PHF7 Antibody

Shipped with Ice Packs
In Stock

Description

PHF7 Antibody: Definition and Applications

PHF7 antibodies are immunoglobulins designed to bind specifically to the PHF7 protein. They enable researchers to:

  • Detect PHF7 expression in tissues and cells via Western blot (WB), immunohistochemistry (IHC), and immunofluorescence (ICC).

  • Study chromatin interactions and histone modifications regulated by PHF7.

  • Investigate PHF7's role in male infertility, germ cell development, and epigenetic regulation.

Key Applications:

MethodPurposeExample Use Cases
Western BlotQuantify PHF7 protein levels in testicular lysates or cell lines.
Immunoprecipitation (IP)Identify PHF7-bound histones (e.g., H3K4me3, H2A) and interacting proteins.
Immunohistochemistry (IHC)Localize PHF7 in spermatids or Sertoli cells during spermatogenesis.

Key Research Findings Involving PHF7 Antibodies

PHF7 antibodies have been pivotal in elucidating PHF7's role in epigenetics and reproductive biology.

PHF7 in Spermatogenesis

  • Histone-to-Protamine Exchange: PHF7 mediates H2A ubiquitination, enabling chromatin compaction during spermiogenesis. Phf7 knockout mice exhibit infertility due to defective histone removal and protamine incorporation .

  • Immune Pathway Dysregulation: Loss of PHF7 upregulates ERVs (Endogenous Retroviruses) and immune-related genes (e.g., PPARα, H2Eb1) in testes, linking PHF7 to immune tolerance in germ cells .

Epigenetic Regulation

  • Histone Binding: PHF7 binds H3K4me3/me2 via its PHD domain and ubiquitinates H2A via its RING domain, marking histones for degradation .

  • Chromatin Accessibility: PHF7 knockout increases chromatin openness, particularly in promoter regions, disrupting gene expression programs .

Clinical Relevance

  • Male Infertility: PHF7 downregulation correlates with non-obstructive azoospermia (NOA) in humans, as confirmed by PHF7 antibody-based studies .

  • Reprogramming Efficiency: PHF7 enhances direct reprogramming of fibroblasts into cardiomyocytes by activating cardiac genes and suppressing inflammatory pathways .

PHF7 Antibody Validation in Experimental Models

ModelKey ObservationsMethodReference
Phf7−/− MiceReduced H2A ubiquitination, impaired spermatogenesis, and ERV-MTD upregulation.WB, ChIP-qPCR
Human Testis SamplesPHF7 downregulation in NOA patients; PPARα upregulation linked to PHF7 loss.IHC, qPCR
DrosophilaPHF7 recognition in germ cells using anti-GST-phf7c antibodies.ELISA

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days after receiving it. Delivery time may vary depending on the purchase method or location. For specific delivery timelines, please consult your local distributor.
Synonyms
DKFZp434L1850 antibody; HSPC045 antibody; HSPC226 antibody; MGC26088 antibody; NYD SP6 antibody; OTTHUMP00000212988 antibody; OTTHUMP00000212989 antibody; OTTHUMP00000212990 antibody; OTTHUMP00000212993 antibody; PHD finger protein 7 antibody; Phf7 antibody; PHF7_HUMAN antibody; Testis development protein NYD SP6 antibody; Testis development protein NYD-SP6 antibody
Target Names
PHF7
Uniprot No.

Target Background

Function
PHF7 Antibody may play a role in spermatogenesis.
Gene References Into Functions
  1. The PHF7 promoter binds H4K12ac in mature spermatozoa. PMID: 22894908
Database Links

HGNC: 18458

KEGG: hsa:51533

STRING: 9606.ENSP00000333024

UniGene: Hs.372719

Subcellular Location
Nucleus.
Tissue Specificity
Highly expressed in Sertoli cells, but not in germ cells in adult testis. Expression in embryonic testis is 30-times lower. Highly expressed in colon, spleen, white blood cells, pancreas, lung, liver, placenta and brain. Detected at lower levels in thymus

Q&A

What is PHF7 and why is it important for reproductive biology research?

PHF7 (PHD finger protein 7) is a nuclear protein that functions as a histone reader, specifically binding to methylated histone H3 (H3K4me2/me3) to control gene expression programs. It plays a critical role in spermatogenesis and male germline development. In humans, canonical PHF7 consists of 381 amino acid residues with a mass of 43.8 kDa, with up to two different isoforms reported . Research has shown that PHF7 is highly expressed in Sertoli cells but not in germ cells of adult testis . Its importance in reproductive biology stems from its role in male fertility, as deletion of Phf7 in mouse models results in significantly compromised spermiogenesis, leading to reduced sperm production and increased immotile sperm . The expression level of PHF7 is also significantly reduced in patients with non-obstructive azoospermia (NOA), particularly in Sertoli cell-only and mature arrest cases, suggesting its involvement in human male spermatogenesis .

What are the key applications for PHF7 antibodies in research?

PHF7 antibodies serve multiple critical applications in reproductive biology and epigenetic research. Western blotting represents the most common application, allowing researchers to detect and quantify PHF7 protein expression in tissue lysates, as demonstrated in studies using mouse testis extracts . Immunofluorescence is another valuable application, particularly for analyzing PHF7's nuclear localization and expression patterns across different cell types within the testis . Additional important applications include flow cytometry for examining PHF7 expression in specific cell populations and immunohistochemistry for visualizing PHF7 distribution in tissue sections . These techniques have been instrumental in revealing PHF7's differential expression between normal testes and those from infertility patients, helping establish its role in spermatogenesis . When designing experiments, researchers should consider proper controls and validation steps for each application to ensure specific detection of PHF7 rather than non-specific binding.

How can PHF7 antibodies be optimized for chromatin immunoprecipitation (ChIP) experiments?

Optimizing PHF7 antibodies for chromatin immunoprecipitation requires careful consideration of several technical factors. First, select an antibody specifically validated for ChIP applications, ideally one recognizing an epitope that remains accessible when PHF7 is bound to chromatin. For crosslinking conditions, start with standard 1% formaldehyde for 10 minutes at room temperature, but optimize time and concentration based on preliminary results. During sonication, aim for chromatin fragments between 200-500bp, checking fragment size by gel electrophoresis. The research indicates that PHF7 functions as a histone reader binding to H3K4me2/me3 , which suggests that PHF7-binding sites significantly overlap with H3K4me3 marks . Therefore, parallel ChIP experiments targeting H3K4me3 can serve as a positive control for regions of interest. The data shows that PHF7 can regulate PPARα transcription by binding to specific locations on the ERV-MTD region , which can be used as a positive control locus during optimization. When analyzing results, compare enrichment at expected binding sites (like ERV-MTD regions) versus control regions lacking H3K4me3 marks to confirm specificity.

What are the best protocols for using PHF7 antibodies in immunofluorescence of reproductive tissues?

For optimal immunofluorescence detection of PHF7 in reproductive tissues, specific fixation and antigen retrieval protocols are crucial. Begin with fresh tissue fixation in 4% paraformaldehyde for 12-24 hours, followed by paraffin embedding or cryopreservation. For paraffin sections (5-7μm thickness), perform heat-induced epitope retrieval using citrate buffer (pH 6.0) for 20 minutes. When working with frozen sections, fix briefly post-sectioning with 4% paraformaldehyde for 10 minutes. Block sections with 5-10% normal serum in PBS containing 0.1-0.3% Triton X-100 for 1-2 hours at room temperature. Apply the primary PHF7 antibody at optimized dilutions (typically 1:100 to 1:500) and incubate overnight at 4°C. Research shows that PHF7 exhibits a distinct nuclear localization pattern in specific cell types like Sertoli cells , so include DAPI nuclear counterstaining for proper localization assessment. When analyzing PHF7 expression in pathological samples, such as from NOA patients, careful comparison with control samples is essential as PHF7 expression is significantly reduced in certain infertility conditions . Include appropriate positive controls (normal testis) and negative controls (antibody omission or tissues with confirmed PHF7 absence) in every experiment.

How can PHF7 antibodies be used to investigate its role in histone modifications and chromatin regulation?

Investigating PHF7's role in histone modifications requires integrating multiple experimental approaches centered around PHF7 antibodies. Begin with co-immunoprecipitation (co-IP) experiments using PHF7 antibodies to identify its interaction partners within chromatin-modifying complexes. Research indicates that PHF7 functions as a histone reader that binds methylated histone H3 (H3K4me2/me3) and may also function as an E3 ligase for H2A ubiquitination . For co-IP, use nuclear extracts from testicular tissue or relevant cell lines, followed by Western blot analysis with antibodies against suspected interacting proteins (H3K4me2/me3, H2A, ubiquitin ligase components). Sequential ChIP (ChIP-reChIP) can determine whether PHF7 and specific histone marks co-occupy the same genomic regions by performing consecutive immunoprecipitations with PHF7 antibodies followed by H3K4me3 antibodies. To assess the functional impact of PHF7 on chromatin accessibility, compare ATAC-seq data between wild-type and Phf7-deficient samples, as research shows PHF7 mutation results in more open chromatin in testicular cells . When analyzing results, pay particular attention to genes involved in immune pathways and DNA repair, as these were significantly enriched in differentially bound sites in Phf7-deficient mice .

How should researchers interpret discrepancies in PHF7 antibody staining patterns between species or experimental conditions?

When encountering discrepancies in PHF7 staining patterns across species or experimental conditions, researchers should implement a systematic analytical approach. First, critically evaluate antibody specificity through western blot validation in each species, confirming the detected protein matches PHF7's expected molecular weight (approximately 43.8 kDa in humans) . Consider that up to two different isoforms of PHF7 have been reported , which may exhibit differential expression patterns. Species-specific differences in PHF7 structure and function are biologically relevant; for instance, Drosophila PHF7 contains an evolutionarily novel C-terminus necessary for complete male programming in early germline that is absent in other species . When comparing results between mouse models and human samples, note that while PHF7 is highly expressed in Sertoli cells but not germ cells in adult human testes , its expression pattern might differ in mice. Experimental conditions significantly impact results—different fixation protocols, antibody incubation times, and antigen retrieval methods can all cause variation in staining intensity and subcellular localization. To resolve discrepancies, perform parallel experiments using multiple PHF7 antibodies targeting different epitopes and include appropriate controls for each species or condition.

What factors might affect the specificity and sensitivity of PHF7 antibody detection in Western blotting?

Multiple factors can influence PHF7 antibody performance in Western blotting, requiring careful optimization for reliable results. Sample preparation significantly impacts detection—complete protein extraction from nuclear compartments where PHF7 resides requires effective nuclear lysis buffers containing DNase treatment. Protein degradation can produce misleading bands, so always include protease inhibitors in lysis buffers. Transfer efficiency affects detection of nuclear proteins like PHF7; optimize transfer conditions using higher methanol concentrations (15-20%) for efficient transfer of nuclear proteins. Regarding antibody factors, epitope accessibility is crucial—antibodies targeting regions involved in protein-protein interactions may show reduced binding in native conditions. The research indicates that available PHF7 antibodies may target different regions, including the C-terminus which contains important functional domains . Blocking conditions significantly impact background levels; test different blocking agents (BSA vs. milk) and concentrations (3-5%). For challenging samples, signal amplification systems like enhanced chemiluminescence plus (ECL+) can improve detection of low-abundance PHF7. Post-translational modifications may alter antibody recognition—research suggests PHF7 is involved in ubiquitination pathways , potentially becoming modified itself. When troubleshooting, include positive controls (testis tissue extracts) which express PHF7 at detectable levels .

How can researchers distinguish between true PHF7 signal and background in immunohistochemistry of testicular tissues?

Distinguishing genuine PHF7 signal from background in testicular immunohistochemistry requires rigorous controls and optimization strategies. Implement a comprehensive panel of controls, including positive control tissues with known PHF7 expression (normal testicular tissue), negative control tissues where PHF7 is absent or significantly reduced (such as certain NOA patient samples) , and technical negative controls where primary antibody is omitted or replaced with non-specific IgG. For antibody validation, perform pre-absorption tests by incubating the PHF7 antibody with excess recombinant PHF7 protein before application to tissue—a significant reduction in signal confirms specificity. When analyzing results, focus on expected subcellular localization; PHF7 should demonstrate predominantly nuclear staining in specific cell populations like Sertoli cells, according to published evidence . Signal intensity should correlate with known expression patterns—reduced in certain pathological conditions like Sertoli cell-only syndrome . Implement counterstains to facilitate cell type identification—hematoxylin for general nuclear morphology or specific cell-type markers through dual immunofluorescence. Multi-spectral imaging and automated analysis can help quantify signal-to-background ratios across different experimental conditions, enabling objective comparison. Always process and stain control and experimental samples simultaneously under identical conditions to minimize technical variation.

What are the optimal sample preparation procedures for PHF7 antibody applications in reproductive tissue research?

Optimal sample preparation for PHF7 detection in reproductive tissues varies by application and requires tissue-specific considerations. For protein extraction and Western blotting, use fresh or flash-frozen testicular tissues and employ specialized nuclear extraction protocols with high-salt buffers (300-400mM NaCl) to efficiently release nuclear proteins like PHF7. Include both phosphatase and protease inhibitor cocktails to preserve native protein state. For immunohistochemistry or immunofluorescence, testicular tissue fixation timing is critical—over-fixation can mask PHF7 epitopes while under-fixation results in poor morphology. Research suggests Bouin's fixative for 4-6 hours provides excellent morphological preservation of testicular architecture while maintaining PHF7 antigenicity. For paraffin sections, implement step-wise dehydration using graded ethanol series to prevent tissue shrinkage. When preparing samples from infertility patients, document clinical parameters and pathological findings to correlate with PHF7 expression, as studies show PHF7 levels are significantly reduced in specific NOA patient subtypes . For RNA extraction to analyze PHF7 transcript variants (like the testis-specific phf7-RC isoform), use specialized RNA extraction protocols optimized for reproductive tissues rich in RNases. Single-cell preparations for flow cytometry require gentle enzymatic digestion protocols to maintain cellular integrity while releasing cells from the seminiferous tubules.

How can researchers validate PHF7 antibody specificity for their particular experimental system?

Validating PHF7 antibody specificity requires a multi-pronged approach tailored to each experimental system. Begin with genetic validation using Phf7 knockout or knockdown models where available—the antibody should show significantly reduced or absent signal in these samples compared to wild-type controls, as demonstrated in research using Phf7-/- mice . Perform peptide competition assays by pre-incubating the antibody with excess purified recombinant PHF7 protein or the immunizing peptide before application to samples—specific binding should be substantially reduced. Implement parallel detection methods for cross-validation, comparing results from different techniques (Western blot, immunoprecipitation, immunofluorescence) using the same antibody. For RNA-protein correlation, compare PHF7 protein levels detected by the antibody with mRNA expression measured by RT-qPCR, particularly focusing on tissue-specific isoforms like phf7-RC that show male-specific expression patterns . When analyzing results in human pathological samples, compare antibody performance across multiple patient cohorts with different testicular pathologies, as PHF7 expression varies significantly between normal samples and different NOA subtypes . For cross-reactivity assessment, test the antibody against related PHD finger proteins to confirm it doesn't recognize other family members with similar structural domains.

What are the best quantification methods for analyzing PHF7 expression in different cellular compartments?

Accurate quantification of PHF7 expression across cellular compartments requires specialized approaches tailored to its nuclear localization and cell type-specific expression pattern. For Western blot quantification, implement subcellular fractionation to isolate nuclear, cytoplasmic, and chromatin-bound fractions, normalizing PHF7 levels to compartment-specific reference proteins (lamin B1 for nuclear fraction, histone H3 for chromatin-bound fraction). Digital image analysis of immunofluorescence data should employ z-stack confocal microscopy to capture the complete nuclear volume where PHF7 resides, followed by 3D reconstruction to measure nuclear PHF7 intensity accurately. For cell type-specific quantification in heterogeneous tissues like testis, combine PHF7 immunostaining with markers for specific cell populations (e.g., SOX9 for Sertoli cells) in multiplexed immunofluorescence assays, as research indicates PHF7 is predominantly expressed in Sertoli cells rather than germ cells . Flow cytometry with nuclear permeabilization protocols can quantify PHF7 expression in specific cell populations isolated from testicular tissue, providing population-level statistics. When analyzing ChIP-seq or ATAC-seq data to assess PHF7's chromatin association, implement peak-calling algorithms optimized for transcription factors and compare binding patterns with histone modifications, particularly H3K4me3 which shows significant overlap with PHF7 binding sites . For all quantification methods, include appropriate biological replicates and statistical analyses to account for natural variation in PHF7 expression.

How can PHF7 antibodies contribute to understanding male infertility mechanisms?

PHF7 antibodies offer powerful tools for investigating male infertility mechanisms through multiple research applications. Implement comparative immunohistochemistry profiling across infertility patient cohorts—research has already demonstrated significantly reduced PHF7 expression in specific NOA subtypes, particularly Sertoli cell-only syndrome and maturation arrest cases . This approach can help stratify patients based on molecular phenotypes rather than morphological criteria alone. Develop diagnostic immunoassays using PHF7 antibodies to analyze testicular biopsies or potentially accessible biomarkers that correlate with PHF7 dysfunction. Multi-parameter flow cytometry combining PHF7 antibodies with markers of germ cell development stages can identify precise points of spermatogenic arrest in patient samples. Single-cell approaches coupling PHF7 immunodetection with transcriptomics can reveal downstream molecular pathways disrupted in PHF7-deficient cells. Research indicates that PHF7 deletion leads to immune pathway activation and aberrant expression of endogenous retroviruses like ERV-MTD , suggesting these as key mechanisms to investigate in infertility patients. For functional studies, complement antibody-based detection with genetic manipulation of PHF7 in relevant cell models, comparing phenotypes with patient observations. Correlation of PHF7 protein levels with histone modification patterns (particularly H3K4me3 and H2A ubiquitination) can elucidate epigenetic mechanisms underlying infertility, as PHF7 has been identified as both a histone reader and potential E3 ligase .

What insights can integrated ChIP-seq and RNA-seq analyses provide when using PHF7 antibodies?

Integrated ChIP-seq and RNA-seq analyses using PHF7 antibodies can reveal comprehensive mechanistic insights into its gene regulatory functions. Begin by performing PHF7 ChIP-seq in relevant reproductive tissues or cell models to map genome-wide binding sites—research indicates PHF7 binding peaks significantly overlap with H3K4me3 marks, providing a useful validation parameter . Parallel RNA-seq of wild-type versus PHF7-deficient samples can identify differentially expressed genes, as demonstrated in studies showing 385 upregulated and 530 downregulated genes following PHF7 deletion . Integrate these datasets to distinguish direct versus indirect regulatory targets by correlating PHF7 binding sites with expression changes. Computational motif analysis of PHF7-bound regions can identify co-occurring transcription factor binding sites, revealing potential collaborative regulatory networks. Studies indicate PHF7 regulates immune pathway genes and endogenous retrovirus expression, particularly ERV-MTD —focus analysis on these genomic regions. Time-course experiments combining ChIP-seq and RNA-seq can track the temporal dynamics of PHF7 binding and subsequent transcriptional changes during key developmental transitions in spermatogenesis. For functional validation of key target genes identified through this integrated approach, use targeted ChIP-qPCR to confirm PHF7 binding to specific regulatory regions, followed by reporter assays to test their functional importance in gene regulation.

How does PHF7's role in histone modifications connect to its function in germline development across species?

PHF7's evolutionarily conserved yet specialized functions in histone modifications and germline development represent a fascinating research area requiring comparative approaches. Implement cross-species immunoprecipitation studies using PHF7 antibodies validated for each organism to identify conserved versus species-specific protein interaction partners in the chromatin regulatory machinery. Research in both mammals and Drosophila indicates PHF7 functions as a histone reader binding to H3K4me2/me3 , but with significant structural and functional differences—particularly in the C-terminus region which is evolutionarily novel and necessary for complete male programming in Drosophila . Comparative ChIP-seq across species can map conservation of PHF7 binding sites and associated histone modifications. Developmental time-course analysis of PHF7 expression and chromatin association during germline specification and differentiation can identify critical windows for its function—research in Drosophila indicates early development is particularly sensitive to PHF7 activity levels for proper germline masculinization . For mechanistic studies, analyze the consequences of PHF7 mutation or deletion on genome-wide histone modification patterns (especially H3K4me2/me3 and H2A ubiquitination) across species. The connection to immune pathway activation observed in mouse models suggests investigating whether this represents a conserved mechanism affecting germline development across evolution. Understanding these species-specific differences is not merely academic—it provides insights into how chromatin-based sex determination mechanisms have evolved and may identify conserved fertility pathways relevant to human reproductive health.

What are the emerging applications of PHF7 antibodies in reproductive medicine and fertility diagnostics?

PHF7 antibodies hold significant promise for advancing reproductive medicine and fertility diagnostics through several emerging applications. Development of standardized immunohistochemical panels incorporating PHF7 antibodies could improve diagnostic precision for male infertility subtypes, particularly given the significant reduction of PHF7 expression observed in specific NOA patient populations . Such panels could help stratify patients beyond current morphological classifications, potentially guiding more personalized treatment approaches. Multi-parameter flow cytometry techniques combining PHF7 with other spermatogenesis markers could enable more detailed analysis of testicular biopsies, quantifying cell populations with abnormal PHF7 expression or localization. Minimally invasive liquid biopsy approaches might detect PHF7-related biomarkers in seminal plasma, potentially offering less invasive diagnostic options. For translational research, high-throughput screening platforms using PHF7 antibodies could identify compounds that modulate PHF7 expression or function, with research suggesting astaxanthin as one such compound affecting the PHF7 pathway through PPARα . Development of companion diagnostics using PHF7 antibodies could eventually help select patients likely to benefit from targeted therapies affecting the pathways regulated by PHF7. As research advances our understanding of PHF7's role in chromatin regulation and immune pathway suppression , diagnostic approaches targeting these downstream mechanisms may complement direct PHF7 detection.

How can researchers address the technical challenges in studying PHF7 across different cell types and developmental stages?

Addressing the technical challenges of studying PHF7 across diverse cellular contexts requires innovative methodological approaches. Develop cell type-specific PHF7 analysis using laser capture microdissection of specific testicular cell populations (Sertoli cells, spermatogonia, etc.) followed by protein extraction and Western blotting or proteomics analysis. For developmental studies, implement stage-specific isolation techniques such as STA-PUT velocity sedimentation or FACS sorting based on developmental markers, followed by PHF7 immunodetection. When analyzing early developmental stages where material is limited, consider proximity ligation assays to detect PHF7 interactions with binding partners or histone modifications with higher sensitivity than conventional co-immunoprecipitation. For in vivo developmental analysis, generate knock-in reporter lines expressing tagged PHF7 variants that can be tracked through development without disrupting function. Research indicates that PHF7 has complex isoform expression (such as phf7-RC in testes) —develop isoform-specific antibodies or detection methods targeting unique regions of each variant. Single-cell approaches combining immunofluorescence with in situ hybridization can correlate PHF7 protein levels with mRNA expression at the individual cell level. For challenging samples like human biopsies with variable fixation quality, implement automated image analysis algorithms trained to recognize PHF7 staining patterns across different preparation conditions. When comparing results across studies, establish standardized reporting of technical parameters including antibody validation methods, tissue processing protocols, and quantification approaches.

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.