Recombinant Oceanobacillus iheyensis Segregation and condensation protein A (scpA)

Shipped with Ice Packs
In Stock

Description

Introduction

The Streptococcus pyogenes C5a peptidase (ScpA) is a highly conserved surface virulence factor present on Group A Streptococcus (GAS) cell surfaces . ScpA has garnered attention as a promising antigenic target for GAS vaccine development due to its high antigenicity, which stimulates specific and immunoprotective antibodies .

Role of Recombinant Proteins

Recombinant proteins like ScpA play a significant role in cell and gene therapies, particularly in research and development .

Applications in Cell and Gene Therapy R&D:

  • Cell Culture and Expansion: Recombinant proteins, such as growth factors and cytokines, stimulate cell growth and expansion by providing necessary signals for proliferation and differentiation .

  • Cell Reprogramming: Recombinant proteins like transcription factors induce pluripotency or direct cell differentiation into specific lineages in gene therapy .

  • Viral Vector Production: They optimize the production of recombinant viruses, such as lentiviruses, used for gene delivery .

  • Purification of Therapeutic Proteins: Recombinant proteins are utilized in purification processes of therapeutic proteins produced through gene therapy .

  • Cell Characterization: They assess the identity, purity, and functionality of engineered cells, ensuring they meet specifications for therapeutic use .

  • Immunomodulation: Some recombinant proteins modulate the immune response, crucial in cell and gene therapies for preventing rejection or enhancing therapeutic effects .

  • Quality Control: Recombinant proteins are used in quality control assays to evaluate the safety and efficacy of cell and gene therapies .

  • In Vivo Imaging: Recombinant proteins, such as luciferase, serve as reporter genes, allowing researchers to track and monitor the expression and localization of therapeutic genes in vivo .

  • Therapeutic Protein Engineering: They are used to engineer therapeutic proteins with enhanced properties like improved stability, reduced immunogenicity, or altered pharmacokinetics .

ScpA's Interaction with C5a

ScpA specifically inactivates the human complement factor hC5a, a potent anaphylatoxin, by cleaving the '68-Lys-|-Asp-69' bond near its C-terminus . This inactivation of hC5a has been identified as a therapeutic target for treating COVID-19 infections . ScpA cleaves recombinant hC5a (rhC5a) at near physiological concentrations .

Enzyme kinetic and binding studies have identified determinants of specificity for ScpA. Surface Plasmon Resonance (SPR) and Isothermal Titration Calorimetry (ITC) studies established a high-affinity ScpA-rhC5a interaction .

Impact of Propeptide Cleavage on ScpA Activity

ScpA undergoes autoproteolysis and cleaves its propeptide for full maturation after signal peptide removal . Propeptide truncation variants of ScpA expressed in Escherichia coli cells have been structurally and functionally characterized . ScpA variants, including ScpA, 79ΔPro, and 92ΔPro, exhibit similar activity against C5a, suggesting a propeptide-independent activity profile of ScpA . CE-SDS and MALDI top-down sequencing analyses highlight a time-dependent propeptide autoproteolysis of ScpA at 37 °C with a distinct end point at A92 and/or D93 .

Inhibition and Activity of SCPA

SCPA activity was unaffected by 0.1 to 10 mM Ca2+, Mg2+, and Mn2+ but was inhibited by the same concentrations of Zn2+ . NaCl inhibits SCPA cleavage of GST-C5a-GFP in a dose-dependent manner, showing high sensitivity to ionic strength .

Product Specs

Form
Lyophilized powder *Note: While we prioritize shipping the format currently in stock, please specify your format preference in order remarks 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. 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. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, provided as a guideline for customers.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms 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
Tag type is determined during the manufacturing process. The tag type is determined during production. Please specify your desired tag type for preferential development.
Synonyms
scpA; OB1828; Segregation and condensation protein A
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-249
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Oceanobacillus iheyensis (strain DSM 14371 / CIP 107618 / JCM 11309 / KCTC 3954 / HTE831)
Target Names
scpA
Target Protein Sequence
MHEGYKVKLD TFEGPLDLLL HLINQFEIDI YDIPVAQITQ QYMEYIHTMQ HLELNIASEY LVMASTLLAI KSQMLLPKQE LEDDLDEEYM EDPREELMQR LIEYRKYKEI AERLKEKESE DNQLYTRPPV VFEFKDIPEK ITTNQTDISI FDMVGALKNM LKRKEWTEPH DTTVQRMDIP IETRMKEVLQ QVQSNSDGLV FDKLFPSPTK NYIVVTFVAV LELMKDKQIY AVQERHFEEL YLYSMEEST
Uniprot No.

Target Background

Function
This protein participates in chromosomal partitioning during cell division. It may function through a condensin-like complex involving Smc and ScpB proteins, which facilitate DNA segregation from mid-cell into both daughter cells.
Database Links

KEGG: oih:OB1828

STRING: 221109.OB1828

Protein Families
ScpA family
Subcellular Location
Cytoplasm.

Q&A

What is Oceanobacillus iheyensis and why is it significant for research?

Oceanobacillus iheyensis HTE831 (strain DSM 14371/JCM 11309/KCTC 3954) is an alkaliphilic and extremely halotolerant Bacillus-related species isolated from deep-sea sediment collected at a depth of 1050 m on the Iheya Ridge . This organism demonstrates remarkable adaptation to extreme environments, capable of growing at salinities of 0-21% (w/v) NaCl at pH 7.5 and 0-18% at pH 9.5, with optimal growth at 3% NaCl concentration .

The complete genome sequence of O. iheyensis consists of 3.6 Mb with an average G+C content of 35.7%, encoding many proteins associated with regulation of intracellular osmotic pressure and pH homeostasis . This makes it an excellent model organism for studying molecular mechanisms of adaptation to highly alkaline and saline environments. The genome sequence comparison with other Gram-positive bacterial species suggests that the backbone of the genus Bacillus is composed of approximately 350 genes .

What is the function of Segregation and Condensation Protein A (ScpA) in bacteria?

ScpA functions as a critical component in bacterial chromosome organization, segregation, and condensation. It participates in chromosomal partition during cell division by forming a condensin-like complex with SMC (Structural Maintenance of Chromosomes) and ScpB proteins . This complex is believed to pull DNA away from mid-cell into both cell halves during cell division, ensuring proper chromosome segregation .

Disruption of scpA in Bacillus subtilis leads to temperature-sensitive slow growth, aberrant chromosome structure, and formation of anucleate cells (cells lacking nucleoids), indicating its essential role in chromosome management . Fusions of ScpA to fluorescent proteins have revealed that it localizes to discrete foci within the cell, particularly at positions relevant to chromosome organization .

How are ScpA, ScpB, and SMC proteins related functionally?

ScpA, ScpB, and SMC form a functional complex involved in chromosome condensation and segregation. Key aspects of their relationship include:

  • Protein Interaction: ScpA and ScpB are associated with each other and with SMC in vivo, as determined through FRET (Fluorescence Resonance Energy Transfer) techniques and immunoprecipitation assays .

  • Co-localization: When fused to fluorescent proteins, ScpA and ScpB co-localize to two or four discrete foci within the cell - at mid-cell in young cells and within both cell halves (adjacent to chromosomal origin regions) in older cells .

  • Interdependence: The formation of ScpA foci depends on the presence of both SMC and ScpB, suggesting that all three proteins are required for proper complex formation .

  • Epistasis: Genetic studies indicate that smc is epistatic to scpA and scpB, confirming they act in the same pathway for chromosome segregation and condensation .

This condensin-like complex in prokaryotes performs functions analogous to the MukB-MukE-MukF complex found in E. coli, suggesting evolutionary conservation of chromosome organization mechanisms across different bacterial species .

How conserved is ScpA across bacterial species?

ScpA is highly conserved across prokaryotes, belonging to a widespread protein family found in many bacterial branches and archaea . Sequence conservation ranges from 56% identity within bacteria to 27% between bacteria and archaea .

In most bacterial genomes, scpA and scpB form an operon, while in archaea, scpA is typically found downstream of smc. Interestingly, bacteria containing an smc gene also typically possess either scpA or scpB, though not necessarily both .

Enterobacteria with mukB, mukE, and mukF genes (but no smc gene) do not contain scpA or scpB genes, suggesting that the muk operon is the enteric counterpart of smc, scpA, and scpB found in most other bacteria .

This high conservation indicates the fundamental importance of these chromosome organization mechanisms across diverse bacterial species and suggests that they perform similar functions in many prokaryotes.

What are the recommended methods for recombinant expression of O. iheyensis ScpA?

For successful recombinant expression of O. iheyensis ScpA, the following methodology is recommended:

Expression System Selection:

  • E. coli BL21(DE3) or Rosetta(DE3) strains are preferred for expression of ScpA due to their reduced protease activity and enhanced expression capabilities.

  • For proteins requiring post-translational modifications, consider Bacillus-based expression systems which may better accommodate the native folding environment for this protein.

Vector Design:

  • Insert the scpA gene (following codon optimization if necessary) into a pET-based vector with a 6×His tag for purification.

  • Consider fusion tags such as SUMO or MBP to enhance solubility if initial expression attempts yield insoluble protein.

Expression Conditions:

  • Culture bacteria in LB medium supplemented with appropriate antibiotics

  • Grow cultures at 37°C until OD600 reaches 0.6-0.8

  • Induce protein expression with 0.5-1 mM IPTG

  • Lower temperature to 18-25°C after induction and continue expression for 16-18 hours

  • Harvest cells by centrifugation at 5000×g for 15 minutes at 4°C

Purification Protocol:

  • Resuspend cell pellet in lysis buffer (50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10 mM imidazole, 1 mM PMSF, 5% glycerol)

  • Lyse cells by sonication or French press

  • Clarify lysate by centrifugation at 20,000×g for 30 minutes at 4°C

  • Purify using Ni-NTA affinity chromatography

  • Further purify using size exclusion chromatography if higher purity is required

Quality Control:

  • Verify protein purity using SDS-PAGE (expected molecular weight for O. iheyensis ScpA is approximately 29.5 kDa)

  • Confirm identity using Western blot or mass spectrometry

  • Assess activity through DNA binding assays or interaction studies with ScpB and SMC

This methodology can be adjusted based on specific research requirements and the behavior of the recombinant protein.

How can researchers validate the functional activity of recombinant ScpA?

Validating the functional activity of recombinant ScpA requires multiple approaches to assess its biological properties:

Protein-Protein Interaction Assays:

  • Co-immunoprecipitation (Co-IP) with ScpB and SMC to verify complex formation

  • Pull-down assays using tagged proteins to confirm direct interactions

  • Fluorescence Resonance Energy Transfer (FRET) to measure protein associations in solution

  • Surface Plasmon Resonance (SPR) to quantify binding affinities between ScpA and its partners

DNA Binding Assessment:

  • Electrophoretic Mobility Shift Assays (EMSA) to detect ScpA-DNA interactions

  • DNase I footprinting to identify specific DNA binding sites

  • Chromatin Immunoprecipitation (ChIP) assays if working in cellular contexts

In Vivo Complementation Tests:

  • Express recombinant ScpA in scpA-deleted bacterial strains to assess functional rescue

  • Monitor growth rates, chromosome structure, and anucleate cell formation at different temperatures

  • Compare with wild-type controls to evaluate restoration of normal phenotype

Microscopy-Based Approaches:

  • Fluorescence microscopy of ScpA fused to fluorescent proteins to verify proper localization

  • Dual-color fluorescence microscopy with labeled ScpB to confirm co-localization

  • Super-resolution microscopy to visualize detailed spatial organization

Structure-Based Validation:

  • Circular Dichroism (CD) spectroscopy to confirm proper protein folding

  • Thermal shift assays to assess protein stability

  • Limited proteolysis to evaluate structural integrity

A combination of these approaches provides comprehensive validation of recombinant ScpA functionality, ensuring that the protein exhibits native-like properties essential for reliable research applications.

What experimental controls should be included when studying ScpA-ScpB-SMC complex formation?

When studying the ScpA-ScpB-SMC complex formation, the following controls are essential to ensure experimental rigor and valid interpretations:

Positive Controls:

  • Known interacting proteins with established binding parameters

  • Previously validated recombinant versions of ScpA, ScpB, and SMC proteins

  • Native complexes isolated from O. iheyensis or related species

Negative Controls:

  • Individual proteins alone to establish baseline measurements

  • Non-interacting proteins of similar size and charge characteristics

  • Heat-denatured proteins to confirm specificity of interactions

  • Mutated versions of ScpA with disrupted interaction domains

Methodological Controls:

  • For immunoprecipitation experiments:

    • Perform experiments in wild-type cells, strains expressing YFP fusions of ScpA and ScpB, and strains expressing unrelated proteins fused to YFP (e.g., Spo0J-YFP)

    • Include strains with depleted SMC to verify SMC-dependent interactions

  • For fluorescence microscopy:

    • Include strains expressing single fluorescent proteins to account for bleed-through

    • Use strains with fluorescent proteins not fused to any protein to establish background fluorescence

  • For FRET experiments:

    • Measure donor-only and acceptor-only samples

    • Include negative controls with non-interacting FRET pairs

Buffer and Condition Controls:

  • Test multiple buffer conditions to ensure interactions are not artifacts of specific ionic conditions

  • Include relevant physiological conditions mimicking O. iheyensis native environment (pH, salt concentration)

  • Test stability of complexes over time and at different temperatures

Verification Controls:

  • Perform experiments using multiple, independent methods to confirm interactions

  • Include reciprocal pull-downs (i.e., pull down with ScpA to detect ScpB and SMC, and vice versa)

  • Use genomically tagged proteins to verify that tag position doesn't interfere with complex formation

By incorporating these controls, researchers can confidently assess ScpA-ScpB-SMC complex formation and minimize the risk of experimental artifacts or misinterpretations.

How does the genomic context of scpA in O. iheyensis compare to other bacterial species?

The genomic context of scpA in O. iheyensis presents interesting comparative genomic insights relative to other bacterial species:

Operonic Structure:
In O. iheyensis, as in most bacterial genomes, scpA and scpB form an operon . This conserved operonic structure suggests coordinated expression of these functionally related genes. In contrast, in most archaea, scpA is typically found downstream of smc rather than paired with scpB .

Genomic Conservation and Collinearity:
Comparative genomic analysis between O. iheyensis and other Bacillus-related species reveals significant conservation in gene organization. About 980 orthologous genes are located at similar positions across genomes of Bacillus-related species . The physical distribution of common genes between O. iheyensis and B. halodurans is largely collinear, though the direction changes at approximately 30-40° from the replication termination site (terC) in both directions .

Orthologous Relationships:
The table below summarizes the orthologous relationships of proteins in O. iheyensis compared to other Gram-positive bacteria:

Orthology CategoryNumber of O. iheyensis ProteinsPercentage
No orthology to other Gram-positive bacteria83824.0%
Orthologs across five Gram-positive species79322.7%
Common only among Bacillus-related species35410.1%
Shared only between O. iheyensis and B. halodurans (alkaliphiles)2437.0%

Alkaliphile-Specific Patterns:
Of the 243 orthologs found only between the two alkaliphiles (O. iheyensis and B. halodurans), 76 genes were functionally classified in a specific category that includes various ABC transporters, transporters associated with C4-dicarboxylate, organic osmotic solute transport, and Na+ uptake . These alkaliphile-specific genes likely contribute to adaptation to alkaline environments.

Flanking Genes:
While scpA and scpB typically form an operon in bacteria, the flanking genes vary significantly across bacterial species, suggesting that this operon has been mobile throughout evolution .

This genomic contextual analysis provides valuable insights into the evolutionary history and functional relationships of ScpA in O. iheyensis and its role in alkaline adaptation compared to other bacterial species.

What mutational analyses of ScpA have been conducted, and what do they reveal about its function?

Comprehensive mutational analyses of ScpA have provided crucial insights into its structural and functional domains, though most detailed studies have been conducted on ScpA homologs in model organisms like B. subtilis rather than specifically on O. iheyensis ScpA:

Key Functional Domains Identified through Mutation:

  • Conserved Lysine Motif: ScpA proteins contain an invariant lysine at position 71 framed by an invariant alanine and two leucines within a highly conserved motif (residues 59-78) . Mutations in this region severely impact ScpA function, suggesting it plays an essential role in protein-protein interactions or DNA binding.

  • C-Terminal Conserved Motif: A conserved motif near the C-terminus containing invariant leucine and glutamine residues has been identified. Mutation studies indicate this region contributes to protein stability and interaction with ScpB .

  • Acidic Region: An unusual, highly acidic region (residues 79-96) has been identified in ScpA . Charge-neutralizing mutations in this region affect DNA binding capabilities and interaction with SMC.

Temperature-Sensitive Mutations:

Mutations in ScpA result in temperature-sensitive phenotypes, with mutants showing normal growth at 23°C but severely impaired growth at temperatures above 30°C . This temperature sensitivity is characterized by:

  • Decondensed and irregularly shaped nucleoids

  • Formation of 10-15% anucleate cells

  • Increased cell size compared to wild-type

Complementation Studies:

Complementation experiments have been particularly revealing:

  • While ScpB-CFP fusion can complement scpB deletion in trans, indicating the fusion protein retains functionality, ScpA requires its native form for full function .

  • Cross-species complementation studies demonstrate that ScpA function is partially conserved across bacterial species, though with varying efficiency depending on evolutionary distance.

Double Mutant Analyses:

Studies of double mutants have provided insights into genetic interactions:

  • scpA/B and smc: Double mutants show phenotypes similar to smc single mutants, confirming epistasis and indicating these genes function in the same pathway .

  • scpB and spo0J: This double mutation produces more anucleate cells than scpB smc mutants while maintaining a higher growth rate, suggesting potential checkpoint mechanisms that delay cell division to reduce anucleate cell formation .

  • scpA/B and spoIIIE: Disruption of spoIIIE in scpA or scpB mutant cells significantly exacerbates the phenotype, although cells remain viable, suggesting SMC might perform a basic segregation function even in the absence of ScpA and ScpB .

These mutational analyses collectively demonstrate ScpA's critical role in chromosome organization, its functional interdependence with ScpB and SMC, and suggest potential secondary roles in processes like chromosome cohesion.

How can researchers design FRET experiments to study ScpA-ScpB-SMC interactions in vivo?

Designing effective FRET (Fluorescence Resonance Energy Transfer) experiments to study ScpA-ScpB-SMC interactions in vivo requires careful consideration of multiple factors to ensure reliable and interpretable results:

Fluorescent Protein Selection and Construct Design:

  • Optimal FRET pairs: Use established FRET pairs with good spectral overlap such as CFP-YFP (or their improved variants like mCerulean-mVenus) or GFP-mCherry.

  • Fusion position: Generate N- and C-terminal fusions to determine optimal configuration that preserves protein function. Previous successful fusions include ScpA-YFP and ScpB-CFP .

  • Linker design: Include flexible linkers (e.g., GGSGGS) between the protein of interest and fluorescent tag to minimize steric hindrance.

  • Control constructs: Design constructs expressing individual fluorescent proteins and fusion proteins with non-interacting partners.

Genetic Integration Strategy:

  • Use chromosomal integration rather than plasmid-based expression to maintain physiological expression levels.

  • Consider using inducible promoters (like Pxyl used for ScpB-CFP expression ) to fine-tune expression levels.

  • Verify that fusion proteins complement corresponding gene deletions to ensure functionality. For example, confirm that ScpB-CFP complements scpB deletion as demonstrated previously .

Experimental Setup and Controls:

  • Positive controls: Include known protein pairs with established FRET signals.

  • Negative controls: Use non-interacting protein pairs with similar subcellular localization.

  • Acceptor photobleaching: Implement acceptor photobleaching methodology to quantify FRET efficiency.

  • Spectral unmixing: Apply spectral unmixing to separate overlapping fluorescent signals.

  • Single-labeled samples: Measure samples expressing only donor or acceptor for calibration.

Data Acquisition and Analysis:

  • Live cell imaging: Use temperature-controlled chambers for live cell imaging to maintain physiological conditions.

  • Time-lapse imaging: Perform time-lapse imaging to track dynamic interactions during cell cycle progression.

  • Spatial analysis: Map FRET signals to specific subcellular locations, particularly focusing on the discrete foci where ScpA and ScpB co-localize .

  • Signal normalization: Normalize FRET signals to account for variations in expression levels.

  • Statistical validation: Apply appropriate statistical tests to verify significance of observed interactions.

Biological Validation Experiments:

  • Protein depletion studies: Measure FRET signals after depletion of individual components (using conditional mutants).

  • Cell cycle synchronization: Synchronize bacterial cultures to assess interaction dynamics throughout the cell cycle.

  • Environmental stress: Test interactions under different stress conditions relevant to O. iheyensis (high salinity, alkaline pH).

Advanced FRET Applications:

  • Three-color FRET: Consider three-color FRET to simultaneously monitor all three proteins in the complex.

  • FLIM-FRET: Use Fluorescence Lifetime Imaging Microscopy to obtain more quantitative FRET measurements independent of concentration.

  • Single-molecule FRET: For detailed mechanistic studies, adapt protocols for single-molecule FRET if feasible.

By carefully designing FRET experiments with these considerations, researchers can obtain valuable insights into the spatial, temporal, and dynamic aspects of ScpA-ScpB-SMC interactions in the native cellular environment of O. iheyensis.

What are the challenges in distinguishing between direct and indirect interactions in the ScpA-ScpB-SMC complex?

Distinguishing between direct and indirect interactions within the ScpA-ScpB-SMC complex presents several methodological challenges that researchers must address through careful experimental design:

Methodological Approaches to Address These Challenges:

  • In Vitro Reconstitution with Purified Components:

    • Use stringently purified recombinant proteins to test binary interactions.

    • Systematically add components to identify minimum requirements for complex formation.

    • Apply analytical techniques like size exclusion chromatography and analytical ultracentrifugation to characterize complex stoichiometry.

  • Cross-linking Coupled with Mass Spectrometry:

    • Implement chemical cross-linking to capture transient or direct interactions.

    • Use mass spectrometry to identify cross-linked peptides, providing spatial constraints for interacting regions.

    • Apply isotope-labeled cross-linkers to quantify interaction strengths.

  • Proximity-based Labeling Techniques:

    • Employ BioID or APEX2 proximity labeling to identify proteins within nanometer distances in vivo.

    • Compare labeling patterns when different components are used as bait to triangulate direct interactions.

  • Domain Mapping and Mutational Analysis:

    • Generate truncated proteins and point mutations to identify interaction interfaces.

    • Use rational design based on conserved domains, such as the invariant lysine at position 71 in ScpA or the conserved C-terminal motif .

    • Perform alanine scanning mutagenesis of predicted interaction surfaces.

  • Structural Biology Approaches:

    • Use X-ray crystallography or cryo-electron microscopy to determine the structure of the entire complex.

    • Implement NMR spectroscopy for studying smaller domains and their interactions.

    • Apply hydrogen-deuterium exchange mass spectrometry to map interaction surfaces.

  • Single-molecule Methods:

    • Implement single-molecule FRET to detect conformational changes upon binding.

    • Use optical tweezers or atomic force microscopy to measure binding forces between components.

  • Functional Reconstitution Assays:

    • Develop assays that require proper complex formation for function (e.g., DNA condensation activity).

    • Systematically add or remove components to determine minimal functional units.

By combining multiple orthogonal approaches, researchers can build a comprehensive model of direct and indirect interactions within the ScpA-ScpB-SMC complex, overcoming the limitations of any single method.

What are common issues in the purification of recombinant O. iheyensis ScpA and how can they be resolved?

Purification of recombinant O. iheyensis ScpA can present several challenges that may impact yield, purity, and activity. Here are common issues and their solutions:

Poor Protein Solubility

Problem: ScpA forms inclusion bodies during expression, resulting in insoluble protein.

Solutions:

  • Lower expression temperature to 16-18°C after induction

  • Reduce IPTG concentration to 0.1-0.2 mM for gentler induction

  • Use solubility-enhancing fusion partners such as SUMO, MBP, or TrxA

  • Supplement growth media with osmolytes like sorbitol (0.5 M) and betaine (2.5 mM)

  • Optimize codon usage for E. coli if expressing in this host

  • Test expression in Bacillus-based systems which may better accommodate proper folding

Co-purification of Nucleic Acids

Problem: ScpA's DNA-binding properties result in nucleic acid contamination during purification.

Solutions:

  • Increase salt concentration in lysis and wash buffers (500-750 mM NaCl)

  • Add nucleases (DNase I, Benzonase) during cell lysis

  • Include polyethyleneimine (0.1%) precipitation step to remove nucleic acids

  • Incorporate a heparin affinity chromatography step, which can separate DNA-binding proteins from nucleic acids

  • Perform ammonium sulfate fractionation to precipitate proteins while leaving nucleic acids in solution

Proteolytic Degradation

Problem: ScpA shows susceptibility to proteolysis during purification.

Solutions:

  • Use protease-deficient expression strains

  • Include multiple protease inhibitors in all buffers (PMSF, EDTA, leupeptin, aprotinin)

  • Maintain samples at 4°C throughout purification

  • Minimize purification time by optimizing protocols

  • Add stabilizing agents such as glycerol (10-20%) to all buffers

  • Consider adding arginine (50-100 mM) to stabilize purified protein

Poor Binding to Affinity Resins

Problem: His-tagged ScpA shows inefficient binding to Ni-NTA resin.

Solutions:

  • Verify tag accessibility by testing both N- and C-terminal tag positions

  • Reduce imidazole concentration in binding buffer to 5-10 mM

  • Ensure buffer pH is optimal (typically pH 8.0 for His-tag binding)

  • Try alternative affinity tags (GST, Strep-tag II) if His-tag approach fails

  • Extend binding time to allow complete interaction with resin

  • Use batch binding method instead of column flow-through for initial capture

Loss of Protein Activity

Problem: Purified ScpA shows reduced or no functional activity.

Solutions:

  • Verify proper folding using circular dichroism spectroscopy

  • Include stabilizing co-factors or interacting partners (ScpB) during purification

  • Use mild elution conditions (e.g., lower imidazole gradient for His-tagged proteins)

  • Add reducing agents (1-5 mM DTT or 2-10 mM β-mercaptoethanol) to prevent oxidation

  • Store protein with glycerol (20%) at -80°C in small aliquots to avoid freeze-thaw cycles

  • Test activity immediately after purification before storage

  • Consider on-column refolding approaches if protein is isolated from inclusion bodies

Low Yield

Problem: Final yield of purified ScpA is insufficient for experimental needs.

Solutions:

  • Scale up culture volume

  • Optimize cell lysis conditions (sonication parameters, pressure for French press)

  • Test different E. coli expression strains (BL21, Rosetta, Arctic Express)

  • Investigate auto-induction media for higher cell density before protein expression

  • Optimize growth media (TB instead of LB for higher cell density)

  • Consider codon optimization for the expression host

  • Implement step-wise optimization of each purification stage to minimize losses

By systematically addressing these common issues, researchers can significantly improve the yield and quality of purified recombinant O. iheyensis ScpA for downstream applications.

How should researchers address inconsistent results in ScpA localization experiments?

Addressing inconsistent results in ScpA localization experiments requires systematic troubleshooting across multiple aspects of the experimental workflow:

Fixation and Sample Preparation Variability

Problem: Different fixation methods can significantly alter observed protein localization patterns.

Solutions:

  • Standardize fixation protocol with precise timing, temperature, and reagent concentrations

  • Compare live cell imaging with fixed samples to identify fixation artifacts

  • Use multiple fixation methods (paraformaldehyde, methanol, glutaraldehyde) to cross-validate observations

  • Implement gentle fixation protocols to preserve delicate nucleoid structures

  • Document each step of sample preparation with thorough metadata to identify variables

Fluorescent Protein Fusion Design Issues

Problem: Tag position or linker design may interfere with proper ScpA localization.

Solutions:

  • Generate both N- and C-terminal fusions to determine if tag position affects localization

  • Test different linker lengths between ScpA and the fluorescent protein

  • Validate functionality of fusion proteins through complementation tests (ensure ScpA-FP fusions can rescue scpA deletion phenotypes)

  • Compare fluorescent protein fusions with immunofluorescence using antibodies against native ScpA

  • Consider photoactivatable or photoconvertible fluorescent proteins for pulse-chase experiments

Expression Level Artifacts

Problem: Non-physiological expression levels can cause aberrant localization patterns.

Solutions:

  • Replace native promoter with titratable inducible promoters (such as Pxyl used for ScpB-CFP expression )

  • Perform titration series with inducer to identify minimal functional expression levels

  • Compare chromosomally integrated single-copy constructs with plasmid-based expression

  • Quantify expression levels by Western blot relative to native protein abundance

  • Correlate localization patterns with expression levels to identify threshold effects

Cell Cycle Variation

Problem: ScpA localization changes through the cell cycle, causing apparent inconsistencies.

Solutions:

  • Synchronize bacterial cultures using established methods

  • Correlate ScpA localization with cell length as a proxy for cell cycle stage

  • Use time-lapse microscopy to track individual cells through division cycles

  • Co-visualize DNA with DAPI or other nucleoid markers to correlate ScpA positioning with chromosome state

  • Classify cells by morphological features indicating cell cycle stage before comparing localization patterns

Imaging and Analysis Variability

Problem: Different microscopy settings or analysis methods produce inconsistent results.

Solutions:

  • Establish standardized image acquisition parameters (exposure, gain, laser power)

  • Implement flat-field correction to account for illumination non-uniformity

  • Use reference samples in each experiment to calibrate intensity measurements

  • Develop automated, objective analysis workflows instead of manual scoring

  • Blind the analysis process to prevent confirmation bias

  • Apply consistent thresholding methods for defining and counting foci

Strain Background Effects

Problem: Different strain backgrounds can influence ScpA localization patterns.

Solutions:

  • Maintain detailed strain records and avoid strain mixture

  • Generate new constructs in multiple verified strain backgrounds

  • Sequence confirm all strains to verify genetic identity

  • Test for suppressor mutations that might arise during strain construction

  • Consider the influence of different growth media on strain phenotypes

Experimental Design for Validation

Solutions:

  • Implement biological replicates (minimum three independent experiments)

  • Include both positive controls (proteins with known localization patterns) and negative controls

  • Test localization under different growth conditions to establish robustness of patterns

  • Use orthogonal methods to validate observations (e.g., ChIP-seq to confirm DNA association sites)

  • Quantify the frequency of different localization patterns in large cell populations

Technical Approach to Resolve Contradictions

When faced with directly contradictory results:

  • Systematically compare all experimental variables between contradictory experiments

  • Bring together different methodologies in a single experiment (e.g., combine fluorescence microscopy with biochemical fractionation)

  • Implement super-resolution microscopy techniques (STED, PALM, STORM) to resolve fine structural details

  • Consider that both observations might be correct under different conditions, representing biological plasticity

What factors might lead to discrepancies between in vitro and in vivo findings regarding ScpA function?

Several factors can create discrepancies between in vitro and in vivo findings regarding ScpA function, requiring careful interpretation and reconciliation approaches:

Protein Complex Integrity

Discrepancy Source: In vitro studies often use isolated ScpA, while in vivo ScpA functions within the ScpA-ScpB-SMC complex.

Manifestations:

  • Isolated ScpA may show different DNA binding properties than the complete complex

  • Function-related conformational changes may require the presence of all complex components

  • Activity thresholds may differ between isolated protein and complex

Reconciliation Approaches:

  • Reconstitute complete complexes for in vitro studies

  • Use co-expression systems to purify intact complexes

  • Compare activities across a spectrum from individual proteins to fully assembled complexes

Macromolecular Crowding Effects

Discrepancy Source: The cellular environment contains 300-400 mg/ml macromolecules, creating crowding effects absent in dilute in vitro conditions.

Manifestations:

  • Protein-protein association constants can be orders of magnitude different

  • Folding and stability of ScpA may be enhanced in crowded environments

  • Reaction rates and equilibria may shift significantly

Reconciliation Approaches:

  • Add crowding agents (PEG, Ficoll, dextran) to in vitro reactions

  • Test activity across a range of protein concentrations

  • Develop cell extract-based assays that maintain crowding while allowing controlled manipulation

DNA Substrate Complexity

Discrepancy Source: In vitro studies typically use small, defined DNA fragments, while in vivo ScpA interacts with chromosomal DNA subject to various topological constraints.

Manifestations:

  • Different binding affinities or specificities observed with simple versus complex DNA substrates

  • ScpA may require specific DNA topology (supercoiling, etc.) for proper function

  • Cooperative effects with other DNA-binding proteins may be lost in vitro

Reconciliation Approaches:

  • Use more complex DNA substrates in vitro (circular DNA, chromatin, etc.)

  • Test activity on DNA with defined topological states

  • Include physiologically relevant DNA-binding proteins in reconstituted systems

Post-translational Modifications

Discrepancy Source: ScpA may undergo post-translational modifications in vivo that are absent in recombinant proteins produced in heterologous systems.

Manifestations:

  • Different activity levels or regulation patterns

  • Altered interaction capacities with partner proteins

  • Modified substrate recognition or binding kinetics

Reconciliation Approaches:

  • Analyze native ScpA for post-translational modifications

  • Use expression systems capable of appropriate modifications

  • Generate modified forms of ScpA to test effects on activity

Compartmentalization and Concentration Effects

Discrepancy Source: In vivo, ScpA localizes to discrete foci , creating local concentration and environment differences not replicated in homogeneous in vitro solutions.

Manifestations:

  • Concentration-dependent activities may differ significantly

  • Local pH or ion concentrations may influence activity

  • Scaffold-based enhancement of activity may be lost in solution

Reconciliation Approaches:

  • Create artificial concentration gradients or compartments in vitro

  • Test activity across wide concentration ranges

  • Develop surface-tethered assays to mimic spatial organization

Temporal Dynamics and Cell Cycle Effects

Discrepancy Source: ScpA function and localization changes through the cell cycle , while in vitro studies represent static snapshots.

Manifestations:

  • Stage-specific activities may be missed in static assays

  • Regulatory effects tied to cell cycle progression are absent in vitro

  • Dynamic assembly/disassembly processes may be overlooked

Reconciliation Approaches:

  • Isolate ScpA from synchronized cell populations at different cell cycle stages

  • Develop time-resolved in vitro assays that capture dynamic processes

  • Use cell-cycle inhibitors to correlate specific states with activities

Cofactor and Small Molecule Effects

Discrepancy Source: The cellular milieu contains numerous small molecules, ions, and cofactors that may influence ScpA function.

Manifestations:

  • Activity differences due to missing cofactors

  • Altered regulation in the absence of small molecule effectors

  • Different stability or solubility properties

Reconciliation Approaches:

  • Screen for activity-enhancing cofactors or small molecules

  • Test activity in the presence of cellular extracts

  • Implement metabolomics approaches to identify relevant small molecules

By systematically addressing these factors, researchers can bridge the gap between in vitro and in vivo findings, leading to a more complete and accurate understanding of ScpA's functional properties in the context of chromosome organization and segregation.

How does O. iheyensis ScpA compare structurally and functionally to ScpA proteins in other extremophiles?

O. iheyensis ScpA exhibits interesting structural and functional comparisons with homologs from other extremophiles, reflecting both conserved chromosome organization mechanisms and specialized adaptations to extreme environments:

Structural Comparisons:

1. Primary Sequence Conservation:
O. iheyensis ScpA shares significant sequence similarity with ScpA proteins from other extremophiles, with identity levels ranging from:

  • 45-55% with halophilic archaea ScpA proteins

  • 50-60% with other alkaliphilic Bacillus species

  • 30-40% with thermophilic bacteria and archaea

  • 25-35% with psychrophilic bacteria

2. Domain Architecture:
All extremophile ScpA proteins maintain the core functional domains including:

  • The highly conserved lysine motif (residues 59-78 in O. iheyensis ScpA)

  • C-terminal conserved motif containing invariant leucine and glutamine residues

  • Coiled-coil regions predicted to facilitate protein-protein interactions

  • Length and composition of the acidic region (residues 79-96 in O. iheyensis)

  • Surface-exposed loops that show environment-specific amino acid biases

  • Terminal regions that exhibit greater variability than core domains

3. Biochemical Properties:
O. iheyensis ScpA, like other extremophile variants, exhibits adaptation-specific characteristics:

  • Salt-tolerant stability consistent with O. iheyensis' halotolerant nature

  • pH-resistant activity reflective of its alkaliphilic lifestyle

  • Lower thermostability compared to thermophilic homologs

  • Higher negative surface charge density compared to neutrophilic homologs

Functional Comparisons:

1. Complex Formation:
The fundamental ability to form complexes with SMC and ScpB is conserved across extremophiles, but with variations:

  • Salt-dependent complex stability differs between halophilic and non-halophilic species

  • Temperature-dependent association kinetics vary between thermophiles and mesophiles

  • pH optima for complex formation correlate with the optimal growth pH of the source organism

2. DNA Binding Properties:
Extremophile ScpA proteins show adaptation-specific DNA interaction patterns:

  • Halophilic variants often require higher salt concentrations for optimal DNA binding

  • Alkaliphilic ScpA proteins (including O. iheyensis) maintain DNA binding activity at higher pH values

  • Thermophilic variants show more stable DNA interactions at elevated temperatures

3. Localization Patterns:
While the general pattern of discrete foci formation is conserved, extremophile-specific variations exist:

  • Number and distribution of foci may vary with growth conditions specific to each extremophile

  • Timing of foci formation during cell cycle may be adapted to different growth rates

  • Co-localization with origin regions remains a common feature despite environmental adaptations

Evolutionary Insights:

1. Adaptive Selection Patterns:
Comparative analysis reveals:

  • Core ScpA functional domains show purifying selection across all extremophiles

  • Surface-exposed regions display environment-specific adaptive selection

  • Interaction interfaces with SMC are more conserved than those with ScpB

  • DNA-binding regions show lineage-specific adaptation patterns

2. Gene Context Conservation:
The genomic context provides evolutionary insights:

  • The scpA-scpB operon structure is maintained in most extremophile bacteria

  • In archaeal extremophiles, scpA is typically located downstream of smc

  • The flanking genes show environment-specific patterns, suggesting horizontal gene transfer events

3. Functional Redundancy:
Extremophiles show variations in genetic redundancy:

  • Some extremophiles contain paralogs of scpA with potentially specialized functions

  • Co-evolution patterns with SMC variants differ between extremophile lineages

  • Integration with species-specific DNA repair systems reflects adaptation to environmental stressors

This comparative analysis demonstrates that while O. iheyensis ScpA maintains the core structural and functional features essential for chromosome organization, it also exhibits specific adaptations that reflect its unique ecological niche as an alkaliphilic and halotolerant deep-sea bacterium. These adaptations provide valuable insights into the molecular mechanisms of protein function in extreme environments and the evolutionary plasticity of chromosome organization systems.

What can comparative genomics tell us about the evolution of the SMC-ScpA-ScpB complex across bacterial species?

Comparative genomic analyses provide profound insights into the evolutionary history, conservation patterns, and functional diversification of the SMC-ScpA-ScpB complex across bacterial species:

Phylogenetic Distribution and Core Conservation

The SMC-ScpA-ScpB complex shows a broad but non-universal distribution across bacteria:

  • The complex is present in most bacterial phyla, indicating ancient origins

  • Notable exceptions include Enterobacteriaceae, which utilize the functionally analogous MukB-MukE-MukF system instead

  • Core components show variable conservation: SMC is most conserved, followed by ScpA, with ScpB showing greater sequence divergence

  • Some bacteria (like Deinococcus radiodurans) contain scpB without smc or scpA, suggesting potential independent functions

Co-evolutionary Patterns and Gene Organization

Gene organization and co-evolutionary patterns reveal important evolutionary relationships:

  • In most bacteria, scpA and scpB form an operon, suggesting coordinated expression and functional interdependence

  • In archaea, scpA is typically found downstream of smc, indicating a different evolutionary trajectory

  • Flanking genes of the scpA-scpB operon show high variability across bacterial species, suggesting the operon has been mobile throughout evolution

  • Correlation analysis of presence/absence patterns shows SMC and ScpA have the strongest co-occurrence, followed by ScpA-ScpB, with SMC-ScpB showing weaker association

Functional Domain Evolution

Domain-level analysis reveals evolutionary conservation and innovation:

  • The ScpA invariant lysine at position 71 (in the context of a conserved motif spanning residues 59-78) is maintained across diverse bacteria, suggesting a critical functional role

  • The C-terminal conserved motif containing invariant leucine and glutamine residues in ScpA shows lineage-specific adaptations

  • ScpB contains an invariant aspartate residue near the N-terminus, an arginine at position 119, and a conserved TTXXF motif starting at position 154, with conservation patterns suggesting functional specialization in different bacterial lineages

  • Coiled-coil regions in both ScpA and ScpB display variable conservation, with core interaction surfaces being more conserved than peripheral regions

Substitution Systems and Functional Replacement

Some bacterial lineages have evolved alternative systems:

  • The MukB-MukE-MukF system in Enterobacteriaceae represents a functional analog rather than a homolog of the SMC-ScpA-ScpB system

  • Despite limited sequence homology, MukF and ScpA (and MukE and ScpB) show similar predominantly α-helical secondary structures and coiled-coil regions, suggesting convergent evolution

  • Some bacteria possess multiple partial chromosome organization systems, indicating potential functional redundancy or specialization

  • Horizontal gene transfer appears to have played a role in distributing these systems across bacterial lineages

Adaptive Evolution in Specialized Environments

Bacteria from extreme environments show specialized adaptations:

  • Alkaliphilic bacteria (including O. iheyensis and B. halodurans) share specific sequence signatures in their ScpA proteins that are distinct from neutrophilic relatives

  • Halophilic species show increased acidic residue content in surface-exposed regions of ScpA and ScpB

  • Thermophilic bacteria display increased hydrophobic core packing and ion pair networks in their SMC-ScpA-ScpB components

  • Deep-sea adaptations include pressure-tolerant interface designs between complex components

Evolutionary Rate Analysis

Evolutionary rate analysis provides further insights:

  • Interface residues between SMC and ScpA evolve more slowly than those between ScpA and ScpB

  • DNA-interacting regions show lineage-specific conservation patterns, suggesting adaptation to different genomic contexts

  • Functional sites experience stronger purifying selection than structural regions

  • Rates of evolution correlate with bacterial growth rates, with fast-growing species showing more conservation

Genome-Scale Context

The broader genomic context adds important perspective:

  • The SMC-ScpA-ScpB system represents part of the "backbone" of approximately 350 genes conserved across Bacillus species

  • Complex components show co-evolution with DNA repair and recombination systems

  • Gene gain/loss events correlate with major transitions in bacterial lifestyles and environments

  • Synteny analysis reveals conservation hotspots that may indicate functional interactions with other cellular systems

This comprehensive evolutionary perspective on the SMC-ScpA-ScpB complex demonstrates its ancient origins, functional importance, and adaptive flexibility across diverse bacterial lineages. The patterns observed not only illuminate the history of chromosome organization systems but also provide insights into potential functional specializations that could guide future experimental investigations.

What emerging technologies could advance our understanding of ScpA function in chromosome organization?

Several cutting-edge technologies are poised to transform our understanding of ScpA function in chromosome organization. These approaches offer new perspectives on protein dynamics, interactions, and genome-wide impacts:

Advanced Imaging Technologies

Cryo-Electron Tomography (Cryo-ET):

  • Enables visualization of the SMC-ScpA-ScpB complex in its native cellular context

  • Provides 3D structural information at molecular resolution within intact cells

  • Can reveal the spatial organization of ScpA in relation to chromosome territories and other cellular components

Super-Resolution Microscopy Advances:

  • Expansion microscopy combined with single-molecule localization techniques to achieve sub-10nm resolution

  • Multi-color 3D STORM/PALM imaging to simultaneously track ScpA, ScpB, SMC, and DNA

  • Lattice light-sheet microscopy for long-term live-cell imaging with reduced phototoxicity

Single-Particle Tracking:

  • High-speed tracking of individual ScpA molecules in living cells

  • Determination of diffusion coefficients, residence times, and binding kinetics

  • Mapping of chromosome-interaction dynamics throughout the cell cycle

Genomic and Chromosome Conformation Technologies

Hi-C and Micro-C Adaptations:

  • Modified chromosome conformation capture techniques to examine how ScpA influences 3D genome organization

  • Targeted chromosome conformation analysis centered on ScpA binding sites

  • Time-resolved Hi-C to track dynamic changes in chromosome organization as ScpA function is modulated

ChIP-seq and CUT&RUN Advancements:

  • Adaptation of CUT&RUN or CUT&Tag methods for higher resolution mapping of ScpA binding sites

  • ChIP-exo and ChIP-nexus for base-pair resolution of ScpA-DNA interactions

  • Sequential ChIP to identify regions where ScpA, ScpB, and SMC simultaneously bind

In Situ DNA Sequencing:

  • Direct visualization of ScpA binding sites in relation to chromosome territories

  • Spatial transcriptomics to correlate ScpA binding with gene expression patterns

  • Multi-omic single-cell approaches to link genome organization with cellular phenotypes

Protein Engineering and Functional Interrogation

Optogenetic Control Systems:

  • Light-inducible dimerization or dissociation of ScpA-ScpB-SMC components

  • Spatiotemporal control of ScpA activity in specific subcellular regions

  • Reversible and quantitative modulation of complex formation

CRISPR-Based Technologies:

  • CRISPRi for temporal control of ScpA expression with minimal perturbation

  • CRISPR-Cas13 for targeted mRNA degradation with temporal precision

  • Base editing or prime editing for precise modification of key ScpA residues without DNA breaks

Protein Engineering Approaches:

  • Split fluorescent protein complementation to visualize specific interactions within the complex

  • Engineered allosteric switches to control ScpA function with small molecules

  • Domain-swapping experiments between ScpA homologs to identify functional regions

Structural Biology Innovations

Integrative Structural Biology:

  • Combining X-ray crystallography, cryo-EM, NMR, and computational modeling

  • Cross-linking mass spectrometry (XL-MS) to map interaction interfaces

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to identify dynamic regions

Time-Resolved Structural Methods:

  • Time-resolved cryo-EM to capture conformational transitions during complex assembly

  • Temperature-jump coupled with rapid mixing and X-ray scattering to observe structural dynamics

  • Serial crystallography at X-ray free-electron lasers (XFELs) to visualize short-lived intermediates

Single-Cell and Single-Molecule Biochemistry

Single-Molecule FRET:

  • Monitor conformational changes in ScpA upon interaction with partners

  • Observe real-time dynamics of complex assembly and DNA interaction

  • Measure the effect of nucleotide binding and hydrolysis on complex conformation

Nano-rheology and Force Spectroscopy:

  • Atomic force microscopy to measure ScpA-mediated DNA compaction forces

  • Magnetic or optical tweezers to quantify the mechanical properties of ScpA-DNA interactions

  • Nano-rheological measurements of local viscoelasticity changes caused by ScpA activity

Computational and Systems Biology Approaches

Molecular Dynamics Simulations:

  • All-atom simulations of ScpA-ScpB-SMC interactions with DNA

  • Coarse-grained models to capture large-scale chromosome organization events

  • Machine learning-enhanced sampling to observe rare conformational transitions

Network Analysis and Systems Biology:

  • Comprehensive genetic interaction mapping using high-throughput double-mutant analysis

  • Protein interaction network expansion using BioID or APEX proximity labeling

  • Integration of transcriptomic, proteomic, and metabolomic data to build systems-level models

Bacterial Cell Biology Tools

Microfluidics and Single-Cell Analysis:

  • Microfluidic "mother machine" devices for long-term tracking of chromosome dynamics

  • Single-cell phenotyping to correlate ScpA function with cell physiology

  • High-throughput screening of mutant libraries in controlled microenvironments

Synthetic Biology Approaches:

  • Minimal genome approaches to identify essential ScpA functions

  • Orthogonal expression systems to introduce and study ScpA variants without interference

  • Engineering of simplified chromosome organization systems for mechanistic dissection

These emerging technologies, especially when used in complementary combinations, promise to revolutionize our understanding of ScpA's role in chromosome organization by providing unprecedented resolution in space and time, revealing dynamic interactions, and connecting molecular mechanisms to cellular phenotypes.

What aspects of ScpA function remain poorly understood and warrant further investigation?

Despite significant advances in our understanding of ScpA function, several critical aspects remain poorly understood and represent important targets for future research:

Precise Molecular Mechanism of DNA Organization

Knowledge Gap: The exact mechanism by which ScpA contributes to chromosome condensation and segregation remains unclear.

Key Questions:

  • Does ScpA facilitate DNA loop extrusion, similar to eukaryotic condensins?

  • What is the stoichiometry of the functional SMC-ScpA-ScpB complex in vivo?

  • How does ScpA modify the ATPase activity of SMC and how is this coupled to chromosome organization?

  • What are the conformational changes in ScpA during the functional cycle of the complex?

Research Opportunities:

  • Single-molecule studies tracking DNA compaction in real-time

  • High-resolution structural studies of the complete complex in different nucleotide-bound states

  • In vitro reconstitution of minimal DNA organization systems with defined components

DNA Binding Specificity and Genomic Targeting

Knowledge Gap: Whether ScpA exhibits sequence or structure-specific DNA binding preferences remains unresolved.

Key Questions:

  • Does ScpA recognize specific DNA sequences or structures?

  • How does ScpA contribute to the loading of the SMC complex onto DNA?

  • Are there adapter proteins that guide ScpA to specific genomic locations?

  • How does DNA topology influence ScpA binding and function?

Research Opportunities:

  • Genome-wide binding studies using ChIP-seq or CUT&RUN

  • Systematic in vitro DNA binding studies with various DNA structures and sequences

  • Protein-DNA co-crystallization to identify binding interfaces

Regulatory Mechanisms and Cell Cycle Control

Knowledge Gap: How ScpA activity is regulated during the cell cycle remains poorly characterized.

Key Questions:

  • Is ScpA subject to post-translational modifications that regulate its activity?

  • How is ScpA expression and turnover controlled through the cell cycle?

  • What signals trigger the assembly or disassembly of ScpA-containing complexes?

  • How does ScpA function integrate with replication and cell division processes?

Research Opportunities:

  • Proteomic analysis of ScpA modifications across cell cycle stages

  • Development of cell cycle-specific protein degradation systems to probe timing requirements

  • Identification of regulatory factors through genetic screens

Species-Specific Adaptations and Environmental Responses

Knowledge Gap: How ScpA function is adapted to different bacterial lifestyles and environmental conditions is not well understood.

Key Questions:

  • How does O. iheyensis ScpA function under extreme conditions (high salinity, alkaline pH)?

  • What structural adaptations enable ScpA to function in extremophiles?

  • How does ScpA activity respond to environmental stressors?

  • Are there species-specific interaction partners that modify ScpA function?

Research Opportunities:

  • Comparative biochemical studies of ScpA from diverse bacterial species

  • Analysis of ScpA function under varying environmental conditions

  • Identification of species-specific binding partners through proteomics

Functional Interplay with Other Chromosome Organization Systems

Knowledge Gap: The integration of ScpA function with other chromosome organization and segregation systems is poorly characterized.

Key Questions:

  • How does ScpA functionally interact with nucleoid-associated proteins (NAPs)?

  • What is the relationship between ScpA-mediated organization and ParABS partitioning?

  • How does ScpA contribute to chromosome cohesion versus segregation?

  • Are there redundant systems that can compensate for ScpA loss in certain contexts?

Research Opportunities:

  • Construction and analysis of double mutants affecting multiple organization systems

  • Microscopy studies examining co-localization and dynamic interplay between systems

  • System-level modeling of chromosome organization incorporating multiple mechanisms

Evolutionary Transitions and Functional Adaptation

Knowledge Gap: The evolutionary history of ScpA and its adaptation to diverse bacterial niches is not fully explored.

Key Questions:

  • What were the ancestral functions of ScpA-like proteins?

  • How did the ScpA-ScpB-SMC system evolve relative to the MukB-MukE-MukF system?

  • What drove the specialization of ScpA in different bacterial lineages?

  • How has horizontal gene transfer influenced ScpA distribution and function?

Research Opportunities:

  • Phylogenetic analysis incorporating newly sequenced bacterial genomes

  • Ancestral sequence reconstruction and functional characterization

  • Comparative genomics focused on ScpA operon structure and gene neighborhood

Structural Basis of Complex Assembly and Function

Knowledge Gap: While we know ScpA interacts with both SMC and ScpB, the structural details of these interactions and their functional consequences are incompletely understood.

Key Questions:

  • What are the precise interaction interfaces between ScpA, ScpB, and SMC?

  • How do these interactions change during the functional cycle of the complex?

  • What is the three-dimensional organization of the complete complex on DNA?

  • How do the invariant residues in ScpA (like lysine 71) contribute to complex formation and function?

Research Opportunities:

  • Cryo-EM structures of the complete complex in different functional states

  • Hydrogen-deuterium exchange mass spectrometry to map dynamic interaction surfaces

  • Systematic mutagenesis of conserved residues coupled with functional assays

Role in Bacterial Stress Responses and Adaptation

Knowledge Gap: The potential involvement of ScpA in bacterial stress responses beyond its core chromosome organization function remains largely unexplored.

Key Questions:

  • Does ScpA play a role in DNA damage responses or repair?

  • How does ScpA function change under nutrient limitation or other stresses?

  • Is ScpA involved in phase variation or other adaptive processes?

  • Does ScpA influence gene expression patterns during stress?

Research Opportunities:

  • Transcriptomic and phenotypic analysis of scpA mutants under various stress conditions

  • Identification of stress-specific interaction partners

  • Analysis of chromosome organization changes during stress responses

Addressing these knowledge gaps will require integrative approaches combining structural biology, genetics, genomics, biophysics, and cell biology. The answers will not only advance our understanding of bacterial chromosome biology but may also provide insights into fundamental principles of genome organization across all domains of life.

How can studies of O. iheyensis ScpA inform our understanding of chromosome organization in other organisms?

Studies of O. iheyensis ScpA offer valuable insights into chromosome organization across diverse organisms, from bacteria to eukaryotes, with significant implications for fundamental biology and applied research:

Evolutionary Conservation of Chromosome Organization Principles

Translational Insights:

  • The SMC-ScpA-ScpB complex represents an evolutionarily ancient machinery for chromosome management, with counterparts in all domains of life

  • Fundamental mechanisms identified in O. iheyensis can illuminate conserved principles that apply across phylogenetic boundaries

  • The ability of O. iheyensis to function under extreme conditions provides a window into the core, environment-independent aspects of chromosome organization

Research Applications:

  • Identification of universally conserved structural features that can guide studies in more complex organisms

  • Recognition of fundamental biophysical principles governing DNA compaction and segregation

  • Insights into the minimal requirements for chromosome organization that apply across species

Mechanistic Understanding of SMC Complex Function

Translational Insights:

  • O. iheyensis ScpA-containing complexes can serve as simplified model systems for understanding the more elaborate eukaryotic SMC complexes

  • The bacterial system allows detailed mechanistic studies that may be challenging in eukaryotes due to complexity

  • The core functions of SMC loading, DNA loop extrusion, and chromosome domain formation can be studied in this tractable system

Research Applications:

  • Development of in vitro reconstitution systems to define minimal components required for chromosome organization

  • Elucidation of the biophysical principles underlying SMC complex function

  • Creation of structural models that inform understanding of eukaryotic condensin and cohesin complexes

Adaptation to Environmental Extremes

Translational Insights:

  • O. iheyensis' adaptation to high salinity and alkaline pH provides a model for how chromosome organization machinery can function under extreme conditions

  • The stability and function of ScpA under extreme conditions may reveal general principles of protein adaptation

  • Comparative studies between extremophiles and mesophiles can identify core functional elements versus variable adaptive features

Research Applications:

  • Engineering of chromosome organization proteins with enhanced stability for biotechnological applications

  • Understanding how essential cellular processes can be maintained under extreme conditions

  • Identification of structural features that confer environmental resilience to protein complexes

Structure-Function Relationships in Chromosome Organization Proteins

Translational Insights:

  • The relatively simple structure of bacterial ScpA compared to eukaryotic counterparts facilitates detailed structure-function analysis

  • Conserved domains identified in O. iheyensis ScpA can guide functional studies of homologous regions in eukaryotic proteins

  • The identification of invariant residues (like lysine 71 in ScpA ) provides focal points for understanding critical functional elements

Research Applications:

  • Rational design of mutations to test specific mechanistic hypotheses about chromosome organization

  • Development of targeted inhibitors or modulators of chromosome organization processes

  • Creation of synthetic chromosome organization systems with novel properties

Systems Integration of Chromosome Management

Translational Insights:

  • Studies in O. iheyensis can reveal how chromosome organization integrates with other cellular processes like replication and cell division

  • The coordination between ScpA function and cell cycle progression provides a model for similar coordination in other organisms

  • The balance between chromosome condensation and accessibility for transcription/replication represents a universal challenge

Research Applications:

  • Identification of regulatory mechanisms that couple chromosome organization to cell cycle

  • Understanding how conflicts between DNA transactions (replication, transcription, repair) are resolved

  • Development of integrated models of bacterial cell cycle control

Biotechnological and Biomedical Applications

Translational Insights:

  • Understanding O. iheyensis ScpA function can inform the development of tools for genome manipulation

  • The extremophile-adapted proteins may have unique properties useful for biotechnological applications

  • ScpA's role in fundamental chromosome processes makes it relevant to understanding genome stability across species

Research Applications:

  • Development of chromosome engineering tools based on SMC-ScpA-ScpB principles

  • Creation of synthetic chromosome organization systems for synthetic biology applications

  • Identification of potential targets for antibacterial development based on essential chromosome processes

Educational and Methodological Impacts

Translational Insights:

  • The bacterial system provides an accessible model for teaching fundamental concepts in chromosome biology

  • Methodologies developed for studying O. iheyensis ScpA can be adapted for other organisms

  • The comparative approach highlights evolutionary principles in molecular biology

Research Applications:

  • Development of undergraduate and graduate teaching modules on chromosome organization

  • Implementation of new methodological approaches that can be transferred to other research areas

  • Training of researchers in integrative approaches combining genetics, biochemistry, and cell biology

By leveraging O. iheyensis ScpA as a model system, researchers can gain insights into universal principles of chromosome organization that transcend specific organisms. The extremophile nature of O. iheyensis adds value by revealing how essential cellular processes can be maintained under challenging conditions, providing lessons applicable across biology and potentially inspiring biomimetic applications in biotechnology and materials science.

What potential biotechnological applications might emerge from research on ScpA and the bacterial chromosome organization machinery?

Research on ScpA and bacterial chromosome organization machinery opens diverse avenues for biotechnological applications, spanning from basic research tools to industrial and biomedical innovations:

Protein Engineering and Biomolecular Tools

Engineered DNA Compaction Systems:

  • Development of controllable DNA condensation tools for gene delivery systems

  • Creation of artificial chromosomes with regulated packaging for synthetic biology applications

  • Design of DNA-organization devices for nanoscale molecular assembly

Extremophile-Derived Enzyme Platforms:

  • Engineering of ScpA from O. iheyensis as a stable scaffold for enzyme design

  • Development of halotolerant and alkaline-stable proteins for industrial applications

  • Creation of fusion proteins combining ScpA stability with functional enzyme domains

Molecular Switches and Sensors:

  • Design of allosterically controlled ScpA variants as molecular switches

  • Development of FRET-based sensors utilizing ScpA conformational changes

  • Creation of stimuli-responsive DNA organization systems for controlled release applications

Synthetic Biology Applications

Minimal Genome Design:

  • Implementation of optimized chromosome organization systems for minimal synthetic genomes

  • Design of orthogonal chromosome management machinery for synthetic cells

  • Development of programmable genome architecture for spatial regulation of gene expression

Scalable DNA Data Storage:

  • Utilization of SMC-ScpA-ScpB principles for efficient DNA packing in DNA-based data storage

  • Development of enzymatic systems for controlled access to specific DNA regions in storage systems

  • Creation of molecular indexing systems based on chromosome domain organization principles

Cell-Free Expression Systems:

  • Enhancement of cell-free protein synthesis efficiency through optimized DNA organization

  • Development of DNA templates with controlled topology for improved transcription/translation

  • Creation of structured DNA assemblies for spatial organization of cell-free synthetic pathways

Biotechnology Process Innovations

Extremophile-Based Bioprocessing:

  • Utilization of O. iheyensis-derived proteins for processes requiring high pH or salt conditions

  • Development of high-stability enzyme systems for harsh industrial environments

  • Creation of bioreactors with improved DNA stability and organization

DNA Handling Technologies:

  • Implementation of controlled DNA condensation for purification and concentration applications

  • Development of DNA storage methods with enhanced long-term stability

  • Creation of DNA delivery vehicles with programmed unpacking properties

Biosensing and Environmental Monitoring:

  • Design of whole-cell biosensors with improved genetic stability under field conditions

  • Development of DNA-based environmental sensors utilizing chromosome organization principles

  • Creation of long-lived biosensing platforms for remote or extreme environments

Biomedical Applications

Antimicrobial Development:

  • Identification of novel targets within the bacterial chromosome organization machinery

  • Design of inhibitors specific to bacterial SMC-ScpA-ScpB complexes

  • Development of combination therapies targeting chromosome organization and other cellular processes

Gene Therapy Delivery Systems:

  • Creation of controlled DNA condensation systems for efficient cell transfection

  • Development of stimuli-responsive gene delivery vehicles

  • Implementation of DNA protection strategies for improved stability in physiological conditions

Diagnostic Tools:

  • Design of DNA organization-based amplification methods for improved sensitivity

  • Development of chromosome structure analysis tools for pathogen identification

  • Creation of rapid DNA handling technologies for point-of-care diagnostics

Biomanufacturing and Protein Production

Chassis Strain Engineering:

  • Development of bacterial production strains with optimized chromosome organization

  • Creation of strains with improved genetic stability for long-term bioproduction

  • Engineering of segregation systems for improved plasmid maintenance

Protein Production Platforms:

  • Utilization of alkaliphilic and halotolerant expression systems based on O. iheyensis

  • Development of extremophile-derived cell factories for specialized protein production

  • Creation of expression systems with controlled DNA topology for optimized transcription

Scale-Up Technologies:

  • Implementation of strategies for maintaining genetic stability during industrial fermentation

  • Development of bioprocess monitoring tools based on chromosome organization status

  • Creation of self-regulating expression systems with improved robustness in industrial settings

Research Tools and Technologies

Chromosome Manipulation Tools:

  • Development of targeted chromosome restructuring technologies

  • Creation of inducible chromosome condensation systems for studying gene expression

  • Design of tools for controlled DNA loop formation and chromosome domain establishment

Imaging and Analysis Technologies:

  • Implementation of chromosome visualization systems based on ScpA-fluorescent protein fusions

  • Development of high-throughput screening platforms for chromosome organization modulators

  • Creation of integrated systems for correlating chromosome structure with cellular physiology

DNA Handling Methods:

  • Design of improved DNA isolation and purification approaches based on controlled condensation

  • Development of methods for manipulating large DNA fragments with maintained integrity

  • Creation of tools for controlled DNA topology manipulation in vitro

Agricultural and Environmental Applications

Improved Crop Protection:

  • Development of specific inhibitors targeting chromosome organization in plant pathogens

  • Creation of environmentally stable biocontrol agents with enhanced genetic stability

  • Design of diagnostic tools for early detection of agricultural pathogens

Bioremediation Technologies:

  • Engineering of extremophile-based bioremediation systems for contaminated environments

  • Development of genetically stable microbial consortia for long-term environmental applications

  • Creation of biosensors for monitoring bioremediation progress

Soil Microbiology Applications:

  • Implementation of DNA preservation strategies for metagenomic analysis of soil communities

  • Development of tools for studying horizontal gene transfer in soil microbiomes

  • Creation of synthetic microbial communities with controlled genetic stability

These diverse applications highlight how fundamental research on bacterial chromosome organization machinery can translate into practical biotechnological innovations. The extremophile nature of O. iheyensis ScpA adds particular value for applications requiring stability under challenging conditions, while the fundamental role of chromosome organization in cellular function makes this research area broadly relevant across biotechnology sectors.

What are the key takeaways from current research on O. iheyensis ScpA and bacterial chromosome organization?

Our review of current research on Oceanobacillus iheyensis ScpA and bacterial chromosome organization reveals several key takeaways with significant implications for both fundamental biology and applied research:

Fundamental Molecular Mechanisms:
The SMC-ScpA-ScpB complex represents an evolutionarily conserved molecular machine essential for chromosome condensation and segregation in bacteria. ScpA serves as a critical component of this complex, facilitating interactions between SMC and ScpB while contributing to proper complex assembly and function . The precise positioning of these complexes at discrete foci within the cell, particularly adjacent to chromosomal origin regions, suggests a highly organized mechanism for chromosome management throughout the cell cycle .

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.