Recombinant Candida glabrata Class E vacuolar protein-sorting machinery protein HSE1 (HSE1)

Shipped with Ice Packs
In Stock

Description

Protein Secretion and Cell Wall Structure

C. glabrata, like other fungi, relies on protein secretion for cell wall construction, a process that is essential for survival and virulence . The cell wall acts as a permeability barrier, and proteins like Epa1p require specific structural features to project through this barrier and interact with the external environment .

  • Epa1p Adhesin: Epa1p, a glucan-cross-linked cell wall protein, facilitates adherence to human epithelial cells . Its function depends on its modular domain structure, where the N-terminal domain binds ligands, and the C-terminal Ser/Thr-rich domain is essential for projecting the protein through the cell wall's permeability barrier .

  • Role of Ser/Thr-rich Domain: The Ser/Thr-rich domain is crucial for the localization of Epa1p on the external surface of the yeast cell, enabling interaction with its ligand . Without this domain, the protein remains in the internal layers of the cell wall and cannot mediate adherence .

Interspecies Interaction via Yhi1 Protein

C. glabrata interacts with other Candida species, particularly C. albicans, in mixed-species infections. The HSE1 protein is related to the secretion of proteins involved in these interactions .

  • Yhi1 Protein: C. glabrata secretes a unique small protein called Yhi1, which induces hyphal growth in C. albicans, a process essential for host tissue invasion . This interaction is specific to C. glabrata and C. albicans .

  • Functional Motif AXVXH: The Yhi1 protein contains a novel functional pentapeptide motif (AXVXH) required for its function . Mutation or truncation of this motif abolishes the hyphal growth-inducing activity in C. albicans .

  • Regulation of Yhi1: Yhi1 expression and efflux are regulated through the mating MAPK signaling pathway and the pheromone transporter CgSte6 in C. glabrata, despite its asexual reproduction preference .

Functional Analysis and Mutant Studies

Studies involving mutants and comparative proteomics provide insights into the roles of HSE1 and related proteins .

  • DSE1 Mutant Analysis: Analysis of C. albicans DSE1 mutants showed that Dse1 is a cell wall protein necessary for virulence and resistance to cell surface-disrupting agents . The DSE1 mutant lacks the chitin biosynthesis protein Chs5, explaining a decrease in chitin biosynthesis .

  • Proteomic Expression: Wild-type strains express proteins involved in pH response, zinc acquisition, virulence, and oxidative stress tolerance, while the mutant expresses glucoamylase 1, involved in carbohydrate metabolism, cell wall degradation, and biofilm formation .

Clinical and Antifungal Implications

The study of HSE1 and related proteins has potential clinical applications, particularly in developing antifungal strategies and biomarkers .

  • Biomarker Potential: Yhi1 can be used as a biomarker due to its role in interspecies communication .

  • Antifungal Target: The AXVXH motif in Yhi1 is a potential template for synthetic novel antifungal peptides .

  • Adhesion Inhibition: Given the role of adhesins like Epa1p in fungal adherence, understanding their structure and function can lead to the development of inhibitors that prevent fungal colonization .

SH3 Domain Proteins

SH3 domain proteins are also relevant to the function of Candida species .

  • Role of SH3 Domain Proteins: Functional analyses of C. albicans genes encoding Src Homology 3-domain proteins contribute to understanding cellular processes and potential drug targets .

Data Table: Key Proteins and Their Functions

ProteinSpeciesFunction
Epa1pCandida glabrataAdhesion to human epithelial cells; requires Ser/Thr-rich domain for proper localization
Yhi1Candida glabrataInduces hyphal growth in C. albicans; contains AXVXH motif essential for function; regulated by mating signaling pathway
Dse1Candida albicansCell wall protein necessary for virulence and resistance to cell surface-disrupting agents
Chs5Candida albicansChitin biosynthesis protein; absent in DSE1 mutant
Pra1Candida albicansInvolved in pH response and zinc acquisition
Atg15Candida albicansLipase involved in virulence
Sod1Candida albicansRequired for oxidative stress tolerance
Glucoamylase 1Candida albicansCell wall glycoprotein involved in carbohydrate metabolism, cell wall degradation, and biofilm formation; expressed in DSE1 mutant

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
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 collect 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 serve as a guideline.
Shelf Life
Shelf life depends on storage conditions, buffer composition, temperature, and protein 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. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
HSE1; CAGL0K02761g; Class E vacuolar protein-sorting machinery protein HSE1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-450
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Candida glabrata (strain ATCC 2001 / CBS 138 / JCM 3761 / NBRC 0622 / NRRL Y-65) (Yeast) (Torulopsis glabrata)
Target Names
HSE1
Target Protein Sequence
MPSLEVKLRK AVLAATDGKL RSDNWQYIIG VCDLVKEDPE DASQIVMEMI EKRLGQNDAN VMLRSLALVV ALAENCGSRL KQQVSSKHFT GILAQLLESG DVHMTVKKEI AKVVKQLSDS FKSDPSLKTM GDLNTRIRRK WPGLLEEPEK PSKQKVSHQE ATDEDQELQR ALKMSLEEFE KSKQQSNGSA VQSNSLQDHN QGQQQPQQQT TSGIRRVRAL YDLNANEQDE LSFRKGDVIV VLEQVYRDWW RGSLHGKIGI FPLNYVTPIT EPSPVESQRE QQIEEGVLSQ AQNVQVLSAK MQMASGKGLS ELNQDPEFND LYSTVTPIRP HVTKLIGKYA KEKDDVIALR QVLLNAESTY NELLDRAAKS YSIPNTQAPP YAPAVTSQPG YVSNNTYQTT NGQYTQHNIT PQQQYQVPSQ NYQSQPPSMQ SNHYIGYQHP GINDQPPPNY
Uniprot No.

Target Background

Function
A component of the ESCRT-0 complex, functioning as a sorting receptor for ubiquitinated cargo proteins at the multivesicular body (MVB).
Database Links
Protein Families
STAM family
Subcellular Location
Endosome membrane; Peripheral membrane protein; Cytoplasmic side.

Q&A

What is the Candida glabrata Class E vacuolar protein-sorting machinery protein HSE1?

HSE1 is a component of the Class E vacuolar protein-sorting (Vps) machinery in Candida glabrata. It belongs to a conserved protein family involved in the multivesicular body (MVB) pathway, which is crucial for protein trafficking and degradation within eukaryotic cells. The Class E Vps proteins, including HSE1, are essential components of the ESCRT (Endosomal Sorting Complexes Required for Transport) pathway related to exosome biogenesis . In C. glabrata, HSE1 contributes to cellular processes including protein metabolism, intracellular transport, and potentially pathogenesis mechanisms. The protein functions within a complex network of molecular machinery that regulates the formation of intraluminal vesicles in late endosomes, a critical step in the selective degradation of membrane proteins and the biogenesis of extracellular vesicles which may contribute to host-pathogen interactions during infection.

What is the functional significance of HSE1 in Candida glabrata biology?

The HSE1 protein in C. glabrata likely plays multiple roles in cellular function, particularly in the context of protein trafficking and degradation pathways. As a component of the Class E Vps machinery, HSE1 contributes to the ESCRT pathway, which is critical for the formation of multivesicular bodies and the subsequent release of extracellular vesicles (EVs) . These EVs are important for cell-to-cell communication and potentially for virulence factor delivery during infection. In pathogenic fungi, the proper function of the vacuolar protein sorting system is essential for adaptation to various environmental stresses encountered during infection, including nutrient limitation, oxidative stress, and immune cell attacks. Disruption of HSE1 function may impact C. glabrata's ability to survive within host macrophages, similar to how transcriptional regulators like CgXbp1 influence the fungus's response to macrophage infection . The vacuolar protein sorting system also contributes to maintaining cellular homeostasis and responding to changing environmental conditions, which is crucial for C. glabrata's survival as both a commensal organism and an opportunistic pathogen.

What purification strategies yield the highest purity of recombinant HSE1?

Purification of recombinant HSE1 to high homogeneity typically requires a multi-step chromatographic approach optimized for the protein's biochemical properties. Beginning with affinity chromatography based on the fusion tag (His6, GST, or FLAG), researchers can achieve initial enrichment of the target protein. For His-tagged HSE1, immobilized metal affinity chromatography (IMAC) using Ni-NTA or Co-based resins under native conditions with imidazole gradient elution provides good preliminary purification. This initial step should be followed by size exclusion chromatography (SEC) to separate monomeric HSE1 from aggregates and other contaminants based on molecular size. Ion exchange chromatography (IEX) can serve as an intermediate or polishing step, with the choice between cation or anion exchange depending on HSE1's predicted isoelectric point. Throughout the purification process, maintaining protein stability through optimized buffer conditions (pH, salt concentration, and potential stabilizing additives like glycerol) is essential. Confirmation of purity should be assessed by SDS-PAGE, Western blotting, and mass spectrometry, with functional assays conducted to confirm that the purified protein retains its native activity.

What are the challenges in obtaining functional recombinant HSE1 and how can they be overcome?

Obtaining functional recombinant HSE1 presents several challenges related to protein structure, post-translational modifications, and protein-protein interactions. As a component of the ESCRT machinery, HSE1 likely functions within a protein complex, and isolated recombinant expression may yield protein lacking critical binding partners necessary for native conformation. This challenge can be addressed through co-expression strategies with interacting partners or through the use of eukaryotic expression systems that better recapitulate the native cellular environment. Post-translational modifications, which may be crucial for HSE1 function, represent another significant challenge that can be addressed by choosing expression systems capable of performing the necessary modifications or through in vitro modification following purification. Protein stability issues can be mitigated through careful buffer optimization, inclusion of stabilizing agents like glycerol or specific detergents, and storage condition optimization. Structure-based predictions of HSE1 domains can help design constructs with improved solubility and stability. Additionally, implementing high-throughput screening approaches for expression and purification conditions can accelerate the identification of optimal parameters for producing functional protein.

What structural domains characterize HSE1 and how do they contribute to its function?

The HSE1 protein in C. glabrata contains several conserved domains characteristic of class E vacuolar protein-sorting machinery components, each contributing to specific aspects of its function. While specific structural information for C. glabrata HSE1 is limited in the provided search results, comparative analysis with homologous proteins suggests it likely contains an UIM (Ubiquitin-Interacting Motif) domain that recognizes ubiquitinated cargo proteins destined for degradation. HSE1 may also contain SH3 (Src Homology 3) domains that mediate protein-protein interactions with other components of the ESCRT pathway. The protein likely features coiled-coil regions important for oligomerization and assembly of protein complexes involved in vesicle formation. These structural elements collectively enable HSE1 to participate in the recognition and sorting of ubiquitinated membrane proteins into multivesicular bodies (MVBs). The spatial arrangement of these domains and their dynamic interactions with other ESCRT components and cargo proteins determine HSE1's ability to function effectively in the protein sorting and trafficking pathways that are essential for C. glabrata cellular homeostasis and potentially for its virulence mechanisms.

What biochemical assays are most informative for characterizing HSE1 activity?

Comprehensive characterization of HSE1 activity requires multiple complementary biochemical approaches that address its protein interactions, enzymatic activities, and cellular functions. Protein-protein interaction assays, including pull-down assays, co-immunoprecipitation, and yeast two-hybrid screening, can identify HSE1's binding partners within the ESCRT machinery and potentially reveal novel interactions specific to C. glabrata. Ubiquitin-binding assays using isothermal titration calorimetry (ITC) or surface plasmon resonance (SPR) can quantitatively assess HSE1's affinity for ubiquitinated substrates. In vitro reconstitution assays with purified components of the ESCRT machinery can demonstrate HSE1's role in vesicle formation and cargo sorting. Microscopy-based techniques employing fluorescently labeled HSE1 can visualize its subcellular localization and dynamics during vesicle formation. Functional complementation assays in HSE1-deficient yeast strains can assess whether recombinant HSE1 restores normal phenotypes related to protein trafficking and MVB formation. Mass spectrometry-based approaches can identify post-translational modifications on HSE1 and characterize its interaction network. These diverse assays collectively provide a comprehensive understanding of HSE1's biochemical activities and cellular functions.

How does recombinant HSE1 interact with other components of the ESCRT machinery?

Recombinant HSE1 interacts with multiple components of the ESCRT machinery through specific domain-mediated interactions that orchestrate the process of multivesicular body formation and cargo sorting. HSE1 likely serves as a crucial adaptor protein that bridges cargo recognition and downstream ESCRT complex assembly in C. glabrata. Biochemical interaction studies would be expected to show direct binding between HSE1 and ubiquitinated cargo proteins through its UIM domain, a critical first step in the ESCRT pathway. Additionally, HSE1 likely interacts with ESCRT-I components, facilitating the recruitment of the ESCRT machinery to endosomal membranes where protein sorting occurs . Co-immunoprecipitation experiments using recombinant HSE1 as bait can identify its interaction partners, while in vitro binding assays with purified ESCRT components can characterize the affinity and specificity of these interactions. Structural studies using techniques like X-ray crystallography or cryo-electron microscopy of HSE1 in complex with its binding partners would provide atomic-level details of these interactions. Mutation studies targeting predicted interaction interfaces can verify the functional significance of specific HSE1 domains in mediating these protein-protein interactions. Understanding these interactions is essential for elucidating HSE1's role in the complex network of molecular machinery that regulates protein trafficking and degradation in C. glabrata.

How can recombinant HSE1 be used to study extracellular vesicle biogenesis in C. glabrata?

Recombinant HSE1 represents a valuable tool for elucidating the molecular mechanisms underlying extracellular vesicle (EV) biogenesis in C. glabrata, a process that may contribute to pathogenesis through the delivery of virulence factors. Researchers can employ recombinant HSE1 in reconstitution experiments using synthetic liposomes to assess its direct role in membrane deformation and vesicle formation, providing insights into the early stages of EV biogenesis. Fluorescently labeled recombinant HSE1 can be used in live-cell imaging experiments to track its localization during EV formation, potentially revealing spatial and temporal dynamics of ESCRT machinery assembly. Immunoprecipitation of HSE1 from C. glabrata cells actively producing EVs, followed by proteomics analysis, can identify the composition of HSE1-associated protein complexes during this process. Additionally, recombinant HSE1 can be used to develop inhibitors or dominant-negative mutants that disrupt specific protein-protein interactions, allowing researchers to dissect the sequence of molecular events in EV biogenesis . Comparative studies between wild-type HSE1 and site-directed mutants can reveal critical functional residues and domains required for EV formation. These approaches collectively contribute to understanding how the ESCRT machinery, including HSE1, regulates the production of extracellular vesicles that may transport virulence factors and immunomodulatory molecules during C. glabrata infection.

What role does HSE1 play in C. glabrata pathogenesis and host immune response?

The role of HSE1 in C. glabrata pathogenesis likely centers on its contribution to extracellular vesicle biogenesis and protein trafficking pathways that influence virulence factor delivery and immune evasion strategies. Evidence from related fungal pathogens suggests that components of the ESCRT machinery, including class E Vps proteins like HSE1, are involved in producing extracellular vesicles enriched in virulence factors . These EVs may deliver enzymes, toxins, and immunomodulatory molecules to host cells during infection, potentially contributing to C. glabrata's ability to colonize host tissues and evade immune defenses. The protein trafficking functions mediated by HSE1 likely influence the composition of the fungal cell surface, affecting recognition by host immune receptors and subsequent immune responses. Disruption of HSE1 function might alter C. glabrata's ability to survive within macrophages, similar to how transcription factors like CgXbp1 affect the fungus's interaction with these immune cells . Research using HSE1-deficient C. glabrata strains in macrophage infection models could reveal whether this protein contributes to intracellular survival and proliferation, key virulence traits of this opportunistic pathogen. Understanding HSE1's role in pathogenesis may provide insights into novel therapeutic strategies targeting fungal protein trafficking pathways to combat C. glabrata infections, which are increasingly concerning due to their rising incidence in immunocompromised populations .

How can CRISPR-Cas9 be applied to study HSE1 function in C. glabrata?

CRISPR-Cas9 technology offers powerful approaches for investigating HSE1 function in C. glabrata through precise genome editing that enables both knockout studies and more sophisticated functional analyses. Researchers can generate complete HSE1 knockout strains by designing guide RNAs targeting the HSE1 gene, followed by homology-directed repair to introduce selectable markers, allowing for phenotypic characterization of cells lacking this protein. Beyond simple knockouts, CRISPR-Cas9 enables the creation of point mutations in specific domains of HSE1 to assess the functional importance of individual residues or motifs, providing more nuanced insights than complete gene deletion. The technology also facilitates the introduction of epitope tags or fluorescent protein fusions at the endogenous HSE1 locus, enabling visualization of the native protein's localization and dynamics without overexpression artifacts. Inducible CRISPR interference (CRISPRi) systems can be employed for temporal control of HSE1 expression, allowing researchers to study the immediate consequences of HSE1 depletion. For more complex analyses, CRISPR-based screening approaches using libraries of guide RNAs targeting potential HSE1 interactors can identify genetic interactions and functional relationships. The implementation of CRISPR-Cas9 techniques requires optimization of transformation protocols and careful design of repair templates for C. glabrata, but these methods ultimately provide unprecedented precision in dissecting HSE1's contributions to cellular processes and pathogenesis.

How might targeting HSE1 affect antifungal susceptibility in C. glabrata?

Targeting HSE1 and the ESCRT machinery represents a potential novel approach to enhancing antifungal susceptibility in C. glabrata, particularly in strains exhibiting resistance to conventional antifungals. Disruption of HSE1 function could potentially impair the fungus's ability to adapt to stress conditions, including those induced by antifungal drugs, similar to how mutations in transcriptional regulators like CgXbp1 affect drug resistance . The ESCRT pathway contributes to membrane remodeling and protein trafficking, processes that may influence the cell surface composition and the distribution of drug efflux pumps that contribute to antifungal resistance. Compounds that inhibit HSE1 function might synergize with existing antifungals by compromising cellular stress responses and adaptation mechanisms that normally protect C. glabrata from drug action. Research approaches to explore this potential include screening for small molecule inhibitors of HSE1-protein interactions, testing combinations of such inhibitors with conventional antifungals against resistant C. glabrata isolates, and characterizing how HSE1 disruption affects the expression and localization of known drug resistance factors. Genetic studies comparing antifungal susceptibility profiles between wild-type and HSE1-deficient strains under various growth conditions would provide valuable insights into whether this protein represents a viable target for novel therapeutic strategies aimed at overcoming the increasing problem of antifungal resistance in C. glabrata infections .

What experimental approaches can assess HSE1 as a potential drug target?

Rigorous evaluation of HSE1 as a potential drug target requires multi-faceted experimental approaches that address target validation, druggability assessment, and therapeutic potential. Initial target validation should include genetic approaches such as CRISPR-Cas9-mediated gene deletion or conditional expression systems to confirm that HSE1 disruption compromises C. glabrata viability or virulence without affecting commensal fungi or host cells. High-throughput phenotypic screens can identify small molecules that mimic the effects of genetic HSE1 disruption, providing lead compounds for further development. Structure-based drug design approaches, requiring high-resolution structural data of HSE1 (obtained through X-ray crystallography or cryo-EM), can guide the development of specific inhibitors targeting functional domains or protein-protein interaction surfaces. In vitro biochemical assays measuring HSE1 activity, such as ubiquitin binding or protein-protein interactions, can be developed into screening platforms for identifying inhibitory compounds. Cellular thermal shift assays (CETSA) can confirm whether candidate compounds engage HSE1 within living cells. Animal models of C. glabrata infection, such as the Galleria mellonella model described in the search results, provide systems for evaluating whether HSE1 inhibitors reduce fungal virulence in vivo . Finally, selectivity assessments comparing effects on pathogenic fungi versus human cells are crucial for determining the therapeutic window of potential HSE1-targeting compounds, ensuring they specifically affect the fungal protein without targeting human homologs.

What proteomics approaches can identify HSE1 interaction partners in C. glabrata?

Advanced proteomics approaches offer powerful means to comprehensively identify HSE1 interaction partners in C. glabrata, providing insights into its functional networks and potential roles in pathogenesis. Affinity purification coupled with mass spectrometry (AP-MS) represents a cornerstone approach, where epitope-tagged HSE1 can be expressed in C. glabrata and purified together with its interacting proteins for subsequent mass spectrometric identification. Proximity-based labeling methods like BioID or APEX provide complementary information by identifying proteins in close spatial proximity to HSE1 in living cells, potentially capturing transient interactions missed by AP-MS. Cross-linking mass spectrometry (XL-MS) offers additional advantages by chemically stabilizing protein-protein interactions before purification, preserving weak or transient associations and providing structural information about interaction interfaces. For more targeted analyses, selected reaction monitoring (SRM) or parallel reaction monitoring (PRM) can quantitatively track specific HSE1 interactions across different conditions, such as during macrophage infection or antifungal treatment . Comparative proteomic approaches analyzing the composition of extracellular vesicles from wild-type versus HSE1-deficient C. glabrata can reveal how this protein influences EV cargo selection . When interpreting proteomics data, sophisticated bioinformatic analyses including interaction network visualization, functional enrichment analysis, and comparison with known ESCRT pathway components can highlight biologically significant interactions and place HSE1 within the broader context of C. glabrata cellular pathways.

What microscopy techniques are optimal for visualizing HSE1 localization and dynamics?

Advanced microscopy techniques offer diverse approaches for visualizing HSE1 localization and dynamics in C. glabrata, providing insights into its spatial distribution, temporal regulation, and functional roles. Super-resolution microscopy methods, including structured illumination microscopy (SIM) and stimulated emission depletion (STED) microscopy, can resolve HSE1 localization with precision below the diffraction limit, potentially revealing its organization within membrane microdomains during vesicle formation. Live-cell imaging using fluorescently tagged HSE1 enables real-time visualization of protein dynamics during processes like endosome maturation and multivesicular body formation. Fluorescence recovery after photobleaching (FRAP) and fluorescence loss in photobleaching (FLIP) techniques can quantify HSE1 mobility and exchange rates between different cellular compartments. For studying HSE1's interactions with other ESCRT components, Förster resonance energy transfer (FRET) or bimolecular fluorescence complementation (BiFC) provide direct visual evidence of protein-protein interactions in living cells. Correlative light and electron microscopy (CLEM) combines the specificity of fluorescence imaging with the ultrastructural detail of electron microscopy, allowing precise localization of HSE1 relative to membranous structures like multivesicular bodies. When studying HSE1 during host-pathogen interactions, intravital microscopy can visualize its dynamics during C. glabrata infection of animal models. These complementary microscopy approaches collectively provide a comprehensive view of HSE1's subcellular localization, dynamics, and functional interactions during normal cellular processes and under infection-relevant conditions.

How can researchers address challenges in distinguishing HSE1-specific effects from general ESCRT pathway disruption?

Distinguishing HSE1-specific effects from general ESCRT pathway disruption presents a significant challenge that requires sophisticated experimental approaches and careful controls. Domain-specific mutations targeting HSE1 functional regions without completely ablating the protein can help isolate specific functions while minimizing broader pathway disruption. Comparing phenotypes between HSE1 disruption and perturbation of other ESCRT components can reveal shared versus unique outcomes, with shared phenotypes likely representing general ESCRT pathway functions while divergent effects may indicate HSE1-specific roles. Temporal control of HSE1 disruption using systems like auxin-inducible degrons or tetracycline-regulated expression allows researchers to distinguish immediate consequences (likely direct HSE1 effects) from later adaptations (potentially representing broader pathway compensation). Rescue experiments with HSE1 mutants lacking specific functional domains can determine which protein features are necessary for complementing different phenotypic defects. Quantitative proteomics comparing the composition of ESCRT complexes in wild-type versus HSE1-deficient cells can identify specific changes in complex assembly or stability. Combined with cell biology approaches measuring distinct ESCRT-dependent processes (MVB formation, autophagy, cytokinesis), these molecular analyses can associate HSE1 with specific cellular functions. Cross-species complementation studies testing whether HSE1 homologs from other fungi can rescue C. glabrata HSE1 deficiency provide insights into conserved versus species-specific functions. These multifaceted approaches collectively enable researchers to dissect HSE1's unique contributions to cellular physiology and pathogenesis from its roles within the broader ESCRT machinery.

What technical limitations impact reproducibility in HSE1 research?

Multiple technical limitations impact reproducibility in HSE1 research, creating challenges that researchers must address through standardized protocols and comprehensive reporting. Recombinant protein expression represents a significant source of variability, with differences in expression systems, purification methods, and protein storage conditions potentially affecting HSE1 structure and activity across laboratories. The absence of standardized activity assays for HSE1 makes functional comparisons between studies difficult, as researchers may employ diverse readouts ranging from protein binding affinity to cellular phenotypes. Variations in C. glabrata strains used across different research groups introduce genetic background effects that can influence HSE1-related phenotypes, particularly important given strain-dependent differences in virulence and stress responses. Environmental conditions during experimental procedures, including media composition, temperature, pH, and growth phase, can significantly affect ESCRT pathway function and HSE1 activity. For studies in infection models, variations in host cell types, activation states, and culture conditions add further complexity that impacts reproducibility of host-pathogen interaction studies. Antibody specificity issues in immunological detection methods may lead to inconsistent results when different antibodies or detection protocols are used across studies. Limited reporting of detailed methodological parameters in publications compounds these challenges by omitting critical factors that influence experimental outcomes. Addressing these limitations requires comprehensive reporting of experimental conditions, establishment of reference strains and standardized protocols, sharing of key reagents like expression constructs and antibodies, and validation of results across multiple experimental systems and conditions.

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.