Recombinant Pseudendoclonium akinetum Photosystem II reaction center protein H (psbH)

Shipped with Ice Packs
In Stock

Description

Recombinant Production and Characterization

While no direct reports exist for Pseudendoclonium akinetum psbH, recombinant production methods for homologous proteins (e.g., Chaetosphaeridium globosum, Arabidopsis thaliana) provide a template:

ParameterExample (Chaetosphaeridium globosum)Example (Arabidopsis thaliana)
Host OrganismE. coli E. coli or Yeast
TagN-terminal His-tag His-tag or GST fusion
Purity>90% (SDS-PAGE) ≥85% (SDS-PAGE)
Expression Yield~2.1 µg/mL culture Not explicitly reported
Reconstitution BufferTris/PBS, 6% trehalose, pH 8.0 Variable (e.g., glycerol added)

Key Challenges:

  • Solubility: PsbH’s hydrophobic transmembrane domain often necessitates fusion partners (e.g., GST) to improve solubility .

  • Stability: Repeated freeze-thaw cycles degrade recombinant psbH; storage at -20°C/-80°C with glycerol is recommended .

Functional Roles in PSII

PsbH contributes to PSII integrity through multiple mechanisms:

Phosphorylation-Dependent Regulation

  • Repair Cycle: Phosphorylation of PsbH’s N-terminal residues facilitates PSII repair under photodamage, enabling migration of damaged complexes to stromal lamellae .

  • Stress Response: In Arabidopsis, the RNA-binding protein HCF107 stabilizes psbH mRNA, ensuring PsbH accumulation during photooxidative stress .

Interaction with Accessory Proteins

  • PsbT and PsbN: PsbH’s proximity to PsbT (4 kDa) and PsbN (controversial subunit) suggests cooperative roles in QB-site stability and electron transport .

Research Gaps and Opportunities

  1. Lack of Direct Studies: No published data exist on Pseudendoclonium akinetum psbH’s recombinant form.

  2. Functional Redundancy: Overexpression of nuclear-encoded psbH in Arabidopsis rescues PSII defects, suggesting potential for synthetic biology applications .

  3. Phosphorylation Dynamics: The interplay between PsbH phosphorylation and PSII repair in Pseudendoclonium remains unexplored.

Comparative Analysis of PsbH Homologs

OrganismPsbH LengthPhosphorylation SitesKey Functional Role
Synechocystis sp. PCC 680374 aa N-terminal serine/threonineQB-site stabilization, photoprotection
Arabidopsis thaliana~8 kDa N-terminal residuesCP47 accumulation, stress tolerance
Chaetosphaeridium globosum74 aa Not explicitly reportedRecombinant production via His-tag

Product Specs

Form
Lyophilized powder
Note: We will prioritize shipping the format currently in stock. However, if you have specific format requirements, please indicate them in your order, and we will accommodate your request.
Lead Time
Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery times.
Note: All our proteins are shipped with standard blue ice packs. If dry ice shipping is required, please inform us in advance as additional fees will apply.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
It is recommended to briefly centrifuge the vial before opening to ensure the contents settle to the bottom. Reconstitute the protein in deionized sterile 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%, which can be used as a reference.
Shelf Life
Shelf life is influenced by various factors including storage conditions, buffer components, temperature, and protein stability.
Generally, liquid forms have a shelf life of 6 months at -20°C/-80°C. Lyophilized forms have a shelf life of 12 months 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 will be determined during the manufacturing process.
The tag type is determined during production. If you have a specific tag type in mind, please inform us, and we will prioritize its development.
Synonyms
psbH; Photosystem II reaction center protein H; PSII-H; Photosystem II 10 kDa phosphoprotein
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-78
Protein Length
full length protein
Species
Tupiella akineta (Green alga) (Pseudendoclonium akinetum)
Target Names
psbH
Target Protein Sequence
MATKTSKSTDDLGMTTALGTLLRPLNSEYGKVAPGWGTATLMGVFMALFAVFLVIILEIY NSSVILDDVQMSWQSLAK
Uniprot No.

Target Background

Function
Photosystem II (PSII) reaction center protein H (psbH) is a crucial component of the core complex in photosystem II. Its presence is essential for the stability and assembly of PSII. PSII is a light-driven water:plastoquinone oxidoreductase that utilizes light energy to extract electrons from H(2)O, generating O(2) and a proton gradient subsequently used for ATP formation. This complex comprises a core antenna complex that captures photons and an electron transfer chain that converts photonic excitation into charge separation.
Protein Families
PsbH family
Subcellular Location
Plastid, chloroplast thylakoid membrane; Single-pass membrane protein.

Q&A

What is the Photosystem II reaction center protein H (psbH) in Pseudendoclonium akinetum?

Photosystem II reaction center protein H (psbH) in Pseudendoclonium akinetum is a small chloroplast-encoded protein that functions as an integral component of the photosystem II complex, critical for photosynthetic electron transport . The psbH gene is conserved across photosynthetic organisms and in P. akinetum is located within the chloroplast genome which spans approximately 195,867 base pairs . Unlike some other photosynthetic proteins, psbH is relatively small but plays an essential role in the assembly, stability, and function of the PSII reaction center. The protein participates in the early steps of photosynthesis, specifically in the water-splitting reactions that generate molecular oxygen.

How does the psbH protein from Pseudendoclonium akinetum compare to those from other green algae?

The psbH protein from Pseudendoclonium akinetum shares structural and functional similarities with those from other green algae, but with notable differences reflecting evolutionary adaptation . Comparative genomic analyses show that P. akinetum psbH is part of conserved gene clusters often found in other Ulvophyceae members, such as the psbE-psbF-psbL-psbJ cluster . When compared with related species like Bryopsis hypnoides and Chlorella vulgaris, the P. akinetum psbH shows conservation of key functional domains while displaying sequence variations that may relate to specific environmental adaptations. Unlike some related algae, the genome structure of P. akinetum has distinctive organizational features, which may influence psbH expression and regulation . The protein's evolutionary conservation suggests its fundamental importance in photosynthesis across diverse algal lineages.

What is the gene structure and organization of psbH in the P. akinetum chloroplast genome?

The psbH gene in Pseudendoclonium akinetum is encoded within the 195,867-bp chloroplast genome . It is located within a functional gene cluster that includes other photosystem components. The chloroplast genome of P. akinetum contains a total of 111 functional genes in Bryopsis hypnoides (a related species), including 69 protein-coding genes, 5 ribosomal RNA genes, and 37 tRNA genes, suggesting similar organization in P. akinetum . The psbH gene is part of conserved gene clusters often maintained across green algae, such as psbE-psbF-psbL-psbJ . Unlike some other algal chloroplast genomes, the P. akinetum genome shows unique structural features which may affect the transcriptional regulation of psbH. The gene arrangement surrounding psbH provides insights into the evolutionary history and functional constraints of photosystem II components in this green alga lineage.

What expression systems are optimal for recombinant production of P. akinetum psbH protein?

For recombinant production of P. akinetum psbH protein, Escherichia coli expression systems have proven most effective due to their high yield and simplified purification protocols . Based on methods used for similar photosystem proteins, the psbH gene should be codon-optimized for E. coli expression and cloned into vectors containing strong inducible promoters such as T7 . A polyhistidine (His) tag fusion, preferably at the N-terminus as demonstrated with similar proteins, facilitates efficient purification through nickel affinity chromatography . Expression conditions typically require optimization with IPTG induction concentrations of 0.5-1.0 mM at lower temperatures (16-25°C) to prevent inclusion body formation. Alternative expression systems including yeast (Pichia pastoris) may be considered for proper folding of this membrane protein, though with potentially lower yields. Chaperone co-expression strategies may improve the solubility and functionality of the recombinant product.

What purification strategies yield the highest purity and functional integrity of recombinant psbH?

Purification of recombinant psbH protein from P. akinetum requires a multi-step approach to maintain structural integrity while achieving high purity . The recommended protocol begins with immobilized metal affinity chromatography (IMAC) using Ni-NTA resins for initial capture of His-tagged protein, followed by size exclusion chromatography to remove aggregates and contaminants of different molecular weights . Buffer composition is critical, with Tris/PBS-based buffers (pH 8.0) containing 6% trehalose proving effective for stability during purification and storage . For membrane proteins like psbH, addition of mild detergents such as n-dodecyl β-D-maltoside (DDM) at 0.03-0.05% helps maintain native conformation during extraction and purification. Ion exchange chromatography may serve as an additional polishing step to achieve >90% purity. To preserve function, purification should be performed at 4°C with protease inhibitors, and the final product is best stored as aliquots in buffer containing glycerol (30-50%) at -80°C to prevent freeze-thaw damage.

How can researchers optimize protein yield and solubility when expressing recombinant psbH?

Optimizing yield and solubility of recombinant P. akinetum psbH involves strategic modifications to expression conditions and protein engineering approaches . Researchers should test multiple E. coli strains (BL21(DE3), C41(DE3), C43(DE3)) specifically designed for membrane protein expression. Growth temperature reduction to 16-18°C post-induction significantly improves soluble protein yield by slowing folding kinetics. Inducer concentration optimization is critical - starting with an IPTG range of 0.1-0.5 mM and monitoring expression through small-scale trials. For this photosystem protein, co-expression with molecular chaperones (GroEL/GroES, DnaK/DnaJ) has demonstrated improved solubility. Fusion tags beyond the standard His-tag, such as MBP (maltose-binding protein) or SUMO, can dramatically enhance solubility, though cleavage options should be incorporated for functional studies. Addition of mild detergents (0.1-0.5% DDM or CHAPS) to lysis buffers effectively solubilizes membrane-associated psbH. Finally, optimizing growth media with supplemental components including 1% glucose, specific metal ions, and osmolytes can further improve expression outcomes.

What structural features distinguish the psbH protein from other photosystem II components?

The psbH protein of Pseudendoclonium akinetum possesses distinctive structural features that differentiate it from other PSII components, though sharing the conservation pattern typical of photosynthetic organisms . Based on comparative analysis with related algal species, psbH is characterized by a single transmembrane α-helix that spans the thylakoid membrane, with small hydrophilic segments extending into the stromal and lumenal sides . The protein's key structural motif includes a phosphorylation site on the N-terminal threonine residue, which is critical for its regulatory function in photosynthesis. Unlike larger PSII proteins such as D1 and D2 that contain multiple transmembrane helices and form the reaction center core, psbH serves as an auxiliary component that interacts with these core proteins to stabilize the complex. The small size (approximately 10 kDa) and high hydrophobicity of psbH reflect its evolutionary adaptation to membrane integration, distinct from the more hydrophilic extrinsic PSII components like the oxygen-evolving complex proteins.

What methods are most effective for analyzing the interaction between psbH and other photosystem II proteins?

For analyzing psbH interactions with other photosystem II proteins, a multi-methodological approach yields the most comprehensive results . Co-immunoprecipitation (Co-IP) using antibodies specific to psbH or its binding partners provides initial evidence of protein-protein interactions in vivo. This should be complemented by pull-down assays with recombinant His-tagged psbH to identify direct binding partners. Crosslinking mass spectrometry (XL-MS) offers a powerful approach for mapping the exact interaction sites, using reagents like DSS or EDC followed by digest and LC-MS/MS analysis . Blue native polyacrylamide gel electrophoresis (BN-PAGE) enables visualization of intact protein complexes and can reveal whether psbH knockouts/mutations alter PSII assembly. For higher resolution structural information, cryo-electron microscopy has proven superior to X-ray crystallography for membrane protein complexes like PSII, potentially revealing the exact position of psbH within the complex. Förster resonance energy transfer (FRET) using fluorescently labeled proteins can demonstrate proximity and dynamic interactions in reconstituted systems or in vivo. Finally, surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC) provides quantitative binding kinetics and thermodynamic parameters of psbH-partner interactions.

How does phosphorylation affect the function of psbH in photosynthetic processes?

Phosphorylation of the psbH protein plays a crucial regulatory role in photosynthetic processes, mediating both short-term responses to light conditions and long-term adaptations . The N-terminal threonine residue of psbH serves as the primary phosphorylation site, with modification catalyzed by redox-sensitive thylakoid protein kinases that respond to the plastoquinone pool reduction state. Upon phosphorylation, psbH undergoes conformational changes that affect its interaction with other PSII components, particularly the D1 protein . This phosphorylation-dependent interaction modulates the rate of D1 turnover during the PSII repair cycle following photodamage, a critical process for maintaining photosynthetic efficiency under variable light conditions. Additionally, phosphorylated psbH contributes to the reorganization of photosystem complexes between stacked and unstacked thylakoid regions during state transitions, facilitating energy distribution between PSI and PSII. Experimental approaches using site-directed mutagenesis to create phospho-mimetic or phospho-null psbH variants have demonstrated that phosphorylation status directly influences PSII function, assembly dynamics, and susceptibility to photoinhibition under high light stress.

How does the psbH gene differ between Pseudendoclonium akinetum and other green algae at the sequence level?

Comparative genomic analysis reveals that the psbH gene in Pseudendoclonium akinetum exhibits both conserved elements and distinctive features when compared to other green algal species . At the sequence level, P. akinetum psbH maintains the conserved functional domains essential for photosystem II activity while displaying nucleotide variations that reflect its evolutionary history within the Ulvophyceae class . Analysis of the 195,867-bp chloroplast genome sequence shows that the gene context surrounding psbH in P. akinetum differs from related species, though certain gene clusters remain conserved, such as the psbE-psbF-psbL-psbJ arrangement found in multiple green algae . The table below summarizes key comparative features:

SpeciespsbH gene length (bp)GC content (%)Conserved domainsNotable sequence variations
P. akinetum~22526-30Transmembrane helix, N-terminal phosphorylation siteExtended N-terminal region
Bryopsis hypnoides~22528-32Similar to P. akinetumVariations in stromal-exposed sequences
Chlorella vulgaris~22233-36Core functional domains preservedHigher sequence conservation
Chaetosphaeridium globosum~21925-30All functional motifs presentDistinct C-terminal sequence

These sequence-level differences likely reflect adaptive evolution to different ecological niches while maintaining the protein's essential photosynthetic function .

What evolutionary insights can be gained from studying psbH across different algal lineages?

Studying psbH across diverse algal lineages provides significant evolutionary insights into both photosynthetic mechanisms and algal phylogeny . The conservation of psbH throughout the green algal lineage indicates its fundamental importance to photosystem II function, despite the protein's relatively small size. Phylogenetic analysis of psbH sequences reveals patterns of selection pressure that distinguish essential functional domains from more variable regions, offering insights into structure-function relationships in photosynthetic apparatuses . In Pseudendoclonium akinetum and related Ulvophyceae, psbH exhibits distinctive sequence signatures that align with the class's evolutionary radiation and adaptation to various ecological niches . Comparative genomic approaches reveal that while the gene's position within the chloroplast genome varies among lineages, certain gene clusters containing psbH are broadly conserved, suggesting functional constraints on genome reorganization during evolution . The presence of psbH in chloroplast genomes across diverse algal groups but with varying sequence conservation rates serves as a valuable marker for reconstructing evolutionary relationships, especially when integrated with data from other chloroplast genes. This evolutionary perspective enhances our understanding of both photosystem development and the broader patterns of plastid genome evolution in eukaryotic photosynthetic organisms.

How do the structural genetics of the psbH region compare between Pseudendoclonium akinetum and other model photosynthetic organisms?

The structural genetics of the psbH region in Pseudendoclonium akinetum reveals distinct organizational patterns when compared with other model photosynthetic organisms, reflecting both functional constraints and lineage-specific evolutionary trajectories . In the 195,867-bp chloroplast genome of P. akinetum, the psbH gene exists within a specific genomic context that differs from that observed in land plants but shares similarities with other green algae . Unlike land plants that typically have the psbH gene located within the inverted repeat regions of their chloroplast genomes, P. akinetum's chloroplast DNA shows a different arrangement more characteristic of algal lineages . Comparative analysis reveals that while certain gene clusters remain conserved across diverse photosynthetic organisms (e.g., psbE-psbF-psbL-psbJ and rps2-atpI-atpH-atpF-atpA), the broader genomic architecture surrounding psbH varies significantly . In Bryopsis hypnoides, a related green alga, the chloroplast genome lacks the typical inverted repeat structure, suggesting evolutionary plasticity in this region across algal lineages . The absence of introns in the P. akinetum psbH gene contrasts with some land plants where introns have been acquired during evolution. Additionally, the regulatory elements flanking the psbH coding region in P. akinetum show distinctive features that may influence expression patterns. These structural genetic differences provide valuable markers for understanding the evolutionary history and functional adaptation of photosynthetic machinery across diverse lineages.

What are the challenges and solutions for investigating psbH protein turnover in vivo?

Investigating psbH protein turnover in vivo presents several methodological challenges that require specialized approaches . The primary difficulty stems from the protein's small size (~10 kDa), membrane localization, and relatively low abundance compared to other photosystem components. To overcome these challenges, researchers should employ pulse-chase experiments using isotopically labeled amino acids (¹⁵N or ¹³C) followed by immunoprecipitation and mass spectrometry to quantify protein synthesis and degradation rates . For temporal resolution of turnover dynamics, an inducible expression system coupled with epitope tagging (FLAG or HA) enables controlled initiation of labeled protein synthesis. The membrane-bound nature of psbH necessitates careful optimization of extraction conditions using non-ionic detergents (0.5-1% n-dodecyl β-D-maltoside) that preserve native protein interactions while enabling efficient solubilization . Fluorescence recovery after photobleaching (FRAP) using GFP-tagged psbH provides spatial information about protein mobility and replacement rates within thylakoid membranes, though care must be taken to ensure the tag doesn't disrupt function. To distinguish between different degradation pathways, specific protease inhibitors targeting chloroplast proteases (FtsH, Deg, ClpP) should be employed. Finally, correlating psbH turnover with photosynthetic activity parameters under varying light conditions reveals the functional significance of the turnover dynamics.

How can CRISPR-Cas9 technology be applied to study psbH function in Pseudendoclonium akinetum?

Applying CRISPR-Cas9 technology to study psbH function in Pseudendoclonium akinetum requires specialized approaches adapted for chloroplast genome editing in algae . The protocol begins with designing guide RNAs (gRNAs) targeting the psbH locus with high specificity, using chloroplast codon optimization and promoters effective in green algae such as the endogenous atpA promoter. Delivery of the CRISPR-Cas9 components presents a significant challenge in algal systems; researchers should optimize biolistic transformation parameters (helium pressure 1100-1350 psi, gold particle size 0.6 μm) for chloroplast targeting, or alternatively employ polyethylene glycol-mediated transformation for protoplasts . Homology-directed repair templates should include selectable markers suitable for chloroplast selection, such as spectinomycin resistance (aadA) or restoration of photosynthetic growth in a psbH-deficient background. For precise functional studies, researchers should create a series of targeted mutations: complete knockouts to assess essentiality, phosphorylation site mutations (T→A or T→E) to study regulatory mechanisms, and sequence substitutions to introduce specific algal variants for evolutionary studies. Post-transformation, heteroplasmy management is critical; repeated selection on increasing antibiotic concentrations promotes homoplasmy. Phenotypic analysis should include photosynthetic efficiency measurements (Fv/Fm, P700 oxidation), photoprotection capacity under high light, and growth rates under varying light regimes to comprehensively characterize the functional impact of psbH modifications.

What role does psbH play in photoprotection mechanisms under high light stress?

The psbH protein plays a multifaceted role in photoprotection mechanisms under high light stress, serving as both a structural stabilizer and regulatory component within the photosystem II complex . Under excessive light conditions, psbH undergoes rapid phosphorylation at its N-terminal threonine residue, which triggers conformational changes critical for the PSII repair cycle. This phosphorylation is mediated by the STN8 kinase (or its algal homolog) in response to the redox state of the plastoquinone pool . Phosphorylated psbH facilitates the migration of photodamaged PSII complexes from grana stacks to stromal thylakoids, where repair occurs through selective degradation and replacement of the D1 protein. Additionally, psbH interacts directly with the PSII reaction center proteins to modulate excitation energy distribution, thereby reducing excess energy pressure on the photosystem during high light exposure . Experimental evidence from mutant studies in related organisms demonstrates that psbH-deficient strains exhibit increased photoinhibition, slower recovery from photodamage, and altered non-photochemical quenching (NPQ) capacity. In Pseudendoclonium akinetum, which inhabits variable light environments, psbH likely contributes to the alga's ability to withstand fluctuating light intensities through these regulatory mechanisms. Furthermore, psbH appears to influence the binding of extrinsic proteins that stabilize the oxygen-evolving complex, thereby maintaining photosynthetic water oxidation efficiency even under stress conditions.

What spectroscopic methods are most informative for analyzing recombinant psbH function?

For analyzing recombinant psbH function, multiple complementary spectroscopic techniques provide comprehensive insights into the protein's structural integrity and activity . Circular dichroism (CD) spectroscopy in the far-UV range (190-250 nm) offers critical information about secondary structure elements, confirming proper folding of the transmembrane α-helical domains characteristic of psbH. Near-UV CD (250-350 nm) provides tertiary structural fingerprints. Fluorescence spectroscopy utilizing the protein's intrinsic tryptophan residues (excitation at 280 nm, emission scan 300-400 nm) reveals conformational states and potential ligand interactions . For functional analysis, oxygen polarography using a Clark-type electrode directly measures oxygen evolution rates in reconstituted PSII complexes containing recombinant psbH. Time-resolved fluorescence decay kinetics using picosecond fluorescence techniques assess energy transfer efficiency within PSII, with psbH-dependent changes in decay lifetimes indicating functional integration. Electron paramagnetic resonance (EPR) spectroscopy provides detailed information about the redox-active centers in PSII and how psbH influences their properties. Fourier-transform infrared (FTIR) difference spectroscopy combined with isotope labeling allows mapping of specific residue contributions to protein function. For monitoring phosphorylation states, phosphorus-31 nuclear magnetic resonance (³¹P-NMR) spectroscopy offers non-invasive detection of phosphorylated residues and their conformational environments, critical for understanding psbH regulatory mechanisms.

How can researchers differentiate between the effects of psbH mutations and assembly defects in photosystem II?

Differentiating between direct effects of psbH mutations and secondary assembly defects in photosystem II requires a systematic analytical approach combining biochemical, spectroscopic, and functional assessments . Researchers should first employ blue native polyacrylamide gel electrophoresis (BN-PAGE) followed by second-dimension SDS-PAGE to visualize the integrity and composition of PSII complexes, comparing wildtype with psbH mutants to identify specific assembly intermediates or aberrant complexes . Complementary immunoblot analysis using antibodies against various PSII subunits can reveal whether particular components fail to incorporate in the absence of functional psbH. For kinetic differentiation, pulse-chase labeling with ³⁵S-methionine followed by time-course analysis distinguishes between initial assembly defects and accelerated degradation of properly assembled but unstable complexes. Spectroscopic approaches provide functional discrimination: prompt fluorescence induction kinetics reflect primary charge separation capacity, while delayed fluorescence specifically indicates recombination reactions within assembled reaction centers. Electron transport measurements using artificial electron acceptors that insert at different points in the electron transport chain can pinpoint where functional blocks occur. Structural information from crosslinking mass spectrometry reveals specific interaction partners missing in mutant complexes. Finally, complementation experiments reintroducing wildtype or mutant psbH variants into null backgrounds provide definitive evidence of direct versus indirect effects, particularly when combined with step-wise assembly analysis using synchronized cell cultures or chloroplast development systems.

What are the best approaches for studying the phosphorylation dynamics of psbH in different light conditions?

Studying the phosphorylation dynamics of psbH under varying light conditions requires an integrated approach combining in vivo monitoring with detailed biochemical analysis . The optimal methodology begins with in vivo radiolabeling using ³²P-orthophosphate under defined light regimes (dark, low light, high light, fluctuating light), followed by immunoprecipitation of psbH to track phosphorylation kinetics. For higher temporal resolution, researchers should employ Phos-tag™ SDS-PAGE, which separates phosphorylated from non-phosphorylated psbH forms without radioactive labeling, enabling rapid sampling during light transitions . Mass spectrometry approaches using either MALDI-TOF or LC-MS/MS with phosphopeptide enrichment (TiO₂ chromatography) provide site-specific phosphorylation information and can identify previously unknown modification sites. Phospho-specific antibodies developed against the known N-terminal phosphorylation site enable immunofluorescence microscopy to visualize the spatial distribution of phosphorylated psbH within thylakoid membranes during light transitions. To correlate phosphorylation with functional consequences, researchers should simultaneously measure photosynthetic parameters (oxygen evolution, fluorescence induction, P700 oxidation) alongside phosphorylation status. Inhibitor studies using kinase inhibitors (e.g., staurosporine derivatives) and phosphatase inhibitors (e.g., NaF, microcystin-LR) help identify the regulatory enzymes involved. For mechanistic insight, reconstitution experiments with purified components (kinases, phosphatases, psbH variants) in liposome systems can establish the minimum requirements for light-responsive phosphorylation. Finally, comparison across different physiological states (circadian time points, developmental stages) reveals how psbH phosphorylation contributes to longer-term photosynthetic adaptations.

How should researchers address protein degradation issues when working with recombinant psbH?

Addressing protein degradation of recombinant psbH requires a comprehensive strategy targeting each stage of the expression and purification process . During expression, researchers should optimize induction parameters, preferably using lower temperatures (16-18°C) and reduced IPTG concentrations (0.1-0.3 mM) to slow production and allow proper folding . The expression host strain is critical - BL21(DE3) pLysS or Rosetta™ strains with reduced protease activity show improved stability for membrane proteins like psbH. Immediately upon cell lysis, a cocktail of protease inhibitors should be employed, including PMSF (1 mM), EDTA (1 mM), leupeptin (10 μg/mL), and aprotinin (2 μg/mL) . Buffer optimization is essential, with data indicating Tris/PBS-based buffers (pH 8.0) containing 6% trehalose significantly enhance psbH stability . The addition of reducing agents (2-5 mM DTT or 1-2 mM β-mercaptoethanol) prevents oxidative damage. Temperature control throughout purification (maintaining 4°C) and minimizing purification duration is crucial. For storage, flash-freezing aliquots in liquid nitrogen with 30-50% glycerol prevents freeze-thaw degradation . If degradation persists despite these measures, researchers should consider fusion partners like thioredoxin or SUMO that enhance stability, or explore detergent screening (testing DDM, LMNG, or digitonin) to better mimic the native membrane environment. Mass spectrometry can identify specific degradation sites, allowing targeted mutagenesis of susceptible residues without compromising function.

What strategies help overcome challenges in obtaining structurally intact and functional psbH protein?

Obtaining structurally intact and functional psbH protein presents unique challenges that require specialized approaches beyond standard protein production techniques . The critical first step involves optimizing the expression construct design with careful consideration of tag position - N-terminal tags typically preserve function better than C-terminal modifications for this membrane protein . Codon optimization specific to the expression host improves translation efficiency and reduces premature termination. For bacterial expression, the inclusion of chloroplast-specific chaperones like cpn60/cpn10 can dramatically improve proper folding. During membrane extraction, detergent selection is crucial; initial screening should include a range of concentrations (0.5-2%) of mild detergents (DDM, LMNG, GDN) with stepwise extraction protocols to maintain native conformation . Amphipols (A8-35) or nanodiscs provide superior stability during purification and downstream applications by mimicking the lipid bilayer environment. Functional validation requires multiple complementary approaches: circular dichroism to confirm secondary structure, phosphorylation assays using recombinant STN7/STN8 kinases to verify regulatory site accessibility, and reconstitution with PSII core components followed by oxygen evolution measurements. For structural studies, limited proteolysis coupled with mass spectrometry identifies stable protein domains and potential flexible regions. If bacterial expression consistently fails to produce functional protein, alternative expression systems including insect cells (baculovirus) or cell-free systems supplemented with lipid nanodiscs may yield superior results, though at higher cost and lower yield.

How can researchers overcome difficulties in detecting and quantifying low-abundance psbH in complex photosynthetic samples?

Detecting and quantifying low-abundance psbH protein in complex photosynthetic samples requires specialized techniques that enhance sensitivity while maintaining specificity . The most effective approach begins with optimized extraction methods using denaturing buffers containing 8M urea or 2% SDS to ensure complete solubilization of membrane-embedded psbH. For enhanced detection sensitivity, researchers should employ western blotting with highly specific antibodies raised against synthetic peptides corresponding to unique psbH epitopes, preferably using chemiluminescent detection with signal enhancers or near-infrared fluorescent secondary antibodies that provide linear quantitation over a broader range than traditional ECL methods . When antibodies are unavailable or cross-reactivity is problematic, targeted proteomics using selected reaction monitoring (SRM) or parallel reaction monitoring (PRM) mass spectrometry enables specific detection of psbH-derived peptides with quantification limits in the femtomole range. Sample preparation for MS-based approaches should include filter-aided sample preparation (FASP) with sequential digestion using both Lys-C and trypsin to improve membrane protein coverage. Enrichment strategies using immobilized metal affinity chromatography (IMAC) or titanium dioxide (TiO₂) enable specific isolation of phosphorylated psbH forms. For highly complex samples, implementing dimethyl labeling or TMT isobaric tags allows multiplexed quantification across different experimental conditions while maintaining detection sensitivity. When absolute quantification is required, isotopically labeled peptide standards (AQUA peptides) corresponding to unique psbH sequences provide precise concentration determinations even in complex backgrounds. For spatial detection in intact tissues or cells, proximity ligation assays offer significantly enhanced sensitivity compared to conventional immunofluorescence.

What are the promising research avenues for understanding psbH post-translational modifications beyond phosphorylation?

While phosphorylation of psbH has been well-studied, several promising research avenues exist for investigating additional post-translational modifications (PTMs) that may regulate this protein's function . Mass spectrometry-based PTM profiling using electron transfer dissociation (ETD) fragmentation represents a powerful approach for comprehensive identification of modifications including acetylation, methylation, and redox-based changes on psbH residues. Preliminary evidence from related photosynthetic proteins suggests that lysine acetylation may play a regulatory role in response to metabolic status, warranting targeted investigation in psbH . S-nitrosylation and glutathionylation of cysteine residues merit exploration as potential redox-sensing mechanisms that could link photosynthetic electron transport to psbH function during oxidative stress. Ubiquitination and SUMOylation pathways, though typically cytosolic, have recently been identified in chloroplasts and may regulate psbH turnover rates. Emerging evidence for crosstalk between different PTMs suggests that phosphorylation may prime psbH for subsequent modifications, creating a complex regulatory code. Comparative PTM profiling across diverse environmental conditions (light intensity, temperature, nutrient availability) could reveal condition-specific modification patterns. Development of site-specific antibodies against newly identified modifications would enable temporal and spatial tracking of these PTMs in vivo. Finally, reconstitution experiments with modified and unmodified psbH variants will be essential to establish the functional consequences of these modifications on photosystem II assembly, stability, and activity.

How might structural biology approaches advance our understanding of psbH interactions within the photosystem II complex?

Advanced structural biology approaches offer transformative potential for elucidating psbH interactions within the photosystem II complex at unprecedented resolution . Cryo-electron microscopy (cryo-EM), which has recently achieved near-atomic resolution for membrane protein complexes, represents the most promising technique for capturing psbH in its native context within PSII. Single-particle cryo-EM analysis could reveal conformational changes associated with psbH phosphorylation states or interactions with assembly factors . Complementary approaches include hydrogen-deuterium exchange mass spectrometry (HDX-MS) to map solvent-accessible regions and conformational dynamics at protein-protein interfaces involving psbH. Cross-linking mass spectrometry (XL-MS) using novel MS-cleavable crosslinkers provides direct evidence of spatial proximity between psbH and other PSII subunits, particularly valuable for capturing transient interactions during assembly or repair processes. Solid-state NMR spectroscopy offers unique advantages for membrane proteins like psbH, potentially revealing dynamic aspects of its transmembrane domain interactions. For higher throughput structural analysis, integrative structural biology combining computational modeling with experimental constraints from various techniques will be particularly valuable. The application of AlphaFold2 and RoseTTAFold to predict psbH interactions, validated by experimental approaches, could accelerate progress. Time-resolved structural methods including time-resolved cryo-EM or X-ray free electron laser (XFEL) studies could capture different states of psbH during the photocycle, connecting structural insights to functional mechanisms. Finally, in situ structural approaches like cryo-electron tomography hold promise for visualizing psbH within native thylakoid membranes, providing contextual information about its organization in different thylakoid domains.

What emerging technologies could revolutionize our ability to study the real-time dynamics of psbH in living photosynthetic systems?

Emerging technologies are poised to revolutionize our understanding of real-time psbH dynamics in living photosynthetic systems, offering unprecedented spatiotemporal resolution . Optogenetic approaches using light-sensitive domains fused to psbH or its interaction partners could enable precise temporal control of protein interactions or conformational changes, allowing researchers to trigger specific events and monitor downstream consequences. CRISPR-based technologies beyond gene editing, including CRISPRi for conditional knockdown and CRISPR activation for enhanced expression, provide temporal control over psbH levels without permanent genetic modifications . For visualization, advances in super-resolution microscopy techniques such as PALM, STORM, and MINFLUX now approach the resolution necessary to track individual protein complexes within thylakoid membranes. When combined with split fluorescent protein systems or FRET pairs, these methods could visualize psbH interactions with unprecedented spatial precision. Genetically encoded biosensors designed to report on psbH phosphorylation state or conformation changes would enable real-time monitoring in living cells. The integration of microfluidic systems with live-cell imaging allows precise control of environmental conditions while simultaneously tracking protein dynamics. Single-molecule tracking approaches using photoactivatable fluorescent proteins could reveal the mobility and diffusion characteristics of psbH within thylakoid membranes under different light conditions. Nanobody-based probes that recognize specific conformational states of psbH could distinguish between functional states in vivo. Finally, the application of adaptive optics and light-sheet microscopy to intact photosynthetic tissues promises to extend these high-resolution approaches from cellular to tissue scales, revealing how psbH dynamics vary across different cell types and developmental stages.

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.