Recombinant Neurospora crassa Molybdopterin synthase catalytic subunit (mocs-2)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized fulfillment.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, which may serve as a guideline for your preparation.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer components, temperature, and inherent 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. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
Tag type is finalized during production. If you require a specific tag, please inform us for prioritized development.
Synonyms
nit-8; 7C14.140; NCU03170; Molybdopterin synthase catalytic subunit; EC 2.8.1.12; Common component for nitrate reductase and xanthine dehydrogenase protein H; Molybdenum cofactor synthesis protein 2 large subunit; Molybdenum cofactor synthesis protein 2B; MOCS2B; Nitrate nonutilizer protein 8
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-206
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987)
Target Names
nit-8
Target Protein Sequence
MATQQPTQTD NSAQAQPPQT NPAKPTEISE PGIYVALTHD HLNYQSVIDR VRSPEAGAIV VFAGTTRNNF NSLPVQHLSY TSYAPLALRT MLTICRSILT KHGLKGIAMV HRLGVVPIGE ESILIAVSSP HRQAAWRAGE EALEECKAKV EVWKREEFGG EGGGVWRANR DGAVGVKVDE PRIGKGEVDE KEDEGDSGNG GNDRKS
Uniprot No.

Target Background

Function

The catalytic subunit of the molybdopterin synthase complex catalyzes the conversion of precursor Z to molybdopterin. Its function involves incorporating two sulfur atoms, derived from thiocarboxylated MOCS2A, into precursor Z, thereby generating a dithiolene group.

Database Links

KEGG: ncr:NCU03170

Protein Families
MoaE family, MOCS2B subfamily
Subcellular Location
Cytoplasm.

Q&A

What is the molybdopterin synthase catalytic subunit in Neurospora crassa and how does it compare to homologs in other organisms?

Molybdopterin synthase catalytic subunit in Neurospora crassa is a critical enzyme in the molybdenum cofactor (Moco) biosynthesis pathway. Similar to other organisms, the N. crassa molybdopterin synthase functions as a heterotetrameric enzyme composed of large (MoaE-like) and small (MoaD-like) subunits . In bacteria, humans, and N. crassa, the enzyme catalyzes the formation of the dithiolene group required for molybdenum ligation in the final Moco structure . Unlike bacterial systems where separate genes encode the two domains, in humans and N. crassa, a single gene locus (MOCS1 and nit-7, respectively) encodes both domains required for catalytic activity . This evolutionary conservation underscores the essential nature of this enzyme across different kingdoms of life.

What is the role of molybdopterin synthase in the molybdenum cofactor biosynthesis pathway?

Molybdopterin synthase catalyzes a crucial step in the molybdenum cofactor biosynthesis pathway by generating the dithiolene group of molybdopterin that is responsible for molybdenum ligation . In the activated form of the enzyme, the MoaD C-terminus is present as a thiocarboxylate, which provides the sulfur atom necessary for dithiolene formation . This reaction transforms a precursor molecule into molybdopterin, which can then bind molybdenum to form the complete molybdenum cofactor. The functional molybdenum cofactor is subsequently incorporated into various molybdoenzymes, including nitrate reductase, which plays essential roles in nitrogen metabolism .

How is the expression of molybdopterin synthase regulated in Neurospora crassa?

The expression of molybdopterin synthase in N. crassa is under nitrogen-dependent transcriptional control, similar to the regulation of nitrate reductase (NR; gene locus nit-3) . Recent research has demonstrated that upon induction of high cellular Moco demand, a specific transcript variant of the nit-7 gene is increasingly formed, suggesting that the encoded enzyme NIT7-A is the key player for Moco biosynthesis activity in Neurospora . The regulation ensures that molybdopterin synthase is expressed at appropriate levels to meet the cellular demand for molybdenum cofactor, particularly under nitrogen-derepressing conditions when nitrate reductase activity is required .

What are the optimal conditions for expressing recombinant N. crassa molybdopterin synthase in heterologous systems?

For effective expression of recombinant N. crassa molybdopterin synthase, E. coli-based expression systems have proven successful, similar to those used for human MOCS2 . When designing expression constructs, researchers should consider:

  • Expression vector selection: pET-based vectors with T7 promoters offer strong, inducible expression.

  • Codon optimization: Adjusting codons for E. coli usage can significantly improve expression yield.

  • Fusion tags: C-terminal His-tags facilitate purification without interfering with the active site formed by the N-terminal domains of the subunits .

  • Growth conditions: Expression at lower temperatures (16-20°C) after IPTG induction often improves protein solubility.

  • Buffer composition: Including glycerol (10%) in purification buffers enhances stability, similar to conditions used for human MOCS2 .

For protein storage, maintaining the purified protein at ≤-70°C in a buffer containing PBS, NaCl, and glycerol (similar to human MOCS2 storage conditions) minimizes freeze-thaw degradation and preserves enzymatic activity .

How can researchers assay molybdopterin synthase activity in vitro?

Assaying molybdopterin synthase activity can be accomplished through several complementary approaches:

  • Converting factor activity assay: This method measures the ability of the high molecular weight fraction from cell extracts to convert a low molecular weight compound from nit-1 and nit-8 mutants into biologically active molybdopterin (MPT) . The assay involves:

    • Preparing protein extracts from wild-type and mutant strains

    • Separating high and low molecular weight fractions

    • Combining fractions in appropriate buffers

    • Measuring the formation of active molybdopterin

  • Reconstitution of nitrate reductase activity: This indirect approach uses the restoration of nitrate reductase activity as a readout for functional molybdopterin formation . This involves:

    • Preparing apo-nitrate reductase from nit-1 mutants

    • Incubating with purified molybdopterin synthase and precursor Z

    • Adding molybdate to form the complete molybdenum cofactor

    • Measuring nitrate reductase activity using standard assays

  • Direct detection of dithiolene formation: Using analytical techniques such as HPLC with fluorescence detection or mass spectrometry to monitor the conversion of precursor Z to molybdopterin.

Assay MethodAdvantagesLimitationsKey Controls
Converting factor activityBiological relevance, measures functional MPTIndirect measurement, requires mutant strainsnit-7 mutant (positive control), nit-8 mutant (negative control)
NR activity reconstitutionQuantitative, established protocolsRequires purified apo-NRMock treatments without molybdopterin synthase
Direct precursor Z conversionDirect measurement of reactionRequires specialized equipmentSynthetic standards of precursor Z and MPT

What are the best methods for purifying native and recombinant molybdopterin synthase from N. crassa?

Purification of native and recombinant molybdopterin synthase from N. crassa requires different approaches:

For native enzyme:

  • Growth conditions: Culture N. crassa under nitrogen-derepressing conditions with nitrate to induce expression .

  • Cell disruption: Grind mycelia in liquid nitrogen followed by extraction in buffer containing protease inhibitors.

  • Initial fractionation: Ammonium sulfate precipitation to concentrate proteins.

  • Chromatographic separation:

    • Ion exchange chromatography (DEAE or Q-Sepharose)

    • Size exclusion chromatography to separate based on the heterotetrameric structure

    • Affinity chromatography using substrate analogs

For recombinant enzyme:

  • Heterologous expression in E. coli with appropriate fusion tags (His-tag recommended) .

  • Lysis under native conditions in buffers containing 300mM NaCl, 10% glycerol, pH 7.4 .

  • Immobilized metal affinity chromatography (IMAC) using Ni-NTA resin.

  • Size exclusion chromatography to ensure tetrameric assembly and remove aggregates.

  • Storage in stabilizing buffer (similar to conditions used for human MOCS2: PBS with 300mM NaCl, 10% glycerol) .

Verification of purified enzyme can be performed using monoclonal antibodies specific to N. crassa molybdopterin synthase subunits, similar to the approach used for generating antibodies against NIT-7A .

How does the structure of N. crassa molybdopterin synthase compare to the solved crystal structures from other organisms?

While the specific crystal structure of N. crassa molybdopterin synthase has not been fully elucidated in the provided search results, comparative analysis with known structures from other organisms reveals important insights:

The heterotetrameric structure observed in bacterial molybdopterin synthase, comprising two large (MoaE) and two small (MoaD) subunits, is likely conserved in N. crassa . The key structural feature across species is the deep insertion of the C-terminus of each small subunit into the large subunit to form the active site . This architecture positions the thiocarboxylate at the C-terminus of the small subunit in the proper orientation for the sulfur transfer reaction.

Based on the crystal structure studies of molybdopterin synthase:

  • The binding pocket for the terminal phosphate of molybdopterin (the product) is formed at the interface between the large and small subunits .

  • The crystal structure also suggests a binding site for the pterin moiety present in precursor Z and molybdopterin .

  • Conformational changes accompany the binding of the small subunit to the large subunit dimer, as revealed by studies of the large subunit homodimer structure .

Given the evolutionary conservation of this pathway, these structural features are likely present in the N. crassa enzyme as well, with species-specific variations that may affect substrate specificity or regulatory interactions.

What is the mechanism of sulfur transfer catalyzed by molybdopterin synthase in N. crassa?

The sulfur transfer mechanism catalyzed by molybdopterin synthase involves the generation of a dithiolene group on the pterin ring of precursor Z to form molybdopterin . Based on structural and biochemical studies:

  • Activation step: The small subunit's C-terminus must first be activated as a thiocarboxylate (-COSX), with the sulfur atom derived from a separate sulfur mobilization pathway .

  • Active site formation: The thiocarboxylate-containing C-terminus of the small subunit is inserted deeply into the large subunit, positioning the reactive sulfur atom in proximity to the substrate (precursor Z) .

  • Nucleophilic attack: The thiocarboxylate sulfur acts as a nucleophile, attacking the appropriate carbon in precursor Z to initiate dithiolene formation.

  • Second sulfur incorporation: A second sulfur atom is incorporated to complete the dithiolene group, though the exact mechanism of this second incorporation varies between organisms.

  • Product release: The formed molybdopterin is released, which can then bind molybdenum to form the active molybdenum cofactor.

In N. crassa, studies of nit-7 and nit-8 mutants provide evidence for the conservation of this mechanism, as these mutants lack converting factor activity necessary for molybdopterin formation . The nit-9 mutants contain a protein-bound precursor form of molybdenum cofactor, presumed to be molybdopterin bound to apo-nitrate reductase, which can be converted to active molybdenum cofactor in the presence of reduced glutathione and high molybdate concentrations .

How do mutations in the catalytic subunit affect enzyme activity and Neurospora phenotype?

Mutations in the molybdopterin synthase catalytic subunit lead to characteristic phenotypes and biochemical defects in N. crassa:

The nit-7 mutation affects the first step of the multi-step Moco biosynthesis pathway . Biochemical characterization reveals that nit-7 mutants contain an activity that fits the functional definition of converting factor activity in E. coli, suggesting that the mutation affects a later step in molybdopterin synthesis rather than the initial converting factor activity .

Phenotypic consequences include:

  • Inability to grow on nitrate as the sole nitrogen source due to lack of functional nitrate reductase

  • Pleiotropic effects on all molybdoenzymes due to the central role of molybdopterin in molybdenum cofactor formation

  • Potential compensatory mechanisms, as indicated by the formation of specific transcript variants under high Moco demand conditions

At the molecular level, mutations may affect:

  • Protein stability and folding

  • Subunit interactions within the heterotetrameric structure

  • Substrate binding or catalytic activity

  • Post-translational modifications or subcellular localization

Research has shown that NIT-7A is localized in the mitochondria, and mitochondrial import of NIT-7B requires fusion of NIT-7A to NIT-7B, suggesting that mutations affecting this fusion or localization could disrupt enzyme function .

How can researchers design genetic complementation experiments to study molybdopterin synthase function?

Designing effective genetic complementation experiments for molybdopterin synthase requires careful consideration of several factors:

  • Construct design considerations:

    • Include the complete gene with native promoter and terminator sequences for proper expression regulation

    • Create targeted mutations to study specific domains or residues

    • Consider epitope or fluorescent protein tagging for localization studies

    • Generate both nit-7A and nit-7AB expression constructs to distinguish their specific roles

  • Transformation methods:

    • Use established N. crassa transformation protocols with selection markers

    • Consider site-specific integration to avoid position effects

    • For heterologous complementation, express N. crassa genes in E. coli or yeast mocs mutants

  • Complementation analysis approaches:

    • Growth assays on media with nitrate as sole nitrogen source

    • Biochemical assays for nitrate reductase activity

    • Direct measurement of molybdopterin and molybdenum cofactor levels

    • Transcript analysis to verify expression patterns

  • Controls and validations:

    • Wild-type strain as positive control

    • Original mutant transformed with empty vector as negative control

    • Rescue with well-characterized orthologous genes from other species

The analysis should include quantitative measurements of growth rates, enzyme activities, and cofactor levels under different nitrogen conditions to fully assess the degree of complementation.

What are the current challenges in studying the transcriptional regulation of molybdopterin synthase genes?

The study of transcriptional regulation of molybdopterin synthase genes presents several challenges:

  • Complex splicing patterns: In N. crassa, the nit-7 gene consists of two exons flanking a single intron, with translation of the non-spliced transcript leading to NIT-7A protein and splicing resulting in the NIT-7AB variant . This complexity requires:

    • Precise RNA analysis techniques to distinguish transcript variants

    • Reporter constructs to monitor alternative splicing events

    • Methods to manipulate splicing to study functional consequences

  • Nitrogen-dependent regulation: The expression of nit-7 is under nitrogen-dependent control, similar to nitrate reductase . Challenges include:

    • Identifying specific transcription factors involved

    • Mapping regulatory elements in the promoter region

    • Understanding crosstalk with other nitrogen metabolism pathways

  • Mitochondrial localization factors: NIT-7A is localized in mitochondria, and mitochondrial import of NIT-7B requires fusion with NIT-7A . Research challenges include:

    • Identifying mitochondrial targeting sequences

    • Understanding the regulation of protein localization

    • Determining how localization affects function

  • Methodological approaches:

    • Chromatin immunoprecipitation (ChIP) to identify transcription factor binding sites

    • Promoter deletion and mutation analysis to map regulatory elements

    • RNA-seq to quantify transcript variants under different conditions

    • Nuclear run-on assays to measure transcription rates

Understanding these regulatory mechanisms is crucial for gaining insights into how N. crassa coordinates molybdoenzyme expression with cofactor biosynthesis under different environmental conditions.

How can researchers investigate the interactions between molybdopterin synthase and other proteins in the molybdenum cofactor biosynthesis pathway?

Investigating protein-protein interactions in the molybdenum cofactor biosynthesis pathway requires multiple complementary approaches:

  • Affinity purification coupled with mass spectrometry:

    • Express tagged versions of molybdopterin synthase subunits

    • Purify under native conditions to maintain interactions

    • Identify co-purifying proteins by mass spectrometry

    • Validate interactions through reciprocal pull-downs

  • Yeast two-hybrid or split-reporter assays:

    • Screen for interactions between molybdopterin synthase subunits and other Moco biosynthesis proteins

    • Map interaction domains through truncation and mutation analysis

    • Test interactions under different nitrogen conditions

  • Bimolecular fluorescence complementation (BiFC):

    • Visualize interactions in living cells

    • Determine subcellular localization of interaction complexes

    • Particularly useful for studying the interactions between NIT-7A and NIT-7B

  • Co-immunoprecipitation with specific antibodies:

    • Use monoclonal antibodies like 4C10 (anti-NIT-7A) to pull down native complexes

    • Identify interacting partners by immunoblotting or mass spectrometry

    • Compare complex formation under different growth conditions

  • Structural biology approaches:

    • Protein crystallography of co-complexes

    • Cryo-EM analysis of larger assemblies

    • Hydrogen-deuterium exchange mass spectrometry to map interaction surfaces

Understanding these interactions is critical for elucidating how molybdopterin synthase is integrated into the broader molybdenum cofactor biosynthesis pathway and how these interactions may be regulated in response to changing cellular demands.

How does the structure and function of N. crassa molybdopterin synthase compare to human MOCS2 in the context of molybdenum cofactor deficiency disorders?

Comparing N. crassa molybdopterin synthase with human MOCS2 reveals important evolutionary and functional insights:

  • Structural organization:

    • Both N. crassa and human enzymes function as heterotetrameric complexes

    • In humans, a single gene locus (MOCS1) encodes both catalytic domains, similar to the nit-7 locus in N. crassa

    • The core catalytic mechanism involving the thiocarboxylate formation and sulfur transfer is conserved

  • Genetic organization differences:

    • In N. crassa, translation of non-spliced nit-7 transcript leads to NIT-7A protein, while splicing results in NIT-7AB

    • Human MOCS2 gene produces two subunits (MOCS2A and MOCS2B) through alternative transcription start sites and splicing

  • Clinical relevance:

    • Mutations in human MOCS genes cause molybdenum cofactor deficiency, a severe and often fatal disorder

    • N. crassa mutants provide valuable models for understanding the biochemical consequences of such mutations

    • Functional studies in N. crassa have helped elucidate the roles of domains that are conserved in human MOCS proteins

  • Therapeutic implications:

    • Understanding substrate binding and catalytic mechanisms in N. crassa can inform therapeutic approaches for human molybdenum cofactor deficiency

    • N. crassa can serve as a platform for screening potential therapeutic compounds that might enhance residual molybdopterin synthase activity

The conservation of this pathway across evolutionary distance underscores its fundamental importance in cellular metabolism and provides valuable opportunities for translational research.

What insights can be gained from studying molybdopterin synthase across different fungal species?

Comparative analysis of molybdopterin synthase across fungal species provides valuable insights into enzyme evolution, specialization, and regulation:

  • Evolutionary conservation and divergence:

    • The core catalytic mechanism is conserved across fungi, but species-specific adaptations exist

    • Variations in domain organization and gene structure may reflect ecological adaptations

    • Regulatory elements in promoters show greater divergence than coding sequences, reflecting adaptations to different niches

  • Nitrogen metabolism adaptations:

    • Species utilizing different nitrogen sources show corresponding adaptations in molybdopterin synthase regulation

    • Saprophytic fungi like N. crassa show nitrogen-dependent regulation

    • Pathogenic fungi may have evolved different regulatory mechanisms based on host interactions

  • Subcellular localization:

    • Mitochondrial localization of NIT-7A in N. crassa suggests compartmentalization of the pathway

    • Variations in targeting sequences across fungal species may reflect differences in mitochondrial import mechanisms

    • Co-localization with other pathway enzymes may vary between species

  • Research approaches:

    • Comparative genomics to identify conserved domains and regulatory elements

    • Heterologous expression to test functional conservation

    • Domain swapping experiments to identify species-specific functional elements

    • Expression pattern analysis across growth conditions and developmental stages

This comparative approach not only enhances our understanding of molybdenum cofactor biosynthesis but also provides insights into fungal evolution and adaptation to diverse ecological niches.

What methodological approaches can be used to study the evolutionary conservation of molybdopterin synthase structure and function?

Multiple methodological approaches can be employed to study the evolutionary conservation of molybdopterin synthase:

  • Sequence and structure analysis:

    • Multiple sequence alignment of molybdopterin synthase genes across species

    • Phylogenetic analysis to trace evolutionary relationships

    • Homology modeling based on known crystal structures

    • Analysis of selection pressure on different domains using dN/dS ratios

  • Functional conservation studies:

    • Heterologous complementation of mutants across species boundaries

    • Expression of recombinant enzymes from different species and comparison of enzymatic properties

    • Structure-function analysis through site-directed mutagenesis of conserved residues

  • Regulatory mechanism comparison:

    • Promoter analysis across species to identify conserved regulatory elements

    • Reporter gene assays to compare transcriptional responses

    • Analysis of splice variants and alternative processing across species

  • Advanced computational approaches:

    • Ancestral sequence reconstruction to infer properties of evolutionary intermediates

    • Molecular dynamics simulations to compare structural flexibility

    • Protein-protein interaction network analysis across species

  • Experimental evolution:

    • Directed evolution of molybdopterin synthase under selective pressure

    • Analysis of natural variants in different strains and species

    • Adaptation studies under different nitrogen source availability

These approaches collectively provide a comprehensive understanding of how molybdopterin synthase has evolved while maintaining its essential catalytic function across diverse organisms from bacteria to fungi and humans.

What are common challenges in purifying active recombinant N. crassa molybdopterin synthase and how can they be addressed?

Researchers frequently encounter several challenges when purifying active recombinant N. crassa molybdopterin synthase:

  • Protein solubility issues:

    • Challenge: Formation of inclusion bodies during overexpression

    • Solution: Lower induction temperature (16-20°C), use solubility-enhancing fusion tags, or optimize expression conditions with auto-induction media

    • Alternative: Develop refolding protocols from inclusion bodies using step-wise dialysis

  • Maintaining heterotetrameric structure:

    • Challenge: Dissociation of subunits during purification

    • Solution: Include stabilizing agents in buffers (10% glycerol, low concentrations of reducing agents)

    • Approach: Consider co-expression of both subunits with compatible tags for tandem affinity purification

  • Loss of thiocarboxylate activation:

    • Challenge: The essential thiocarboxylate at the C-terminus of the small subunit is lost during purification

    • Solution: Co-express with the sulfur transfer system or develop in vitro reconstitution methods

    • Validation: Confirm thiocarboxylate presence using mass spectrometry

  • Storage stability:

    • Challenge: Activity loss during storage

    • Solution: Store at ≤-70°C in buffer containing cryoprotectants like glycerol

    • Best practice: Aliquot protein to avoid repeated freeze-thaw cycles

  • Verification of activity:

    • Challenge: Difficulty in assessing functional activity

    • Solution: Develop complementation assays using nit-7 mutant extracts

    • Alternative: Establish in vitro assays using precursor Z and analytical detection methods

ProblemSymptomsSolutionsValidation Method
Inclusion bodiesLow soluble yieldLower temperature, solubility tagsSDS-PAGE of soluble fraction
Subunit dissociationActivity loss, size shiftStabilizing buffer additivesSize exclusion chromatography
Inactive enzymeNo catalytic activityCo-expression with sulfur transfer systemMass spectrometry for thiocarboxylate
Protein instabilityDegradation bandsOptimize buffer, add protease inhibitorsWestern blot, activity time course

How can researchers optimize conditions for assaying molybdopterin synthase activity in crude extracts?

Optimizing assay conditions for molybdopterin synthase in crude extracts requires addressing several key factors:

  • Extract preparation:

    • Use rapid extraction methods to preserve enzyme activity

    • Include protease inhibitor cocktails to prevent degradation

    • Prepare extracts under anaerobic conditions to prevent oxidation of the thiocarboxylate

    • Fractionate extracts to separate high and low molecular weight components as described for converting factor activity assays

  • Assay buffer optimization:

    • Test various pH ranges (typically pH 7.0-8.0)

    • Include reducing agents (DTT, β-mercaptoethanol, or reduced glutathione)

    • Add stabilizing agents (glycerol, PEG)

    • Determine optimal salt concentration (typically 50-300 mM NaCl)

  • Substrate considerations:

    • For direct assays, ensure precursor Z is protected from oxidation

    • For coupled assays with nitrate reductase, use freshly prepared apo-NR from nit-1 mutants

    • Consider substrate concentration ranges carefully, as high concentrations may be inhibitory

  • Detection methods:

    • Indirect detection via nitrate reductase activity using established assays

    • Direct detection of molybdopterin formation using fluorescence-based HPLC methods

    • Consider using specific antibodies for detecting protein-bound molybdopterin intermediates

  • Controls and normalization:

    • Include positive controls (wild-type extracts) and negative controls (extracts from known molybdopterin synthase mutants)

    • Account for background activity from endogenous enzymes

    • Normalize activity to protein concentration or to internal standards

These optimizations should be systematically evaluated to establish a robust and reproducible assay protocol suitable for comparative studies across different experimental conditions.

What analytical methods can be used to detect and quantify molybdopterin and its precursors in N. crassa extracts?

Several analytical methods can be employed to detect and quantify molybdopterin and its precursors in N. crassa extracts:

  • Chromatographic methods:

    • HPLC with fluorescence detection: Molybdopterin and its derivatives exhibit natural fluorescence that can be used for detection

    • Anion exchange chromatography: Separates molecules based on charge differences

    • Reversed-phase HPLC: Useful for separating molybdopterin and precursors based on hydrophobicity

    • Size exclusion chromatography: Separates free molybdopterin from protein-bound forms

  • Mass spectrometry approaches:

    • LC-MS/MS: Provides both separation and structural identification

    • MALDI-TOF: Useful for analyzing protein-bound forms

    • Targeted multiple reaction monitoring (MRM): For quantitative analysis of specific known compounds

    • Ion mobility MS: Helps distinguish isomers with similar masses

  • Biological activity assays:

    • Reconstitution of nitrate reductase activity: Indirect measurement of molybdopterin content

    • Converting factor activity assays: Measures the ability to convert precursors to active molybdopterin

    • In vitro molybdate repair assays: Detects protein-bound precursor forms

  • Specialized detection methods:

    • Derivatization techniques: Chemical modification to enhance detection sensitivity

    • Isotope labeling: For tracking biosynthetic pathways

    • Antibody-based detection: Using specific antibodies against molybdopterin or protein-bound forms

  • Sample preparation considerations:

    • Rapid extraction under anaerobic conditions to prevent oxidation

    • Acidic extraction for release of protein-bound forms

    • Selective precipitation or ultrafiltration to concentrate samples

    • Use of internal standards for quantification

These methods can be combined in complementary approaches to provide both qualitative and quantitative information about molybdopterin and its precursors in complex biological samples.

What emerging technologies could advance our understanding of molybdopterin synthase function and regulation?

Several emerging technologies hold promise for advancing our understanding of molybdopterin synthase:

  • CRISPR/Cas9 genome editing in N. crassa:

    • Precise manipulation of the nit-7 locus to create specific mutations

    • Introduction of reporter tags at endogenous loci

    • Creation of conditional alleles to study essential functions

    • Multiplexed editing to study pathway interactions

  • Single-cell analysis technologies:

    • Single-cell RNA-seq to study cell-to-cell variation in expression

    • Single-molecule imaging to track protein localization and dynamics

    • Microfluidics-based approaches for real-time monitoring of single cells

    • Correlative microscopy to link localization with function

  • Structural biology advancements:

    • Cryo-electron microscopy for high-resolution structures without crystallization

    • Integrative structural biology combining multiple data sources

    • Time-resolved structural methods to capture catalytic intermediates

    • Hydrogen-deuterium exchange mass spectrometry to study dynamics

  • Systems biology approaches:

    • Metabolic flux analysis to understand molybdenum cofactor pathway dynamics

    • Multi-omics integration (transcriptomics, proteomics, metabolomics)

    • Mathematical modeling of the complete biosynthetic pathway

    • Network analysis to identify regulatory hubs

  • Synthetic biology tools:

    • Engineered biosensors for molybdenum cofactor levels

    • Orthogonal translation systems for site-specific incorporation of modified amino acids

    • Cell-free expression systems for rapid prototyping

    • Minimal synthetic pathways to study essential components

These technologies will enable researchers to address longstanding questions about molybdopterin synthase with unprecedented precision and to develop new hypotheses based on systems-level insights.

What are unresolved questions about the catalytic mechanism of molybdopterin synthase that require further investigation?

Despite significant progress, several key questions about the catalytic mechanism of molybdopterin synthase remain unresolved:

  • Thiocarboxylate formation and transfer:

    • What is the precise mechanism of sulfur transfer from the thiocarboxylate to precursor Z?

    • How is the thiocarboxylate group protected from oxidation during catalysis?

    • Is there a conformational change in the enzyme during sulfur transfer?

    • What is the role of specific conserved residues in coordinating the reaction?

  • Second sulfur incorporation:

    • What is the source of the second sulfur atom in the dithiolene group?

    • Is the incorporation of two sulfur atoms concerted or sequential?

    • Are there intermediate states that can be isolated and characterized?

    • How does the enzyme ensure correct stereochemistry of the dithiolene group?

  • Substrate recognition and binding:

    • What specific residues are involved in precursor Z recognition?

    • How does the enzyme discriminate between similar pterin derivatives?

    • Is there allosteric regulation of substrate binding?

    • What is the order of substrate binding and product release?

  • Species-specific variations:

    • Are there fundamental differences in catalytic mechanism between N. crassa and other organisms?

    • How do these differences relate to ecological adaptations?

    • Can mechanism variations explain differences in sensitivity to inhibitors?

  • Methodological approaches for future studies:

    • Time-resolved spectroscopy to capture transient intermediates

    • Site-directed mutagenesis of conserved residues

    • Trapping of reaction intermediates through mechanism-based inhibitors

    • Quantum mechanical/molecular mechanical (QM/MM) calculations to model the reaction coordinate

Resolving these questions will require interdisciplinary approaches combining structural biology, enzymology, computational modeling, and advanced spectroscopic techniques.

How might insights from N. crassa molybdopterin synthase research contribute to therapeutic strategies for molybdenum cofactor deficiency in humans?

Research on N. crassa molybdopterin synthase has significant translational potential for addressing human molybdenum cofactor deficiency disorders:

  • Model system advantages:

    • N. crassa provides a genetically tractable system for studying conserved aspects of molybdopterin synthesis

    • Mutations in N. crassa can model specific human disease variants

    • The rapid growth and established genetic tools facilitate high-throughput screening approaches

  • Therapeutic strategy development:

    • Structure-guided design of small molecules that could stabilize mutant human MOCS proteins

    • Identification of bypass mechanisms or alternative pathways observed in suppressor mutants

    • Development of enzyme replacement strategies based on recombinant protein production systems

    • Screening for compounds that enhance residual molybdopterin synthase activity

  • Mechanistic insights for treatment approaches:

    • Understanding precursor Z binding could guide development of modified precursors for therapy

    • Insights into protein-protein interactions might reveal new therapeutic targets

    • Detailed knowledge of the reaction mechanism could identify rate-limiting steps amenable to intervention

  • Experimental testing platforms:

    • Humanized N. crassa strains expressing human MOCS variants

    • Heterologous complementation systems to evaluate therapeutic candidates

    • Cell-free systems incorporating purified recombinant enzymes for high-throughput screening

    • Validation in mammalian cell models prior to clinical translation

  • Translational research directions:

    • Investigation of small molecule chaperones to improve folding of mutant proteins

    • Development of cyclic pyranopterin monophosphate (cPMP) derivatives with enhanced stability

    • Exploration of gene therapy approaches informed by fungal expression systems

    • Design of targeted protein degradation strategies for dominant negative mutants

By leveraging the fundamental knowledge gained from N. crassa studies, researchers can develop more effective therapeutic interventions for human patients suffering from these devastating disorders.

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.