Recombinant Protochlamydia amoebophila Chromosomal replication initiator protein DnaA 1 (dnaA1)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for fulfillment based on availability.
Lead Time
Delivery times vary depending on the purchase method and location. 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 consolidate the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and may serve as a reference.
Shelf Life
Shelf life depends on storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is finalized during production. Specify your required tag type for preferential development.
Synonyms
dnaA1; pc1082Chromosomal replication initiator protein DnaA 1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-451
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Protochlamydia amoebophila (strain UWE25)
Target Names
dnaA1
Target Protein Sequence
MQAWEEFLKA QEVELGLETV QKWLRTLKIQ RFDACNLYLE AKDSFQALWF EEHIRNKAQQ KFINGNNKRI KIHLSVANTP QRAKKTKTAN KEKDFKAPFE LTFDELDPLC LFPYFISTEE NLLSHQLLLE IAGLSPQIHS TQLGTFNPIY LYGSGGSGKT HLLMSLAHAL KAQGLKVIYV RAETFTDHVV TAIRAGEMSV FRQAYRNIDV LLVDDVHVFS RKGATQEEFF HTFNTLHLEG KQIILASECS PQDLQLIEPR LVSRFEWGIV LPLKPLRPGE MRNLLIAKAK ALHFELPLKI ADYLIETFKS NAKALIKGLE ALVLRLHLDA KHSITALSVT ATKALLIDLI EEEQKTAITP QKIIQAVAEQ YGIRTEDILG KAQTRECALP RQLAMHLCRE QLKMPFMKIG DLFSRDHSTV MSSVKSIQKS LEQDDREISG ICHIILKKLQ G
Uniprot No.

Target Background

Function
Recombinant Protochlamydia amoebophila Chromosomal replication initiator protein DnaA 1 (dnaA1) plays a crucial role in initiating and regulating chromosomal replication. It binds to the origin of replication, specifically interacting with double-stranded DNA at a 9-bp consensus sequence (dnaA box): 5'-TTATC[CA]A[CA]A-3'. DnaA also binds ATP and acidic phospholipids.
Database Links

KEGG: pcu:pc1082

STRING: 264201.pc1082

Protein Families
DnaA family
Subcellular Location
Cytoplasm.

Q&A

What is Protochlamydia amoebophila and why is its DnaA1 protein significant for research?

Protochlamydia amoebophila is a Chlamydia-related bacterium that thrives as an obligate intracellular symbiont within Acanthamoeba species . Its significance stems from being an environmental chlamydial organism that shares evolutionary relationships with pathogenic Chlamydiaceae while maintaining distinct biological properties. The DnaA1 protein is particularly important as a chromosomal replication initiator, essential for the bacterium's DNA replication process. This protein represents an interesting research target for understanding the fundamental biology of obligate intracellular bacteria, particularly how organisms with reduced genomes coordinate their replication cycles with host cells .

Studying P. amoebophila DnaA1 provides insights into:

  • The molecular mechanisms of bacterial chromosome replication initiation in obligate intracellular bacteria

  • Evolutionary adaptations in replication systems of symbiotic bacteria

  • Potential targets for controlling or manipulating chlamydial growth

What are the standard methods for expressing recombinant P. amoebophila DnaA1 protein?

Recombinant P. amoebophila DnaA1 is typically expressed using heterologous expression systems, primarily in E. coli, as demonstrated in multiple studies with other P. amoebophila proteins . Based on established protocols for similar P. amoebophila proteins, a methodological approach includes:

  • Gene amplification and cloning:

    • PCR amplification of the dnaA1 gene (pc1082) from purified P. amoebophila DNA

    • Introduction of appropriate restriction sites (e.g., XhoI at the start and BamHI after the stop codon)

    • Cloning into an expression vector such as pET16b containing a histidine tag

  • Expression and purification:

    • Transformation into an E. coli expression strain (e.g., BL21)

    • IPTG induction of protein expression

    • Purification using affinity chromatography, typically Ni-NTA for His-tagged proteins

    • Validation by SDS-PAGE and Western blotting

This approach yields recombinant protein with >90% purity that can be stored in liquid form containing glycerol at -20°C for short-term storage or -80°C for long-term storage .

How can I verify the functionality of recombinant P. amoebophila DnaA1 protein in vitro?

Verifying the functionality of recombinant DnaA1 requires assessing its primary activity as a replication initiator. Based on established approaches for similar proteins, recommended methodological steps include:

  • DNA binding assays:

    • Electrophoretic mobility shift assays (EMSA) with labeled DNA fragments containing putative DnaA boxes

    • DNase I footprinting to identify specific binding sites

  • ATPase activity assessment:

    • Measuring ATP hydrolysis using colorimetric assays (e.g., malachite green)

    • Monitoring the protein's ability to bind ATP using filter-binding assays

  • Helicase loading assays:

    • Testing the ability of DnaA1 to facilitate the loading of replicative helicases onto DNA

    • Assessing unwinding of DNA at origin sequences

  • Functional complementation:

    • Attempting complementation of temperature-sensitive dnaA mutants in model systems

    • Observing restoration of replication function

These methodological approaches provide comprehensive assessment of the protein's functional capabilities in relation to its role in DNA replication initiation.

How do the biochemical properties of P. amoebophila DnaA1 compare to DnaA proteins from other obligate intracellular bacteria?

Comparative analysis of DnaA proteins from various obligate intracellular bacteria reveals important evolutionary adaptations in replication systems. While specific data for P. amoebophila DnaA1 is limited in the search results, a methodological approach for this comparison would include:

  • Sequence and structural analysis:

    • Multiple sequence alignment of DnaA proteins from P. amoebophila, pathogenic Chlamydiaceae, and free-living bacteria

    • Identification of conserved domains (DNA binding, ATP binding) and unique sequence features

    • Homology modeling to predict structural differences

  • Biochemical property comparison:

    • Expression and purification of multiple DnaA proteins under identical conditions

    • Side-by-side assessment of:

      • DNA binding affinity and specificity

      • ATP binding and hydrolysis rates

      • Oligomerization properties

      • pH and temperature optima

  • Origin recognition specificity:

    • Analysis of DnaA box sequences from different bacteria

    • Cross-recognition experiments testing whether DnaA from one species can bind to origin sequences from others

This comparative approach would highlight how P. amoebophila DnaA1 has adapted to the specialized niche of an amoeba symbiont, potentially revealing unique features linked to its obligate intracellular lifestyle .

What is known about the interaction between P. amoebophila DnaA1 and host cell factors during infection?

Understanding the interaction between bacterial replication proteins and host factors is critical for obligate intracellular organisms like P. amoebophila. Based on related research on P. amoebophila's intracellular lifestyle, methodological approaches to study these interactions include:

  • Identification of potential host interaction partners:

    • Co-immunoprecipitation (Co-IP) with tagged DnaA1 in infected Acanthamoeba cells

    • Mass spectrometry analysis of pulled-down complexes

    • Yeast two-hybrid screening using DnaA1 as bait against host cDNA libraries

  • Validation of interactions:

    • Bimolecular fluorescence complementation (BiFC)

    • Förster resonance energy transfer (FRET) measurements

    • Surface plasmon resonance (SPR) for direct binding kinetics

  • Functional significance assessment:

    • RNAi knockdown of identified host factors in Acanthamoeba

    • Generation of DnaA1 mutants lacking interaction domains

    • Microscopy-based assessment of replication timing when interactions are disrupted

These approaches would help determine whether P. amoebophila DnaA1 directly interacts with host factors to coordinate bacterial replication with host cell cycles, potentially contributing to the successful establishment of its intracellular niche within amoebae .

How does P. amoebophila coordinate DnaA1-mediated replication with its unique metabolic dependencies on the host?

P. amoebophila, like other obligate intracellular bacteria, relies heavily on host metabolism. P. amoebophila is known to have specialized nucleotide transport systems for acquiring essential building blocks from its host . A comprehensive methodological approach to study the coordination between replication and metabolism would include:

  • Metabolic profiling during different replication stages:

    • Synchronized infection models in Acanthamoeba

    • Metabolomic analysis at different time points correlating with DnaA1 expression/activity

    • Isotope labeling experiments to track nucleotide flux from host to bacteria

  • Investigation of regulatory mechanisms:

    • Transcriptional analysis of dnaA1 and metabolic genes under different nutrient conditions

    • ChIP-seq to identify potential DnaA1 binding throughout the genome beyond origin regions

    • Protein modification analysis (phosphorylation, acetylation) of DnaA1 in response to metabolic signals

  • Experimental manipulation of host-pathogen metabolic interface:

    • Inhibition of specific nucleotide transporters (NTTs) and assessment of DnaA1 activity

    • Creation of conditional dnaA1 mutants to study metabolic changes when replication is altered

    • Heterologous expression systems combining P. amoebophila DnaA1 with different NTT proteins

This research approach would help elucidate how P. amoebophila coordinates its replication initiation through DnaA1 with the acquisition of essential metabolites from its host, a critical adaptation for obligate intracellular lifestyle .

What are the methodological challenges in studying the in vivo function of P. amoebophila DnaA1 and how can they be overcome?

Studying P. amoebophila proteins in vivo presents significant challenges due to the organism's obligate intracellular lifestyle and lack of genetic manipulation systems. Based on approaches used for similar challenging systems, methodological strategies include:

  • Development of cell-free transcription-translation systems:

    • Creation of P. amoebophila-specific extracts from purified bacteria

    • Supplementation with host factors to recreate the intracellular environment

    • Direct observation of DnaA1 activity on template DNA

  • Heterologous in vivo systems:

    • Expression of fluorescently tagged DnaA1 in host amoebae

    • Live-cell imaging to track localization during the infection cycle

    • Complementation studies in related, genetically tractable chlamydial species

  • Advanced microscopy approaches:

    • Super-resolution microscopy of fixed infected cells

    • Immunogold electron microscopy to precisely localize DnaA1

    • Correlative light and electron microscopy (CLEM) for context-specific visualization

  • Host cell manipulation strategies:

    • Generation of Acanthamoeba cell lines expressing modified DnaA1 binding partners

    • CRISPR interference to modify host pathways that interact with bacterial replication

These methodological approaches address the limitations of working with obligate intracellular bacteria while providing valuable insights into DnaA1 function in its natural environment .

What are the optimal conditions for expressing soluble recombinant P. amoebophila DnaA1 protein?

Obtaining soluble recombinant bacterial proteins can be challenging, especially for those from obligate intracellular organisms. Based on successful approaches with other P. amoebophila proteins, the following methodology is recommended:

  • Expression system optimization:

    • Testing multiple E. coli strains (BL21, Rosetta, Arctic Express)

    • Comparing different expression vectors with various fusion tags (His, GST, MBP)

    • Evaluation of codon-optimized synthetic genes to overcome potential codon bias

  • Expression condition matrix:

    ParameterRange to TestNotes
    Temperature10-30°CLower temperatures often increase solubility
    IPTG concentration0.1-1.0 mMLower concentrations may improve folding
    Duration4-24 hoursExtended expression at low temperature
    MediaLB, TB, M9TB provides richer nutrient source
    AdditivesGlycerol (5-10%), Glucose (0.5-1%)Can reduce basal expression
  • Lysis and purification optimization:

    • Testing different lysis buffers with varying salt concentrations (100-500 mM)

    • Addition of solubility enhancers (0.1% Triton X-100, 10% glycerol)

    • Inclusion of ATP (1-5 mM) which may stabilize DnaA1

    • Gentle lysis procedures (sonication with cooling intervals)

  • Refolding strategies if inclusion bodies form:

    • On-column refolding during affinity purification

    • Stepwise dialysis with decreasing denaturant concentrations

    • Addition of chaperone-expressing plasmids to expression strain

These optimized conditions should be determined empirically through systematic testing, as the specific requirements for P. amoebophila DnaA1 may differ from other bacterial proteins .

How can I design experiments to study the role of P. amoebophila DnaA1 in bacterial persistence and stress response?

P. amoebophila, like other chlamydial organisms, can enter persistent states under stress conditions . Investigating DnaA1's role in this process requires specialized experimental approaches:

  • Stress induction model development:

    • Establishment of defined stress conditions (nutrient limitation, antibiotics, temperature)

    • Confirmation of aberrant body formation through microscopy

    • Quantification of bacterial viability and replication during stress

  • DnaA1 expression and activity analysis:

    • qRT-PCR to measure dnaA1 transcript levels under various stress conditions

    • Western blotting with anti-DnaA1 antibodies to track protein levels

    • ChIP-qPCR to measure DnaA1 binding to the origin during persistence

  • Functional interference strategies:

    • Development of DnaA-targeting peptide inhibitors that can penetrate bacterial cells

    • Construction of antisense oligonucleotides targeting dnaA1 mRNA

    • Conditional expression systems regulated by tetracycline-responsive elements

  • Recovery dynamics assessment:

    • Time-course analysis of DnaA1 activity upon stress removal

    • Correlation between DnaA1 function restoration and resumption of bacterial division

    • Blocking DnaA1 function during recovery phase to assess essentiality

This experimental framework would help determine whether regulation of DnaA1 activity is a key mechanism in the establishment and maintenance of persistent states in P. amoebophila, potentially revealing new targets for controlling chlamydial infections .

What bioinformatic approaches should I use to identify potential regulatory elements affecting P. amoebophila DnaA1 function?

Understanding the regulation of DnaA1 requires comprehensive bioinformatic analysis. Based on approaches used in similar bacterial systems, the following methodological framework is recommended:

  • Promoter and regulatory region analysis:

    • Identification of the dnaA1 promoter region using RNA-seq data and consensus sequence analysis

    • Investigation of potential transcription factor binding sites using position weight matrices

    • Comparative genomics across chlamydial species to identify conserved regulatory elements

  • Post-transcriptional regulation prediction:

    • Secondary structure prediction of the dnaA1 mRNA using MFold or similar algorithms

    • Identification of potential small RNA binding sites using IntaRNA or TargetRNA

    • Analysis of the 5' UTR for potential riboswitches or attenuators

  • Protein modification site identification:

    • Prediction of phosphorylation, acetylation, and other post-translational modification sites

    • Structural modeling to assess how modifications might affect DnaA1 function

    • Comparison with experimental proteomics data from related species

  • Regulatory network reconstruction:

    • Integration of transcriptomic data to identify genes co-regulated with dnaA1

    • Metabolic pathway analysis to link DnaA1 regulation with key metabolic processes

    • Protein-protein interaction prediction using homology-based approaches

This comprehensive bioinformatic approach would generate testable hypotheses about the regulation of P. amoebophila DnaA1, guiding subsequent experimental validation and providing insights into how this obligate intracellular bacterium coordinates its replication with its host .

What are the most effective methods for generating antibodies against P. amoebophila DnaA1 for experimental applications?

Generating specific antibodies against P. amoebophila proteins requires careful consideration of several factors. Based on successful approaches with other P. amoebophila proteins, the following methodological strategy is recommended:

  • Antigen design and production:

    • Identification of immunogenic epitopes using prediction algorithms

    • Expression of full-length DnaA1 versus specific domains (particularly hydrophilic regions)

    • Production of multiple peptide antigens spanning different protein regions

    • Ensuring high purity (>95%) of recombinant protein or synthetic peptides

  • Immunization strategy:

    • Selection of appropriate animals (rabbits, guinea pigs, chickens) for antibody production

    • Implementation of prime-boost protocols with multiple immunization timepoints

    • Careful adjuvant selection to maximize response while minimizing non-specific reactivity

    • Consideration of alternative hosts (e.g., llamas for nanobody production)

  • Antibody purification and validation:

    • Affinity purification against the immunizing antigen

    • Extensive cross-reactivity testing against host (Acanthamoeba) proteins

    • Western blot analysis against both recombinant protein and native DnaA1 from P. amoebophila

    • Immunofluorescence microscopy to confirm specificity in infected amoeba cells

  • Application-specific optimization:

    • Determination of optimal antibody dilutions for each application (Western, IF, IP)

    • Fixation method testing (methanol versus PFA) for immunofluorescence applications

    • Development of blocking conditions to minimize background in complex samples

Following this methodological approach has been demonstrated to generate highly specific antibodies against P. amoebophila proteins, as shown in previous studies with other targets such as inclusion membrane proteins .

How can I set up an in vitro DNA replication system to study P. amoebophila DnaA1 function?

Establishing an in vitro replication system for studying P. amoebophila DnaA1 requires reconstitution of the essential components of bacterial replication machinery. The methodological approach includes:

  • Identification and production of core replisome components:

    • Recombinant expression of key P. amoebophila replication proteins (DnaA1, DNA polymerase III, helicase, primase)

    • Purification under conditions that maintain protein-protein interactions

    • Verification of individual protein activities before reconstitution

  • Template DNA preparation:

    • Cloning of the P. amoebophila origin of replication region

    • Construction of template plasmids containing the origin and reporter sequences

    • Preparation of both supercoiled and linear DNA templates

  • Assay development and optimization:

    ComponentConcentration RangeOptimization Parameter
    DnaA110-500 nMTitration to determine minimal active concentration
    ATP1-5 mMRequired for DnaA1 activation
    Mg²⁺5-15 mMCritical for enzymatic activities
    Template DNA1-50 nMLow concentrations improve assay sensitivity
    dNTPs40-100 μM eachInclude labeled dNTPs for detection
    Other proteinsVariableSequential addition to determine requirements
  • Detection and analysis methods:

    • Incorporation of radiolabeled or fluorescently labeled nucleotides

    • Gel-based separation of replication products

    • Real-time monitoring using intercalating dyes

    • Electron microscopy visualization of replication bubbles

This methodological framework provides a system to dissect the molecular mechanisms of DnaA1-initiated replication and could reveal unique features of the P. amoebophila replication process compared to model organisms .

What approaches should I consider for studying the impact of P. amoebophila DnaA1 on host cell metabolic pathways?

The interaction between bacterial replication and host metabolism is particularly important for obligate intracellular bacteria like P. amoebophila. A comprehensive methodological approach would include:

  • Metabolic profiling during infection:

    • Targeted metabolomics focusing on nucleotide pools in infected versus uninfected amoebae

    • Isotope labeling to track metabolite flux between host and bacteria

    • Temporal correlation of metabolic changes with DnaA1 expression and activity

  • Manipulative experimental designs:

    • Controlled expression of recombinant DnaA1 in host cells in the absence of bacteria

    • Comparison of metabolic impacts between wild-type and mutated DnaA1 variants

    • Development of small molecule inhibitors specifically targeting DnaA1

  • Integration with known host-pathogen interfaces:

    • Analysis of nucleotide transporter (NTT) expression and activity in relation to DnaA1

    • Assessment of inclusion membrane protein interactions that might mediate metabolic signaling

    • Investigation of host cell cycle impacts on bacterial DNA replication

  • Systems biology approaches:

    • Transcriptomic analysis of host cells in response to DnaA1 activity

    • Network analysis integrating metabolomic and transcriptomic data

    • Mathematical modeling of metabolite exchange between host and pathogen

This research approach would help elucidate the complex metabolic coordination between P. amoebophila and its host, potentially revealing how bacterial replication is synchronized with host metabolic states through DnaA1 activity .

How might P. amoebophila DnaA1 be utilized as a target for controlling intracellular bacterial infections?

DnaA1 represents a potential target for antimicrobial development given its essential role in bacterial replication. A research strategy to explore this application would include:

  • Inhibitor design and screening approaches:

    • Structure-based design of small molecules targeting the ATP-binding domain

    • High-throughput screening of compound libraries using ATPase activity assays

    • Fragment-based drug discovery targeting multiple pockets on DnaA1

    • Peptide inhibitors designed to disrupt DnaA1 oligomerization

  • Validation in cellular models:

    • Testing candidate inhibitors in P. amoebophila-infected Acanthamoeba cultures

    • Demonstration of specific effects on bacterial replication without host toxicity

    • Time-kill studies to determine bacteriostatic versus bactericidal effects

    • Resistance development assessment through prolonged exposure

  • Translational considerations:

    • Evaluation of cross-reactivity with DnaA proteins from other pathogenic Chlamydiales

    • Assessment of inhibitor effects on human DnaA-related proteins (MCM complex)

    • Development of drug delivery strategies to reach intracellular bacteria

    • Combination approaches with existing antibiotics

  • Broader applications:

    • Extension to other obligate intracellular pathogens with similar replication mechanisms

    • Potential for inhibiting horizontal gene transfer between environmental and pathogenic Chlamydiae

    • Development as research tools to study bacterial persistence mechanisms

This approach builds on the understanding that while P. amoebophila itself is not pathogenic to humans, insights from this system could be applied to related pathogenic chlamydiae for which DnaA inhibition would represent a novel therapeutic strategy .

What role might P. amoebophila DnaA1 play in horizontal gene transfer and evolution of Chlamydiales?

P. amoebophila and related Chlamydia-related bacteria have been implicated in horizontal gene transfer events, including significant contributions to plant genomes . A research approach to investigate DnaA1's potential role would include:

  • Comparative genomic analysis:

    • Identification of genomic islands in P. amoebophila through nucleotide composition analysis

    • Assessment of DnaA binding sites within or near regions of potential horizontal gene transfer

    • Phylogenetic analysis of dnaA1 genes across Chlamydiales to identify recombination events

  • Experimental models of gene transfer:

    • Co-culture systems with potential recipient organisms

    • Detection of DnaA1 binding to foreign DNA elements

    • Investigation of stress conditions that might promote DNA transfer and DnaA1 involvement

  • Integration with other transfer mechanisms:

    • Analysis of potential interaction between DnaA1 and the F-type conjugative system found in P. amoebophila

    • Investigation of CRISPR system interactions in species like P. naegleriophila

    • Examination of DnaA1's potential role in mobilizing genomic islands

  • Evolutionary implications:

    • Dating of horizontal gene transfer events using molecular clock approaches

    • Correlation between DnaA1 sequence divergence and horizontal gene transfer capabilities

    • Assessment of selective pressures on dnaA1 in different chlamydial lineages

This research would help determine whether DnaA1 plays any role in facilitating the significant horizontal gene transfer events documented between Chlamydiae and other organisms, including the transfer of at least 55 genes to Plantae .

How can insights from P. amoebophila DnaA1 inform our understanding of bacterial adaptation to intracellular lifestyles?

P. amoebophila represents an excellent model for studying evolutionary adaptations to intracellular lifestyles. A comprehensive research approach would include:

  • Comparative analysis across lifestyles:

    • Detailed comparison of DnaA1 structure and function between free-living bacteria, facultative intracellular bacteria, and obligate intracellular bacteria

    • Identification of specific adaptations in the ATP-binding and DNA-binding domains

    • Assessment of regulatory mechanisms across the lifestyle spectrum

  • Integration with metabolic adaptations:

    • Investigation of how DnaA1 activity is coordinated with the specialized metabolic systems in P. amoebophila (e.g., nucleotide transporters)

    • Correlation between genome reduction events and changes in DNA replication machinery

    • Modeling of the energetic requirements for DNA replication in different bacterial lifestyles

  • Experimental evolution approaches:

    • Long-term evolution experiments in variable host conditions

    • Tracking of mutations in dnaA1 during adaptation to new hosts or stress conditions

    • Assessment of replication timing and efficiency as markers of adaptation

  • Broader biological implications:

    • Extension of findings to other symbiotic systems, including organelles with bacterial origins

    • Comparison with symbiont transition models in other bacterial phyla

    • Development of general principles regarding replication system evolution during adaptation to intracellular lifestyles

This research direction would provide fundamental insights into the evolutionary processes underlying bacterial adaptation to intracellular niches, with potential implications for understanding both symbiotic and pathogenic relationships between bacteria and eukaryotic cells .

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.