Recombinant Neosartorya fumigata Histone transcription regulator 3 homolog (hir3), partial

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Neosartorya fumigata Histone Transcription Regulator 3 Homolog (hir3), Partial

The Recombinant Neosartorya fumigata Histone transcription regulator 3 homolog (hir3), partial is a protein derived from the fungus Neosartorya fumigata, which is closely related to Aspergillus fumigatus. This protein is involved in the regulation of histone dynamics and chromatin structure, playing a crucial role in transcriptional regulation. Despite the lack of direct information on this specific protein, understanding its function requires insight into histone regulators and their roles in fungi.

Background on Histone Regulators

Histone regulators, such as those involved in the deposition of histone variants like H3.3, are essential for maintaining chromatin structure and facilitating transcription. In eukaryotes, histone variants are deposited by specific chaperones, which influence chromatin accessibility and transcription factor binding . For instance, HIRA is a chaperone that deposits H3.3, promoting chromatin accessibility and transcriptional activity .

Role of Histone Regulators in Fungi

In fungi, histone modifications and regulators are crucial for developmental processes, including mating and pathogenicity. For example, in Aspergillus, regulators like NsdD are involved in sexual development and cell wall integrity . While specific data on Neosartorya fumigata histone regulators is limited, understanding their function can be inferred from studies on related species.

Potential Functions of Recombinant Neosartorya fumigata hir3

Given the role of histone regulators in other organisms, the Recombinant Neosartorya fumigata Histone transcription regulator 3 homolog (hir3), partial likely participates in:

  • Chromatin Remodeling: Facilitating changes in chromatin structure to allow or block transcription.

  • Transcriptional Regulation: Influencing gene expression by modifying histone marks or recruiting transcription factors.

  • Developmental Processes: Contributing to fungal development, such as sporulation or pathogenicity.

Research Findings and Data

While specific research findings on Recombinant Neosartorya fumigata hir3 are not available, related studies provide insights into the broader functions of histone regulators:

OrganismHistone RegulatorFunction
ArabidopsisH3.3Involved in transcription and resetting histone marks .
DrosophilaH3.3-K27MImpairs chromatin marks, leading to transcriptional deregulation .
MagnaportheH3K27 modificationsRegulates transcription of in planta induced genes .

References PLOS Genetics: Dynamic Deposition of Histone Variant H3.3 Accompanies... Society for Developmental Biology: Histone H3.3A PMC: Histone modification dynamics at H3K27 are associated with altered transcription... bioRxiv: H3.3 contributes to chromatin accessibility and transcription factor binding... ASM: Conserved Regulators of Mating Are Essential for Aspergillus...

Product Specs

Form
Lyophilized powder.
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless dry ice is specifically requested. Advance notification is required, and additional fees will apply.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and can be used as a reference.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer components, temperature, and the protein's inherent stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.
The tag type will be determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
hir3; AFUA_3G12810; Histone transcription regulator 3 homolog
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Neosartorya fumigata (strain ATCC MYA-4609 / Af293 / CBS 101355 / FGSC A1100) (Aspergillus fumigatus)
Target Names
hir3
Uniprot No.

Target Background

Function
This protein plays a critical role in a nucleosome assembly pathway essential for maintaining heterochromatin integrity and ensuring proper chromosome segregation.
Database Links
Protein Families
HIR3 family
Subcellular Location
Nucleus.

Q&A

What is Neosartorya fumigata and its taxonomic relationship to Aspergillus fumigatus?

Neosartorya fumigata is a heat-resistant fungus that belongs to the genus Neosartorya, with Aspergillus fumigatus being its anamorphic (asexual) form. The taxonomic relationship between these organisms has been established through phylogenetic analysis of β-tubulin and calmodulin genes, which has shown that they are extremely close in both phylogenetic and morphological characteristics . This relationship creates challenges in identification and distinction for researchers working with these organisms. The primary difference is that Neosartorya species are known to cause spoilage in heat-processed acidic foods due to their heat-resistant ascospores, while A. fumigatus has not been reported as a spoilage agent in heat-processed food products . Understanding this taxonomic relationship is essential when designing experiments involving these organisms or when considering target genes for recombinant protein expression.

What are the key considerations when expressing recombinant proteins from Neosartorya fumigata?

When expressing recombinant proteins from Neosartorya fumigata, researchers should consider several critical factors. First, selection of an appropriate expression system is crucial - E. coli systems are commonly used for fungal proteins, as demonstrated with the ASPF3 protein . Second, protein tagging strategy significantly impacts purification efficiency and protein functionality; N-terminal tags such as His-SUMO are often employed to enhance solubility and facilitate purification . Third, researchers must carefully optimize buffer conditions to maintain protein stability; Tris/PBS-based buffers with 5-50% glycerol are typically suitable for storage . Fourth, expression range and target sequence selection require consideration of protein domains and functional regions to ensure biological activity is preserved. Finally, researchers should implement rigorous quality control measures, including SDS-PAGE analysis, to confirm purity (typically >90% is desirable for research applications) . For histones and transcription regulators specifically, special consideration must be given to DNA-binding properties and nuclear localization signals that might impact expression and purification strategies.

How can researchers distinguish between Neosartorya fumigata and related Aspergillus species in laboratory settings?

Researchers can reliably distinguish between Neosartorya fumigata and related Aspergillus species through a combination of molecular and morphological techniques. PCR-based identification using specific primer sets targeting the β-tubulin and calmodulin genes provides the most reliable differentiation, as these genes contain regions that specifically detect these species . This method has been demonstrated to have extremely high specificity, not detecting other fungi involved in food spoilage or environmental contamination . Additional distinguishing techniques include examination of ascospore heat resistance patterns, as Neosartorya species produce characteristically heat-resistant ascospores that can survive temperatures that would kill Aspergillus conidia. Microscopic examination focusing on sexual reproductive structures can also help differentiate these fungi, as Neosartorya species produce distinctive cleistothecia (sexual fruiting bodies) under appropriate conditions, while the asexual Aspergillus form does not typically exhibit these structures under standard laboratory conditions . For definitive identification, researchers should employ a combination of these approaches rather than relying on a single method.

What molecular mechanisms govern histone transcription regulation in Neosartorya fumigata, and how does hir3 contribute to this process?

The molecular mechanisms governing histone transcription regulation in Neosartorya fumigata involve a complex interplay of transcription factors, chromatin remodeling complexes, and cell cycle control elements. The histone transcription regulator 3 homolog (hir3) likely functions as part of a conserved histone regulatory complex that represses histone gene transcription outside of S-phase, similar to its orthologs in other fungi. In Neosartorya/Aspergillus species, transcription factors like the GATA-type transcriptional activator NsdD play essential roles in regulating various cellular processes including hyphal extension, cell wall integrity, and heterokaryon formation . The hir3 protein presumably interacts with these transcription networks to coordinate histone gene expression with DNA replication and cell division. Evidence from related fungi suggests that hir3 likely collaborates with other histone regulatory proteins to form repressive chromatin structures at histone gene promoters during G1 and G2 phases, with this repression being lifted during S-phase to allow histone synthesis coincident with DNA replication. Mutations in hir3 would likely disrupt this coordination, potentially leading to aberrant nuclear division, altered stress responses, and compromised cell wall integrity, similar to what is observed with other transcriptional regulators in this organism .

How can researchers optimize heterokaryon formation assays when studying transcription regulators in Neosartorya fumigata?

Optimizing heterokaryon formation assays when studying transcription regulators in Neosartorya fumigata requires careful consideration of several methodological factors. Researchers should implement the histone-assisted merged fluorescence (HAMF) technique, which utilizes fluorescent protein-tagged histone variants to visualize and monitor nuclear dynamics during heterokaryon formation . This approach requires expressing GFP-H2A in one strain and a red fluorescent variant (such as mCherry-H2A) in the mating partner to definitively identify heterokaryotic hyphae containing both types of labeled nuclei . For optimal results, strains should be co-cultivated on minimal medium to promote hyphal fusion, with regular microscopic monitoring beginning 24-48 hours after inoculation. When studying transcription regulators specifically, researchers should consider constructing deletion mutants and complemented strains to assess the impact of specific regulators on heterokaryon formation efficiency. The experimental design should include appropriate controls, including individual strains grown alone to establish baseline fluorescence patterns and known compatible strains as positive controls . Quantitative analysis should include measuring the frequency of heterokaryotic hyphae formation and the distribution pattern of different nuclear types within these hyphae. This approach can reveal defects in hyphal fusion that may be linked to transcriptional regulation of cell wall integrity genes, as demonstrated with the NsdD transcription factor in A. fumigatus .

What strategies can be employed to investigate the role of hir3 in stress response and virulence in Neosartorya fumigata?

To investigate the role of hir3 in stress response and virulence in Neosartorya fumigata, researchers should employ a multi-faceted experimental approach. First, gene deletion or conditional expression systems should be established using homologous recombination or CRISPR-Cas9 technologies to create hir3 mutant strains. These mutants should then be subjected to a comprehensive phenotypic analysis including growth under various stress conditions (oxidative, cell wall, osmotic, and temperature stresses), with particular attention to cell wall integrity, as transcription factors in N. fumigata have been shown to influence cell wall synthesis and resistance to cell wall-targeting compounds like nikkomycin Z . For oxidative stress studies, researchers can leverage protocols established for other N. fumigata proteins such as peroxiredoxins, which play crucial roles in oxidative stress response . Virulence assessment should include both in vitro models (macrophage interaction assays, epithelial cell adhesion/invasion) and in vivo models (Galleria mellonella larvae, murine pulmonary aspergillosis). Transcriptomic analysis (RNA-seq) comparing wild-type and hir3 mutant strains under various conditions would help identify genes regulated by hir3, while chromatin immunoprecipitation (ChIP-seq) using tagged hir3 would reveal direct binding targets. Protein interaction studies (co-immunoprecipitation, yeast two-hybrid) would elucidate hir3's role in transcriptional complexes. This comprehensive approach would establish whether hir3, like other transcription regulators in N. fumigata, contributes to stress adaptation and virulence through regulation of cell wall integrity, stress response pathways, or other virulence mechanisms.

What are the recommended purification protocols for recombinant Neosartorya fumigata histone-related proteins?

The recommended purification protocol for recombinant Neosartorya fumigata histone-related proteins involves a multi-step process optimized for nuclear proteins with DNA-binding properties. Begin with expressing the target protein (such as hir3) in E. coli with an N-terminal His-SUMO tag, which enhances solubility and facilitates purification of potentially challenging nuclear proteins . Following cell lysis (typically using sonication in a buffer containing 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10 mM imidazole, 1 mM PMSF, and 0.1% Triton X-100), perform the initial purification using immobilized metal affinity chromatography (IMAC) with Ni-NTA resin, washing with increasing imidazole concentrations (20-50 mM) before elution with 250-300 mM imidazole . The His-SUMO tag should then be cleaved using SUMO protease (Ulp1) during overnight dialysis against a buffer containing 50 mM Tris-HCl pH 8.0, 150 mM NaCl, and 1 mM DTT. A second IMAC step will remove the cleaved tag and protease, with the flow-through containing the target protein. Further purification via ion exchange chromatography (typically using a HiTrap Q column for histone-related proteins) and size exclusion chromatography will yield highly pure protein. Throughout purification, monitor protein quality using SDS-PAGE to achieve >90% purity . For long-term storage, add glycerol to a final concentration of 5-50% and store aliquots at -80°C to prevent repeated freeze-thaw cycles, which are particularly damaging to DNA-binding proteins .

How can researchers effectively analyze the interaction between hir3 and chromatin in Neosartorya fumigata?

To effectively analyze the interaction between hir3 and chromatin in Neosartorya fumigata, researchers should employ a comprehensive approach combining in vivo and in vitro techniques. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) represents the gold standard for identifying genomic binding sites of hir3 in vivo. This requires either developing antibodies against native hir3 or creating strains expressing epitope-tagged versions of hir3 (e.g., FLAG, HA, or GFP fusions). Following crosslinking, sonication, immunoprecipitation, and sequencing, researchers can map hir3 binding sites across the genome and correlate these with specific histone modifications or gene expression patterns. For studying the dynamics of hir3-chromatin interactions, fluorescence recovery after photobleaching (FRAP) with GFP-tagged hir3 can reveal association/dissociation kinetics at chromatin regions. In vitro approaches should include electrophoretic mobility shift assays (EMSAs) with recombinant hir3 protein and DNA fragments from putative target genes to confirm direct binding. Researchers can also perform in vitro reconstitution assays using recombinant histones and purified hir3 to study how hir3 affects nucleosome assembly or stability. Microscopy-based approaches using fluorescently tagged histones and hir3 can reveal their colocalization patterns in the nucleus under different conditions, similar to the histone-assisted merged fluorescence technique used to study nuclear dynamics during heterokaryon formation in Aspergillus . Combining these approaches will provide comprehensive insights into how hir3 interacts with and regulates chromatin structure and function in Neosartorya fumigata.

What are common challenges in expressing recombinant Neosartorya fumigata proteins and strategies to overcome them?

Common challenges in expressing recombinant Neosartorya fumigata proteins include protein insolubility, improper folding, low yield, and post-translational modification discrepancies. Insolubility is particularly problematic with nuclear proteins like hir3, which often form inclusion bodies in bacterial expression systems. To overcome this, researchers should employ solubility-enhancing fusion tags such as His-SUMO, which has proven effective for other N. fumigata proteins . Reducing expression temperature (to 16-18°C) and inducer concentration can also improve solubility by slowing protein synthesis and allowing more time for proper folding. For proteins that remain insoluble, denaturing purification followed by step-wise refolding may be necessary, though this often results in lower yields of functional protein. Low expression yields can be addressed by codon optimization for the expression host, as fungal codon usage differs significantly from bacterial systems. For proper protein folding, co-expression with fungal chaperones or expression in eukaryotic systems (yeast, insect, or mammalian cells) may be required for complex proteins. Post-translational modifications present in native N. fumigata proteins are often absent in bacterial systems; for proteins requiring specific modifications, consider yeast expression systems like Pichia pastoris. During purification, protein stability issues can be mitigated through buffer optimization, with many N. fumigata proteins showing enhanced stability in Tris/PBS-based buffers with 5-50% glycerol . For specialized applications requiring highly pure protein, additional chromatography steps beyond initial affinity purification are recommended to achieve >90% purity .

How can researchers validate the specificity and functionality of recombinant hir3 in experimental systems?

Validating the specificity and functionality of recombinant hir3 requires a comprehensive suite of biochemical, biophysical, and cellular assays. First, researchers should confirm structural integrity through circular dichroism (CD) spectroscopy to assess secondary structure composition and thermal stability assays to verify proper folding. DNA-binding specificity should be evaluated using electrophoretic mobility shift assays (EMSAs) with both predicted target sequences and control DNA fragments, followed by more quantitative techniques such as surface plasmon resonance (SPR) or bio-layer interferometry (BLI) to determine binding kinetics and affinities. For functional validation, in vitro chromatin assembly assays can assess hir3's ability to facilitate histone deposition onto DNA templates, while in vitro transcription assays using recombinant RNA polymerase II and chromatin templates can evaluate hir3's effects on transcriptional regulation. Cellular validation is equally important—researchers should perform complementation experiments in hir3-deficient strains to determine if the recombinant protein can rescue mutant phenotypes related to growth, stress response, or cell wall integrity, similar to studies conducted with other transcription regulators in N. fumigata . Localization studies using fluorescently tagged recombinant hir3 can confirm proper nuclear targeting and distribution, while ChIP experiments followed by qPCR can verify binding to predicted genomic targets. Finally, researchers should conduct protein-protein interaction studies using pull-down assays or co-immunoprecipitation to confirm that recombinant hir3 maintains appropriate interactions with binding partners such as other components of histone regulatory complexes.

What quality control measures should be implemented when working with recombinant Neosartorya fumigata proteins?

Implementing rigorous quality control measures is essential when working with recombinant Neosartorya fumigata proteins to ensure reliability and reproducibility of research findings. Researchers should establish a comprehensive quality control workflow beginning with purity assessment via SDS-PAGE, which should demonstrate >90% purity for most research applications . This should be complemented by mass spectrometry analysis to confirm protein identity, detect potential contaminants, and verify the presence of expected post-translational modifications. For oligomeric proteins, size exclusion chromatography coupled with multi-angle light scattering (SEC-MALS) should be employed to determine the oligomeric state and homogeneity of the protein preparation. Functional integrity assessment is critical and should be tailored to the protein's known biochemical activities; for histone-related proteins like hir3, this might include DNA-binding assays, histone interaction studies, or transcriptional regulation assays. Thermal stability analysis using differential scanning fluorimetry (DSF) can provide valuable information about protein folding and stability under various buffer conditions, helping optimize storage conditions. Endotoxin testing is essential for proteins intended for cell-based assays to prevent experimental artifacts from bacterial contaminants. Batch-to-batch consistency should be monitored through standardized activity assays and biophysical characterization. For long-term storage, researchers should conduct stability studies to establish optimal conditions (typically Tris/PBS-based buffers with 5-50% glycerol at -80°C for most N. fumigata proteins) , and implement aliquoting strategies to avoid repeated freeze-thaw cycles, which can significantly compromise protein integrity .

How does hir3 contribute to pathogenicity and stress response in Neosartorya fumigata infections?

The contribution of histone transcription regulator 3 homolog (hir3) to pathogenicity and stress response in Neosartorya fumigata likely involves multiple molecular mechanisms that parallel those of other transcriptional regulators in this organism. Based on studies of related transcription factors like NsdD, hir3 presumably influences pathogenicity through regulation of cell wall integrity, stress adaptation, and possibly heterokaryon formation . Cell wall integrity is crucial for fungal pathogenesis, as it mediates interactions with host cells and provides protection against host defense mechanisms. Transcription factors in N. fumigata have been shown to affect sensitivity to cell wall stressors, with deletion mutants exhibiting altered phenotypes including reduced hyphal extension and increased sensitivity to antifungal compounds . Studies of NsdD deletion mutants have demonstrated that transcription factors can influence cell wall synthesis pathways, which directly impacts virulence potential . Hir3 likely regulates genes involved in oxidative stress response, which is essential for survival within host macrophages, similar to how the peroxiredoxin Asp f3 contributes to virulence by detoxifying reactive oxygen species . Additionally, the potential role of hir3 in regulating heterokaryon formation, a process facilitated by hyphal fusion, may influence the organism's ability to adapt to challenging host environments through genetic complementation between different nuclei . Future studies employing hir3 deletion mutants in infection models will be necessary to fully elucidate its specific contributions to N. fumigata pathogenicity.

What are the potential applications of recombinant hir3 in developing novel antifungal strategies?

Recombinant hir3 offers several promising avenues for developing novel antifungal strategies against Neosartorya fumigata infections. As a transcription regulator likely involved in critical cellular processes, hir3 represents a potential target for small molecule inhibitors that could disrupt fungal growth and virulence. The recombinant protein can be utilized in high-throughput screening assays to identify compounds that specifically interfere with hir3's DNA-binding activity or its interactions with other components of transcriptional complexes. Structure-based drug design approaches become feasible once the three-dimensional structure of recombinant hir3 is determined through X-ray crystallography or cryo-electron microscopy. Additionally, recombinant hir3 could serve as an antigen for developing immunotherapeutic approaches, similar to how other Aspergillus proteins have been explored as vaccine candidates. By understanding hir3's role in regulating stress response pathways, researchers might identify synthetic lethal interactions that could be exploited therapeutically—for example, combining inhibition of hir3 with compounds that induce specific stresses that would normally be countered by hir3-regulated pathways. The protein could also be used to generate high-affinity aptamers or antibodies for diagnostic applications, enabling earlier detection of invasive aspergillosis. Comparative studies between human histone regulators and fungal hir3 could identify structural or functional differences that might be exploited to develop highly selective antifungal agents with minimal host toxicity. Finally, knowledge gained from studying recombinant hir3 could inform broader strategies targeting epigenetic regulation in pathogenic fungi, an emerging area in antifungal drug development.

How can CRISPR-Cas9 technology be optimized for studying hir3 function in Neosartorya fumigata?

Optimizing CRISPR-Cas9 technology for studying hir3 function in Neosartorya fumigata requires addressing several technical challenges specific to filamentous fungi while implementing cutting-edge modifications to enhance precision and efficiency. Researchers should begin by designing multiple guide RNAs targeting different regions of the hir3 gene, with careful analysis using algorithms that predict off-target effects while accounting for the GC-rich genome of N. fumigata. For delivery of CRISPR components, Agrobacterium-mediated transformation offers advantages for filamentous fungi, though protoplast transformation with ribonucleoprotein (RNP) complexes (pre-assembled Cas9 protein and sgRNA) can provide higher efficiency and reduce off-target effects. To enhance homology-directed repair (HDR) for precise gene editing, repair templates should include homology arms of at least 1 kb flanking the target site. Conditional gene expression systems should be implemented when studying essential genes like transcriptional regulators—consider using the tetracycline-inducible (Tet-On) system, which has been successfully applied in Aspergillus species. For studying complex phenotypes related to histone regulation, multiplexed CRISPR systems enabling simultaneous editing of multiple genes can reveal functional redundancy or synergistic effects between hir3 and related factors. Base editing (using catalytically impaired Cas9 fused to deaminases) and prime editing technologies can introduce precise point mutations without creating double-strand breaks, enabling nuanced studies of hir3 domain functions. Finally, CRISPR interference (CRISPRi) using deactivated Cas9 fused to repressor domains offers an alternative approach for studying hir3 function through targeted transcriptional repression rather than complete gene knockout, which may be particularly valuable when studying dosage-dependent effects of transcriptional regulators.

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.