    • Apply single-cell RNA sequencing to identify cell-specific transcriptional responses.

    • Use mass cytometry (CyTOF) to simultaneously measure multiple activation markers across cell types.

These approaches, often used in combination, help delineate the complex cascade of events following TLR7 activation and identify direct receptor-mediated effects versus secondary consequences of the inflammatory milieu.

What emerging technologies are likely to advance TLR7 antibody research in the coming years?

Several cutting-edge technologies are poised to significantly advance TLR7 antibody research:

  • Advanced CRISPR-Cas9 applications:

    • Optimized protocols for CRISPR-Cas9 knockout in primary human plasmacytoid dendritic cells have already revealed new insights into TLR7 biology, including a previously undescribed role for RNase 6 in degrading oligoribonucleotides into TLR ligands .

    • Base editing and prime editing technologies will enable precise introduction of specific TLR7 variants to study their functional consequences.

    • CRISPR activation/interference systems will allow for controlled modulation of TLR7 expression without genetic modification.

  • Structural biology advances:

    • Cryo-electron microscopy will provide detailed insights into TLR7 structure, including ligand binding sites and conformational changes upon activation.

    • This structural information will enable rational design of more specific antibodies targeting functional domains of TLR7.

  • Single-cell multi-omics:

    • Integration of transcriptomics, proteomics, and epigenomics at the single-cell level will reveal cell-specific TLR7 signaling networks.

    • Spatial transcriptomics will map TLR7 expression and activation patterns within tissue microenvironments.

  • Advanced imaging techniques:

    • Super-resolution microscopy will visualize TLR7 trafficking and interactions with signaling components at nanoscale resolution.

    • Intravital microscopy will track TLR7-expressing cells in living organisms during immune responses.

  • Antibody engineering platforms:

    • Bispecific antibodies targeting TLR7 and tumor antigens may enhance therapeutic efficacy.

    • Antibody fragments with enhanced tissue penetration will improve targeting of TLR7 in specific anatomical niches.

These technological advances will provide unprecedented insights into TLR7 biology and accelerate the development of TLR7-targeted therapeutics for autoimmune diseases and cancer.

How might TLR7 research contribute to understanding and treating emerging viral infections?

TLR7 research has significant implications for understanding and addressing emerging viral infections:

  • Viral sensing mechanisms:

    • As a sensor of viral RNA, TLR7 plays a crucial role in detecting RNA viruses, including emerging pathogens .

    • Understanding the specific viral RNA motifs recognized by TLR7 may help predict immune responses to novel viral threats.

    • Research into nucleotide modifications that affect TLR7 recognition could reveal how viruses evade immune detection.

  • Adjuvant development for viral vaccines:

    • TLR7 agonists show promise as vaccine adjuvants, particularly for enhancing CD8 T cell responses .

    • Rational design of TLR7-selective ligands through nucleotide modifications offers opportunities for developing tailored adjuvants for specific viral vaccines.

    • Understanding how 2' sugar-modified bases prevent RNase-mediated degradation while preserving TLR7 activation can inform adjuvant optimization .

  • Understanding severe viral disease pathogenesis:

    • TLR7 signaling may contribute to immunopathology in some viral infections through excessive inflammation.

    • Conversely, defective TLR7 responses might explain increased susceptibility to severe viral disease in certain populations.

    • Research into TLR7 gain-of-function variants may provide insights into hyperinflammatory responses observed in some viral infections .

  • Therapeutic targeting:

    • TLR7 agonist-antibody conjugates could potentially be adapted to target viral reservoirs in persistent infections .

    • Conversely, TLR7 antagonists might help manage cytokine storm syndromes associated with severe viral infections.

  • Human genetic variation effects:

    • Understanding how TLR7 genetic variants influence antiviral responses may help identify individuals at risk for severe viral disease outcomes.

    • This knowledge could inform personalized treatment approaches for viral infections.

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.